Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37679549

ABSTRACT

Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including ß-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.

2.
Nature ; 620(7972): 200-208, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37407815

ABSTRACT

Cancer cells evade T cell-mediated killing through tumour-immune interactions whose mechanisms are not well understood1,2. Dendritic cells (DCs), especially type-1 conventional DCs (cDC1s), mediate T cell priming and therapeutic efficacy against tumours3. DC functions are orchestrated by pattern recognition receptors3-5, although other signals involved remain incompletely defined. Nutrients are emerging mediators of adaptive immunity6-8, but whether nutrients affect DC function or communication between innate and adaptive immune cells is largely unresolved. Here we establish glutamine as an intercellular metabolic checkpoint that dictates tumour-cDC1 crosstalk and licenses cDC1 function in activating cytotoxic T cells. Intratumoral glutamine supplementation inhibits tumour growth by augmenting cDC1-mediated CD8+ T cell immunity, and overcomes therapeutic resistance to checkpoint blockade and T cell-mediated immunotherapies. Mechanistically, tumour cells and cDC1s compete for glutamine uptake via the transporter SLC38A2 to tune anti-tumour immunity. Nutrient screening and integrative analyses show that glutamine is the dominant amino acid in promoting cDC1 function. Further, glutamine signalling via FLCN impinges on TFEB function. Loss of FLCN in DCs selectively impairs cDC1 function in vivo in a TFEB-dependent manner and phenocopies SLC38A2 deficiency by eliminating the anti-tumour therapeutic effect of glutamine supplementation. Our findings establish glutamine-mediated intercellular metabolic crosstalk between tumour cells and cDC1s that underpins tumour immune evasion, and reveal glutamine acquisition and signalling in cDC1s as limiting events for DC activation and putative targets for cancer treatment.


Subject(s)
Amino Acid Transport System A , Dendritic Cells , Glutamine , Neoplasms , Signal Transduction , Amino Acid Transport System A/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glutamine/metabolism , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism
3.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31668641

ABSTRACT

Regulatory T (Treg) cells are critical mediators of immune tolerance whose activity depends upon T cell receptor (TCR) and mTORC1 kinase signaling, but the mechanisms that dictate functional activation of these pathways are incompletely understood. Here, we showed that amino acids license Treg cell function by priming and sustaining TCR-induced mTORC1 activity. mTORC1 activation was induced by amino acids, especially arginine and leucine, accompanied by the dynamic lysosomal localization of the mTOR and Tsc complexes. Rag and Rheb GTPases were central regulators of amino acid-dependent mTORC1 activation in effector Treg (eTreg) cells. Mice bearing RagA-RagB- or Rheb1-Rheb2-deficient Treg cells developed a fatal autoimmune disease and had reduced eTreg cell accumulation and function. RagA-RagB regulated mitochondrial and lysosomal fitness, while Rheb1-Rheb2 enforced eTreg cell suppressive gene signature. Together, these findings reveal a crucial requirement of amino acid signaling for licensing and sustaining mTORC1 activation and functional programming of Treg cells.


Subject(s)
Arginine/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Cycle , Cell Differentiation/physiology , Cell Line , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Ras Homolog Enriched in Brain Protein/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/cytology
4.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413360

ABSTRACT

Interleukin-2 (IL-2) and downstream transcription factor STAT5 are important for maintaining regulatory T (Treg) cell homeostasis and function. Treg cells can respond to low IL-2 levels, but the mechanisms of STAT5 activation during partial IL-2 deficiency remain uncertain. We identified the serine-threonine kinase Mst1 as a signal-dependent amplifier of IL-2-STAT5 activity in Treg cells. High Mst1 and Mst2 (Mst1-Mst2) activity in Treg cells was crucial to prevent tumor resistance and autoimmunity. Mechanistically, Mst1-Mst2 sensed IL-2 signals to promote the STAT5 activation necessary for Treg cell homeostasis and lineage stability and to maintain the highly suppressive phosphorylated-STAT5+ Treg cell subpopulation. Unbiased quantitative proteomics revealed association of Mst1 with the cytoskeletal DOCK8-LRCHs module. Mst1 deficiency limited Treg cell migration and access to IL-2 and activity of the small GTPase Rac, which mediated downstream STAT5 activation. Collectively, IL-2-STAT5 signaling depends upon Mst1-Mst2 functions to maintain a stable Treg cell pool and immune tolerance.


