Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37679549

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-37407815

RESUMO

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.


Assuntos
Sistema A de Transporte de Aminoácidos , Células Dendríticas , Glutamina , Neoplasias , Transdução de Sinais , Sistema A de Transporte de Aminoácidos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Glutamina/metabolismo , Neoplasias/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo
3.
Nat Cell Biol ; 24(11): 1642-1654, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36302969

RESUMO

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.


Assuntos
Interleucina-17 , Linfócitos T , Humanos , Feminino , Camundongos , Animais , Linfócitos T/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais , Homeostase , Interleucina-23
4.
Nature ; 595(7869): 724-729, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34234346

RESUMO

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.


Assuntos
Imunidade Humoral , Fosfatidiletanolaminas/metabolismo , Receptores CXCR5/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linfócitos B/imunologia , Sistemas CRISPR-Cas , Diferenciação Celular , Cistina Difosfato , Feminino , Regulação da Expressão Gênica , Humanos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfotransferases (Aceptor do Grupo Álcool) , RNA Nucleotidiltransferases , Transdução de Sinais
5.
Cell Metab ; 32(6): 996-1011.e7, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33207246

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Reguladores , Terpenos , Animais , Feminino , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prenilação de Proteína , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Terpenos/imunologia , Terpenos/metabolismo
6.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31668641

RESUMO

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.


Assuntos
Arginina/metabolismo , Leucina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Ciclo Celular , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia
7.
Nature ; 565(7737): 101-105, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30568299

RESUMO

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.


Assuntos
Transdiferenciação Celular , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Th17/citologia , Células Th17/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Feminino , Memória Imunológica/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Proteína Regulatória Associada a mTOR/deficiência , Proteína Regulatória Associada a mTOR/genética , Análise de Sequência de RNA , Transdução de Sinais , Análise de Célula Única , Células-Tronco/imunologia , Fator 1 de Transcrição de Linfócitos T/biossíntese , Fator 1 de Transcrição de Linfócitos T/metabolismo , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/metabolismo , Células Th17/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
8.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30413360

RESUMO

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.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Interleucina-2/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Animais , Autoimunidade/genética , Autoimunidade/imunologia , Linhagem da Célula/genética , Fator de Crescimento de Hepatócito/genética , Via de Sinalização Hippo , Interleucina-2/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Serina-Treonina Quinase 3 , Linfócitos T Reguladores/imunologia , Proteínas rac de Ligação ao GTP/metabolismo
9.
Nature ; 548(7669): 602-606, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28847007

RESUMO

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.


Assuntos
Homeostase , Tolerância Imunológica , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Apoptose , Sobrevivência Celular/genética , Citocinas/metabolismo , Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/biossíntese , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Receptores OX40/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/citologia , Células Th2/imunologia , beta Catenina/metabolismo , Linfopoietina do Estroma do Timo
10.
Immunity ; 46(3): 488-503, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28285833

RESUMO

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.


Assuntos
Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Alquil e Aril Transferases/imunologia , Animais , Metabolismo Energético , Espectrometria de Massas , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteômica , Receptores de Antígenos de Linfócitos T/imunologia
11.
Cancer Res ; 77(1): 123-133, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27815386

RESUMO

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.


Assuntos
Neoplasias Cerebelares/genética , Meduloblastoma/genética , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/patologia , PTEN Fosfo-Hidrolase/genética , Animais , Neoplasias Cerebelares/patologia , Hibridização Genômica Comparativa , Modelos Animais de Doenças , Imunofluorescência , Imuno-Histoquímica , Meduloblastoma/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/genética
12.
Elife ; 42015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26633882

RESUMO

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.


Assuntos
Epilepsia/genética , Megalencefalia/complicações , Megalencefalia/genética , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Animais , Encéfalo/patologia , Classe I de Fosfatidilinositol 3-Quinases , Modelos Animais de Doenças , Epilepsia/patologia , Humanos , Megalencefalia/patologia , Camundongos , Proteínas Mutantes/genética
13.
Nat Genet ; 46(5): 444-450, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24705251

RESUMO

Pediatric high-grade glioma (HGG) is a devastating disease with a less than 20% survival rate 2 years after diagnosis. We analyzed 127 pediatric HGGs, including diffuse intrinsic pontine gliomas (DIPGs) and non-brainstem HGGs (NBS-HGGs), by whole-genome, whole-exome and/or transcriptome sequencing. We identified recurrent somatic mutations in ACVR1 exclusively in DIPGs (32%), in addition to previously reported frequent somatic mutations in histone H3 genes, TP53 and ATRX, in both DIPGs and NBS-HGGs. Structural variants generating fusion genes were found in 47% of DIPGs and NBS-HGGs, with recurrent fusions involving the neurotrophin receptor genes NTRK1, NTRK2 and NTRK3 in 40% of NBS-HGGs in infants. Mutations targeting receptor tyrosine kinase-RAS-PI3K signaling, histone modification or chromatin remodeling, and cell cycle regulation were found in 68%, 73% and 59% of pediatric HGGs, respectively, including in DIPGs and NBS-HGGs. This comprehensive analysis provides insights into the unique and shared pathways driving pediatric HGG within and outside the brainstem.


