Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Nucleic Acids Res ; 48(19): 10924-10939, 2020 11 04.
Article En | MEDLINE | ID: mdl-33010171

NBS1 is a critical component of the MRN (MRE11/RAD50/NBS1) complex, which regulates ATM- and ATR-mediated DNA damage response (DDR) pathways. Mutations in NBS1 cause the human genomic instability syndrome Nijmegen Breakage Syndrome (NBS), of which neuronal deficits, including microcephaly and intellectual disability, are classical hallmarks. Given its function in the DDR to ensure proper proliferation and prevent death of replicating cells, NBS1 is essential for life. Here we show that, unexpectedly, Nbs1 deletion is dispensable for postmitotic neurons, but compromises their arborization and migration due to dysregulated Notch signaling. We find that Nbs1 interacts with NICD-RBPJ, the effector of Notch signaling, and inhibits Notch activity. Genetic ablation or pharmaceutical inhibition of Notch signaling rescues the maturation and migration defects of Nbs1-deficient neurons in vitro and in vivo. Upregulation of Notch by Nbs1 deletion is independent of the key DDR downstream effector p53 and inactivation of each MRN component produces a different pattern of Notch activity and distinct neuronal defects. These data indicate that neuronal defects and aberrant Notch activity in Nbs1-deficient cells are unlikely to be a direct consequence of loss of MRN-mediated DDR function. This study discloses a novel function of NBS1 in crosstalk with the Notch pathway in neuron development.


Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neurogenesis , Neurons/metabolism , Receptors, Notch/metabolism , Acid Anhydride Hydrolases/metabolism , Animals , Cells, Cultured , DNA Damage , DNA Repair , Embryo, Mammalian , Fibroblasts , MRE11 Homologue Protein/metabolism , Mice , Neurons/cytology
2.
Mol Cell Neurosci ; 78: 1-8, 2017 01.
Article En | MEDLINE | ID: mdl-27825984

Microglial motility is tightly controlled by multitude of agonistic and antagonistic factors. Chemoattractants, released after infection or damage of the brain, provoke directed migration of microglia to the pathogenic incident. In contrast, noradrenaline and other stress hormones have been shown to suppress microglial movement. Here we asked for the signaling reactions involved in the positive and negative control of microglial motility. Using pharmacological and genetic approaches we identified the lipid kinase activity of phosphoinositide 3-kinase species γ (PI3Kγ) as an essential mediator of microglial migration provoked by the complement component C5a and other chemoattractants. Inhibition of PI3Kγ lipid kinase activity by protein kinase A was disclosed as mechanism causing suppression of microglial migration by noradrenaline. Together these data characterize PI3Kγ as a nodal point in the control of microglial motility.


Adrenergic alpha-Agonists/pharmacology , Chemotactic Factors/pharmacology , Chemotaxis , Microglia/metabolism , Norepinephrine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line , Cells, Cultured , Complement C5a/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/physiology , Phosphatidylinositol 3-Kinases/genetics
3.
Glia ; 65(2): 416-428, 2017 02.
Article En | MEDLINE | ID: mdl-27859601

Microglia cells are brain macrophages whose proper functioning is essential for maintenance and repair processes of the central nervous system (CNS). Migration and phagocytosis are critical aspects of microglial activity. By using genetically modified cell lines and knockout mice we demonstrate here that the receptor protein-tyrosine phosphatase (PTP) DEP-1 (also known as PTPRJ or CD148) acts as a positive regulator of both processes in vitro and in vivo. Notably, reduced microglial migration was detectable in brains of Ptprj-/- mice using a wounding assay. Mechanistically, density-enhanced phosphatase-1 (DEP-1) may in part function by inhibiting the activity of the Src family kinase Fyn. In the microglial cell line BV2 DEP-1 depletion by shRNA-mediated knockdown resulted in enhanced phosphorylation of the Fyn activating tyrosine (Tyr420 ) and elevated specific Fyn-kinase activity in immunoprecipitates. Moreover, Fyn mRNA and protein levels were reduced in DEP-1 deficient microglia cells. Consistent with a negative regulatory role of Fyn for microglial functions, which is inhibited by DEP-1, microglial cells from Fyn-/- mice exhibited elevated migration and phagocytosis. Enhanced microglia migration to a site of injury was also observed in Fyn-/- mice in vivo. Taken together our data revealed a previously unrecognized role of DEP-1 and suggest the existence of a potential DEP-1-Fyn axis in the regulation of microglial functions. GLIA 2017;65:416-428.


