Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (69)

Search Parameters:
Keywords = CD44 expression in astrocytes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 2152 KB  
Article
Spp1 Appears to Be a Key Gene for Sporadic Obstructive Hydrocephalus in the Absence of AQP4
by Miriam Echevarría, Laura Hiraldo-González, José Luis Trillo-Contreras, Francisco D. Rodríguez-Gómez, Francisco Mayo, Elaheh Sobh-Doush, Carmen Ortiz-Salguero, Javier Villadiego and Reposo Ramírez-Lorca
Int. J. Mol. Sci. 2025, 26(21), 10290; https://doi.org/10.3390/ijms262110290 - 22 Oct 2025
Viewed by 220
Abstract
Aquaporin-4 (AQP4) is expressed in ependymal cells bordering the ventricles, the glia limitans, and pericapillary astrocyte endfeet forming the blood–brain barrier. The sporadic occurrence of obstructive congenital hydrocephalus (OH) has been observed in the offspring of AQP4/ mice generated in [...] Read more.
Aquaporin-4 (AQP4) is expressed in ependymal cells bordering the ventricles, the glia limitans, and pericapillary astrocyte endfeet forming the blood–brain barrier. The sporadic occurrence of obstructive congenital hydrocephalus (OH) has been observed in the offspring of AQP4/ mice generated in the CD1 strain background. Here, we used microarray analysis to explore gene expression profiles in the periaqueductal area from littermate AQP4/ pups at postnatal day 12. We compared wild-type (WT) animals with AQP4/ animals that developed OH (AQP4/-OH) and those that did not (AQP4/-NH). Bioinformatic analysis identified gene sets associated with proliferation and migration of microglia, ependymal cell adhesion, extracellular matrix components, axon myelination, and neuronal synapsis. Among the differentially expressed genes, Spp1—expressed by neonatal CD11c+ microglia—was highlighted in the triple comparison. Spp1 was significantly upregulated in AQP4/-NH and downregulated in AQP4/-OH mice. These findings suggest that CD11c+ microglia, via Spp1 expression, play a key morphogenic role in the aqueduct of Sylvius and their absence, occurring in a small subset of AQP4/-CD1 animals, leads to obstructive hydrocephalus. Full article
(This article belongs to the Special Issue Aquaporins in Brain Disease, 2nd Edition)
Show Figures

Figure 1

24 pages, 2986 KB  
Article
Transcriptional Profiling Defines Unique Subtypes of Transit Amplifying Neural Progenitors Within the Neonatal Mouse Subventricular Zone
by Rebecca Zaritsky, Ekta Kumari, Fernando Janczur Velloso, Alexander Lemenze, Seema Husain and Steven W. Levison
Biomolecules 2025, 15(10), 1438; https://doi.org/10.3390/biom15101438 - 11 Oct 2025
Viewed by 298
Abstract
While significant progress has been made in understanding the heterogeneity of Neural Stem Cells (NSCs), our understanding of similar heterogeneity among the more abundant transit amplifying progenitors is lagging. Our work on the neural progenitors (NPs) of the neonatal subventricular zone (SVZ) began [...] Read more.
While significant progress has been made in understanding the heterogeneity of Neural Stem Cells (NSCs), our understanding of similar heterogeneity among the more abundant transit amplifying progenitors is lagging. Our work on the neural progenitors (NPs) of the neonatal subventricular zone (SVZ) began over a decade ago, when we used antibodies to the four antigens, CD133, LeX, CD140a, and NG2 to perform Fluorescence-activated cell sorting to classify subsets of the neonatal mouse SVZ as either multi-potential (MP1, MP2, MP3, MP4 and PFMPs), glial-restricted (GRP1, GRP2, and GRP3), or neuron-astrocyte restricted (BNAP). Using RNA sequencing, we have characterized the distinctive molecular fingerprints of four SVZ neural progenitor subtypes and compared their gene expression profiles to those of the NSCs. We performed bioinformatic analyses to provide insights into each NP type’s unique interactome and the transcription factors regulating their development. Overall, we identified 1581 genes upregulated in at least one NP subset compared to the NSCs. Of these genes, 796 genes were upregulated in BNAP/GRP1 compared to NSCs; 653 in GRP2/MP3; 440 in GRP3; and 527 in PFMPs. One gene that emerged from our analysis that can be used to distinguish the NPs from the NSCs is Etv1, also known as Er81. Also notable is that the NSCs downregulated cilia formation genes as they differentiated to become multipotential progenitors. Among the NPs, both PFMP and GRP3 subtypes differentially expressed genes related to neuron and oligodendrocyte development, including Matn4, Lhfpl3 and Olig2. GRP3s uniquely expressed Etv5, a transcription factor known to promote glial cell fate specification, while PFMPs uniquely expressed Lhx6, a transcription factor that regulates interneuron specification. PFMPs also expressed transcripts for olfactory receptors. Unlike the other NPs, the GRP1 and GRP2 NPs upregulated expression of genes for proteins involved in immune function. The present work will serve as an important resource for investigators interested in further defining the transit amplifying progenitors of the mammalian SVZ. Full article
(This article belongs to the Special Issue Cellular and Molecular Biology of Neurodevelopment)
Show Figures