Subject(s)
Hepatocyte Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Lineage/genetics , Hepatocyte Growth Factor/genetics , Hippo Signaling Pathway , Interleukin-2/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Serine-Threonine Kinase 3 , T-Lymphocytes, Regulatory/immunology , rac GTP-Binding Proteins/metabolism
5.
Nature ; 595(7869): 724-729, 2021 07.
Article in English | MEDLINE | ID: mdl-34234346

ABSTRACT

T follicular helper (TFH) cells are crucial for B cell-mediated humoral immunity1. Although transcription factors such as BCL6 drive the differentiation of TFH cells2,3, it is unclear whether and how post-transcriptional and metabolic programs enforce TFH cell programming. Here we show that the cytidine diphosphate (CDP)-ethanolamine pathway co-ordinates the expression and localization of CXCR5 with the responses of TFH cells and humoral immunity. Using in vivo CRISPR-Cas9 screening and functional validation in mice, we identify ETNK1, PCYT2, and SELENOI-enzymes in the CDP-ethanolamine pathway for de novo synthesis of phosphatidylethanolamine (PE)-as selective post-transcriptional regulators of TFH cell differentiation that act by promoting the surface expression and functional effects of CXCR5. TFH cells exhibit unique lipid metabolic programs and PE is distributed to the outer layer of the plasma membrane, where it colocalizes with CXCR5. De novo synthesis of PE through the CDP-ethanolamine pathway co-ordinates these events to prevent the internalization and degradation of CXCR5. Genetic deletion of Pcyt2, but not of Pcyt1a (which mediates the CDP-choline pathway), in activated T cells impairs the differentiation of TFH cells, and this is associated with reduced humoral immune responses. Surface levels of PE and CXCR5 expression on B cells also depend on Pcyt2. Our results reveal that phospholipid metabolism orchestrates post-transcriptional mechanisms for TFH cell differentiation and humoral immunity, highlighting the metabolic control of context-dependent immune signalling and effector programs.


Subject(s)
Immunity, Humoral , Phosphatidylethanolamines/metabolism , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Differentiation , Cytidine Diphosphate , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphotransferases (Alcohol Group Acceptor) , RNA Nucleotidyltransferases , Signal Transduction
6.
Immunity ; 46(3): 488-503, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28285833

ABSTRACT

The molecular circuits by which antigens activate quiescent T cells remain poorly understood. We combined temporal profiling of the whole proteome and phosphoproteome via multiplexed isobaric labeling proteomics technology, computational pipelines for integrating multi-omics datasets, and functional perturbation to systemically reconstruct regulatory networks underlying T cell activation. T cell receptors activated the T cell proteome and phosphoproteome with discrete kinetics, marked by early dynamics of phosphorylation and delayed ribosome biogenesis and mitochondrial activation. Systems biology analyses identified multiple functional modules, active kinases, transcription factors and connectivity between them, and mitochondrial pathways including mitoribosomes and complex IV. Genetic perturbation revealed physiological roles for mitochondrial enzyme COX10-mediated oxidative phosphorylation in T cell quiescence exit. Our multi-layer proteomics profiling, integrative network analysis, and functional studies define landscapes of the T cell proteome and phosphoproteome and reveal signaling and bioenergetics pathways that mediate lymphocyte exit from quiescence.


Subject(s)
Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Alkyl and Aryl Transferases/immunology , Animals , Energy Metabolism , Mass Spectrometry , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Phosphorylation , Proteomics , Receptors, Antigen, T-Cell/immunology
7.
Nature ; 565(7737): 101-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30568299

ABSTRACT

A defining feature of adaptive immunity is the development of long-lived memory T cells to curtail infection. Recent studies have identified a unique stem-like T-cell subset amongst exhausted CD8-positive T cells in chronic infection1-3, but it remains unclear whether CD4-positive T-cell subsets with similar features exist in chronic inflammatory conditions. Amongst helper T cells, TH17 cells have prominent roles in autoimmunity and tissue inflammation and are characterized by inherent plasticity4-7, although how such plasticity is regulated is poorly understood. Here we demonstrate that TH17 cells in a mouse model of autoimmune disease are functionally and metabolically heterogeneous; they contain a subset with stemness-associated features but lower anabolic metabolism, and a reciprocal subset with higher metabolic activity that supports transdifferentiation into TH1-like cells. These two TH17-cell subsets are defined by selective expression of the transcription factors TCF-1 and T-bet, and by discrete levels of CD27 expression. We also identify signalling via the kinase complex mTORC1 as a central regulator of TH17-cell fate decisions by coordinating metabolic and transcriptional programmes. TH17 cells with disrupted mTORC1 signalling or anabolic metabolism fail to induce autoimmune neuroinflammation or to develop into TH1-like cells, but instead upregulate TCF-1 expression and acquire stemness-associated features. Single-cell RNA sequencing and experimental validation reveal heterogeneity in fate-mapped TH17 cells, and a developmental arrest in the TH1 transdifferentiation trajectory upon loss of mTORC1 activity or metabolic perturbation. Our results establish that the dichotomy of stemness and effector function underlies the heterogeneous TH17 responses and autoimmune pathogenesis, and point to previously unappreciated metabolic control of plasticity in helper T cells.


