676 results on '"Reactive gliosis"'
Search Results
2. The dopamine analogue CA140 alleviates AD pathology, neuroinflammation, and rescues synaptic/cognitive functions by modulating DRD1 signaling or directly binding to Abeta.
- Author
-
Chae, Sehyun, Lee, Hyun-Ju, Lee, Ha-Eun, Kim, Jieun, Jeong, Yoo, Lin, Yuxi, Kim, Hye, Leriche, Geoffray, Ehrlich, Rachel, Lingl, Sascha, Seo, Min-Duk, Lee, Young-Ho, Yang, Jerry, Kim, Jae-Ick, and Hoe, Hyang-Sook
- Subjects
Aβ ,CA140 ,Dopamine D1 receptor ,LTP ,Learning and memory ,Reactive gliosis ,Tau ,Animals ,Alzheimer Disease ,Mice ,Mice ,Transgenic ,Amyloid beta-Peptides ,Neuroinflammatory Diseases ,Signal Transduction ,Receptors ,Dopamine D1 ,Synapses ,Cognition ,Dopamine ,Mice ,Inbred C57BL ,Male ,Humans - Abstract
BACKGROUND: We recently reported that the dopamine (DA) analogue CA140 modulates neuroinflammatory responses in lipopolysaccharide-injected wild-type (WT) mice and in 3-month-old 5xFAD mice, a model of Alzheimers disease (AD). However, the effects of CA140 on Aβ/tau pathology and synaptic/cognitive function and its molecular mechanisms of action are unknown. METHODS: To investigate the effects of CA140 on cognitive and synaptic function and AD pathology, 3-month-old WT mice or 8-month-old (aged) 5xFAD mice were injected with vehicle (10% DMSO) or CA140 (30 mg/kg, i.p.) daily for 10, 14, or 17 days. Behavioral tests, ELISA, electrophysiology, RNA sequencing, real-time PCR, Golgi staining, immunofluorescence staining, and western blotting were conducted. RESULTS: In aged 5xFAD mice, a model of AD pathology, CA140 treatment significantly reduced Aβ/tau fibrillation, Aβ plaque number, tau hyperphosphorylation, and neuroinflammation by inhibiting NLRP3 activation. In addition, CA140 treatment downregulated the expression of cxcl10, a marker of AD-associated reactive astrocytes (RAs), and c1qa, a marker of the interaction of RAs with disease-associated microglia (DAMs) in 5xFAD mice. CA140 treatment also suppressed the mRNA levels of s100β and cxcl10, markers of AD-associated RAs, in primary astrocytes from 5xFAD mice. In primary microglial cells from 5xFAD mice, CA140 treatment increased the mRNA levels of markers of homeostatic microglia (cx3cr1 and p2ry12) and decreased the mRNA levels of a marker of proliferative region-associated microglia (gpnmb) and a marker of lipid-droplet-accumulating microglia (cln3). Importantly, CA140 treatment rescued scopolamine (SCO)-mediated deficits in long-term memory, dendritic spine number, and LTP impairment. In aged 5xFAD mice, these effects of CA140 treatment on cognitive/synaptic function and AD pathology were regulated by dopamine D1 receptor (DRD1)/Elk1 signaling. In primary hippocampal neurons and WT mice, CA140 treatment promoted long-term memory and dendritic spine formation via effects on DRD1/CaMKIIα and/or ERK signaling. CONCLUSIONS: Our results indicate that CA140 improves neuronal/synaptic/cognitive function and ameliorates Aβ/tau pathology and neuroinflammation by modulating DRD1 signaling in primary hippocampal neurons, primary astrocytes/microglia, WT mice, and aged 5xFAD mice.
- Published
- 2024
3. Co-targeting of glial activation and inflammation by tsRNA-Gln-i-0095 for treating retinal ischemic pathologies.
- Author
-
Zhang, Ying, Ma, Yan, Ji, Yu-Ke, Jiang, Yi-Fei, Li, Duo, Mu, Wan, Yao, Mu-Di, Yao, Jin, and Yan, Biao
- Subjects
- *
RETINAL ganglion cells , *MEDICAL sciences , *VISION , *GENE expression , *REGULATOR genes , *TRANSFER RNA - Abstract
Ischemic retinopathies are the major causes of blindness, yet effective early-stage treatments remain limited due to an incomplete understanding of the underlying molecular mechanisms. Significant changes in gene expression often precede structural and functional alterations. Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are emerging as novel gene regulators, involved in various biological processes and human diseases. In this study, tsRNA-Gln-i-0095 was identified as a novel regulator, which was significantly upregulated in retinal ischemia/reperfusion (I/R) injury. Reducing the levels of tsRNA-Gln-i-0095 suppressed reactive gliosis, lowered inflammatory cytokine levels, and protected retinal ganglion cells from I/R injury. These effects led to reduced structural and functional damage, inhibited glial activation and inflammation, and enhanced neuronal function. Mechanistically, tsRNA-Gln-i-0095 downregulated the expression of NFIA and TGFBR2 through a miRNA-like mechanism. Collectively, this study highlights the potential of targeting tsRNA-Gln-i-0095 as a novel therapeutic approach to reduce retinal I/R injury and preserve visual function. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
4. Lycium barbarum glycopeptide (wolfberry extract) slows N-methyl-N-nitrosourea-induced degradation of photoreceptors.
- Author
-
Qihang Kong, Xiu Han, Haiyang Cheng, Jiayu Liu, Huijun Zhang, Tangrong Dong, Jiansu Chen, Kwok-Fai So, Xuesong Mi, Ying Xu, and Shibo Tang
- Published
- 2024
- Full Text
- View/download PDF
5. Механізми розвитку ранньої діабетичної ретинопатії (експериментальне дослідження).
- Author
-
К. О., Усенко
- Abstract
Background. The study of the diabetic retinopathy (DR) mechanisms should be comprehensive and include the assessment of various interconnected cellular and molecular processes initiated by hyperglycemia. The purpose was to study the mechanisms for the development of the initial stage of DR in an experiment in order to determine the main and secondary pathological processes in the retina. Materials and methods. Diabetes mellitus and DR were modeled in male Wistar rats by a single injection of streptozotocin (50 mg/kg; Sigma-Aldrich Co, China). On the 28th day of the experiment, immunohistochemical studies were performed using monoclonal antibodies to glial fibrillary acidic protein (GFAP), neuron-specific enolase (NSE), heavy neurofilaments, caspase-3 (Thermo Fisher Scientific, USA), S100 protein (Master Diagnostica, USA) and vascular endothelial growth factor (VEGF; Invitrogen, USA). Results. Early manifestations of DR included edema and detached retinal layers, dilation of the venous bed with microthrombosis, formation of diffuse zones of ischemia, foci of pathological angiogenesis (microaneurysms), degeneration of ganglion cells, retinal nuclear layer thinning. Astrocytes, Müller cells and their processes actively expressed GFAP and S100 protein, which indicated the development of reactive gliosis. Calcium overload in these cells could contribute to their death through apoptosis, which was confirmed by an elevated caspase-3 expression. A significant increase in the VEGF expression by macroglia whose processes formed tight couplings around the retinal capillaries could stimulate pathological angiogenesis. The development of neurodegeneration was confirmed by a significant decrease in the expression of neurofilaments in the nerve fiber layers and an increase in the neuronal damage marker, NSE. Conclusions. Excessive activation of macroglia (reactive gliosis) can be considered a primary link in the pathogenesis of DR whose correction can complement anti-VEGF therapy or be used separately to prevent the development of DR in the early stages. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Ultrasound inhibits tumor growth and selectively eliminates malignant brain tumor in vivo.
- Author
-
Buaron, Nitsa, Mangraviti, Antonella, Wang, Yuan, Liu, Ann, Pedone, Mariangela, Sankey, Eric, Adar, Itay, Nyska, Abraham, Goldbart, Riki, Traitel, Tamar, Brem, Henry, Tyler, Betty, and Kost, Joseph
- Subjects
- *
GLIOMAS , *TUMOR growth , *BRAIN tumors , *THERAPEUTICS , *PAIN measurement - Abstract
Glioma is one of the most common primary malignant brain tumors. Despite progress in therapeutic approaches, the median survival of patients with glioma remains less than 2 years, generating the need for new therapeutic approaches. Ultrasound (US) is widely used in medical fields and is used as a therapeutic tool mainly for improving the performance of therapeutic entities. In this study, we examined a novel approach using low frequency US (20 kHz) (LFUS) as an independent treatment tool for malignant glioma, since primary studies showed that cancer cells are more susceptible to LFUS than healthy cells. LFUS safety and efficacy were examined in a 9L gliosarcoma‐bearing female Fischer 344 rats. Two LFUS protocols were examined: a one‐time treatment (US1X), and two treatments 24 h apart (US2X). For safety evaluation, rats were monitored for weight change and pain measurements. For efficacy, tumor volume was measured as a function of time and the tumor structural chances were examined histopathologically. LFUS treatment showed rapid inhibition of tumor growth, seen as soon as 12 h after US application. In addition, LFUS was found to affect the tumor structure, which was more extensive (>60% of tumor area) in smaller tumors. In US2X, the tumor tissue was completely destroyed, and an extensive immune response was observed. Importantly, the treatment was highly selective, keeping the healthy tissue surrounding the tumor unharmed. We developed a highly efficient and selective therapeutic protocol for treating malignant glioma with minimal side effects based solely on LFUS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Pathological remodeling of reactive astrocytes: Involvement of DNA methylation and downregulation of homeostatic genes.
- Author
-
Cuautle, Dante Gómez, Donna, Soledad, Cieri, María Belén, Villarreal, Alejandro, and Ramos, Alberto Javier
- Subjects
- *
GENE expression , *BRAIN injuries , *DNA methylation , *GENE silencing , *ASTROCYTES , *AQUAPORINS - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Cytokine profiling in senescent and reactive astrocytes: A systematic review.