Figure 1

23 pages, 1704 KB  
Review
Expression of CD44 and Its Spliced Variants: Innate and Inducible Roles in Nervous Tissue Cells and Their Environment
by Maria Concetta Geloso, Francesco Ria, Valentina Corvino and Gabriele Di Sante
Int. J. Mol. Sci. 2025, 26(17), 8223; https://doi.org/10.3390/ijms26178223 - 24 Aug 2025
Viewed by 1478
Abstract
CD44, a structurally diverse cell-surface glycoprotein, plays a multifaceted and indispensable role in neural tissue across both physiological and pathological conditions. It orchestrates complex cell–extracellular matrix interactions and intracellular signaling through its variant isoforms and post-translational modifications and is broadly expressed in neural [...] Read more.
CD44, a structurally diverse cell-surface glycoprotein, plays a multifaceted and indispensable role in neural tissue across both physiological and pathological conditions. It orchestrates complex cell–extracellular matrix interactions and intracellular signaling through its variant isoforms and post-translational modifications and is broadly expressed in neural stem/progenitor cells, microglia, astrocytes, and selected neuronal populations. The interactions of CD44 with ligands such as hyaluronan and osteopontin regulate critical cellular functions, including migration, differentiation, inflammation, and synaptic plasticity. In microglia and macrophages, CD44 mediates immune signaling and phagocytic activity, and it is dynamically upregulated in neuroinflammatory diseases, particularly through pathways involving Toll-like receptor 4. CD44 expression in astrocytes is abundant during central nervous system development and in diseases, contributing to glial differentiation, reactive astrogliosis, and scar formation. Though its expression is less prominent in mature neurons, CD44 supports neural plasticity, circuit organization, and injury-induced repair mechanisms. Additionally, its expression at nervous system barriers, such as the blood–brain barrier, underscores its role in regulating vascular permeability during inflammation and ischemia. Collectively, CD44 emerges as a critical integrator of neural cell function and intercellular communication. Although the roles of CD44 in glial cells appear to be similar to those explored in other tissues, the expression of this molecule and its variants on neurons reveals peculiar functions. Elucidating the cell-type-specific roles and regulation of CD44 variants may offer novel therapeutic strategies for diverse neurological disorders. Full article
(This article belongs to the Collection Feature Papers in Molecular Neurobiology)
Show Figures

Graphical abstract

18 pages, 3217 KB  
Article
The Biflavonoid Agathisflavone Regulates Microglial and Astrocytic Inflammatory Profiles via Glucocorticoid Receptor
by Áurea Maria Alves Nunes Almeida, Cleonice Creusa dos Santos, Daniele Takahashi, Larissa Pereira da Silva, Verônica Moreira de Sousa, Monique Reis de Santana, Ana Elisa Del Arco, Balbino Lino dos Santos, Jorge Mauricio David, Victor Diogenes Amaral da Silva, Suzana Braga-de-Souza and Silvia Lima Costa
Molecules 2025, 30(5), 1014; https://doi.org/10.3390/molecules30051014 - 22 Feb 2025
Viewed by 1037
Abstract
Nuclear receptors such as glucocorticoid receptors (GRs) are transcription factors with prominent regulatory effects on neuroinflammation. Agathisflavone is a biflavonoid that demonstrates neurogenic, neuroprotective, anti-inflammatory, antioxidant, and pro-myelinogenic effects in vitro. This study investigated whether the control of glial reactivity by agathisflavone is [...] Read more.
Nuclear receptors such as glucocorticoid receptors (GRs) are transcription factors with prominent regulatory effects on neuroinflammation. Agathisflavone is a biflavonoid that demonstrates neurogenic, neuroprotective, anti-inflammatory, antioxidant, and pro-myelinogenic effects in vitro. This study investigated whether the control of glial reactivity by agathisflavone is mediated by GRs. Primary cultures of astrocytes and microglia were induced to neuroinflammation by lipopolysaccharides (LPSs) and exposed to agathisflavone or not in the presence or absence of mifepristone, a GR antagonist. The microglia morphology and reactivity were evaluated by immunofluorescence against calcium-binding ionized adapter (Iba-1) and CD68. The astrocyte morphology and reactivity were evaluated by immunofluorescence against glial fibrillary acidic protein (GFAP). The inflammatory profile was evaluated by RT-qPCR. Molecular docking was performed to characterize agathisflavone and GR interactions. Microglial branching was increased in response to agathisflavone, an effect that was inhibited by mifepristone. CD68 and GFAP expression was decreased by agathisflavone but not in the presence of mifepristone. Agathisflavone decreased the expression of the pro-inflammatory cytokine IL-1β and increased the expression of the regulatory cytokine IL-10. The increase in IL-10 mRNA was inhibited by the GR antagonist. The in silico analysis showed that agathisflavone binds to a pocket at the glucocorticoid receptor. These interactions were stronger than mifepristone, dexamethasone, and the agathisflavone monomer apigenin. These results indicate that the GR is involved in the regulatory effects of agathisflavone on microglia and astrocyte inflammation, contributing to the elucidation of the molecular mechanisms of agathisflavone’s effects in the nervous system. Full article
(This article belongs to the Special Issue Phytochemistry, Human Health and Molecular Mechanisms)
Show Figures

Graphical abstract

21 pages, 7648 KB  
Article
CX3CR1–Fractalkine Dysregulation Affects Retinal GFAP Expression, Inflammatory Gene Induction, and LPS Response in a Mouse Model of Hypoxic Retinopathy
by Colin Rorex, Sandra M. Cardona, Kaira A. Church, Derek Rodriguez, Difernando Vanegas, Reina A. Saldivar, Amira El-Sheikh, Yufeng Wang, Stefka Gyoneva, Anne C. Cotleur and Astrid E. Cardona
Int. J. Mol. Sci. 2025, 26(3), 1131; https://doi.org/10.3390/ijms26031131 - 28 Jan 2025
Cited by 1 | Viewed by 1881
Abstract
Diabetic retinopathy (DR) causes vision loss due to sustained inflammation and vascular damage. The vascular damage is evident by fibrinogen leakage, angiogenesis, and hypoxia. Neuronal regulation of microglia via the CX3CL1 (Fractalkine or FKN)-CX3CR1 pathway plays a significant role in retinal pathology. Defects [...] Read more.
Diabetic retinopathy (DR) causes vision loss due to sustained inflammation and vascular damage. The vascular damage is evident by fibrinogen leakage, angiogenesis, and hypoxia. Neuronal regulation of microglia via the CX3CL1 (Fractalkine or FKN)-CX3CR1 pathway plays a significant role in retinal pathology. Defects in FKN or CX3CR1 exacerbate inflammation, vascular damage, and vision impairment. However, the contribution of hypoxic astrocytes to the pathological process of DR is unclear. A hypoxic model (7 days of systemic 7.5% O2) was utilized to induce retinal damage in adult mice in the absence of systemic inflammatory signals. This model induced vascular and microglial responses similar to 10 weeks of STZ-induced hyperglycemia. The goal of this study is to characterize retinal damage in WT and mice with defects in the FKN-CX3CR1 signaling axis and hence assess the impact of the microglial inflammatory responses to hypoxic retinopathy. Tissues were analyzed by immunostaining, RNA sequencing, and cytokine quantification. We found that CX3CR1 deficiency in hypoxic animals induced reactive astrogliosis and that Müller glial responses to hypoxia and systemic inflammation were dependent on FKN signaling. Exacerbated microglial reactivity to hypoxic conditions significantly altered the expression of HIF transcripts. Microglial dysregulation was found to reduce the anti-inflammatory response to hypoxic conditions, downregulate hypoxia-responsive gene expression, and restrained LPS-induced inflammatory responses. We found that microglia dysregulation alters the hypoxic response by inhibiting the upregulation of HIF2α/3α, increasing CD31 immunoreactivity, and altering the expression of ECM-associated transcripts such as type I, III, and XVIII collagens to hypoxic conditions. Full article
(This article belongs to the Special Issue Advanced Research in Retina: 3rd Edition)
Show Figures

Figure 1

20 pages, 2651 KB  
Article
Alterations in Blood and Hippocampal mRNA and miRNA Expression, Along with Fat Deposition in Female B6C3F1 Mice Continuously Exposed to Prenatal Low-Dose-Rate Radiation and Their Comparison with Male Mice
by Hong Wang, Ignacia Braga Tanaka, Salihah Lau, Satoshi Tanaka, Amanda Tan and Feng Ru Tang
Cells 2025, 14(3), 173; https://doi.org/10.3390/cells14030173 - 23 Jan 2025
Viewed by 1432
Abstract
Our recent study revealed that continuous prenatal low-dose-rate irradiation did not induce cellular changes in the dentate gyrus of the hippocampus in male B6C3F1 mice exposed to gamma rays during prenatal development. However, changes in body weight, body mass index (BMI), locomotor ability, [...] Read more.
Our recent study revealed that continuous prenatal low-dose-rate irradiation did not induce cellular changes in the dentate gyrus of the hippocampus in male B6C3F1 mice exposed to gamma rays during prenatal development. However, changes in body weight, body mass index (BMI), locomotor ability, and mRNA and miRNA expressions in the hippocampus and blood were observed. To investigate potential sex differences in the effects of prenatal gamma irradiation, we conducted a parallel study on female B6C3F1 mice. The results showed significant reductions in the weight of the lungs and left kidney in prenatally irradiated female offspring, accompanied by significantly increased fat deposits in the mesentery, retroperitoneal, and left perigonadal areas. Despite these systemic changes, no cellular alterations were observed in the subgranular zone (immature neurons) or the hilus of the dentate gyrus (mature neurons and glial cells, including astrocytes, microglia, and oligodendrocyte progenitor cells). However, significant increases in hippocampal mRNA expression were detected for genes such as H2bc24, Fos, Cd74, Tent5a, Traip, and Sap25. Conversely, downregulation of mRNAs Inpp5j and Gdf3 was observed in whole blood. A Venn diagram highlighted the differential expression of two mRNAs, Ttn and Slc43a3, between the hippocampus and whole blood. Comparisons between prenatally irradiated male and female B6C3F1 mice revealed sex-specific differences. In whole blood, 4 mRNAs (Scd1, Cd59b, Vmn1r58, and Gm42427) and 1 miRNA (mmu-miR-8112) exhibited differential expression. In the hippocampus, 12 mRNAs and 2 novel miRNAs were differentially expressed between the sexes. qRT-PCR analysis validated the upregulation of H2bc24, Fos, Cd74, and Tent5a in the female hippocampus. These gene expression changes may be associated with the increased fat deposition observed following chronic low-dose-rate gamma irradiation exposure. This study underscores the importance of investigating sex-specific biological responses to prenatal gamma irradiation and highlights potential molecular pathways linked to observed physiological changes. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

14 pages, 8907 KB  
Article
Agathisflavone Modulates Reactive Gliosis After Trauma and Increases the Neuroblast Population at the Subventricular Zone
by Juliana Helena Castro e Silva, Francesca Pieropan, Andrea Domenico Rivera, Arthur Morgan Butt and Silvia Lima Costa
Nutrients 2024, 16(23), 4053; https://doi.org/10.3390/nu16234053 - 26 Nov 2024
Cited by 3 | Viewed by 1363
Abstract
Background: Reactive astrogliosis and microgliosis are coordinated responses to CNS insults and are pathological hallmarks of traumatic brain injury (TBI). In these conditions, persistent reactive gliosis can impede tissue repopulation and limit neurogenesis. Thus, modulating this phenomenon has been increasingly recognized as potential [...] Read more.
Background: Reactive astrogliosis and microgliosis are coordinated responses to CNS insults and are pathological hallmarks of traumatic brain injury (TBI). In these conditions, persistent reactive gliosis can impede tissue repopulation and limit neurogenesis. Thus, modulating this phenomenon has been increasingly recognized as potential therapeutic approach. Methods: In this study, we investigated the potential of the flavonoid agathisflavone to modulate astroglial and microglial injury responses and promote neurogenesis in the subventricular zone (SVZ) neurogenic niche. Agathisflavone, or the vehicle in controls, was administered directly into the lateral ventricles in postnatal day (P)8-10 mice by twice daily intracerebroventricular (ICV) injections for 3 days, and brains were examined at P11. Results: In the controls, ICV injection caused glial reactivity along the needle track, characterised immunohistochemically by increased astrocyte expression of glial fibrillary protein (GFAP) and the number of Iba-1+ microglia at the lesion site. Treatment with agathisflavone decreased GFAP expression, reduced both astrocyte reactivity and the number of Iba-1+ microglia at the core of the lesion site and the penumbra, and induced a 2-fold increase on the ratio of anti-inflammatory CD206+ to pro-inflammatory CD16/32+ microglia. Notably, agathisflavone increased the population of neuroblasts (GFAP+ type B cells) in all SVZ microdomains by up to double, without significantly increasing the number of neuronal progenitors (DCX+). Conclusions: Although future studies should investigate the underlying molecular mechanisms driving agathisflavone effects on microglial polarization and neurogenesis at different timepoints, these data indicate that agathisflavone could be a potential adjuvant treatment for TBI or central nervous system disorders that have reactive gliosis as a common feature. Full article
Show Figures