Assuntos
Receptores de Ativinas Tipo I/genética , Neoplasias do Tronco Encefálico/genética , Glioma/genética , Transdução de Sinais/genética , Animais , Criança , Estudos de Coortes , Biologia Computacional , Perfilação da Expressão Gênica , Fusão Gênica/genética , Humanos , Immunoblotting , Imuno-Histoquímica , Análise em Microsséries , Receptor trkA/genética , Receptor trkB/genética , Receptor trkC/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Estatísticas não Paramétricas , Peixe-Zebra
14.
Hepatology ; 54(3): 1043-50, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21656538

RESUMO

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.


Assuntos
Receptor de Asialoglicoproteína/fisiologia , Citotoxicidade Imunológica , Hepatócitos/imunologia , Animais , Células Hep G2 , Humanos , Ativação Linfocitária , Camundongos , Neuraminidase/farmacologia , Receptor fas/fisiologia
15.
Cancer Cell ; 19(3): 305-16, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21397855

RESUMO

Mutations in the PTEN, TP53, and RB1 pathways are obligate events in the pathogenesis of human glioblastomas. We induced various combinations of deletions in these tumor suppressors in astrocytes and neural precursors in mature mice, resulting in astrocytomas ranging from grade III to grade IV (glioblastoma). There was selection for mutation of multiple genes within a pathway, shown by somatic amplifications of genes in the PI3K or Rb pathway in tumors in which Pten or Rb deletion was an initiating event. Despite multiple mutations within PI3K and Rb pathways, elevated Mapk activation was not consistent. Gene expression profiling revealed striking similarities to subclasses of human diffuse astrocytoma. Astrocytomas were found within and outside of proliferative niches in the adult brain.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , PTEN Fosfo-Hidrolase/genética , Proteína do Retinoblastoma/genética , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , Animais , Astrocitoma/metabolismo , Astrocitoma/patologia , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Feminino , Amplificação de Genes , Perfilação da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/metabolismo , Receptores Proteína Tirosina Quinases/genética , Proteína do Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
16.
Biochem Biophys Res Commun ; 379(3): 721-5, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19114034

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Neurônios/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Receptor de Fator de Crescimento Neural/genética , Fator de Transcrição Sp1/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , DNA/metabolismo , Regulação para Baixo , Ensaio de Desvio de Mobilidade Eletroforética , Laminina/metabolismo , Laminina/farmacologia , Mutação , Neurônios/efeitos dos fármacos , PTEN Fosfo-Hidrolase/genética , Ratos , Transcrição Gênica/efeitos dos fármacos
17.
J Neurochem ; 107(3): 799-813, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18786176

RESUMO

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.


Assuntos
Laminina/metabolismo , Regeneração Nervosa/fisiologia , Neuritos/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Células Cultivadas , Regulação para Baixo , Imuno-Histoquímica , Proteínas do Tecido Nervoso , Células PC12 , PTEN Fosfo-Hidrolase/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas rho de Ligação ao GTP/metabolismo
18.
Brain Res ; 1217: 10-24, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18511024

RESUMO

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.


Assuntos
Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Proteína Quinase C-delta/metabolismo , Receptores de Fator de Crescimento Neural/biossíntese , Transdução de Sinais/fisiologia , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Fator de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptores de Fatores de Crescimento , Transfecção , Regulação para Cima
19.
Hepatology ; 47(5): 1691-701, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18393317

RESUMO

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.


Assuntos
Morte Celular/fisiologia , Hepatócitos/fisiologia , Perforina/fisiologia , Animais , Southern Blotting , Carcinoma Hepatocelular , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Primers do DNA , Granzimas/genética , Hepatócitos/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Neoplasias Hepáticas , Marmota , Camundongos , Camundongos Endogâmicos ICR , Perforina/genética , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/genética , Receptor fas/fisiologia
20.
J Neurosci Methods ; 156(1-2): 55-63, 2006 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16554096

RESUMO

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.


Assuntos
Movimento Celular/fisiologia , Células PC12/fisiologia , Animais , Adesão Celular/genética , Adesão Celular/fisiologia , Movimento Celular/genética , Imuno-Histoquímica , Laminina/farmacologia , Microscopia Confocal , Mutação/fisiologia , Fatores de Crescimento Neural/farmacologia , Células PC12/ultraestrutura , Ratos , Receptor trkA/antagonistas & inibidores , Receptor trkA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...