Subject(s)
Cell Transdifferentiation , Stem Cells/cytology , Stem Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Disease Models, Animal , Female , Immunologic Memory/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Regulatory-Associated Protein of mTOR/deficiency , Regulatory-Associated Protein of mTOR/genetics , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Stem Cells/immunology , T Cell Transcription Factor 1/biosynthesis , T Cell Transcription Factor 1/metabolism , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/metabolism , Th17 Cells/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
8.
Nature ; 548(7669): 602-606, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28847007

ABSTRACT

Regulatory T cells (Treg cells) have a pivotal role in the establishment and maintenance of immunological self-tolerance and homeostasis. Transcriptional programming of regulatory mechanisms facilitates the functional activation of Treg cells in the prevention of diverse types of inflammatory responses. It remains unclear how Treg cells orchestrate their homeostasis and interplay with environmental signals. Here we show that liver kinase B1 (LKB1) programs the metabolic and functional fitness of Treg cells in the control of immune tolerance and homeostasis. Mice with a Treg-specific deletion of LKB1 developed a fatal inflammatory disease characterized by excessive TH2-type-dominant responses. LKB1 deficiency disrupted Treg cell survival and mitochondrial fitness and metabolism, but also induced aberrant expression of immune regulatory molecules including the negative co-receptor PD-1 and the TNF receptor superfamily proteins GITR and OX40. Unexpectedly, LKB1 function in Treg cells was independent of conventional AMPK signalling or the mTORC1-HIF-1α axis, but contributed to the activation of ß-catenin signalling for the control of PD-1 and TNF receptor proteins. Blockade of PD-1 activity reinvigorated the ability of LKB1-deficient Treg cells to suppress TH2 responses and the interplay with dendritic cells primed by thymic stromal lymphopoietin. Thus, Treg cells use LKB1 signalling to coordinate their metabolic and immunological homeostasis and to prevent apoptotic and functional exhaustion, thereby orchestrating the balance between immunity and tolerance.


Subject(s)
Homeostasis , Immune Tolerance , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , AMP-Activated Protein Kinases , Animals , Apoptosis , Cell Survival/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Mice , Mitochondria/metabolism , Mitochondria/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, OX40/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Th2 Cells/immunology , beta Catenin/metabolism , Thymic Stromal Lymphopoietin
9.
Hepatology ; 54(3): 1043-50, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21656538

ABSTRACT

UNLABELLED: It has been recently identified that hepatocytes can act as cytotoxic effectors and can kill contacted cells by way of CD95 ligand-CD95 and perforin-dependent pathways. However, it remained unknown whether hepatocyte-mediated cell killing is indiscriminant or is directed toward targets with particular cell surface characteristics, as well as whether hepatocytes have the capacity to directly eliminate contacted lymphocytes. In this study, we found that desialylation of surface glycoproteins significantly augments cell susceptibility to hepatocyte-mediated killing. Using asialofetuin as a competitive ligand, and by silencing gene transcription with specific small interfering RNA, we found that the asialoglycoprotein receptor (ASGPR) is involved in hepatocyte recognition of cells predestined for killing, including activated autologous T lymphocytes. CONCLUSION: Hepatocytes are constitutively equipped in the molecular machinery capable of eliminating cells brought into contact with their surface in a manner that is reliant, at least in part, upon the recognition of terminally desialylated glycoproteins by hepatocyte ASGPR. The study adds a new dimension to the physiological role of hepatic ASGPR and provides further evidence that hepatocytes can actively contribute to intrahepatic immune regulation and moderation of the local inflammatory response.