- Author
-
López-Teros, Michel, Alarcón-Aguilar, Adriana, Castillo-Aragón, Alejandra, Königsberg, Mina, and Luna-López, Armando
- Subjects
- *
ASTROCYTES , *CYTOKINES , *NEURODEGENERATION , *GLIOSIS , *COGNITION disorders - Abstract
Astrocytes play an important role in neuroinflammation by producing proinflammatory molecules. In response to various stressful stimuli, astrocytes can become senescent or reactive, both are present in age-associated cognitive impairment and other neurodegenerative diseases, and contribute to neuroinflammation. However, there are no studies that compare the cytokines secreted by these types of astrocytes in the brain during aging. Hence, we aimed to broaden the picture of the secretory profiles and to differentiate the variability between them. Therefore, a systematic review was conducted following the guidelines of the "Reporting Items for Systematic Review and Meta-Analyses". Only three studies that met the inclusion terms evaluated age-related cytokine secretion, however, no evaluation of senescence or gliosis was performed. Consequently, to increase the spectrum of the review, studies where those phenotypes were induced and cytokines determined were included. Although some cytokines were common for gliosis and senescence, some interesting differences were also found. The dissimilarities in cytokines secretion between these phenotypes could be studied in the future as potential markers. [Display omitted] • In response to stress, astrocytes become senescent or reactive. • Senescent or reactive astrocytes induce neuroinflammation by releasing cytokines. • A systematic review was done to compare these secretions during the brain aging. • Some cytokines were common for gliosis and senescence. • Both astrocyte´s phenotypes decreased anti-inflammatory secretion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Effect of Sonic hedgehog gene-modified bone marrow mesenchymal stem cells on graft-induced retinal gliosis and retinal ganglion cells survival in diabetic mice
- Author
-
Tong Wang, Hai-Chun Li, Jin Ma, and Xi-Ling Yu
- Subjects
mesenchymal stem cells ,sonic hedgehog signaling ,reactive gliosis ,diabetic retinopathy ,retinal ganglion cells ,Ophthalmology ,RE1-994 - Abstract
AIM: To investigate the effects of Sonic hedgehog (Shh) gene-modified bone marrow mesenchymal stem cells (MSCs) on graft-induced retinal gliosis and retinal ganglion cells (RGCs) survival in diabetic mice. METHODS: Bone marrow-derived MSCs were genetically modified with the Shh gene to generate a stably transfected cell line of Shh-modified MSCs (MSC-Shh). Intravitreal injections of MSC-Shh and green fluorescent protein-modified MSCs (MSC-Gfp; control) were administered in diabetic mice. After 4wk, the effects of MSC-Shh on retinal gliosis were evaluated using fundus photography, and markers of gliosis were examined by immunofluorescence and Western blotting. The neurotrophic factors expression and RGCs survival in the host retina were evaluated using Western blotting and immunofluorescence. The mechanisms underlying the effects of MSC-Shh was investigated. RESULTS: A significant reduction of proliferative vitreoretinopathy (PVR) was observed after intravitreal injection of MSC-Shh compared to MSC-Gfp. Significant downregulation of glial fibrillary acidic protein (GFAP) was demonstrated in the host retina after MSC-Shh administration compared to MSC-Gfp. The extracellular signal-regulated kinase 1/2 (ERK1/2), protein kinase B (AKT) and phosphatidylin-ositol-3-kinase (PI3K) pathways were significantly downregulated after MSC-Shh administration compared to MSC-Gfp. Brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) levels were significantly increased in the host retina, and RGCs loss was significantly prevented after MSC-Shh administration. CONCLUSION: MSC-Shh administration reduces graft-induced reactive gliosis following intravitreal injection in diabetic mice. The ERK1/2, AKT and PI3K pathways are involved in this process. MSC-Shh also increases the levels of neurotrophic factors in the host retina and promoted RGCs survival in diabetic mice.
- Published
- 2024
- Full Text
- View/download PDF
10. Metabolic Enzyme Alterations and Astrocyte Dysfunction in a Murine Model of Alexander Disease With Severe Reactive Gliosis
- Author
-
Heaven, Michael R, Herren, Anthony W, Flint, Daniel L, Pacheco, Natasha L, Li, Jiangtao, Tang, Alice, Khan, Fatima, Goldman, James E, Phinney, Brett S, and Olsen, Michelle L
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Neurosciences ,Neurodegenerative ,Genetics ,Rare Diseases ,Brain Disorders ,2.1 Biological and endogenous factors ,Neurological ,Alexander Disease ,Animals ,Astrocytes ,Disease Models ,Animal ,Gliosis ,Humans ,Mice ,Mice ,Transgenic ,Mutation ,Proteomics ,Alexander disease ,Fabp7 ,Ugt8 ,astrocytes ,reactive gliosis ,Biochemistry & Molecular Biology - Abstract
Alexander disease (AxD) is a rare and fatal neurodegenerative disorder caused by mutations in the gene encoding glial fibrillary acidic protein (GFAP). In this report, a mouse model of AxD (GFAPTg;Gfap+/R236H) was analyzed that contains a heterozygous R236H point mutation in murine Gfap as well as a transgene with a GFAP promoter to overexpress human GFAP. Using label-free quantitative proteomic comparisons of brain tissue from GFAPTg;Gfap+/R236H versus wild-type mice confirmed upregulation of the glutathione metabolism pathway and indicated proteins were elevated in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which had not been reported previously in AxD. Relative protein-level differences were confirmed by a targeted proteomics assay, including proteins related to astrocytes and oligodendrocytes. Of particular interest was the decreased level of the oligodendrocyte protein, 2-hydroxyacylsphingosine 1-beta-galactosyltransferase (Ugt8), since Ugt8-deficient mice exhibit a phenotype similar to GFAPTg;Gfap+/R236H mice (e.g., tremors, ataxia, hind-limb paralysis). In addition, decreased levels of myelin-associated proteins were found in the GFAPTg;Gfap+/R236H mice, consistent with the role of Ugt8 in myelin synthesis. Fabp7 upregulation in GFAPTg;Gfap+/R236H mice was also selected for further investigation due to its uncharacterized association to AxD, critical function in astrocyte proliferation, and functional ability to inhibit the anti-inflammatory PPAR signaling pathway in models of amyotrophic lateral sclerosis (ALS). Within Gfap+ astrocytes, Fabp7 was markedly increased in the hippocampus, a brain region subjected to extensive pathology and chronic reactive gliosis in GFAPTg;Gfap+/R236H mice. Last, to determine whether the findings in GFAPTg;Gfap+/R236H mice are present in the human condition, AxD patient and control samples were analyzed by Western blot, which indicated that Type I AxD patients have a significant fourfold upregulation of FABP7. However, immunohistochemistry analysis showed that UGT8 accumulates in AxD patient subpial brain regions where abundant amounts of Rosenthal fibers are located, which was not observed in the GFAPTg;Gfap+/R236H mice.
- Published
- 2022
11. Regional Microglial Response in Entorhino–Hippocampal Slice Cultures to Schaffer Collateral Lesion and Metalloproteinases Modulation.
- Author
-
Virtuoso, Assunta, Galanis, Christos, Lenz, Maximilian, Papa, Michele, and Vlachos, Andreas
- Subjects
- *
METALLOPROTEINASES , *MICROGLIA , *EXTRACELLULAR matrix , *NEUROGLIA , *CENTRAL nervous system , *THETA rhythm - Abstract
Microglia and astrocytes are essential in sustaining physiological networks in the central nervous system, with their ability to remodel the extracellular matrix, being pivotal for synapse plasticity. Recent findings have challenged the traditional view of homogenous glial populations in the brain, uncovering morphological, functional, and molecular heterogeneity among glial cells. This diversity has significant implications for both physiological and pathological brain states. In the present study, we mechanically induced a Schaffer collateral lesion (SCL) in mouse entorhino–hippocampal slice cultures to investigate glial behavior, i.e., microglia and astrocytes, under metalloproteinases (MMPs) modulation in the lesioned area, CA3, and the denervated region, CA1. We observed distinct response patterns in the microglia and astrocytes 3 days after the lesion. Notably, GFAP-expressing astrocytes showed no immediate changes post-SCL. Microglia responses varied depending on their anatomical location, underscoring the complexity of the hippocampal neuroglial network post-injury. The MMPs inhibitor GM6001 did not affect microglial reactions in CA3, while increasing the number of Iba1-expressing cells in CA1, leading to a withdrawal of their primary branches. These findings highlight the importance of understanding glial regionalization following neural injury and MMPs modulation and pave the way for further research into glia-targeted therapeutic strategies for neurodegenerative disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Seizures
- Author
-
Walz, Wolfgang and Walz, Wolfgang
- Published
- 2023
- Full Text
- View/download PDF
13. Emerging Insights into the Interstitial Distribution of Neuraxial Therapeutics via the Cerebrospinal Fluid Compartment
- Author
-
Jansson, Deidre J., Iliff, Jeffrey J., Yaksh, Tony, editor, and Hayek, Salim, editor
- Published
- 2023
- Full Text
- View/download PDF
14. The effects of Src tyrosine kinase inhibitor, saracatinib, on the markers of epileptogenesis in a mixed-sex cohort of adult rats in the kainic acid model of epilepsy.
- Author
-
Rao, Nikhil S., Putra, Marson, Meyer, Christina, Almanza, Aida, and Thippeswamy, Thimmasettappa
- Subjects
PROTEIN-tyrosine kinase inhibitors ,KAINIC acid ,ESTRUS ,TEMPORAL lobe epilepsy ,SPRAGUE Dawley rats ,VAGUS nerve ,EPILEPSY - Abstract
Neurodegeneration and neuroinflammation are key processes of epileptogenesis in temporal lobe epilepsy (TLE). A considerable number (~30%) of patients with epilepsy are resistant to currently available antiseizure drugs and thus there is a need to develop adjunct therapies to modify disease progression. A vast majority of interventional strategies to treat TLE have utilized males which limits the translational nature of the studies. In this study, we investigated the effects of repeated low-dose kainic acid (KA) injection on the initial status epilepticus (SE) and the effects of Src kinase inhibitor, saracatinib (SAR/AZD0530; 20 mg/kg, oral, daily for 7 days), in a mixed-sex cohort of adult Sprague Dawley rats during early epileptogenesis. There were no sex differences in response to KAinduced SE, and neither did the stage of estrus influence SE severity. KA-induced SE caused significant astrogliosis and microgliosis across the hippocampus, piriform cortex, and amygdala. SAR treatment resulted in a significant reduction of microgliosis across brain regions. Microglial morphometrics such as branch length and the endpoints strongly correlated with CD68 expression in the vehicle-treated group but not in the SAR-treated group, indicating mitigation by SAR. KA-induced SE caused significant neuronal loss, including parvalbuminpositive inhibitory neurons, in both vehicle (VEH) and SAR-treated groups. SAR treatment significantly mitigated FJB-positive neuronal counts as compared to the VEH group. There was an increase in C3-positive reactive astrocytes in the VEH-treated group, and SAR treatment significantly reduced the increase in the piriform cortex. C3-positive astrogliosis significantly correlated with CD68 expression in the amygdala (AMY) of VEH-treated rats, and SAR treatment mitigated this relationship. There was a significant increase of pSrc(Y419)-positive microglia in both KA-treated groups with a statistically insignificant reduction by SAR. KA-induced SE caused the development of classical glial scars in the piriform cortex (PIR) in both KA-treated groups, while SAR treatment led to a 42.17% reduction in the size of glial scars. We did not observe sex differences in any of the parameters in this study. SAR, at the dose tested in the rat kainate model for a week in this study mitigated some of the markers of epileptogenesis in both sexes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Вплив бензодіазепінових рецепторів на стан глії при розвитку діабетичної ретинопатії
- Author
-
Зябліцев, С. В., Жупан, Д. Б., and Дядик, О. О.