Cell Movement/physiology , Gene Expression Regulation/genetics , Microglia/physiology , Phagocytosis/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Animals , Animals, Newborn , Cell Line, Transformed , Cell Movement/genetics , Cells, Cultured , Cerebral Cortex/cytology , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/physiology , Proto-Oncogene Proteins c-fyn/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism
4.
Immunol Lett ; 180: 9-16, 2016 12.
Article En | MEDLINE | ID: mdl-27720677

Due to their ability to phagocytise invading microbes macrophages play a key role in the innate and acquired immune system. In this article the role of phosphoinositide 3-kinase gamma (PI3Kγ) for phagocytosis was studied in bone marrow derived macrophages (BMDM). By using genetic and pharmacological approaches our data clearly demonstrate PI3Kγ is acting as a mediator of macrophage phagocytosis. Phagocytosis of LPS activated BMDM was reduced in PI3Kγ depleted primary BMDM or macrophage cell line J774. Depletion of other class I phosphoinositide 3-kinases did not alter phagocytic activity. Partial reduction of the phagocytic index of BMDM expressing kinase inactive PI3Kγ indicate a lipid-kinase independent role of the PI3Kγ protein. Since inhibition of PI3Kγ interaction partner phosphodiesterase PDE3B reduced BMDM phagocytosis and PI3Kγ knock out super stimulated cAMP level, our data reveal that PI3Kγ protein mediated suppression of cAMP signalling is a critical for efficient phagocytosis of macrophages.


Bone Marrow/metabolism , Bone Marrow/physiology , Macrophages/metabolism , Macrophages/physiology , Phagocytosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line , Mice , Mice, Knockout , Signal Transduction/physiology
5.
Mol Neurobiol ; 53(8): 5468-79, 2016 10.
Article En | MEDLINE | ID: mdl-26452362

Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and phagocytosis. This study was conducted to elucidate conjectural differences of lipid kinase-dependent and kinase-independent functions of PI3Kγ in the evolvement of brain damage induced by focal cerebral ischemia/reperfusion. Therefore, PI3Kγ wild-type, knockout, and kinase-dead mice were subjected to middle cerebral artery occlusion followed by reperfusion. Tissue damage and cellular composition were assessed by immunohistochemical stainings. In addition, microglial cells derived from respective mouse genotypes were used for analysis of PI3Kγ effects on phagocytic activity, matrix metalloproteinase-9 release, and cAMP content under conditions of oxygen/glucose deprivation and recovery. Brain infarction was more pronounced in PI3Kγ-knockout mice compared to wild-type and kinase-dead mice 48 h after reperfusion. Immunohistochemical analyses revealed a reduced amount of galectin-3/MAC-2-positive microglial cells indicating that activated phagocytosis was reduced in ischemic brains of knockout mice. Cell culture studies disclosed enhanced metalloproteinase-9 secretion in supernatants derived from microglia of PI3Kγ-deficient mice after 2-h oxygen/glucose deprivation and 48-h recovery. Furthermore, PI3Kγ-deficient microglial cells showed a failed phagocytic activation throughout the observed recovery period. Lastly, PI3Kγ-deficient microglia exhibited strongly increased cAMP levels in comparison with wild-type microglia or cells expressing kinase-dead PI3Kγ after oxygen/glucose deprivation and recovery. Our data suggest PI3Kγ kinase activity-independent control of cAMP phosphodiesterase as a crucial mediator of microglial cAMP regulation, MMP-9 expression, and phagocytic activity following focal brain ischemia/recirculation. The suppressive effect of PI3Kγ on cAMP levels appears critical for the restriction of ischemia-induced immune cell functions and in turn tissue damage.