Subject(s)
Asialoglycoprotein Receptor/physiology , Cytotoxicity, Immunologic , Hepatocytes/immunology , Animals , Hep G2 Cells , Humans , Lymphocyte Activation , Mice , Neuraminidase/pharmacology , fas Receptor/physiology
10.
Nat Cell Biol ; 24(11): 1642-1654, 2022 11.
Article in English | MEDLINE | ID: mdl-36302969

ABSTRACT

Phosphatase and tensin homologue (PTEN) is frequently mutated in human cancer, but its roles in lymphopoiesis and tissue homeostasis remain poorly defined. Here we show that PTEN orchestrates a two-step developmental process linking antigen receptor and IL-23-Stat3 signalling to type-17 innate-like T cell generation. Loss of PTEN leads to pronounced accumulation of mature IL-17-producing innate-like T cells in the thymus. IL-23 is essential for their accumulation, and ablation of IL-23 or IL-17 signalling rectifies the reduced survival of female PTEN-haploinsufficient mice that model human patients with PTEN mutations. Single-cell transcriptome and network analyses revealed the dynamic regulation of PTEN, mTOR and metabolic activities that accompanied type-17 cell programming. Furthermore, deletion of mTORC1 or mTORC2 blocks PTEN loss-driven type-17 cell accumulation, and this is further shaped by the Foxo1 and Stat3 pathways. Collectively, our study establishes developmental and metabolic signalling networks underpinning type-17 cell fate decisions and their functional effects at coordinating PTEN-dependent tissue homeostasis.


Subject(s)
Interleukin-17 , T-Lymphocytes , Humans , Female , Mice , Animals , T-Lymphocytes/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Homeostasis , Interleukin-23
11.
Cell Metab ; 32(6): 996-1011.e7, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33207246

ABSTRACT

Effector regulatory T (eTreg) cells are essential for immune tolerance and depend upon T cell receptor (TCR) signals for generation. The immunometabolic signaling mechanisms that promote the differentiation and maintenance of eTreg cells remain unclear. Here, we show that isoprenoid-dependent posttranslational lipid modifications dictate eTreg cell accumulation and function by intersecting with TCR-induced intracellular signaling. We find that isoprenoids are essential for activated Treg cell suppressive activity, and Treg cell-specific deletion of the respective farnesylation- and geranylgeranylation-promoting enzymes Fntb or Pggt1b leads to the development of fatal autoimmunity, associated with reduced eTreg cell accumulation. Mechanistically, Fntb promotes eTreg cell maintenance by regulating mTORC1 activity and ICOS expression. In contrast, Pggt1b acts as a rheostat of TCR-dependent transcriptional programming and Rac-mediated signaling for establishment of eTreg cell differentiation and immune tolerance. Therefore, our results identify bidirectional metabolic signaling, specifically between immunoreceptor signaling and metabolism-mediated posttranslational lipid modifications, for the differentiation and maintenance of eTreg cells.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory , Terpenes , Animals , Female , Immune Tolerance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Prenylation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Terpenes/immunology , Terpenes/metabolism
12.
Biochem Biophys Res Commun ; 379(3): 721-5, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19114034

ABSTRACT

p75NTR is expressed throughout the nervous system and its dysregulation is associated with pathological conditions. We have recently demonstrated a signalling cascade initiated by laminin (LN), which upregulates PTEN and downregulates p75NTR. Here we investigate the mechanism by which PTEN modulates p75NTR. Studies using PTEN mutants show that its protein phosphatase activity directly modulates p75NTR protein expression. Nuclear relocalization of PTEN subsequent to LN stimulation suggests transcriptional control of p75NTR expression, which was confirmed following EMSA and ChIP analysis of Sp1 transcription factor binding activity. LN and PTEN independently decrease the DNA-binding ability of PTEN to the p75NTR promoter. Sp1 regulation of p75NTR occurs via dephosphorylation of Sp1, thus reducing p75NTR transcription and protein expression. This mechanism represents a novel regulatory pathway which controls the expression level of a receptor with broad implications not only for the development of the nervous system but also for progression of pathological conditions.