- Abstract
Diabetic retinopathy is a progressive tissue-specific neurovascular complication of diabetes with a multifactorial pathogenesis, in which microvascular disorders are preceded by damage to nerve elements. The latter begin with the early involvement of glia, including astrocytes and Müller cells. Taking into account the establishment of GABA-ergic deficiency, the use of modulators of the GABA-benzodiazepine receptor complex, for example, Carbacetam, which has shown satisfactory neuroprotective properties, seems promising. Diabetes mellitus was modeled by a single administration of streptozotocin (50 mg/kg; "Sigma-Aldrich", China) to threemonth-old male Wistar rats. Already after 7 days, according to immunohistochemical detection of glial fibrillary acidic protein (GFAP), reactive gliosis of astrocytes of the inner retina layers was detected, to which Müller cells joined from the 14th day. The content of GFAP in retinal tissues increased significantly. GFAP-positive cells were in close contact with foci of pathological angiogenesis in the inner layers of the retina and also took part in the formation of fibrous proliferates in the outer layers. Detection of caspase-3 showed the activation of apoptosis in astrocytes and radial processes of Müller cells in the inner plexiform layer. Carbacetam in combination with insulin reduced the expression of GFAP and caspase-3 in the retina and prevented the development of reactive gliosis, angiogenesis, and the formation of fibrous proliferates, which makes it a candidate for further studies in the treatment of diabetic retinopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
16. Reactive gliosis and neuroinflammation: prime suspects in the pathophysiology of post-acute neuroCOVID-19 syndrome.
- Author
-
Saucier, Jacob, Comeau, Dominique, Robichaud, Gilles A., and Chamard-Witkowski, Ludivine
- Subjects
CENTRAL nervous system injuries ,GLIOSIS ,NEUROINFLAMMATION ,PATHOLOGICAL physiology ,POST-acute COVID-19 syndrome ,CENTRAL nervous system - Abstract
Introduction: As the repercussions from the COVID-19 pandemic continue to unfold, an ever-expanding body of evidence suggests that infection also elicits pathophysiological manifestations within the central nervous system (CNS), known as neurological symptoms of post-acute sequelae of COVID infection (NeuroPASC). Although the neurological impairments and repercussions associated with NeuroPASC have been well described in the literature, its etiology remains to be fully characterized. Objectives: This mini-review explores the current literature that elucidates various mechanisms underlining NeuroPASC, its players, and regulators, leading to persistent neuroinflammation of affected individuals. Specifically, we provide some insights into the various roles played by microglial and astroglial cell reactivity in NeuroPASC and how these cell subsets potentially contribute to neurological impairment in response to the direct or indirect mechanisms of CNS injury. Discussion: A better understanding of themechanisms and biomarkers associated with this maladaptive neuroimmune response will thus provide better diagnostic strategies for NeuroPASC and reveal new potential mechanisms for therapeutic intervention. Altogether, the elucidation of NeuroPASC pathogenesis will improve patient outcomes and mitigate the socioeconomic burden of this syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Transient inhibition of microsomal prostaglandin E synthase-1 after status epilepticus blunts brain inflammation and is neuroprotective
- Author
-
Nelufar Yasmen, Madison N. Sluter, Lexiao Li, Ying Yu, and Jianxiong Jiang
- Subjects
Antiseizure drugs (ASDs) ,Epilepsy ,Epileptogenesis ,Reactive gliosis ,Inhibitor ,Microsomal prostaglandin E synthase-1 (mPGES-1) ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Status epilepticus (SE) in humans is characterized by prolonged convulsive seizures that are generalized and often difficult to control. The current antiseizure drugs (ASDs) aim to stop seizures quickly enough to prevent the SE-induced brain inflammation, injury, and long-term sequelae. However, sole reliance on acute therapies is imprudent because prompt treatment may not always be possible under certain circumstances. The pathophysiological mechanisms underlying the devastating consequences of SE are presumably associated with neuroinflammatory reactions, where prostaglandin E2 (PGE2) plays a pivotal role. As the terminal synthase for pathogenic PGE2, the microsomal prostaglandin E synthase-1 (mPGES-1) is rapidly and robustly induced by prolonged seizures. Congenital deletion of mPGES-1 in mice is neuroprotective and blunts gliosis following chemoconvulsant seizures, suggesting the feasibility of mPGES-1 as a potential antiepileptic target. Herein, we investigated the effects of a dual species mPGES-1 inhibitor in a mouse pilocarpine model of SE. Treatment with the mPGES-1 inhibitor in mice after SE that was terminated by diazepam, a fast-acting benzodiazepine, time-dependently abolished the SE-induced PGE2 within the brain. Its negligible effects on cyclooxygenases, the enzymes responsible for the initial step of PGE2 biosynthesis, validated its specificity to mPGES-1. Post-SE inhibition of mPGES-1 also blunted proinflammatory cytokines and reactive gliosis in the hippocampus and broadly prevented neuronal damage in a number of brain areas. Thus, pharmacological inhibition of mPGES-1 by small-molecule inhibitors might provide an adjunctive strategy that can be implemented hours after SE, together with first-line ASDs, to reduce SE-provoked brain inflammation and injury.
- Published
- 2023
- Full Text
- View/download PDF
18. The effects of Src tyrosine kinase inhibitor, saracatinib, on the markers of epileptogenesis in a mixed-sex cohort of adult rats in the kainic acid model of epilepsy
- Author
-
Nikhil S. Rao, Marson Putra, Christina Meyer, Aida Almanza, and Thimmasettappa Thippeswamy
- Subjects
status epilepticus ,reactive gliosis ,neurodegeneration ,microglia morphology ,CD68 correlation ,glial scars ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Neurodegeneration and neuroinflammation are key processes of epileptogenesis in temporal lobe epilepsy (TLE). A considerable number (∼30%) of patients with epilepsy are resistant to currently available antiseizure drugs and thus there is a need to develop adjunct therapies to modify disease progression. A vast majority of interventional strategies to treat TLE have utilized males which limits the translational nature of the studies. In this study, we investigated the effects of repeated low-dose kainic acid (KA) injection on the initial status epilepticus (SE) and the effects of Src kinase inhibitor, saracatinib (SAR/AZD0530; 20 mg/kg, oral, daily for 7 days), in a mixed-sex cohort of adult Sprague Dawley rats during early epileptogenesis. There were no sex differences in response to KA-induced SE, and neither did the stage of estrus influence SE severity. KA-induced SE caused significant astrogliosis and microgliosis across the hippocampus, piriform cortex, and amygdala. SAR treatment resulted in a significant reduction of microgliosis across brain regions. Microglial morphometrics such as branch length and the endpoints strongly correlated with CD68 expression in the vehicle-treated group but not in the SAR-treated group, indicating mitigation by SAR. KA-induced SE caused significant neuronal loss, including parvalbumin-positive inhibitory neurons, in both vehicle (VEH) and SAR-treated groups. SAR treatment significantly mitigated FJB-positive neuronal counts as compared to the VEH group. There was an increase in C3-positive reactive astrocytes in the VEH-treated group, and SAR treatment significantly reduced the increase in the piriform cortex. C3-positive astrogliosis significantly correlated with CD68 expression in the amygdala (AMY) of VEH-treated rats, and SAR treatment mitigated this relationship. There was a significant increase of pSrc(Y419)-positive microglia in both KA-treated groups with a statistically insignificant reduction by SAR. KA-induced SE caused the development of classical glial scars in the piriform cortex (PIR) in both KA-treated groups, while SAR treatment led to a 42.17% reduction in the size of glial scars. We did not observe sex differences in any of the parameters in this study. SAR, at the dose tested in the rat kainate model for a week in this study mitigated some of the markers of epileptogenesis in both sexes.
- Published
- 2023
- Full Text
- View/download PDF
19. Reactive gliosis and neuroinflammation: prime suspects in the pathophysiology of post-acute neuroCOVID-19 syndrome
- Author
-
Jacob Saucier, Dominique Comeau, Gilles A. Robichaud, and Ludivine Chamard-Witkowski
- Subjects
post-COVID syndrome ,NeuroPASC ,reactive gliosis ,neuroinflammation ,microglial reactivity ,reactive astrocytes ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
IntroductionAs the repercussions from the COVID-19 pandemic continue to unfold, an ever-expanding body of evidence suggests that infection also elicits pathophysiological manifestations within the central nervous system (CNS), known as neurological symptoms of post-acute sequelae of COVID infection (NeuroPASC). Although the neurological impairments and repercussions associated with NeuroPASC have been well described in the literature, its etiology remains to be fully characterized.ObjectivesThis mini-review explores the current literature that elucidates various mechanisms underlining NeuroPASC, its players, and regulators, leading to persistent neuroinflammation of affected individuals. Specifically, we provide some insights into the various roles played by microglial and astroglial cell reactivity in NeuroPASC and how these cell subsets potentially contribute to neurological impairment in response to the direct or indirect mechanisms of CNS injury.DiscussionA better understanding of the mechanisms and biomarkers associated with this maladaptive neuroimmune response will thus provide better diagnostic strategies for NeuroPASC and reveal new potential mechanisms for therapeutic intervention. Altogether, the elucidation of NeuroPASC pathogenesis will improve patient outcomes and mitigate the socioeconomic burden of this syndrome.
- Published
- 2023
- Full Text
- View/download PDF
20. Astrocytes and the Psychiatric Sequelae of COVID-19: What We Learned from the Pandemic.
- Author
-
Steardo Jr., Luca, Steardo, Luca, and Scuderi, Caterina
- Subjects
- *
PSYCHOLOGICAL manifestations of general diseases , *ASTROCYTES , *COVID-19 , *BRAIN physiology , *AUTOPSY , *NEUROGLIA , *NEUROENDOCRINE cells , *AVIAN influenza - Abstract
COVID-19, initially regarded as specific lung disease, exhibits an extremely broad spectrum of symptoms. Extrapulmonary manifestations of the disease also include important neuropsychiatric symptoms with atypical characteristics. Are these disturbances linked to stress accompanying every systemic infection, or are due to specific neurobiological changes associated with COVID-19? Evidence accumulated so far indicates that the pathophysiology of COVID-19 is characterized by systemic inflammation, hypoxia resulting from respiratory failure, and neuroinflammation (either due to viral neurotropism or in response to cytokine storm), all affecting the brain. It is reasonable to hypothesize that all these events may initiate or worsen psychiatric and cognitive disorders. Damage to the brain triggers a specific type of reactive response mounted by neuroglia cells, in particular by astrocytes which are the homeostatic cell par excellence. Astrocytes undergo complex morphological, biochemical, and functional remodeling aimed at mobilizing the regenerative potential of the central nervous system. If the brain is not directly damaged, resolution of systemic pathology usually results in restoration of the physiological homeostatic status of neuroglial cells. The completeness and dynamics of this process in pathological conditions remain largely unknown. In a subset of patients, glial cells could fail to recover after infection thus promoting the onset and progression of COVID-19-related neuropsychiatric diseases. There is evidence from post-mortem examinations of the brains of COVID-19 patients of alterations in both astrocytes and microglia. In conclusion, COVID-19 activates a huge reactive response of glial cells, that physiologically act as the main controller of the inflammatory, protective and regenerative events. However, in some patients the restoration of glial physiological state does not occur, thus compromising glial function and ultimately resulting in homeostatic failure underlying a set of specific neuropsychiatric symptoms related to COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. STAT3 Drives GFAP Accumulation and Astrocyte Pathology in a Mouse Model of Alexander Disease.
