Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Immunol ; 25(7): 1158-1171, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38902519

ABSTRACT

Up to 25% of individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibit postacute cognitive sequelae. Although millions of cases of coronavirus disease 2019 (COVID-19)-mediated memory dysfunction are accumulating worldwide, the underlying mechanisms and how vaccination lowers risk are unknown. Interleukin-1 (IL-1), a key component of innate immune defense against SARS-CoV-2 infection, is elevated in the hippocampi of individuals with COVID-19. Here we show that intranasal infection of C57BL/6J mice with SARS-CoV-2 Beta variant leads to central nervous system infiltration of Ly6Chi monocytes and microglial activation. Accordingly, SARS-CoV-2, but not H1N1 influenza virus, increases levels of brain IL-1ß and induces persistent IL-1R1-mediated loss of hippocampal neurogenesis, which promotes postacute cognitive deficits. Vaccination with a low dose of adenoviral-vectored spike protein prevents hippocampal production of IL-1ß during breakthrough SARS-CoV-2 infection, loss of neurogenesis and subsequent memory deficits. Our study identifies IL-1ß as one potential mechanism driving SARS-CoV-2-induced cognitive impairment in a new mouse model that is prevented by vaccination.


Subject(s)
COVID-19 , Hippocampus , Interleukin-1beta , Memory Disorders , Mice, Inbred C57BL , Neurogenesis , SARS-CoV-2 , Animals , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Mice , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Hippocampus/immunology , Hippocampus/metabolism , Memory Disorders/immunology , Neurogenesis/immunology , Vaccination , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Vaccines/immunology , Male , Humans , Microglia/immunology , Microglia/metabolism , Disease Models, Animal , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-1 Type I/genetics , Monocytes/immunology , Monocytes/metabolism , Female
2.
J Neuroimmune Pharmacol ; 14(3): 375-382, 2019 09.
Article in English | MEDLINE | ID: mdl-30905008

ABSTRACT

HIV-1 infection causes chronic neuroinflammation resulting in cognitive decline associated with diminution of survival of neural stem cells (NSC). In part, this is attributable to production of toxic viral proteins (gp120 and tat) by infected cells in the brain that can activate microglia. Here, we evaluated a novel model for HIV-1 neuropathogenesis by direct administration of viral proteins into the hippocampus. Chronic administration of either HIV-1 gp120 or tat over 14 days significantly decreased NSC proliferation, survival and neuroblast formation (by 32-37%) within the hippocampal subgranular zone as detected by doublecortin/BrdU or Ki67-positive cells. Intrahippocampal administration of gp120 or tat induced microglial activation within the hippocampus as determined by increases in microglial number and increases in the volume of the microglia (2.5-3-fold, evaluated by double IBA-1/CD68 staining). We further assessed inflammatory responses within the hippocampus by RNAseq and Ingenuity Pathway Analysis. There was a significant mRNA upregulation of numerous inflammatory mediators including Il1b, Icam1, Il12a, Ccl2, and Ccl4. These data suggest that chronic administration induces a prolonged inflammatory state within the hippocampus that negatively affects NSC survival potentially leading to cognitive dysfunction. Graphical Abstract.


Subject(s)
AIDS Dementia Complex/etiology , Disease Models, Animal , HIV Envelope Protein gp120/toxicity , HIV-1/pathogenicity , Hippocampus/drug effects , Inflammation/chemically induced , Neural Stem Cells/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , HIV Envelope Protein gp120/administration & dosage , Hippocampus/metabolism , Hippocampus/pathology , Inflammation Mediators/metabolism , Infusions, Parenteral , Mice , Mice, Inbred C57BL , Microglia/physiology , Neural Stem Cells/pathology , Neurogenesis , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Sequence Analysis, RNA , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
3.
Brain Behav Immun ; 76: 165-181, 2019 02.
Article in English | MEDLINE | ID: mdl-30465881

ABSTRACT

New neurons are continuously produced by neural stem cells (NSCs) within the adult hippocampus. Numerous diseases, including major depressive disorder and HIV-1 associated neurocognitive disorder, are associated with decreased rates of adult neurogenesis. A hallmark of these conditions is a chronic release of neuroinflammatory mediators by activated resident glia. Recent studies have shown a neuroprotective role on NSCs of cannabinoid receptor activation. Yet, little is known about the effects of GPR55, a candidate cannabinoid receptor, activation on reductions of neurogenesis in response to inflammatory insult. In the present study, we examined NSCs exposed to IL-1ß in vitro to assess inflammation-caused effects on NSC differentiation and the ability of GPR55 agonists to attenuate NSC injury. NSC differentiation and neurogenesis was determined via immunofluorescence and flow cytometric analysis of NSC markers (Nestin, Sox2, DCX, S100ß, ßIII Tubulin, GFAP). GPR55 agonist treatment protected against IL-1ß induced reductions in neurogenesis rates. Moreover, inflammatory cytokine receptor mRNA expression was down regulated by GPR55 activation in a neuroprotective manner. To determine inflammatory responses in vivo, we treated C57BL/6 and GPR55-/- mice with LPS (0.2 mg/kg/day) continuously for 14 days via osmotic mini-pump. Reductions in NSC survival (as determined by BrdU incorporation), immature neurons, and neuroblast formation due to LPS were attenuated by concurrent direct intrahippocampal administration of the GPR55 agonist, O-1602 (4 µg/kg/day). Molecular analysis of the hippocampal region showed a suppressed ability to regulate immune responses by GPR55-/- animals manifesting in a prolonged inflammatory response (IL-1ß, IL-6, TNFα) after chronic, systemic inflammation as compared to C57BL/6 animals. Taken together, these results suggest a neuroprotective role of GPR55 activation on NSCs in vitro and in vivo and that GPR55 provides a novel therapeutic target against negative regulation of hippocampal neurogenesis by inflammatory insult.


Subject(s)
Hippocampus/metabolism , Inflammation/metabolism , Neural Stem Cells/immunology , Neurogenesis/physiology , Receptors, Cannabinoid/metabolism , Animals , Cannabidiol/analogs & derivatives , Cannabidiol/pharmacology , Cell Differentiation/drug effects , Doublecortin Protein , Female , Hippocampus/immunology , Hippocampus/pathology , Immunity, Active , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotection/drug effects , Neuroprotection/immunology , Receptors, Cannabinoid/genetics , Receptors, Cannabinoid/immunology
4.
Br J Pharmacol ; 175(16): 3407-3421, 2018 08.
Article in English | MEDLINE | ID: mdl-29888782

ABSTRACT

BACKGROUND AND PURPOSE: The cannabinoid system exerts functional regulation of neural stem cell (NSC) proliferation and adult neurogenesis, yet not all effects of cannabinoid-like compounds seen can be attributed to the cannabinoid 1 (CB1 ) or CB2 receptor. The recently de-orphaned GPR55 has been shown to be activated by numerous cannabinoid ligands suggesting that GPR55 is a third cannabinoid receptor. Here, we examined the role of GPR55 activation in NSC proliferation and early adult neurogenesis. EXPERIMENTAL APPROACH: The effects of GPR55 agonists (LPI, O-1602, ML184) on human (h) NSC proliferation in vitro were assessed by flow cytometry. Human NSC differentiation was determined by flow cytometry, qPCR and immunohistochemistry. Immature neuron formation in the hippocampus of C57BL/6 and GPR55-/- mice was evaluated by immunohistochemistry. KEY RESULTS: Activation of GPR55 significantly increased proliferation rates of hNSCs in vitro. These effects were attenuated by ML193, a selective GPR55 antagonist. ML184 significantly promoted neuronal differentiation in vitro while ML193 reduced differentiation rates as compared to vehicle treatment. Continuous administration of O-1602 into the hippocampus via a cannula connected to an osmotic pump resulted in increased Ki67+ cells within the dentate gyrus. O-1602 increased immature neuron generation, as assessed by DCX+ and BrdU+ cells, as compared to vehicle-treated animals. GPR55-/- animals displayed reduced rates of proliferation and neurogenesis within the hippocampus while O-1602 had no effect as compared to vehicle controls. CONCLUSIONS AND IMPLICATIONS: Together, these findings suggest GPR55 activation as a novel target and strategy to regulate NSC proliferation and adult neurogenesis.


Subject(s)
Hippocampus/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Receptors, Cannabinoid/physiology , Receptors, G-Protein-Coupled/agonists , Animals , Cannabidiol/analogs & derivatives , Cannabidiol/pharmacology , Cell Line , Cell Proliferation/drug effects , Doublecortin Protein , Hippocampus/drug effects , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Receptors, G-Protein-Coupled/physiology
5.
Stem Cells Dev ; 24(12): 1377-89, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25892136

ABSTRACT

Recent reports indicate that neural stem cells (NSCs) exist in a cluster-like formation in close proximity to cerebral microvessels. Similar appearing clusters can be seen ex vivo in NSC cultures termed neurospheres. It is known that this neurosphere configuration is important for preserving stemness and a proliferative state. How NSCs form neurospheres or neuroclusters remains largely undetermined. In this study, we show that primary human NSCs express the tight junction proteins (TJPs): zonula occludens-1 (ZO-1), occludin, claudin-1, -3, -5, and -12. The relative mRNA expression was measured by quantitative polymerase chain reaction, and protein expression was confirmed by flow cytometry and immunofluorescence microscopy. Our results show that downregulation of TJPs occurs as neuronal differentiation is induced, suggesting that control of TJPs may be tied to the neuronal differentiation program. Importantly, upon specific knockdown of the accessory TJP, ZO-1, undifferentiated NSCs showed decreased levels of key stem cell markers. Taken together, our results indicate that TJPs possibly aid in maintaining the intercellular configuration of NSCs and that reduction in TJP expression consequently affects the stemness status.


Subject(s)
Cell Differentiation/genetics , Neural Stem Cells/metabolism , Tight Junction Proteins/biosynthesis , Zonula Occludens-1 Protein/genetics , Claudin-1/biosynthesis , Claudin-3/biosynthesis , Claudin-5/biosynthesis , Claudins/biosynthesis , Flow Cytometry , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/metabolism , Humans , Occludin/biosynthesis , Tight Junctions/metabolism , Zonula Occludens-1 Protein/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...