Subject(s)
Gene Expression Regulation , Neurons/enzymology , PTEN Phosphohydrolase/metabolism , Receptor, Nerve Growth Factor/genetics , Sp1 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/enzymology , DNA/metabolism , Down-Regulation , Electrophoretic Mobility Shift Assay , Laminin/metabolism , Laminin/pharmacology , Mutation , Neurons/drug effects , PTEN Phosphohydrolase/genetics , Rats , Transcription, Genetic/drug effects
13.
Hepatology ; 47(5): 1691-701, 2008 May.
Article in English | MEDLINE | ID: mdl-18393317

ABSTRACT

UNLABELLED: The liver displays unique immunological properties including the ability to remove aberrant cells and pathogens and to induce peripheral immunotolerance. We have previously demonstrated that hepatocytes can cause cell death by a CD95 ligand-mediated mechanism. Here, we provide evidence that hepatocytes can kill other cells via a perforin-dependent pathway. Using cultured woodchuck hepatocytes and human liver cells as well as freshly isolated woodchuck, mouse, and human hepatocytes, we show that hepatocyte-mediated death of CD95-deficient target cells requires microtubule polymerization, a feature of the granule exocytosis-mediated cytotoxicity. Neutralizing anti-perforin antibodies and short-hairpin RNA directed against perforin messenger RNA confirmed the involvement of perforin in hepatocyte-mediated cell killing. CONCLUSION: This study shows that hepatocytes express biologically competent perforin capable of killing susceptible cells and emphasizes the role of hepatocytes as cytotoxic effectors. This also is the first demonstration of perforin in a non-lymphoid cell type.


Subject(s)
Cell Death/physiology , Hepatocytes/physiology , Perforin/physiology , Animals , Blotting, Southern , Carcinoma, Hepatocellular , Cell Communication/drug effects , Cell Communication/physiology , Cell Death/drug effects , Cell Line, Tumor , DNA Primers , Granzymes/genetics , Hepatocytes/drug effects , Humans , Interferon-gamma/pharmacology , Liver Neoplasms , Marmota , Mice , Mice, Inbred ICR , Perforin/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/genetics , fas Receptor/physiology
14.
J Neurochem ; 107(3): 799-813, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18786176

ABSTRACT

Laminin (LN), an extracellular matrix component, is a key factor in promoting axonal regeneration, coordinately regulating growth in conjunction with trophic signals provided by the neurotrophins, including nerve growth factor (NGF). This study investigated potential interactions between the LN and NGF-mediated signaling pathways in PC12 cells and primary neurons. Neurite outgrowth stimulated by NGF was enhanced on a LN substrate. Western blot analysis of pertinent signal transduction components revealed both enhanced phosphorylation of early signaling intermediates upon co-stimulation, and a LN-induced down-regulation of p75NTR which could be prevented by the addition of integrin inhibitory arginine-glycine-aspartate (RGD) peptides. This p75NTR down-regulation was associated with a LN-mediated up-regulation of PTEN and resulted in a decrease in Rho activity. Studies using over-expression or siRNA-mediated knock-down of PTEN demonstrate a consistent inverse relationship with p75NTR, and the over-expression of p75NTR impaired neurite outgrowth on a LN substrate, as well as resulting in sustained activation of Rho which is inhibitory to neurite outgrowth. p75NTR is documented for its role in the transduction of inhibitory myelin-derived signals, and our results point to extracellular matrix regulation of p75NTR as a potential mechanism to ameliorate inhibitory signaling leading to optimized neurite outgrowth.


Subject(s)
Laminin/metabolism , Nerve Regeneration/physiology , Neurites/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Down-Regulation , Immunohistochemistry , Nerve Tissue Proteins , PC12 Cells , PTEN Phosphohydrolase/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Reverse Transcriptase Polymerase Chain Reaction , rho GTP-Binding Proteins/metabolism
15.
Brain Res ; 1217: 10-24, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18511024

ABSTRACT

Neurotrophins exert their biological effects via p75NTR and Trk receptors. Functional interplay between these two receptors has been widely explored with respect to p75NTR enhancing the activation and signalling of Trk, but few studies address the bidirectional aspects. We have previously demonstrated that the expression of p75NTR can be differentially modulated by different Trk receptor mutations. Here we investigate the mechanism of Nerve Growth Factor (NGF)-induced upregulation of p75NTR expression. We utilize pharmacological inhibition to investigate the role of various TrkA-associated signalling intermediates in this regulatory cascade. Notably, the inhibition of phospholipase C-gamma (PLC-gamma) using U73122, prevented the NGF-induced upregulation of p75NTR protein and mRNA. The inhibition of protein kinase C-delta (PKC-delta) activation by rottlerin, a selective PKC-delta inhibitor, and by small interfering RNA (siRNA) directed against PKC-delta also inhibited this NGF-induced upregulation. Finally, we also show that in cerebellar granule neurons, BDNF acting via TrkB increases p75NTR expression in a PKC-delta dependent manner. These results indicate the importance of Trk-dependent PLC-gamma and PKC-delta activation for downstream regulation of p75NTR protein expression in response to neurotrophin stimulation, a process that has implications to the survival and growth of the developing nervous system.