- Author
-
Hagemann, Tracy L., Coyne, Sierra, Levin, Alder, Wang, Liqun, Feany, Mel B., and Messing, Albee
- Subjects
- *
KNOCKOUT mice , *MICE , *GLIAL fibrillary acidic protein , *STAT proteins , *LABORATORY mice , *ANIMAL disease models , *MUTANT proteins - Abstract
Alexander disease (AxD) is caused by mutations in the gene for glial fibrillary acidic protein (GFAP), an intermediate filament expressed by astrocytes in the central nervous system. AxD-associated mutations cause GFAP aggregation and astrogliosis, and GFAP is elevated with the astrocyte stress response, exacerbating mutant protein toxicity. Studies in mouse models suggest disease severity is tied to Gfap expression levels, and signal transducer and activator of transcription (STAT)-3 regulates Gfap during astrocyte development and in response to injury and is activated in astrocytes in rodent models of AxD. In this report, we show that STAT3 is also activated in the human disease. To determine whether STAT3 contributes to GFAP elevation, we used a combination of genetic approaches to knockout or reduce STAT3 activation in AxD mouse models. Conditional knockout of Stat3 in cells expressing Gfap reduced Gfap transactivation and prevented protein accumulation. Astrocyte-specific Stat3 knockout in adult mice with existing pathology reversed GFAP accumulation and aggregation. Preventing STAT3 activation reduced markers of reactive astrocytes, stress-related transcripts, and microglial activation, regardless of disease stage or genetic knockout approach. These results suggest that pharmacological inhibition of STAT3 could potentially reduce GFAP toxicity and provide a therapeutic benefit in patients with AxD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Purinergic signaling systems across comparative models of spinal cord injury.
- Author
-
Stefanova, Eva E. and Scott, Angela L.
- Published
- 2023
- Full Text
- View/download PDF
23. Modulating sonic hedgehog (SHH) pathway to create a rapid CNS-TB model: Facilitating drug discovery.
- Author
-
Mubarak, Mohamad Mosa, Majeed, Shahnawaz, Wani, Zubair Ahmad, Kantroo, Hadiya Amin, Malik, Abbass, Baba, Ishfaq Ahmad, Mhatre, Radhika, and Ahmad, Zahoor
- Subjects
- *
MYCOBACTERIUM tuberculosis , *DRUG discovery , *CENTRAL nervous system diseases , *TUBERCULOUS meningitis , *CENTRAL nervous system - Abstract
Tuberculous meningitis, a severe complication of Mycobacterium tuberculosis (M. tb) infection, involves the dissemination of bacilli in the brain. This study explored the role of the sonic hedgehog (SHH) signaling pathway in regulating blood-brain barrier (BBB) integrity, M. tb invasion into the central nervous system (CNS), and disease progression of Central Nervous System Tuberculosis (CNS-TB) in a Balb/c mouse model. The modulation of the SHH pathway using agonist Purmorphamine (PUR) and antagonist Cyclopamine (CYC) revealed that CYC treatment led to a rapid and extensive invasion of M. tb in the brain, with bacterial loads increasing by 99 % compared to the untreated-infected group. In contrast, PUR reduced M. tb loads by 50 % and delayed disease progression. Histopathological analysis showed that CYC exacerbated inflammation and immune cell infiltration, while PUR mitigated these responses. Immunohistochemistry demonstrated that CYC caused severe BBB breakdown and reactive gliosis, while PUR partially attenuated this response. Further analysis revealed that CYC upregulated Matrix Metalloproteinase-9 (MMP-9) secretion, a key contributor to BBB disruption. These findings highlight the critical role of the SHH pathway in maintaining BBB integrity and regulating the immunopathological response during CNS-TB, opening up future scope for drug discovery. This Cyclopamine-induced model of rapid M. tb invasion and chronic inflammation provides a new tool for studying CNS-TB pathogenesis and evaluating potential therapeutic interventions targeting the SHH signaling axis. Understanding how tuberculosis (TB) infection can spread to the brain is crucial, as this "central nervous system TB" (CNS-TB) is a serious and potentially life-threatening health complication. However, studying CNS-TB in humans is very difficult. Animal models are needed to better understand how TB gets into the brain and the resulting damage. This study in mice showed that blocking a signaling pathway called Sonic Hedgehog (SHH) allowed TB to rapidly spread to the brain, damaging the blood-brain barrier and causing severe inflammation. In contrast, activating the SHH pathway helped protect the brain from TB. These findings provide important insights that could lead to new ways to prevent or treat this dangerous form of TB. Modulating the sonic hedgehog (SHH) pathway to regulate the blood-brain barrier (BBB) integrity and Mycobacterium tuberculosis (M. tb) invasion in a mouse CNS-TB model. Cyclopamine, an SHH antagonist, caused rapid M. tb brain invasion and high bacterial loads, exacerbated inflammation, and disrupted the BBB. In contrast, the SHH agonist Purmorphamine reduced M. tb loads, mitigated inflammation, and protected BBB integrity. These findings underscore the SHH pathway's critical role in CNS-TB and suggest the Cyclopamine-induced model as a useful tool for studying pathogenesis and evaluating therapeutics in CNS-TB. [Display omitted] • Findings highlight the SHH pathway's critical role in maintaining BBB integrity and regulating immunopathology in CNS-TB. • CYC SHH antagonist, caused rapid M. tb brain invasion, while SHH agonist PUR reduced M. tb loads, suggesting protection. • CYC exacerbated inflammation and immune cell infiltration, whereas PUR mitigated these. • CYC upregulated MMP-9, a key BBB disruptor, and PUR partially attenuated this. • CYC-induced rapid M. tb invasion/chronic inflammation model can be useful for studying CNS-TB and SHH-targeted therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Therapeutic Hypothermia after Cardiac Arrest Attenuates Hindlimb Paralysis and Damage of Spinal Motor Neurons and Astrocytes through Modulating Nrf2/HO-1 Signaling Pathway in Rats.
- Author
-
Ahn, Ji Hyeon, Lee, Tae-Kyeong, Kim, Dae Won, Shin, Myoung Cheol, Cho, Jun Hwi, Lee, Jae-Chul, Tae, Hyun-Jin, Park, Joon Ha, Hong, Seongkweon, Lee, Choong-Hyun, Won, Moo-Ho, and Kim, Yang Hee
- Subjects
- *
HINDLIMB , *NUCLEAR factor E2 related factor , *ASTROCYTES , *THERAPEUTIC hypothermia , *RETURN of spontaneous circulation , *MOTOR neurons , *CARDIAC arrest , *CELLULAR signal transduction - Abstract
Cardiac arrest (CA) and return of spontaneous circulation (ROSC), a global ischemia and reperfusion event, lead to neuronal damage and/or death in the spinal cord as well as the brain. Hypothermic therapy is reported to protect neurons from damage and improve hindlimb paralysis after resuscitation in a rat model of CA induced by asphyxia. In this study, we investigated roles of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in the lumbar spinal cord protected by therapeutic hypothermia in a rat model of asphyxial CA. Male Sprague-Dawley rats were subjected to seven minutes of asphyxial CA (induced by injection of 2 mg/kg vecuronium bromide) and hypothermia (four hours of cooling, 33 ± 0.5 °C). Survival rate, hindlimb motor function, histopathology, western blotting, and immunohistochemistry were examined at 12, 24, and 48 h after CA/ROSC. The rats of the CA/ROSC and hypothermia-treated groups had an increased survival rate and showed an attenuated hindlimb paralysis and a mild damage/death of motor neurons located in the anterior horn of the lumbar spinal cord compared with those of the CA/ROSC and normothermia-treated groups. In the CA/ROSC and hypothermia-treated groups, expressions of cytoplasmic and nuclear Nrf2 and HO-1 were significantly higher in the anterior horn compared with those of the CA/ROSC and normothermia-treated groups, showing that cytoplasmic and nuclear Nrf2 was expressed in both motor neurons and astrocytes. Moreover, in the CA/ROSC and hypothermia-treated group, interleukin-1β (IL-1β, a pro-inflammatory cytokine) expressed in the motor neurons was significantly reduced, and astrocyte damage was apparently attenuated compared with those found in the CA/ROSC and normothermia group. Taken together, our results indicate that hypothermic therapy after CA/ROSC attenuates CA-induced hindlimb paralysis by protecting motor neurons in the lumbar spinal cord via activating the Nrf2/HO-1 signaling pathway and attenuating pro-inflammation and astrocyte damage (reactive astrogliosis). [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Transient inhibition of microsomal prostaglandin E synthase-1 after status epilepticus blunts brain inflammation and is neuroprotective.
- Author
-
Yasmen, Nelufar, Sluter, Madison N., Li, Lexiao, Yu, Ying, and Jiang, Jianxiong
- Subjects
ENCEPHALITIS ,STATUS epilepticus ,PROSTAGLANDIN receptors ,PROSTAGLANDINS ,SEIZURES (Medicine) ,NEUROPROTECTIVE agents - Abstract
Status epilepticus (SE) in humans is characterized by prolonged convulsive seizures that are generalized and often difficult to control. The current antiseizure drugs (ASDs) aim to stop seizures quickly enough to prevent the SE-induced brain inflammation, injury, and long-term sequelae. However, sole reliance on acute therapies is imprudent because prompt treatment may not always be possible under certain circumstances. The pathophysiological mechanisms underlying the devastating consequences of SE are presumably associated with neuroinflammatory reactions, where prostaglandin E2 (PGE
2 ) plays a pivotal role. As the terminal synthase for pathogenic PGE2 , the microsomal prostaglandin E synthase-1 (mPGES-1) is rapidly and robustly induced by prolonged seizures. Congenital deletion of mPGES-1 in mice is neuroprotective and blunts gliosis following chemoconvulsant seizures, suggesting the feasibility of mPGES-1 as a potential antiepileptic target. Herein, we investigated the effects of a dual species mPGES-1 inhibitor in a mouse pilocarpine model of SE. Treatment with the mPGES-1 inhibitor in mice after SE that was terminated by diazepam, a fast-acting benzodiazepine, time-dependently abolished the SE-induced PGE2 within the brain. Its negligible effects on cyclooxygenases, the enzymes responsible for the initial step of PGE2 biosynthesis, validated its specificity to mPGES-1. Post-SE inhibition of mPGES-1 also blunted proinflammatory cytokines and reactive gliosis in the hippocampus and broadly prevented neuronal damage in a number of brain areas. Thus, pharmacological inhibition of mPGES-1 by small-molecule inhibitors might provide an adjunctive strategy that can be implemented hours after SE, together with first-line ASDs, to reduce SE-provoked brain inflammation and injury. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
26. Dexmedetomidine postconditioning alleviates spinal cord ischemia-reperfusion injury in rats via inhibiting neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation.