Graphical abstract

29 pages, 6971 KB  
Article
Honey Enriched with Additives Alleviates Behavioral, Oxidative Stress, and Brain Alterations Induced by Heavy Metals and Imidacloprid in Zebrafish
by Emanuela Paduraru, Roxana Jijie, Ira-Adeline Simionov, Cristina-Maria Gavrilescu, Tudor Ilie, Diana Iacob, Andreea Lupitu, Cristian Moisa, Claudia Muresan, Lucian Copolovici, Dana M. Copolovici, Gabriela Mihalache, Florin Daniel Lipsa, Gheorghe Solcan, Gabriela-Alexandra Danelet, Mircea Nicoara, Alin Ciobica and Carmen Solcan
Int. J. Mol. Sci. 2024, 25(21), 11730; https://doi.org/10.3390/ijms252111730 - 31 Oct 2024
Cited by 4 | Viewed by 3674
Abstract
Environmental concerns have consistently been a focal point for the scientific community. Pollution is a critical ecological issue that poses significant threats to human health and agricultural production. Contamination with heavy metals and pesticides is a considerable concern, a threat to the environment, [...] Read more.
Environmental concerns have consistently been a focal point for the scientific community. Pollution is a critical ecological issue that poses significant threats to human health and agricultural production. Contamination with heavy metals and pesticides is a considerable concern, a threat to the environment, and warrants special attention. In this study, we investigated the significant issues arising from sub-chronic exposure to imidacloprid (IMI), mercury (Hg), and cadmium (Cd), either alone or in combination, using zebrafish (Danio rerio) as an animal model. Additionally, we assessed the potential protective effects of polyfloral honey enriched with natural ingredients, also called honey formulation (HF), against the combined sub-chronic toxic effects of the three contaminants. The effects of IMI (0.5 mg·L−1), Hg (15 μg·L−1), and Cd (5 μg·L−1), both individually and in combination with HF (500 mg·L−1), on zebrafish were evaluated by quantifying acetylcholinesterase (AChE) activity, lipid peroxidation (MDA), various antioxidant enzyme activities like superoxide dismutase and glutathione peroxidase (SOD and GPx), 2D locomotor activity, social behavior, histological and immunohistochemical factors, and changes in body element concentrations. Our findings revealed that all concentrations of pollutants may disrupt social behavior, diminish swimming performances (measured by total distance traveled, inactivity, and swimming speed), and elevate oxidative stress (OS) biomarkers of SOD, GPx, and MDA in zebrafish over the 21-day administration period. Fish exposed to IMI and Hg + Cd + IMI displayed severe lesions and increased GFAP (Glial fibrillary acidic protein) and S100B (S100 calcium-binding protein B) protein expression in the optic tectum and cerebellum, conclusively indicating astrocyte activation and neurotoxic effects. Furthermore, PCNA (Proliferating cell nuclear antigen) staining revealed reduced cell proliferation in the IMI-exposed group, contrasting with intensified proliferation in the Hg + Cd group. The nervous system exhibited significant damage across all studied concentrations, confirming the observed behavioral changes. Moreover, HF supplementation significantly mitigated the toxicity induced by contaminants and reduced OS. Therefore, the exposure to chemical mixtures offers a more complete picture of adverse impacts on aquatic ecosystems and the supplementation with bioactive compounds can help to reduce the toxicity induced by exposure to environmental pollutants. Full article
(This article belongs to the Special Issue Mechanisms of Neurotoxicity)
Show Figures

Graphical abstract

13 pages, 1185 KB  
Article
The Absence of CXCL10 Activity Does Not Affect the Capability of CD8+ T Cells to Migrate and Eliminate the Tissue Cysts of Toxoplasma gondii from the Brains of Chronically Infected Mice
by Rajesh Mani and Yasuhiro Suzuki
Microorganisms 2024, 12(11), 2172; https://doi.org/10.3390/microorganisms12112172 - 29 Oct 2024
Viewed by 1228
Abstract
Toxoplasma gondii forms tissue cysts in neurons and astrocytes in the brain to establish chronic infection, and astrocytes express the CXCL10 chemokine in chronically infected mice. Since chemokines mediate the migration of T cells to attack their targets, and since CXCL10 plays key [...] Read more.
Toxoplasma gondii forms tissue cysts in neurons and astrocytes in the brain to establish chronic infection, and astrocytes express the CXCL10 chemokine in chronically infected mice. Since chemokines mediate the migration of T cells to attack their targets, and since CXCL10 plays key roles in T cell-mediated control of the proliferation of tachyzoites (the acute stage form) of T. gondii during the acute stage of infection, we examined whether CXCL10 is involved in recruiting anti-cyst CD8+ cytotoxic T cells to eliminate the cysts in their brains. We employed adoptive transfer of CD8+ immune T cells to infected, T cell-deficient SCID and RAG1−/− mice in combination with blocking CXCL10 activity by neutralizing antibody or a deletion of this chemokine gene. The treatment of chronically infected (infected and treated with sulfadiazine) SCID mice with the anti-CXCL10 antibody did not inhibit the recruitment of the transferred CD8+ T cells into their brains and the removal of cerebral T. gondii cysts by the T cells. In addition, the neutralization of CXCL10 did not reduce the cerebral expression of mRNA for the mediators (perforin and granzyme B [GzmB]) of the cytotoxic activity of CD8+ T cells in the SCID mice. Consistently, the adoptive transfer of CD8+ immune T cells to chronically infected RAG1−/−CXCL10−/− mice did not show any defects in recruiting the CD8+ T cells into their brains and eliminating the cysts when compared to infected RAG1−/− mice. The former rather displayed enhanced cyst removal with increased cerebral expression of GzmB mRNA. These results indicate that the absence of CXCL10 activity does not ablate the capability of CD8+ cytotoxic T cells to migrate into the brain and eliminate T. gondii cysts from the brains of chronically infected mice. These results also suggest that the immune system utilizes distinct chemokines to control T. gondii depending on the two different life cycle stages, tachyzoite and cyst, of this protozoan parasite. Full article
(This article belongs to the Special Issue Parasitic Diseases in Humans and Animals)
Show Figures

Figure 1

31 pages, 3191 KB  
Article
In Vitro Assessment of the Neuroprotective Effects of Pomegranate (Punica granatum L.) Polyphenols Against Tau Phosphorylation, Neuroinflammation, and Oxidative Stress
by Mehdi Alami, Kaoutar Boumezough, Echarki Zerif, Nada Zoubdane, Abdelouahed Khalil, Ton Bunt, Benoit Laurent, Jacek M. Witkowski, Charles Ramassamy, Samira Boulbaroud, Tamas Fulop and Hicham Berrougui
Nutrients 2024, 16(21), 3667; https://doi.org/10.3390/nu16213667 - 28 Oct 2024
Cited by 9 | Viewed by 3626
Abstract
Background: Oxidative stress and chronic inflammation, at both the systemic and the central level, are critical early events in atherosclerosis and Alzheimer’s disease (AD). Purpose: To investigate the oxidative stress-, inflammation-, and Tau-phosphorylation-lowering effects of pomegranate polyphenols (PPs) (punicalagin, ellagic acid, peel, and [...] Read more.
Background: Oxidative stress and chronic inflammation, at both the systemic and the central level, are critical early events in atherosclerosis and Alzheimer’s disease (AD). Purpose: To investigate the oxidative stress-, inflammation-, and Tau-phosphorylation-lowering effects of pomegranate polyphenols (PPs) (punicalagin, ellagic acid, peel, and aril extracts). Methods: We used flow cytometry to quantify the protein expression of proinflammatory cytokines (IL-1β) and anti-inflammatory mediators (IL-10) in THP-1 macrophages, as well as M1/M2 cell-specific marker (CD86 and CD163) expression in human microglia HMC3 cells. The IL-10 protein expression was also quantified in U373-MG human astrocytes. The effect of PPs on human amyloid beta 1-42 (Aβ1-42)-induced oxidative stress was assessed in the microglia by measuring ROS generation and lipid peroxidation, using 2′,7′-dichlorofluorescein diacetate (DCFH-DA) and thiobarbituric acid reactive substance (TBARS) tests, respectively. Neuronal viability and cell apoptotic response to Aβ1-42 toxicity were assayed using the MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay and the annexin-V-FITC apoptosis detection kit, respectively. Finally, flow cytometry analysis was also performed to evaluate the ability of PPs to modulate Aβ1-42-induced Tau-181 phosphorylation (pTau-181). Results: Our data indicate that PPs are significantly (p < 0.05) effective in countering Aβ1-42-induced inflammation through increasing the anti-inflammatory cytokines (IL-10) in U373-MG astrocytes and THP1 macrophages and decreasing proinflammatory marker (IL-1β) expression in THP1 macrophages. The PPs were also significantly (p < 0.05) effective in inducing the phenotypic transition of THP-1 macrophages and microglial cells from M1 to M2 by decreasing CD86 and increasing CD163 surface receptor expression. Moreover, our treatments have a significant (p < 0.05) beneficial impact on oxidative stress, illustrated in the reduction in TBARS and ROS generation. Our treatments have significant (p < 0.05) cell viability improvement capacities and anti-apoptotic effects on human H4 neurons. Furthermore, our results suggest that Aβ1-42 significantly (p < 0.05) increases pTau-181. This effect is significantly (p < 0.05) attenuated by arils, peels, and punicalagin and drastically reduced by the ellagic acid treatment. Conclusion: Overall, our results attribute to PPs anti-inflammatory, antioxidant, anti-apoptotic, and anti-Tau-pathology potential. Future studies should aim to extend our knowledge of the potential role of PPs in Aβ1-42-induced neurodegeneration, particularly concerning its association with the tauopathy involved in AD. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