Subject(s)
Nerve Growth Factors/metabolism , Neurons/metabolism , Protein Kinase C-delta/metabolism , Receptors, Nerve Growth Factor/biosynthesis , Signal Transduction/physiology , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Nerve Growth Factor/metabolism , Nerve Tissue Proteins , Neurons/drug effects , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptors, Growth Factor , Transfection , Up-Regulation
16.
Cancer Res ; 77(1): 123-133, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27815386

ABSTRACT

Loss of the tumor suppressor gene PTEN exerts diverse outcomes on cancer in different developmental contexts. To gain insight into the effect of its loss on outcomes in the brain, we conditionally inactivated the murine Pten gene in neonatal neural stem/progenitor cells. Pten inactivation created an abnormal perivascular proliferative niche in the cerebellum that persisted in adult animals but did not progress to malignancy. Proliferating cells showed undifferentiated morphology and expressed the progenitor marker Nestin but not Math1, a marker of committed granule neuron progenitors. Codeletion of Pten and Trp53 resulted in fully penetrant medulloblastoma originating from the perivascular niche, which exhibited abnormal blood vessel networks and advanced neuronal differentiation of tumor cells. EdU pulse-chase experiments demonstrated a perivascular cancer stem cell population in Pten/Trp53 double mutant medulloblastomas. Genetic analyses revealed recurrent somatic inactivations of the tumor suppressor gene Ptch1 and a recapitulation of the sonic hedgehog subgroup of human medulloblastomas. Overall, our results showed that PTEN acts to prevent the proliferation of a progenitor niche in postnatal cerebellum predisposed to oncogenic induction of medulloblastoma. Cancer Res; 77(1); 123-33. ©2016 AACR.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , PTEN Phosphohydrolase/genetics , Animals , Cerebellar Neoplasms/pathology , Comparative Genomic Hybridization , Disease Models, Animal , Fluorescent Antibody Technique , Immunohistochemistry , Medulloblastoma/pathology , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Signal Transduction/physiology , Stem Cell Niche/physiology , Tumor Suppressor Protein p53/genetics
17.
J Neurosci Methods ; 156(1-2): 55-63, 2006 Sep 30.
Article in English | MEDLINE | ID: mdl-16554096

ABSTRACT

Cellular migration is central to a wide range of biological and pathological processes in vivo. In vitro cell migration assays can be used to obtain invaluable information relating to the mechanism of cell movement, but current available methods can be limiting. Here we describe a novel motility assay that allows the simultaneous investigation of both quantitative and qualitative aspects of a population of motile cells as they move across a variety of substrates. By plating cells in a confluent monolayer on a coverslip, the monolayer can then be inverted to migrate over a larger substrate-coated coverslip, which can subsequently be reliably quantified, and subjected to immunocytochemistry and confocal imaging. This assay can be used to assess multiple aspects of motility, including distance, quantity, morphology, polarization and component colocalization. To demonstrate the utility of this assay, it was applied to the study of a stimulator of PC12 cell migration, nerve growth factor (NGF), and how this migration is influenced by the extracellular substrate, laminin. Furthermore, since mutations to the NGF receptor, TrkA, have been noted to alter the behaviour of PC12 cells in response to NGF, a PC12 subline that expresses a mutated TrkA receptor was utilized to illustrate that a Y785F mutation in the cytoplasmic tail of TrkA results in increased migration in response to the stimulus compared to the control PC12s.