- Author
-
Chen, Fengshou, Wang, Dan, Jiang, Yanhua, Ma, Hong, Li, Xiaoqian, and Wang, He
- Subjects
- *
GLIAL fibrillary acidic protein , *DEXMEDETOMIDINE , *SPINAL cord injuries , *CHEMOKINE receptors , *NEUTROPHILS , *GLIOSIS - Abstract
Spinal cord ischemia-reperfusion (I/R) injury is an unresolved complication and its mechanisms are still not completely understood. Here, we studied the neuroprotective effects of dexmedetomidine (DEX) postconditioning against spinal cord I/R injury in rats and explored the possible mechanisms. In the study, rats were randomly divided into five groups: sham group, I/R group, DEX0.5 group, DEX2.5 group, and DEX5 group. I/R injury was induced in experimental rats; 0.5 μg/kg, 2.5 μg/kg, 5 μg/kg DEX were intravenously injected upon reperfusion respectively. Neurological function, histological assessment, and the disruption of blood-spinal cord barrier (BSCB) were evaluated via the BBB scoring, hematoxylin and eosin staining, Evans Blue (EB) extravasation and spinal cord edema, respectively. Neutrophil infiltration was evaluated via Myeloperoxidase (MPO) activity. Microglia activation and reactive gliosis was evaluated via ionized calcium-binding adapter molecule-1(IBA-1) and glial fibrillary acidic protein (GFAP) immunofluorescence, respectively. The expression of C-X-C motif ligand 13 (CXCL13), C-X-C chemokine receptor type 5(CXCR5), caspase-3 was determined by western blotting. The expression levels of interleukin 6(IL-6), tumor necrosis factor-α(TNF-α), IL-1β were determined by ELISA assay. DEX postconditioning preserved neurological assessment scores, improved histological assessment scores, attenuated BSCB leakage after spinal cord I/R injury. Neutrophil infiltration, microglia activation and reactive gliosis were also inhibited by DEX postconditioning. The expression of CXCL13, CXCR5, caspase-3, IL-6, TNF-α, IL-1β were reduced by DEX postconditioning. DEX postconditioning alleviated spinal cord I/R injury, which might be mediated via inhibition of neutrophil infiltration, microglia activation, reactive gliosis and CXCL13/CXCR5 axis activation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Characteristics of Epileptiform Spike-wave Discharges and Chronic Histopathology in Controlled Cortical Impact Model of Sprague–Dawley Rats.
- Author
-
Sun, Lei, Liu, Ru, Yang, Huajun, Yu, Tingting, Wu, Jianping, and Wang, Qun
- Subjects
- *
EPILEPTIFORM discharges , *SPRAGUE Dawley rats , *BRAIN injuries , *PATHOLOGICAL physiology , *GLIOSIS - Abstract
Post-traumatic epilepsy (PTE) is a serious complication that can occur following traumatic brain injury (TBI). Sustained secondary changes after TBI promote the process of PTE. Here, we aim to evaluate changes in behavior, electrocorticogram, and histomorphology in rats following chronic TBI models. We observed intensive 7–8 Hz spike-wave-discharges (SWDs) at frontal recording sites and quantified them in SD rats with different degrees of TBI and compared them with age-matched sham rats to evaluate the association between SWDs and injury severity. Notably, although SWDs were even presented in the sham group, the number and duration of events were much lower than those in the TBI groups. SWDs have numerous similarities to absence seizures, such as abrupt onset, termination, and lack of postictal suppression, which may be the nonconvulsive characteristics of PTE. Retigabine, a novel antiepileptic drug, is ineffective in reducing SWDs. In addition, we examined chronic histopathological changes in TBI rats. Rats subjected to moderate and severe TBI exhibited significantly impaired neurological function, which was accompanied by marked cortical injury, hippocampus deformation, reactive gliosis, and mossy fiber sprouting. Long-term progressive structural changes in the brain are one of the characteristics of epileptogenesis after TBI. Our study provided the potential value of epileptiform SWDs in reflecting the nonconvulsive characteristic of PTE and highlighted the vital role of chronic pathological changes, such as reactive gliosis, in promoting the epileptogenesis following TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. Lutein delays photoreceptor degeneration in a mouse model of retinitis pigmentosa
- Author
-
Hui-Jun Zhang, Xiao-Bin Liu, Xiong-Min Chen, Qi-Hang Kong, Yu-Sang Liu, Kwok-Fai So, Jian-Su Chen, Ying Xu, Xue-Song Mi, and Shi-Bo Tang
- Subjects
anti-inflammation ,glial fibrillary acidic protein ,lutein ,microglia ,pde6brd10 (rd10) mouse ,photoreceptor ,reactive gliosis ,retinal degeneration ,retinal disease ,retinitis pigmentosa ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Retinitis pigmentosa is a retinal disease characterized by photoreceptor degeneration. There is currently no effective treatment for retinitis pigmentosa. Although a mixture of lutein and other antioxidant agents has shown promising effects in protecting the retina from degeneration, the role of lutein alone remains unclear. In this study, we administered intragastric lutein to Pde6brd10 model mice, which display degeneration of retinal photoreceptors, on postnatal days 17 (P17) to P25, when rod apoptosis reaches peak. Lutein at the optimal protective dose of 200 mg/kg promoted the survival of photoreceptors compared with vehicle control. Lutein increased rhodopsin expression in rod cells and opsin expression in cone cells, in line with an increased survival rate of photoreceptors. Functionally, lutein improved visual behavior, visual acuity, and retinal electroretinogram responses in Pde6brd10 mice. Mechanistically, lutein reduced the expression of glial fibrillary acidic protein in Müller glial cells. The results of this study confirm the ability of lutein to postpone photoreceptor degeneration by reducing reactive gliosis of Müller cells in the retina and exerting anti-inflammatory effects. This study was approved by the Laboratory Animal Ethics Committee of Jinan University (approval No. LACUC-20181217-02) on December 17, 2018.
- Published
- 2022
- Full Text
- View/download PDF
29. Purinergic signaling systems across comparative models of spinal cord injury
- Author
-
Eva E Stefanova and Angela L Scott
- Subjects
cell death ,differenriation ,glia ,inflammation ,neurogenesis ,proliferation ,purinergic signaling ,reactive gliosis ,regeneration ,spinal cord injury ,teleost ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Within the last several decades, the scientific community has made substantial progress in elucidating the complex pathophysiology underlying spinal cord injury. However, despite the many advances using conventional mammalian models, both cellular and axonal regeneration following spinal cord injury have remained out of reach. In this sense, turning to non-mammalian, regenerative species presents a unique opportunity to identify pro-regenerative cues and characterize a spinal cord microenvironment permissive to re-growth. Among the signaling pathways hypothesized to be dysregulated during spinal cord injury is the purinergic signaling system. In addition to its well-known role as energy currency in cells, ATP and its metabolites are small molecule neurotransmitters that mediate many diverse cellular processes within the central nervous system. While our understanding of the roles of the purinergic system following spinal cord injury is limited, this signaling pathway has been implicated in all injury-induced secondary processes, including cellular death, inflammation, reactive gliosis, and neural regeneration. Given that the purinergic system is also evolutionarily conserved between mammalian and non-mammalian species, comparisons of these roles may provide important insights into conditions responsible for recovery success. Here, we compare the secondary processes between key model species and the influence of purinergic signaling in each context. As our understanding of this signaling system and pro-regenerative conditions continues to evolve, so does the potential for the development of novel therapeutic interventions for spinal cord injury.
- Published
- 2022
- Full Text
- View/download PDF
30. Neuronal reprogramming in treating spinal cord injury
- Author
-
Xuanyu Chen and Hedong Li
- Subjects
astrocyte ,microrna ,neurod1 ,neuronal relay ,neuronal reprogramming ,ng2 glia ,pericyte ,reactive gliosis ,sox2 ,spinal cord injury ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Spinal cord injury represents a devastating central nervous system injury that could impair the mobility and sensory function of afflicted patients. The hallmarks of spinal cord injury include neuroinflammation, axonal degeneration, neuronal loss, and reactive gliosis. Furthermore, the formation of a glial scar at the injury site elicits an inhibitory environment for potential neuroregeneration. Besides axonal regeneration, a significant challenge in treating spinal cord injury is to replenish the neurons lost during the pathological process. However, despite decades of research efforts, current strategies including stem cell transplantation have not resulted in a successful clinical therapy. Furthermore, stem cell transplantation faces serious hurdles such as immunorejection of the transplanted cells and ethical issues. In vivo neuronal reprogramming is a recently developed technology and leading a major breakthrough in regenerative medicine. This innovative technology converts endogenous glial cells into functional neurons for injury repair in the central nervous system. The feasibility of in vivo neuronal reprogramming has been demonstrated successfully in models of different neurological disorders including spinal cord injury by numerous laboratories. Several reprogramming factors, mainly the pro-neural transcription factors, have been utilized to reprogram endogenous glial cells into functional neurons with distinct phenotypes. So far, the literature on in vivo neuronal reprogramming in the model of spinal cord injury is still small. In this review, we summarize a limited number of such reports and discuss several questions that we think are important for applying in vivo neuronal reprogramming in the research field of spinal cord injury as well as other central nervous system disorders.
- Published
- 2022
- Full Text
- View/download PDF
31. Beyond hypertrophy: Changing views of astrocytes in glaucoma.
- Author
-
Cooper, Melissa L. and Calkins, David J.
- Subjects
- *
HYPERTROPHY , *ASTROCYTES , *GLAUCOMA , *AXONS , *OPTIC nerve - Abstract
Astrocytes serve multiple roles in helping to maintain homeostatic physiology of central nervous system tissue, ranging from metabolic support to coupling between vascular and neural elements. Astrocytes are especially critical in axonal tracts such as the optic nerve, where axons propagate energy-demanding action potentials great distances. In disease, astrocyte remodeling is a dynamic, multifaceted process that is often over-simplified between states of quiescence and reactivity. In glaucoma, axon degeneration in the optic nerve is characterized by progressive stages. So too is astrocyte remodeling. Here, using quantitative analysis of light and electron micrographs of myelinated optic nerve sections from the DBA/2J mouse model of glaucoma, we offer further insight into how astrocyte organization reflects stages of degeneration. This analysis indicates that even as axons degenerate, astrocyte gliosis in the nerve increases without abject proliferation, similar to results in the DBA/2J retina. Gliosis is accompanied by reorganization. As axons expand prior to frank degeneration, astrocyte processes retract from the extra-axonal space and reorient towards the nerve edge. After a critical threshold of expansion, axons drop out, and astrocyte processes distribute more evenly across the nerve reflecting gliosis. This multi-stage process likely reflects local rather than global cues from axons and the surrounding tissue that induce rapid reorganization to promote axon survival and extend functionality of the nerve. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. High-Contrast Stimulation Potentiates the Neurotrophic Properties of Müller Cells and Suppresses Their Pro-Inflammatory Phenotype.
- Author
-
Zloh, Miloslav, Kutilek, Patrik, and Stofkova, Andrea
- Subjects
- *
NF-kappa B , *BRAIN-derived neurotrophic factor , *VISION , *PHENOTYPES , *NEUROTROPHINS - Abstract
High-contrast visual stimulation promotes retinal regeneration and visual function, but the underlying mechanism is not fully understood. Here, we hypothesized that Müller cells (MCs), which express neurotrophins such as brain-derived neurotrophic factor (BDNF), could be key players in this retinal plasticity process. This hypothesis was tested by conducting in vivo and in vitro high-contrast stimulation of adult mice and MCs. Following stimulation, we examined the expression of BDNF and its inducible factor, VGF, in the retina and MCs. We also investigated the alterations in the expression of VGF, nuclear factor kappa B (NF-κB) and pro-inflammatory mediators in MCs, as well as their capacity to proliferate and develop a neurogenic or reactive gliosis phenotype after high-contrast stimulation and treatment with BDNF. Our results showed that high-contrast stimulation upregulated BDNF levels in MCs in vivo and in vitro. The additional BDNF treatment significantly augmented VGF production in MCs and their neuroprotective features, as evidenced by increased MC proliferation, neurodifferentiation, and decreased expression of the pro-inflammatory factors and the reactive gliosis marker GFAP. These results demonstrate that high-contrast stimulation activates the neurotrophic and neuroprotective properties of MCs, suggesting their possible direct involvement in retinal neuronal survival and improved functional outcomes in response to visual stimulation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
33. Advance of neurodegeneration in diabetic retinopathy
- Author
-
Meng-Meng Jiang, Lin Liu, and Jing-Fa Zhang
- Subjects
diabetic retinopathy ,neurodegeneration ,neuronal apoptosis ,reactive gliosis ,microangiopathy ,Ophthalmology ,RE1-994 - Abstract
Diabetic retinopathy(DR), one of the most common complications of diabetes, has been widely reported as microangiopathy. However, retinal neurodegeneration was reported to occur early in DR and played a significant role in DR progression. Retinal neurodegeneration in DR was characterized as neuronal apoptosis and reactive gliosis. The mechanisms for its pathogenesis include hyperglycemia, oxidative stress, glutamate excitotoxicity, and inflammation etc. Furthermore, retinal neurodegeneration has a close relationship with the microangiopathy in the pathogenesis of DR.
- Published
- 2021
- Full Text
- View/download PDF
34. Luteolin delays photoreceptor degeneration in a mouse model of retinitis pigmentosa
- Author
-
Xiao-Bin Liu, Feng Liu, Yi-Yao Liang, Gang Yin, Hui-Jun Zhang, Xue-Song Mi, Zai-Jun Zhang, Kwok-Fai So, Ang Li, and Ying Xu
- Subjects
anti-inflammation ,apoptosis ,flavonoid ,jnk pathway ,luteolin ,photoreceptor ,reactive gliosis ,reactive oxygen species ,retinal degeneration ,retinitis pigmentosa ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Luteolin is neuroprotective for retinal ganglion cells and retinal pigment epithelial cells after oxidative injury, whereby it can inhibit microglial neurotoxicity. Therefore, luteolin holds the potential to be useful for treatment of retinal diseases. The purpose of this study was to investigate whether luteolin exhibits neuroprotective effects on rod cells in rd10 mice, a slow photoreceptor-degenerative model of retinitis pigmentosa. Luteolin (100 mg/kg) intraperitoneally injected daily from postnatal day 14 (P14) to P25 significantly enhanced the visual performance and retinal light responses of rd10 mice at P25. Moreover, it increased the survival of photoreceptors and improved retinal structure. Mechanistically, luteolin treatment attenuated increases in reactive oxygen species, photoreceptor apoptosis, and reactive gliosis; increased mRNA levels of anti-inflammatory cytokines while lowering that of pro-inflammatory and chemoattractant cytokines; and lowered the ratio of phospho-JNK/JNK. Application of the JNK inhibitor SP600125 exerted a similar protective effect to luteolin, suggesting that luteolin delays photoreceptor degeneration and functional deterioration in rd10 mice through regulation of retinal oxidation and inflammation by inhibiting the JNK pathway. Therefore, luteolin may be useful as a supplementary treatment for retinitis pigmentosa. This study was approved by the Qualified Ethics Committee of Jinan University, China (approval No. IACUC-20181217-02) on December 17, 2018.
- Published
- 2021
- Full Text
- View/download PDF
35. Neurons and Astrocytes Elicit Brain Region Specific Transcriptional Responses to Prion Disease in the Murine CA1 and Thalamus.
- Author
-
Slota, Jessy A., Medina, Sarah J., Frost, Kathy L., and Booth, Stephanie A.
- Subjects
PRION diseases ,THALAMUS ,NEURONS ,ASTROCYTES ,PRIONS ,MICROGLIA - Abstract
Progressive dysfunction and loss of neurons ultimately culminates in the symptoms and eventual fatality of prion disease, yet the pathways and mechanisms that lead to neuronal degeneration remain elusive. Here, we used RNAseq to profile transcriptional changes in microdissected CA1 and thalamus brain tissues from prion infected mice. Numerous transcripts were altered during clinical disease, whereas very few transcripts were reliably altered at pre-clinical time points. Prion altered transcripts were assigned to broadly defined brain cell types and we noted a strong transcriptional signature that was affiliated with reactive microglia and astrocytes. While very few neuronal transcripts were common between the CA1 and thalamus, we described transcriptional changes in both regions that were related to synaptic dysfunction. Using transcriptional profiling to compare how different neuronal populations respond during prion disease may help decipher mechanisms that lead to neuronal demise and should be investigated with greater detail. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
36. Matrix metalloproteinases, purinergic signaling, and epigenetics: hubs in the spinal neuroglial network following peripheral nerve injury.
- Author
-
De Luca, Ciro, Virtuoso, Assunta, Cerasuolo, Michele, Gargano, Francesca, Colangelo, Anna Maria, Lavitrano, Marialuisa, Cirillo, Giovanni, and Papa, Michele
- Subjects
- *
MATRIX metalloproteinases , *PURINERGIC receptors , *EPIGENETICS , *PERIPHERAL nerve injuries , *SPINAL cord - Abstract
Activation of glial cells (reactive gliosis) and the purinergic pathway, together with metalloproteinase (MMP)-induced remodeling of the neural extracellular matrix (nECM), drive maladaptive changes in the spinal cord following peripheral nerve injury (PNI). We evaluated the effects on spinal maladaptive plasticity through administration of oxidized ATP (oxATP), an antagonist of P2X receptors (P2XR), and/or GM6001, an inhibitor of MMPs, in rats following spared nerve injury (SNI) of the sciatic nerve. With morpho-molecular techniques, we demonstrated a reduction in spinal reactive gliosis and changes in the neuro-glial-nECM crosstalk via expression remodeling of P2XR, nerve growth factor (NGF) receptors (TrkA and p75), and histone deacetylase 2 (HDAC2) after treatments with oxATP/GM6001. Altogether, our data suggest that MMPs and purinergic inhibition have a modulatory impact on key proteins in the neuro-glial-nECM network, acting at different levels from intracellular signaling to epigenetic modifications. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
37. Function of astrocyte MyD88 in high-fat-diet-induced hypothalamic inflammation
- Author
-
Sungho Jin, Kwang Kon Kim, Byong Seo Park, Dong Hee Kim, Bora Jeong, Dasol Kang, Tae Hwan Lee, Jeong Woo Park, Jae Geun Kim, and Byung Ju Lee
- Subjects
Myeloid differentiation primary response 88 ,Hypothalamus ,Reactive gliosis ,Obesity ,Leptin resistance ,High-fat diet ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background A growing body of evidence shows that hypothalamic inflammation is an important factor in the initiation of obesity. In particular, reactive gliosis accompanied by inflammatory responses in the hypothalamus are pivotal cellular events that elicit metabolic abnormalities. In this study, we examined whether MyD88 signaling in hypothalamic astrocytes controls reactive gliosis and inflammatory responses, thereby contributing to the pathogenesis of obesity. Methods To analyze the role of astrocyte MyD88 in obesity pathogenesis, we used astrocyte-specific Myd88 knockout (KO) mice fed a high-fat diet (HFD) for 16 weeks or injected with saturated free fatty acids. Astrocyte-specific gene expression in the hypothalamus was determined using real-time PCR with mRNA purified by the Ribo-Tag system. Immunohistochemistry was used to detect the expression of glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, phosphorylated signal transducer and activator of transcription 3, and α-melanocyte-stimulating hormone in the hypothalamus. Animals’ energy expenditure was measured using an indirect calorimetry system. Results The astrocyte-specific Myd88 KO mice displayed ameliorated hypothalamic reactive gliosis and inflammation induced by injections of saturated free fatty acids and a long-term HFD. Accordingly, the KO mice were resistant to long-term HFD-induced obesity and showed an improvement in HFD-induced leptin resistance. Conclusions These results suggest that MyD88 in hypothalamic astrocytes is a critical molecular unit for obesity pathogenesis that acts by mediating HFD signals for reactive gliosis and inflammation.
- Published
- 2020
- Full Text
- View/download PDF
38. Neurons and Astrocytes Elicit Brain Region Specific Transcriptional Responses to Prion Disease in the Murine CA1 and Thalamus
- Author
-
Jessy A. Slota, Sarah J. Medina, Kathy L. Frost, and Stephanie A. Booth
- Subjects
prion ,neurodegeneration ,pathophysiology ,synaptic dysfunction ,neuroinflammation ,reactive gliosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Progressive dysfunction and loss of neurons ultimately culminates in the symptoms and eventual fatality of prion disease, yet the pathways and mechanisms that lead to neuronal degeneration remain elusive. Here, we used RNAseq to profile transcriptional changes in microdissected CA1 and thalamus brain tissues from prion infected mice. Numerous transcripts were altered during clinical disease, whereas very few transcripts were reliably altered at pre-clinical time points. Prion altered transcripts were assigned to broadly defined brain cell types and we noted a strong transcriptional signature that was affiliated with reactive microglia and astrocytes. While very few neuronal transcripts were common between the CA1 and thalamus, we described transcriptional changes in both regions that were related to synaptic dysfunction. Using transcriptional profiling to compare how different neuronal populations respond during prion disease may help decipher mechanisms that lead to neuronal demise and should be investigated with greater detail.