14 pages, 970 KB  
Article
Increased Pan-Type, A1-Type, and A2-Type Astrocyte Activation and Upstream Inflammatory Markers Are Induced by the P2X7 Receptor
by Keith E. Campagno, Puttipong Sripinun, Lily P. See, Jiaqi Li, Wennan Lu, Assraa Hassan Jassim, Néstor Más Gómez and Claire H. Mitchell
Int. J. Mol. Sci. 2024, 25(16), 8784; https://doi.org/10.3390/ijms25168784 - 13 Aug 2024
Cited by 6 | Viewed by 2794
Abstract
This study asked whether the P2X7 receptor was necessary and sufficient to trigger astrocyte polarization into neuroinflammatory activation states. Intravitreal injection of agonist BzATP increased gene expression of pan-astrocyte activation markers Gfap, Steap4, and Vim and A1-type astrocyte activation markers C3 [...] Read more.
This study asked whether the P2X7 receptor was necessary and sufficient to trigger astrocyte polarization into neuroinflammatory activation states. Intravitreal injection of agonist BzATP increased gene expression of pan-astrocyte activation markers Gfap, Steap4, and Vim and A1-type astrocyte activation markers C3, Serping1, and H2T23, but also the Cd14 and Ptx3 genes usually associated with the A2-type astrocyte activation state and Tnfa, IL1a, and C1qa, assumed to be upstream of astrocyte activation in microglia. Correlation analysis of gene expression suggested the P2X7 receptor induced a mixed A1/A2-astrocyte activation state, although A1-state genes like C3 increased the most. A similar pattern of mixed glial activation genes occurred one day after intraocular pressure (IOP) was elevated in wild-type mice, but not in P2X7-/- mice, suggesting the P2X7 receptor is necessary for the glial activation that accompanies IOP elevation. In summary, this study suggests stimulation of the P2X7R is necessary and sufficient to trigger the astrocyte activation in the retina following IOP elevation, with a rise in markers for pan-, A1-, and A2-type astrocyte activation. The P2X7 receptor is expressed on microglia, optic nerve head astrocytes, and retinal ganglion cells (RGCs) in the retina, and can be stimulated by the mechanosensitive release of ATP that accompanies IOP elevation. Whether the P2X7 receptor connects this mechanosensitive ATP release to microglial and astrocyte polarization in glaucoma remains to be determined. Full article
Show Figures

Figure 1

24 pages, 8623 KB  
Article
Loss of Mitochondrial Tusc2/Fus1 Triggers a Brain Pro-Inflammatory Microenvironment and Early Spatial Memory Impairment
by Tonie Farris, Salvador González-Ochoa, Muna Mohammed, Harshana Rajakaruna, Jane Tonello, Thanigaivelan Kanagasabai, Olga Korolkova, Akiko Shimamoto, Alla Ivanova and Anil Shanker
Int. J. Mol. Sci. 2024, 25(13), 7406; https://doi.org/10.3390/ijms25137406 - 5 Jul 2024
Viewed by 2706
Abstract
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to [...] Read more.
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2−/− female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2−/− female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2−/− brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2−/− hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2−/− mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health. Full article
(This article belongs to the Special Issue Immunometabolic Disorders in Aging and Disease)
Show Figures

Figure 1

25 pages, 4973 KB  
Article
Adult Human Brain Tissue Cultures to Study NeuroHIV
by Rachel Van Duyne, Elena Irollo, Angel Lin, James A. Johnson, Alain M. Guillem, Erick V. O’Brien, Laura Merja, Bradley Nash, Joshua G. Jackson, Atom Sarkar, Zachary A. Klase and Olimpia Meucci
Cells 2024, 13(13), 1127; https://doi.org/10.3390/cells13131127 - 29 Jun 2024
Cited by 3 | Viewed by 3057
Abstract
HIV-associated neurocognitive disorders (HAND) persist under antiretroviral therapy as a complex pathology that has been difficult to study in cellular and animal models. Therefore, we generated an ex vivo human brain slice model of HIV-1 infection from surgically resected adult brain tissue. Brain [...] Read more.
HIV-associated neurocognitive disorders (HAND) persist under antiretroviral therapy as a complex pathology that has been difficult to study in cellular and animal models. Therefore, we generated an ex vivo human brain slice model of HIV-1 infection from surgically resected adult brain tissue. Brain slice cultures processed for flow cytometry showed >90% viability of dissociated cells within the first three weeks in vitro, with parallel detection of astrocyte, myeloid, and neuronal populations. Neurons within brain slices showed stable dendritic spine density and mature spine morphologies in the first weeks in culture, and they generated detectable activity in multi-electrode arrays. We infected cultured brain slices using patient-matched CD4+ T-cells or monocyte-derived macrophages (MDMs) that were exposed to a GFP-expressing R5-tropic HIV-1 in vitro. Infected slice cultures expressed viral RNA and developed a spreading infection up to 9 days post-infection, which were significantly decreased by antiretrovirals. We also detected infected myeloid cells and astrocytes within slices and observed minimal effect on cellular viability over time. Overall, this human-centered model offers a promising resource to study the cellular mechanisms contributing to HAND (including antiretroviral toxicity, substance use, and aging), infection of resident brain cells, and new neuroprotective therapeutics. Full article
Show Figures