Subject(s)
Cell Movement/physiology , PC12 Cells/physiology , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/genetics , Immunohistochemistry , Laminin/pharmacology , Microscopy, Confocal , Mutation/physiology , Nerve Growth Factors/pharmacology , PC12 Cells/ultrastructure , Rats , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics
18.
Brain Res ; 1068(1): 34-48, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16376863

ABSTRACT

Heat shock protein 27 (Hsp27), a molecular chaperone ubiquitously expressed in many cell types, has been shown to play a role in protecting neurons from cellular stresses. Unlike adult DRG neurons in vitro, neonatal DRG neurons require NGF for survival; withdrawal of NGF results in apoptosis of a majority of neonatal neurons. We hypothesized that Hsp27 contributes to the neurotrophin-independent survival of adult DRG neurons. Constitutive Hsp27 expression is higher in adult DRG neurons compared to neonates, although both upregulate Hsp27 expression after heat shock (HS). We found that increasing endogenous Hsp27 by HS in neonatal neurons was able to inhibit NGF withdrawal-induced apoptosis. Heat shock of adult and neonatal neurons also resulted in Akt activation, which could be a mechanism for the increased survival. Hsp27 siRNA treatment of adult neurons effected a decreased expression of Hsp27, which correlated with increased apoptosis in these neurons. Downregulation of Hsp27 via siRNA also blocked the HS-induced rescue of neonatal neurons after NGF withdrawal. These results indicate that physiologically induced upregulation of Hsp27 is sufficient to provide some degree of neuronal protection. Further, this induction appears to be regulated by the transcriptional activation of HSF1 as shown by HSF1 nuclear translocation and by EMSA analyses of HSF1 binding to nuclear protein.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/physiology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Neurons/physiology , Animals , Animals, Newborn/physiology , Blotting, Western , Cell Survival/physiology , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , HSP27 Heat-Shock Proteins , Heat Shock Transcription Factors , Hot Temperature , Immunohistochemistry , In Situ Nick-End Labeling , Neuroprotective Agents , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation/physiology
19.
Neurosci Lett ; 383(3): 305-10, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-15955426

ABSTRACT

The cellular response to nerve growth factor (NGF) is mediated by two structurally unrelated receptors, TrkA and p75 neurotrophin receptor (p75NTR), which have been shown to interact resulting in reciprocal modulation of function. In this study, we have examined the modulation of p75NTR protein expression by specific TrkA autophosphorylation sites in the presence or absence of NGF. We have used cell lines derived from PC12 cells that express either no endogenous TrkA (PC12nnr5) or TrkA receptors mutated via site-directed mutagenesis to abrogate individual tyrosine autophosphorylation sites on the cytoplasmic tail (Y490F, Y785F and Y490/785F). Results indicate that in the absence of TrkA in PC12nnr5 cells there is reduced constitutive p75NTR expression, which can be restored to different degrees by transfection of the Y490F TrkA or the Y490/785F TrkA, but not by transfection of the Y785F TrkA. In addition, the expression of p75NTR was upregulated in the presence of NGF in the parental and Y490F cell lines only. Together these results indicate a role for the individual tyrosine autophosphorylation sites of TrkA in regulating p75NTR expression.


Subject(s)
Gene Expression Regulation/physiology , Receptor, trkA/physiology , Receptors, Nerve Growth Factor/metabolism , Analysis of Variance , Animals , Blotting, Western/methods , Gene Expression Regulation/drug effects , Immunohistochemistry/methods , Mutagenesis/physiology , Mutation/physiology , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphorylation , RNA, Messenger/biosynthesis , Rats , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods
20.
Elife ; 42015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633882

ABSTRACT

Mutations in the catalytic subunit of phosphoinositide 3-kinase (PIK3CA) and other PI3K-AKT pathway components have been associated with cancer and a wide spectrum of brain and body overgrowth. In the brain, the phenotypic spectrum of PIK3CA-related segmental overgrowth includes bilateral dysplastic megalencephaly, hemimegalencephaly and focal cortical dysplasia, the most common cause of intractable pediatric epilepsy. We generated mouse models expressing the most common activating Pik3ca mutations (H1047R and E545K) in developing neural progenitors. These accurately recapitulate all the key human pathological features including brain enlargement, cortical malformation, hydrocephalus and epilepsy, with phenotypic severity dependent on the mutant allele and its time of activation. Underlying mechanisms include increased proliferation, cell size and altered white matter. Notably, we demonstrate that acute 1 hr-suppression of PI3K signaling despite the ongoing presence of dysplasia has dramatic anti-epileptic benefit. Thus PI3K inhibitors offer a promising new avenue for effective anti-epileptic therapy for intractable pediatric epilepsy patients.


Subject(s)
Epilepsy/genetics , Megalencephaly/complications , Megalencephaly/genetics , Mutation, Missense , Phosphatidylinositol 3-Kinases/genetics , Animals , Brain/pathology , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Epilepsy/pathology , Humans , Megalencephaly/pathology , Mice , Mutant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL