Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res Commun ; 3(12): 2430-2446, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37971169

RESUMO

Understanding the intricate dynamics between adoptively transferred immune cells and the brain tumor immune microenvironment (TIME) is crucial for the development of effective T cell-based immunotherapies. In this study, we investigated the influence of the TIME and chimeric antigen receptor (CAR) design on the anti-glioma activity of B7-H3-specific CAR T-cells. Using an immunocompetent glioma model, we evaluated a panel of seven fully murine B7-H3 CARs with variations in transmembrane, costimulatory, and activation domains. We then investigated changes in the TIME following CAR T-cell therapy using high-dimensional flow cytometry and single-cell RNA sequencing. Our results show that five out of six B7-H3 CARs with single costimulatory domains demonstrated robust functionality in vitro. However, these CARs had significantly varied levels of antitumor activity in vivo. To enhance therapeutic effectiveness and persistence, we incorporated 41BB and CD28 costimulation through transgenic expression of 41BBL on CD28-based CAR T-cells. This CAR design was associated with significantly improved anti-glioma efficacy in vitro but did not result in similar improvements in vivo. Analysis of the TIME revealed that CAR T-cell therapy influenced the composition of the TIME, with the recruitment and activation of distinct macrophage and endogenous T-cell subsets crucial for successful antitumor responses. Indeed, complete brain macrophage depletion using a CSF1R inhibitor abrogated CAR T-cell antitumor activity. In sum, our study highlights the critical role of CAR design and its modulation of the TIME in mediating the efficacy of adoptive immunotherapy for high-grade glioma. SIGNIFICANCE: CAR T-cell immunotherapies hold great potential for treating brain cancers; however, they are hindered by a challenging immune environment that dampens their effectiveness. In this study, we show that the CAR design influences the makeup of the immune environment in brain tumors, underscoring the need to target specific immune components to improve CAR T-cell performance, and highlighting the significance of using models with functional immune systems to optimize this therapy.


Assuntos
Neoplasias Encefálicas , Glioma , Receptores de Antígenos Quiméricos , Camundongos , Animais , Receptores de Antígenos Quiméricos/genética , Linfócitos T , Macrófagos Associados a Tumor/metabolismo , Antígenos CD28/genética , Glioma/terapia , Neoplasias Encefálicas/terapia , Encéfalo/metabolismo , Microambiente Tumoral
2.
Cell Mol Life Sci ; 79(10): 531, 2022 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-36169732

RESUMO

In response to infection or sterile insults, inflammatory programmed cell death is an essential component of the innate immune response to remove infected or damaged cells. PANoptosis is a unique innate immune inflammatory cell death pathway regulated by multifaceted macromolecular complexes called PANoptosomes, which integrate components from other cell death pathways. Growing evidence shows that PANoptosis can be triggered in many physiological conditions, including viral and bacterial infections, cytokine storms, and cancers. However, PANoptosomes at the single cell level have not yet been fully characterized. Initial investigations have suggested that key pyroptotic, apoptotic, and necroptotic molecules including the inflammasome adaptor protein ASC, apoptotic caspase-8 (CASP8), and necroptotic RIPK3 are conserved components of PANoptosomes. Here, we optimized an immunofluorescence procedure to probe the highly dynamic multiprotein PANoptosome complexes across various innate immune cell death-inducing conditions. We first identified and validated antibodies to stain endogenous mouse ASC, CASP8, and RIPK3, without residual staining in the respective knockout cells. We then assessed the formation of PANoptosomes across innate immune cell death-inducing conditions by monitoring the colocalization of ASC with CASP8 and/or RIPK3. Finally, we established an expansion microscopy procedure using these validated antibodies to image the organization of ASC, CASP8, and RIPK3 within the PANoptosome. This optimized protocol, which can be easily adapted to study other multiprotein complexes and other cell death triggers, provides confirmation of PANoptosome assembly in individual cells and forms the foundation for a deeper molecular understanding of the PANoptosome complex and PANoptosis to facilitate therapeutic targeting.


Assuntos
Inflamassomos , Análise de Célula Única , Animais , Apoptose , Caspase 8/metabolismo , Inflamassomos/metabolismo , Camundongos , Microscopia , Piroptose
3.
Mol Cell ; 82(13): 2401-2414.e9, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35597236

RESUMO

Activated CD8+ T lymphocytes differentiate into heterogeneous subsets. Using super-resolution imaging, we found that prior to the first division, dynein-dependent vesicular transport polarized active TORC1 toward the microtubule-organizing center (MTOC) at the proximal pole. This active TORC1 was physically associated with active eIF4F, required for the translation of c-myc mRNA. As a consequence, c-myc-translating polysomes polarized toward the cellular pole proximal to the immune synapse, resulting in localized c-myc translation. Upon division, the TORC1-eIF4A complex preferentially sorted to the proximal daughter cell, facilitating asymmetric c-Myc synthesis. Transient disruption of eIF4A activity at first division skewed long-term cell fate trajectories to memory-like function. Using a genetic barcoding approach, we found that first-division sister cells often displayed differences in transcriptional profiles that largely correlated with c-Myc and TORC1 target genes. Our findings provide mechanistic insights as to how distinct T cell fate trajectories can be established during the first division.


Assuntos
Linfócitos T CD8-Positivos , Fator de Iniciação 4F em Eucariotos , Diferenciação Celular , Ativação Linfocitária , Alvo Mecanístico do Complexo 1 de Rapamicina/genética
4.
Stat Methods Med Res ; 31(8): 1484-1499, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35450486

RESUMO

Spatial data from high-resolution images abound in many scientific disciplines. For example, single-molecule localization microscopy, such as stochastic optical reconstruction microscopy, provides super-resolution images to help scientists investigate co-localization of proteins and hence their interactions inside cells, which are key events in living cells. However, there are few accurate methods for analyzing co-localization in super-resolution images. The current methods and software are prone to produce false-positive errors and are restricted to only 2-dimensional images. In this paper, we develop a novel statistical method to effectively address the problems of unbiased and robust quantification and comparison of protein co-localization for multiple 2- and 3-dimensional image datasets. This method significantly improves the analysis of protein co-localization using super-resolution image data, as shown by its excellent performance in simulation studies and an analysis of co-localization of protein light chain 3 and lysosomal-associated membrane protein 1 in cell autophagy. Moreover, this method is directly applicable to co-localization analyses in other disciplines, such as diagnostic imaging, epidemiology, environmental science, and ecology.


Assuntos
Software , Simulação por Computador
5.
Cell Rep ; 38(10): 110482, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263574

RESUMO

Infection and vaccination repeatedly expose individuals to antigens that are conserved between influenza virus subtypes. Nevertheless, antibodies recognizing variable influenza epitopes greatly outnumber antibodies reactive against conserved epitopes. Elucidating factors contributing to the paucity of broadly reactive influenza antibodies remains a major obstacle for developing a universal influenza vaccine. Here, we report that inducing broadly reactive influenza antibodies increases autoreactive antibodies in humans and mice and exacerbates disease in four distinct models of autoimmune disease. Importantly, transferring broadly reactive influenza antibodies augments disease in the presence of inflammation or autoimmune susceptibility. Further, broadly reactive influenza antibodies spontaneously arise in mice with defects in B cell tolerance. Together, these data suggest that self-tolerance mechanisms limit the prevalence of broadly reactive influenza antibodies, which can exacerbate disease in the context of additional risk factors.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Autoimunidade , Epitopos , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Humanos , Camundongos
6.
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
7.
Cancer Res ; 81(19): 5047-5059, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34301764

RESUMO

Immune cells regulate tumor growth by mirroring their function as tissue repair organizers in normal tissues. To understand the different facets of immune-tumor collaboration through genetics, spatial transcriptomics, and immunologic manipulation with noninvasive, longitudinal imaging, we generated a penetrant double oncogene-driven autochthonous model of neuroblastoma. Spatial transcriptomic analysis showed that CD4+ and myeloid populations colocalized within the tumor parenchyma, while CD8+ T cells and B cells were peripherally dispersed. Depletion of CD4+ T cells or CCR2+ macrophages, but not B cells, CD8+ T cells, or natural killer (NK) cells, prevented tumor formation. Tumor CD4+ T cells displayed unconventional phenotypes and were clonotypically diverse and antigen independent. Within the myeloid fraction, tumor growth required myeloid cells expressing arginase-1. Overall, these results demonstrate how arginine-metabolizing myeloid cells conspire with pathogenic CD4+ T cells to create permissive conditions for tumor formation, suggesting that these protumorigenic pathways could be disabled by targeting myeloid arginine metabolism. SIGNIFICANCE: A new model of human neuroblastoma provides ways to track tumor formation and expansion in living animals, allowing identification of CD4+ T-cell and macrophage functions required for oncogenesis.


Assuntos
Arginase/genética , Linfócitos T CD4-Positivos/metabolismo , Suscetibilidade a Doenças , Células Mieloides/metabolismo , Neuroblastoma/etiologia , Neuroblastoma/metabolismo , Animais , Arginase/metabolismo , Biomarcadores , Células da Medula Óssea/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Biologia Computacional/métodos , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Neuroblastoma/patologia , Oncogenes , Análise de Célula Única , Transcriptoma
10.
Nature ; 587(7834): 466-471, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33116313

RESUMO

Severe respiratory infections can result in acute respiratory distress syndrome (ARDS)1. There are no effective pharmacological therapies that have been shown to improve outcomes for patients with ARDS. Although the host inflammatory response limits spread of and eventually clears the pathogen, immunopathology is a major contributor to tissue damage and ARDS1,2. Here we demonstrate that respiratory viral infection induces distinct fibroblast activation states, which we term extracellular matrix (ECM)-synthesizing, damage-responsive and interferon-responsive states. We provide evidence that excess activity of damage-responsive lung fibroblasts drives lethal immunopathology during severe influenza virus infection. By producing ECM-remodelling enzymes-in particular the ECM protease ADAMTS4-and inflammatory cytokines, damage-responsive fibroblasts modify the lung microenvironment to promote robust immune cell infiltration at the expense of lung function. In three cohorts of human participants, the levels of ADAMTS4 in the lower respiratory tract were associated with the severity of infection with seasonal or avian influenza virus. A therapeutic agent that targets the ECM protease activity of damage-responsive lung fibroblasts could provide a promising approach to preserving lung function and improving clinical outcomes following severe respiratory infections.


Assuntos
Proteína ADAMTS4/metabolismo , Fibroblastos/enzimologia , Fibroblastos/patologia , Vírus da Influenza A/patogenicidade , Pulmão/patologia , Pulmão/fisiopatologia , Proteína ADAMTS4/antagonistas & inibidores , Animais , Aves/virologia , Matriz Extracelular/enzimologia , Perfilação da Expressão Gênica , Humanos , Influenza Aviária/virologia , Influenza Humana/patologia , Influenza Humana/terapia , Influenza Humana/virologia , Interferons/imunologia , Interferons/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Pulmão/enzimologia , Pulmão/virologia , Camundongos , Síndrome do Desconforto Respiratório/enzimologia , Síndrome do Desconforto Respiratório/fisiopatologia , Síndrome do Desconforto Respiratório/terapia , Síndrome do Desconforto Respiratório/virologia , Estações do Ano , Análise de Célula Única , Células Estromais/metabolismo
11.
Immunity ; 52(6): 994-1006.e8, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32428502

RESUMO

Cell death pathways regulate various homeostatic processes. Autoimmune lymphoproliferative syndrome (ALPS) in humans and lymphoproliferative (LPR) disease in mice result from abrogated CD95-induced apoptosis. Because caspase-8 mediates CD95 signaling, we applied genetic approaches to dissect the roles of caspase-8 in cell death and inflammation. Here, we describe oligomerization-deficient Caspase-8F122GL123G/F122GL123G and non-cleavable Caspase-8D387A/D387A mutant mice with defective caspase-8-mediated apoptosis. Although neither mouse developed LPR disease, removal of the necroptosis effector Mlkl from Caspase-8D387A/D387A mice revealed an inflammatory role of caspase-8. Ablation of one allele of Fasl, Fadd, or Ripk1 prevented the pathology of Casp8D387A/D387AMlkl-/- animals. Removing both Fadd alleles from these mice resulted in early lethality prior to post-natal day 15 (P15), which was prevented by co-ablation of either Ripk1 or Caspase-1. Our results suggest an in vivo role of the inflammatory RIPK1-caspase-8-FADD (FADDosome) complex and reveal a FADD-independent inflammatory role of caspase-8 that involves activation of an inflammasome.


Assuntos
Caspase 8/genética , Suscetibilidade a Doenças , Proteína de Domínio de Morte Associada a Fas/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Necroptose/genética , Animais , Apoptose/genética , Biomarcadores , Caspase 8/química , Caspase 8/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Imunofluorescência , Regulação da Expressão Gênica , Inflamassomos/metabolismo , Inflamação/mortalidade , Inflamação/patologia , Lipopolissacarídeos/efeitos adversos , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Mortalidade , Fenótipo , Multimerização Proteica
12.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32150572

RESUMO

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Assuntos
Infecções por Burkholderia/imunologia , Burkholderia/imunologia , Proteínas de Ligação ao GTP/imunologia , Células Gigantes/imunologia , Macrófagos/imunologia , Doenças Nasais/imunologia , Prenilação de Proteína/imunologia , Animais , Infecções por Burkholderia/genética , Infecções por Burkholderia/patologia , Fusão Celular , Proteínas de Ligação ao GTP/genética , Células Gigantes/microbiologia , Células Gigantes/patologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Doenças Nasais/genética , Doenças Nasais/microbiologia , Doenças Nasais/patologia
13.
Nature ; 573(7775): 590-594, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31511697

RESUMO

The cellular stress response has a vital role in regulating homeostasis by modulating cell survival and death. Stress granules are cytoplasmic compartments that enable cells to survive various stressors. Defects in the assembly and disassembly of stress granules are linked to neurodegenerative diseases, aberrant antiviral responses and cancer1-5. Inflammasomes are multi-protein heteromeric complexes that sense molecular patterns that are associated with damage or intracellular pathogens, and assemble into cytosolic compartments known as ASC specks to facilitate the activation of caspase-1. Activation of inflammasomes induces the secretion of interleukin (IL)-1ß and IL-18 and drives cell fate towards pyroptosis-a form of programmed inflammatory cell death that has major roles in health and disease6-12. Although both stress granules and inflammasomes can be triggered by the sensing of cellular stress, they drive contrasting cell-fate decisions. The crosstalk between stress granules and inflammasomes and how this informs cell fate has not been well-studied. Here we show that the induction of stress granules specifically inhibits NLRP3 inflammasome activation, ASC speck formation and pyroptosis. The stress granule protein DDX3X interacts with NLRP3 to drive inflammasome activation. Assembly of stress granules leads to the sequestration of DDX3X, and thereby the inhibition of NLRP3 inflammasome activation. Stress granules and the NLRP3 inflammasome compete for DDX3X molecules to coordinate the activation of innate responses and subsequent cell-fate decisions under stress conditions. Induction of stress granules or loss of DDX3X in the myeloid compartment leads to a decrease in the production of inflammasome-dependent cytokines in vivo. Our findings suggest that macrophages use the availability of DDX3X to interpret stress signals and choose between pro-survival stress granules and pyroptotic ASC specks. Together, our data demonstrate the role of DDX3X in driving NLRP3 inflammasome and stress granule assembly, and suggest a rheostat-like mechanistic paradigm for regulating live-or-die cell-fate decisions under stress conditions.


Assuntos
Morte Celular/genética , RNA Helicases DEAD-box/metabolismo , Inflamassomos/genética , Macrófagos/citologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Fisiológico/genética , Animais , Linhagem Celular , Sobrevivência Celular/genética , RNA Helicases DEAD-box/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Células HEK293 , Humanos , Inflamassomos/imunologia , Macrófagos/imunologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética
14.
Cell ; 178(3): 536-551.e14, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31257024