Figure 1

13 pages, 3238 KB  
Article
Derivatization of Hyaluronan to Target Neuroblastoma and Neuroglioma Expressing CD44
by Giau Van Vo, Kummara Madhusudana Rao, Ildoo Chung, Chang-Sik Ha, Seong Soo A. An and Yang H. Yun
Pharmaceutics 2024, 16(6), 836; https://doi.org/10.3390/pharmaceutics16060836 - 20 Jun 2024
Cited by 2 | Viewed by 2299
Abstract
Therapeutics for actively targeting over-expressed receptors are of great interest because the majority of diseased tissues originate from normal cells and do not possess a unique receptor from which they can be differentiated. One such receptor is CD44, which has been shown to [...] Read more.
Therapeutics for actively targeting over-expressed receptors are of great interest because the majority of diseased tissues originate from normal cells and do not possess a unique receptor from which they can be differentiated. One such receptor is CD44, which has been shown to be highly overexpressed in many breast cancers and other types of cancer cells. While CD44 has been documented to express low levels in normal adult neurons, astrocytes, and microglia, this receptor may be overexpressed by neuroblastoma and neuroglioma. If differential expression exists between normal and cancerous cells, hyaluronan (HA) could be a useful carrier that targets carcinomas. Thus, HA was conjugated with resveratrol (HA-R), and its efficacy was tested on cortical–neuroblastoma hybrid, neuroblastoma, and neuroglioma cells. Confocal and flow cytometry showed these cells express CD44 and are able to bind and uptake HA-R. The toxicity of HA-R correlated well with CD44 expression in this study. Therefore, conjugating resveratrol and other chemotherapeutics to HA could minimize the side effects for normal cells within the brain and nervous system and could be a viable strategy for developing targeted therapies. Full article
(This article belongs to the Special Issue Carbohydrate-Based Carriers for Drug Delivery)
Show Figures

Graphical abstract

15 pages, 4760 KB  
Article
Astrogliosis in the GFAP-CreERT2:Rosa26iDTR Mouse Model Does Not Exacerbate Retinal Microglia Activation or Müller Cell Gliosis under Hypoxic Conditions
by Colin Rorex, Sandra M. Cardona, Kaira A. Church, Derek Rodriguez, Difernando Vanegas, Reina Saldivar, Brianna Faz and Astrid E. Cardona
Biomolecules 2024, 14(5), 567; https://doi.org/10.3390/biom14050567 - 10 May 2024
Cited by 3 | Viewed by 3751
Abstract
Diabetic retinopathy (DR) affects over 140 million people globally. The mechanisms that lead to blindness are still enigmatic but there is evidence that sustained inflammation and hypoxia contribute to vascular damage. Despite efforts to understand the role of inflammation and microglia in DR’s [...] Read more.
Diabetic retinopathy (DR) affects over 140 million people globally. The mechanisms that lead to blindness are still enigmatic but there is evidence that sustained inflammation and hypoxia contribute to vascular damage. Despite efforts to understand the role of inflammation and microglia in DR’s pathology, the contribution of astrocytes to hypoxic responses is less clear. To investigate the role of astrocytes in hypoxia-induced retinopathy, we utilized a 7-day systemic hypoxia model using the GFAP-CreERT2:Rosa26iDTR transgenic mouse line. This allows for the induction of inflammatory reactive astrogliosis following tamoxifen and diphtheria toxin administration. We hypothesize that DTx-induced astrogliosis is neuroprotective during hypoxia-induced retinopathy. Glial, neuronal, and vascular responses were quantified using immunostaining, with antibodies against GFAP, vimentin, IBA-1, NeuN, fibrinogen, and CD31. Cytokine responses were measured in both the brain and serum. We report that while both DTx and hypoxia induced a phenotype of reduced microglia morphological activation, DTx, but not hypoxia, induced an increase in the Müller glia marker vimentin. We did not observe that the combination of DTx and hypoxic treatments exacerbated the signs of reactive glial cells, nor did we observe a significant change in the expression immunomodulatory mediators IL-1β, IL2, IL-4, IL-5, IL-6, IL-10, IL-18, CCL17, TGF-β1, GM-CSF, TNF-α, and IFN-γ. Overall, our results suggest that, in this hypoxia model, reactive astrogliosis does not alter the inflammatory responses or cause vascular damage in the retina. Full article
(This article belongs to the Special Issue Molecular Interventions in Ocular Disorders)
Show Figures

Figure 1

Back to TopTop