Brain Ischemia/enzymology , Brain Ischemia/pathology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Microglia/enzymology , Neurotoxins/toxicity , Animals , Brain Infarction/pathology , Brain Ischemia/complications , Cell Count , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Cyclic AMP/metabolism , Glucose/deficiency , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Models, Biological , Neutrophils/metabolism , Oxygen , Phagocytosis , Time Factors
6.
Neuromolecular Med ; 16(4): 704-13, 2014 Dec.
Article En | MEDLINE | ID: mdl-25033932

The breakdown of the blood-brain barrier (BBB) is a key event in the development of sepsis-induced brain damage. BBB opening allows blood-born immune cells to enter the CNS to provoke a neuroinflammatory response. Abnormal expression and activation of matrix metalloproteinases (MMP) was shown to contribute to BBB opening. Using different mouse genotypes in a model of LPS-induced systemic inflammation, our present report reveals phosphoinositide 3-kinase γ (PI3Kγ) as a mediator of BBB deterioration and concomitant generation of MMP by microglia. Unexpectedly, microglia expressing lipid kinase-deficient mutant PI3Kγ exhibited similar MMP regulation as wild-type cells. Our data suggest kinase-independent control of cAMP phosphodiesterase activity by PI3Kγ as a crucial mediator of microglial cell activation, MMP expression and subsequent BBB deterioration. The results identify the suppressive effect of PI3Kγ on cAMP as a critical mediator of immune cell functions.


Blood-Brain Barrier/metabolism , Class Ib Phosphatidylinositol 3-Kinase/physiology , Cyclic AMP/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Sepsis-Associated Encephalopathy/metabolism , Systemic Inflammatory Response Syndrome/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Biological Transport, Active , Capillary Permeability , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Coloring Agents/pharmacokinetics , Evans Blue/pharmacokinetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/enzymology , RNA, Messenger/biosynthesis , Second Messenger Systems
7.
Br J Pharmacol ; 171(13): 3328-37, 2014 Jul.
Article En | MEDLINE | ID: mdl-24654606

BACKGROUND AND PURPOSE: The µ-opioid receptor has been characterized as the main mediator of opioid signalling in neuronal cells. Opioid-induced pain suppression was originally proposed to be mediated by µ-opioid receptor-induced inhibitory effects on cAMP, which is known to mediate inflammatory hypernociception. Recent investigations revealed PI3Kγ and Akt (PKB) as additional elements of µ-opioid receptor signalling. Hence, we investigated the interaction between pronociceptive cAMP and antinociceptive PI3K/Akt signalling pathways. EXPERIMENTAL APPROACH: The human neuroblastoma cell line SK-N-LO and primary dorsal root ganglia (DRG) cells from mice were used to elucidate mediators of µ-opioid receptor signalling. In both cellular systems cAMP was manipulated by stimulation of adenylate cyclase and consequent effects on PI3K/Akt signalling were analysed. KEY RESULTS: Morphine stimulated Akt phosphorylation on Ser(473) and Thr(308) in a dose- and time-dependent manner indicating a functional µ-opioid receptor/Akt signalling pathway in µ-SK-N-LO cells. This effect of morphine was suppressed by the µ-opioid receptor inhibitor, naloxone, Pertussis toxin, an inhibitor of Gi heterotrimeric G-proteins, and the pan PI3K inhibitor wortmannin. cAMP-elevating agents also suppressed µ-opioid receptor-dependent stimulation of PI3Kγ lipid kinase and Akt activities in SK-N-LO cells and DRG. CONCLUSIONS AND IMPLICATIONS: The data unveil a hitherto unknown interaction of pronociceptive cAMP and antinociceptive PI3K/Akt signalling pathways in neuronal cells. PI3Kγ was identified as a mediator of the inhibitory action of cAMP on Akt in SK-N-LO cells and DRG. The data indicate that PI3Kγ has a critical role in cAMP-mediated inflammatory hypernociception and analgesic signalling via µ-opioid receptors and PI3K/Akt in neuronal cells.


Class Ib Phosphatidylinositol 3-Kinase/metabolism , Cyclic AMP/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Neuroblastoma/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects , Time Factors
...