- Published
- 2022
- Full Text
- View/download PDF
39. Targeting long noncoding RNA-AQP4-AS1 for the treatment of retinal neurovascular dysfunction in diabetes mellitus
- Author
-
Xiumiao Li, Junya Zhu, Yuling Zhong, Chang Liu, Mudi Yao, Yanan Sun, Wen Yao, Xisen Ni, Fen Zhou, Jin Yao, and Qin Jiang
- Subjects
Long noncoding RNA ,Müller cell ,Reactive gliosis ,Neurovascular dysfunction ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Diabetic retinopathy (DR) is a leading cause of blindness in the working-age population, which is characterized by retinal neurodegeneration and vascular dysfunction. Long non-coding RNAs (LncRNAs) have emerged as critical regulators in several biological processes and disease progression. Here we investigated the role of lncRNA AQP4-AS1 in retinal neurovascular dysfunction induced by diabetes. Methods: Quantitative RT-PCR was used to detect the AQP4-AS1 expression pattern upon diabetes mellitus-related stresses. Visual electrophysiology examination, TUNEL staining, Evans blue staining, retinal trypsin digestion and immunofluorescent staining were conducted to detect the role of AQP4-AS1 in retinal neurovascular dysfunction in vivo. MTT assays, TUNEL staining, PI/Calcein-AM staining, EdU incorporation assay transwell assay and tube formation were conducted to detect the role of AQP4-AS1 in retinal cells function in vitro. qRT-PCR, western blot and in vivo studies were conducted to reveal the mechanism of AQP4-AS1-mediated retinal neurovascular dysfunction. Findings: AQP4-AS1 was significantly increased in the clinical samples of diabetic retinopathy patients, high glucose-treated Müller cells, and diabetic retinas of a murine model. AQP4-AS1 silencing in vivo alleviated retinal neurodegeneration and vascular dysfunction as shown by improved retinal capillary degeneration, decreased reactive gliosis, and reduced RGC loss. AQP4-AS1 directly regulated Müller cell function and indirectly affected endothelial cell and RGC function in vitro. Mechanistically, AQP4-AS1 regulated retinal neurovascular dysfunction through affecting AQP4 levels. Interpretation: This study reveals AQP4-AS1 is involved in retinal neurovascular dysfunction and expected to become a promising target for the treatment of neurovascular dysfunction in DR. Funding: This work was generously supported by the grants from the National Natural Science Foundation of China (Grant No. 81800858, 82070983, 81870679 and 81970823), grants from the Medical Science and Technology Development Project Fund of Nanjing (Grant No ZKX17053 and YKK19158), grants from Innovation Team Project Fund of Jiangsu Province (No. CXTDB2017010), and the Science and Technology Development Plan Project Fund of Nanjing (Grant No 201716007, 201805007 and 201803058).
- Published
- 2022
- Full Text
- View/download PDF
40. Why Has the Ability to Regenerate Following CNS Injury Been Repeatedly Lost Over the Course of Evolution?
- Author
-
Seth Blackshaw
- Subjects
regeneration ,neurogenesis ,vertebrate ,mammal ,zebrafish ,reactive gliosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
While many vertebrates can regenerate both damaged neurons and severed axons in the central nervous system (CNS) following injury, others, including all birds and mammals, have lost this ability for reasons that are still unclear. The repeated evolutionary loss of regenerative competence seems counterintuitive, and any explanation must account for the fact that regenerative competence is lost in both cold-blooded and all warm-blooded clades, that both injury-induced neurogenesis and axonal regeneration tend to be lost in tandem, and that mammals have evolved dedicated gene regulatory networks to inhibit injury-induced glia-to-neuron reprogramming. Here, different hypotheses that have been proposed to account for evolutionary loss of regenerative competence are discussed in the light of new insights obtained into molecular mechanisms that control regeneration in the central nervous system. These include pleiotropic effects of continuous growth, enhanced thyroid hormone signaling, prevention of neoplasia, and improved memory consolidation. Recent evidence suggests that the most compelling hypothesis, however, may be selection for greater resistance to the spread of intra-CNS infections, which has led to both enhanced reactive gliosis and a loss of injury-induced neurogenesis and axonal regeneration. Means of testing these hypotheses, and additional data that are urgently needed to better understand the evolutionary pressures and mechanisms driving loss of regenerative competence, are also discussed.
- Published
- 2022
- Full Text
- View/download PDF
41. Why Has the Ability to Regenerate Following CNS Injury Been Repeatedly Lost Over the Course of Evolution?
- Author
-
Blackshaw, Seth
- Subjects
NERVOUS system regeneration ,CENTRAL nervous system injuries ,GENE regulatory networks ,CENTRAL nervous system - Abstract
While many vertebrates can regenerate both damaged neurons and severed axons in the central nervous system (CNS) following injury, others, including all birds and mammals, have lost this ability for reasons that are still unclear. The repeated evolutionary loss of regenerative competence seems counterintuitive, and any explanation must account for the fact that regenerative competence is lost in both cold-blooded and all warm-blooded clades, that both injury-induced neurogenesis and axonal regeneration tend to be lost in tandem, and that mammals have evolved dedicated gene regulatory networks to inhibit injury-induced glia-to-neuron reprogramming. Here, different hypotheses that have been proposed to account for evolutionary loss of regenerative competence are discussed in the light of new insights obtained into molecular mechanisms that control regeneration in the central nervous system. These include pleiotropic effects of continuous growth, enhanced thyroid hormone signaling, prevention of neoplasia, and improved memory consolidation. Recent evidence suggests that the most compelling hypothesis, however, may be selection for greater resistance to the spread of intra-CNS infections, which has led to both enhanced reactive gliosis and a loss of injury-induced neurogenesis and axonal regeneration. Means of testing these hypotheses, and additional data that are urgently needed to better understand the evolutionary pressures and mechanisms driving loss of regenerative competence, are also discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
42. Retinal Astrocytes and Microglia Activation in Diabetic Retinopathy Rhesus Monkey Models.
- Author
-
Xia, Yu, Luo, Qihui, Chen, Jingfei, Huang, Chao, Jahangir, Asad, Pan, Ting, Wei, Xiaoli, Liu, Wentao, and Chen, Zhengli
- Subjects
- *
RHESUS monkeys , *DIABETIC retinopathy , *GLIAL fibrillary acidic protein , *MICROGLIA , *ASTROCYTES - Abstract
To assess the retinal neurodegeneration in type-1 diabetes mellitus (T1DM) and type-2 diabetes mellitus (T2DM) rhesus monkeys, and to investigate whether alterations of glial cells occur in the early stage of diabetic retinopathy (DR). T1DM rhesus monkeys were established by daily intravenous injections of streptozotocin (STZ, 25 mg/kg body weight) in citrate buffer (pH 4.5) for 5 days, while T2DM rhesus monkeys were induced by feeding with high-fat diet. The period of DR in rhesus monkeys was evaluated by fundoscopy and optical coherence tomography (OCT). Afterward, the morphological changes of inner neurons and glial cells in the retina were detected by immunofluorescence (IF). When compared with the control groups, no difference was observed in both T1DM and T2DM by fundus photographs, while slight exudation and effusion in the blood vessels of retina of rhesus monkeys were found by OCT in DM rhesus monkeys. In addition, the expression of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule (Iba1) were significantly increased in both T1DM (P <.01) and T2DM (P <.05) rhesus monkeys. Moreover, the positive expression of PKC-α, parvalbumin, and NeuN were significantly decreased, while the positive expression of calbindin showed no difference in T1DM group. However, only the expression cells of PKC-α were reduced in T2DM group when compared with that of the control group. Astrocytes activation, reactive gliosis, and neurodegeneration were observed in both T1DM and T2DM rhesus monkey models at the early stage of DR. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. C3a Receptor Signaling Inhibits Neurodegeneration Induced by Neonatal Hypoxic-Ischemic Brain Injury
- Author
-
Andrea Pozo-Rodrigálvarez, YiXian Li, Anna Stokowska, Jingyun Wu, Verena Dehm, Hana Sourkova, Harry Steinbusch, Carina Mallard, Henrik Hagberg, Milos Pekny, and Marcela Pekna
- Subjects
developing brain ,neonatal encephalopathy ,hypoxia-ischemia ,complement system: neurodegeneration ,reactive gliosis ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Hypoxic-ischemic neonatal encephalopathy due to perinatal asphyxia is the leading cause of brain injury in newborns. Clinical data suggest that brain inflammation induced by perinatal insults can persist for years. We previously showed that signaling through the receptor for complement peptide C3a (C3aR) protects against cognitive impairment induced by experimental perinatal asphyxia. To investigate the long-term neuropathological effects of hypoxic-ischemic injury to the developing brain and the role of C3aR signaling therein, we subjected wildtype mice, C3aR deficient mice, and mice expressing biologically active C3a in the CNS to mild hypoxic-ischemic brain injury on postnatal day 9. We found that such injury triggers neurodegeneration and pronounced reactive gliosis in the ipsilesional hippocampus both of which persist long into adulthood. Transgenic expression of C3a in reactive astrocytes reduced hippocampal neurodegeneration and reactive gliosis. In contrast, neurodegeneration and microglial cell density increased in mice lacking C3aR. Intranasal administration of C3a for 3 days starting 1 h after induction of hypoxia-ischemia reduced neurodegeneration and reactive gliosis in the hippocampus of wildtype mice. We conclude that neonatal hypoxic-ischemic brain injury leads to long-lasting neurodegeneration. This neurodegeneration is substantially reduced by treatment with C3aR agonists, conceivably through modulation of reactive gliosis.
- Published
- 2021
- Full Text
- View/download PDF
44. Systemic Inflammation and Astrocyte Reactivity in the Neuropsychiatric Sequelae of COVID-19: Focus on Autism Spectrum Disorders
- Author
-
Marta Valenza, Luca Steardo, Alexei Verkhratsky, and Caterina Scuderi
- Subjects
astrocytes ,autism spectrum disorders ,COVID-19 ,microglia ,neuroinflammation ,reactive gliosis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2021
- Full Text
- View/download PDF
45. Systemic Inflammation and Astrocyte Reactivity in the Neuropsychiatric Sequelae of COVID-19: Focus on Autism Spectrum Disorders.
- Author
-
Valenza, Marta, Steardo Jr., Luca, Steardo, Luca, Verkhratsky, Alexei, and Scuderi, Caterina
- Subjects
COVID-19 ,AUTISM spectrum disorders ,CHILDREN with autism spectrum disorders ,CENTRAL nervous system infections ,MATERNALLY acquired immunity ,DISEASE complications ,SYMPTOMS ,POST-acute COVID-19 syndrome - Abstract
Keywords: astrocytes; autism spectrum disorders; COVID-19; microglia; neuroinflammation; reactive gliosis; astrogliosis; SARS-CoV-2 EN astrocytes autism spectrum disorders COVID-19 microglia neuroinflammation reactive gliosis astrogliosis SARS-CoV-2 1 7 7 12/02/21 20211129 NES 211129 Introduction The Neurotropism of the SARS-CoV-2 The coronavirus disease (COVID)-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was initially regarded as a specific lung disease. Therefore, some authors have speculated that ASD could be a risk factor for SARS-CoV-2 infection and COVID-19 outcome (Lima et al., [46]; Brown et al., [11]). The effect of coronavirus infection (SARS-CoV-2, MERS-CoV, and SARS-CoV) during pregnancy and the possibility of vertical maternal-fetal transmission: a systematic review and meta-analysis. COVID-19 and Neuropsychiatric Sequelae: Focus on Autism Spectrum Disorders Epidemiologic data correlate maternal infections with several neuropsychiatric disorders, including autism spectrum disorders (ASD) (Minakova and Warner, [49]). [Extracted from the article]
- Published
- 2021
- Full Text
- View/download PDF
46. Pectic Galactan Polysaccharide‐Based Gene Delivery System for Targeting Neuroinflammation.
- Author
-
Buaron, Nitsa, Mangraviti, Antonella, Volpin, Francesco, Liu, Ann, Pedone, Mariangela, Sankey, Eric, Aranovich, Dina, Adar, Itay, Rodriguez, Fausto J., Nyska, Abraham, Goldbart, Riki, Traitel, Tamar, Brem, Henry, Tyler, Betty, and Kost, Joseph
- Subjects
- *
NEUROINFLAMMATION , *GENE therapy , *GALACTANS , *NEUROGLIA , *GALECTINS - Abstract
Treating neuroinflammation‐related injuries and disorders through manipulation of neuroinflammation functions is being heralded as a new therapeutic strategy. In this study, a novel pectic galactan (PG) polysaccharide based gene therapy approach is developed for targeting reactive gliosis in neuroinflammation. Galectin‐3 (Gal‐3) is a cell protein with a high affinity to β‐galactoside sugars and is highly expressed in reactive gliosis. Since PG carries galactans, it can target reactive gliosis via specific carbohydrate interaction between galactan and Gal‐3 on the cell membrane, and therefore can be utilized as a carrier for delivering genes to these cells. The carrier is synthesized by modifying quaternary ammonium groups on the PG. The resulting quaternized PG (QPG) is found to form complexes with plasmid DNA with a mean diameter of 100 nm and have the characteristics required for targeted gene therapy. The complexes efficiently condense large amounts of plasmid per particle and successfully bind to Gal‐3. The in vivo study shows that the complexes are biocompatible and safe for administration and can selectively transfect reactive glial cells of an induced cortical lesion. The results confirm that this PG‐based delivery system is a promising platform for targeting Gal‐3 overexpressing neuroinflammation cells for treating neuroinflammation‐related injuries and neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Autism-associated CHD8 controls reactive gliosis and neuroinflammation via remodeling chromatin in astrocytes.
- Author
-
Megagiannis, Platon, Mei, Yuan, Yan, Rachel E., Yuan, Lin, Wilde, Jonathan J., Eckersberg, Hailey, Suresh, Rahul, Tan, Xinzhu, Chen, Hong, Farmer, W. Todd, Cha, Kuwook, Le, Phuong Uyen, Catoire, Helene, Rochefort, Daniel, Kwan, Tony, Yee, Brian A., Dion, Patrick, Krishnaswamy, Arjun, Cloutier, Jean-Francois, and Stifani, Stefano
- Abstract
Reactive changes of glial cells during neuroinflammation impact brain disorders and disease progression. Elucidating the mechanisms that control reactive gliosis may help us to understand brain pathophysiology and improve outcomes. Here, we report that adult ablation of autism spectrum disorder (ASD)-associated CHD8 in astrocytes attenuates reactive gliosis via remodeling chromatin accessibility, changing gene expression. Conditional Chd8 deletion in astrocytes, but not microglia, suppresses reactive gliosis by impeding astrocyte proliferation and morphological elaboration. Astrocyte Chd8 ablation alleviates lipopolysaccharide-induced neuroinflammation and septic-associated hypothermia in mice. Astrocytic CHD8 plays an important role in neuroinflammation by altering the chromatin landscape, regulating metabolic and lipid-associated pathways, and astrocyte-microglia crosstalk. Moreover, we show that reactive gliosis can be directly mitigated in vivo using an adeno-associated virus (AAV)-mediated Chd8 gene editing strategy. These findings uncover a role of ASD-associated CHD8 in the adult brain, which may warrant future exploration of targeting chromatin remodelers in reactive gliosis and neuroinflammation in injury and neurological diseases. [Display omitted] • Astrocytic CHD8 in the adult brain regulates injury-induced reactive gliosis • Astrocytic CHD8 in the adult brain controls LPS-induced neuroinflammation • Astrocytic CHD8 regulates chromatin accessibility and transcription during neuroinflammation • AAV- and CRISPR-mediated direct Chd8 editing in astrocytes in vivo attenuates reactive gliosis Megagiannis et al. revealed the role of autism-associated CHD8 in reactive gliosis and neuroinflammation through regulating chromatin landscape and gene transcription in astrocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Regional brain susceptibility to neurodegeneration: what is the role of glial cells?
- Author
-
Andrea Beatriz Cragnolini, Giorgia Lampitella, Assunta Virtuoso, Immacolata Viscovo, Fivos Panetsos, Michele Papa, and Giovanni Cirillo
- Subjects
astrocytes ,glial cells ,microglia ,neurodegenerative diseases ,neuroinflammation ,parkinson’s disease ,reactive gliosis ,selective neuronal degeneration ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
The main pathological feature of the neurodegenerative diseases is represented by neuronal death that represents the final step of a cascade of adverse/hostile events. Early in the neurodegenerative process, glial cells (including astrocytes, microglial cells, and oligodendrocytes) activate and trigger an insidious neuroinflammatory reaction, metabolic decay, blood brain barrier dysfunction and energy impairment, boosting neuronal death. How these mechanisms might induce selective neuronal death in specific brain areas are far from being elucidated. The last two decades of neurobiological studies have provided evidence of the main role of glial cells in most of the processes of the central nervous system, from development to synaptogenesis, neuronal homeostasis and integration into, highly specific neuro-glial networks. In this mini-review, we moved from in vitro and in vivo models of neurodegeneration to analyze the putative role of glial cells in the early mechanisms of neurodegeneration. We report changes of transcriptional, genetic, morphological, and metabolic activity in astrocytes and microglial cells in specific brain areas before neuronal degeneration, providing evidence in experimental models of neurodegenerative disorders, including Parkinson’s and Alzheimer’s diseases. Understanding these mechanisms might increase the insight of these processes and pave the way for new specific glia-targeted therapeutic strategies for neurodegenerative disorders.
- Published
- 2020
- Full Text
- View/download PDF
49. Dysregulated Microglial Cell Activation and Proliferation Following Repeated Antigen Stimulation
- Author
-
Sujata Prasad, Wen S. Sheng, Shuxian Hu, Priyanka Chauhan, and James R. Lokensgard
- Subjects
viral reactivation ,microglia ,neurotoxicity ,reactive gliosis ,resident memory T-cells ,immune activation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Upon reactivation of quiescent neurotropic viruses antigen (Ag)-specific brain resident-memory CD8+ T-cells (bTRM) may respond to de novo-produced viral Ag through the rapid release of IFN-γ, which drives subsequent interferon-stimulated gene expression in surrounding microglia. Through this mechanism, a small number of adaptive bTRM may amplify responses to viral reactivation leading to an organ-wide innate protective state. Over time, this brain-wide innate immune activation likely has cumulative neurotoxic and neurocognitive consequences. We have previously shown that HIV-1 p24 Ag-specific bTRM persist within the murine brain using a heterologous prime-CNS boost strategy. In response to Ag restimulation, these bTRM display rapid and robust recall responses, which subsequently activate glial cells. In this study, we hypothesized that repeated challenges to viral antigen (Ag) (modeling repeated episodes of viral reactivation) culminate in prolonged reactive gliosis and exacerbated neurotoxicity. To address this question, mice were first immunized with adenovirus vectors expressing the HIV p24 capsid protein, followed by a CNS-boost using Pr55Gag/Env virus-like particles (HIV-VLPs). Following the establishment of the bTRM population [>30 days (d)], prime-CNS boost animals were then subjected to in vivo challenge, as well as re-challenge (at 14 d post-challenge), using the immunodominant HIV-1 AI9 CD8+ T-cell epitope peptide. In these studies, Ag re-challenge resulted in prolonged expression of microglial activation markers and an increased proliferative response, longer than the challenge group. This continued expression of MHCII and PD-L1 (activation markers), as well as Ki67 (proliferative marker), was observed at 7, 14, and 30 days post-AI9 re-challenge. Additionally, in vivo re-challenge resulted in continued production of inducible nitric oxide synthase (iNOS) with elevated levels observed at 7, 14 and 30 days post re-challenge. Interestingly, iNOS expression was significantly lower among challenged animals when compared to re-challenged groups. Furthermore, in vivo specific Ag re-challenge produced lower levels of arginase (Arg)-1 when compared with the challenged group. Taken together, these results indicate that repeated Ag-specific stimulation of adaptive immune responses leads to cumulative dysregulated microglial cell activation.
- Published
- 2021
- Full Text
- View/download PDF
50. Differential Regenerative Capacity of the Optic Tectum of Adult Medaka and Zebrafish
- Author
-
Yuki Shimizu and Takashi Kawasaki
- Subjects
radial glia ,stab wound injury ,optic tectum ,neuronal differentiation ,reactive gliosis ,zebrafish ,Biology (General) ,QH301-705.5 - Abstract
Zebrafish have superior regenerative capacity in the central nervous system (CNS) compared to mammals. In contrast, medaka were shown to have low regenerative capacity in the adult heart and larval retina, despite the well-documented high tissue regenerative ability of teleosts. Nevertheless, medaka and zebrafish share similar brain structures and biological features to those of mammals. Hence, this study aimed to compare the neural stem cell (NSC) responses and regenerative capacity in the optic tectum of adult medaka and zebrafish after stab wound injury. Limited neuronal differentiation was observed in the injured medaka, though the proliferation of radial glia (RG) was induced in response to tectum injury. Moreover, the expression of the pro-regenerative transcriptional factors ascl1a and oct4 was not enhanced in the injured medaka, unlike in zebrafish, whereas expression of sox2 and stat3 was upregulated in both fish models. Of note, glial scar-like structures composed of GFAP+ radial fibers were observed in the injured area of medaka at 14 days post injury (dpi). Altogether, these findings suggest that the adult medaka brain has low regenerative capacity with limited neuronal generation and scar formation. Hence, medaka represent an attractive model for investigating and evaluating critical factors for brain regeneration.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.