RESUMO

The expression of some proteins in the autophagy pathway declines with age, which may impact neurodegeneration in diseases, including Alzheimer's Disease. We have identified a novel non-canonical function of several autophagy proteins in the conjugation of LC3 to Rab5+, clathrin+ endosomes containing ß-amyloid in a process of LC3-associated endocytosis (LANDO). We found that LANDO in microglia is a critical regulator of immune-mediated aggregate removal and microglial activation in a murine model of AD. Mice lacking LANDO but not canonical autophagy in the myeloid compartment or specifically in microglia have a robust increase in pro-inflammatory cytokine production in the hippocampus and increased levels of neurotoxic ß-amyloid. This inflammation and ß-amyloid deposition were associated with reactive microgliosis and tau hyperphosphorylation. LANDO-deficient AD mice displayed accelerated neurodegeneration, impaired neuronal signaling, and memory deficits. Our data support a protective role for LANDO in microglia in neurodegenerative pathologies resulting from ß-amyloid deposition.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Endocitose , Proteínas Associadas aos Microtúbulos/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/deficiência , Proteína 5 Relacionada à Autofagia/genética , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Antígenos CD36/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/citologia , Microglia/metabolismo , Células RAW 264.7 , Receptores Imunológicos/metabolismo , Receptor 4 Toll-Like/metabolismo
15.
Immunity ; 49(3): 515-530.e5, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231985

RESUMO

Fungi represent a significant proportion of the gut microbiota. Aberrant immune responses to fungi are frequently observed in inflammatory bowel diseases (IBD) and colorectal cancer (CRC), and mutations in the fungal-sensing pathways are associated with the pathogenesis of IBD. Fungal recognition receptors trigger downstream signaling via the common adaptor protein CARD9 and the kinase SYK. Here we found that commensal gut fungi promoted inflammasome activation during AOM-DSS-induced colitis. Myeloid cell-specific deletion of Card9 or Syk reduced inflammasome activation and interleukin (IL)-18 maturation and increased susceptibility to colitis and CRC. IL-18 promoted epithelial barrier restitution and interferon-γ production by intestinal CD8+ T cells. Supplementation of IL-18 or transfer of wild-type myeloid cells reduced tumor burden in AOM-DSS-treated Card9-/- and Sykfl/flLysMCre/+ mice, whereas treatment with anti-fungal agents exacerbated colitis and CRC. CARD9 deletion changes the gut microbial landscape, suggesting that SYK-CARD9 signaling maintains a microbial ecology that promotes inflammasome activation and thereby restrains colitis and colon tumorigenesis.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Colite/imunologia , Neoplasias do Colo/imunologia , Fungos/imunologia , Microbioma Gastrointestinal/imunologia , Inflamassomos/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/fisiologia , Células Mieloides/fisiologia , Quinase Syk/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Células Cultivadas , Colite/induzido quimicamente , Modelos Animais de Doenças , Humanos , Interleucina-18/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Dodecilsulfato de Sódio , Quinase Syk/genética
16.
J Neurosci ; 38(10): 2615-2630, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29437892

RESUMO

Members of the SCY1-like (SCYL) family of protein kinases are evolutionarily conserved and ubiquitously expressed proteins characterized by an N-terminal pseudokinase domain, centrally located Huntingtin, elongation factor 3, protein phosphatase 2A, yeast kinase TOR1 repeats, and an overall disorganized C-terminal segment. In mammals, three family members encoded by genes Scyl1, Scyl2, and Scyl3 have been described. Studies have pointed to a role for SCYL1 and SCYL2 in regulating neuronal function and viability in mice and humans, but little is known about the biological function of SCYL3. Here, we show that the biochemical and cell biological properties of SCYL3 are similar to those of SCYL1 and both proteins work in conjunction to maintain motor neuron viability. Specifically, although lack of Scyl3 in mice has no apparent effect on embryogenesis and postnatal life, it accelerates the onset of the motor neuron disorder caused by Scyl1 deficiency. Growth abnormalities, motor dysfunction, hindlimb paralysis, muscle wasting, neurogenic atrophy, motor neuron degeneration, and loss of large-caliber axons in peripheral nerves occurred at an earlier age in Scyl1/Scyl3 double-deficient mice than in Scyl1-deficient mice. Disease onset also correlated with the mislocalization of TDP-43 in spinal motor neurons, suggesting that SCYL1 and SCYL3 regulate TDP-43 proteostasis. Together, our results demonstrate an overlapping role for SCYL1 and SCYL3 in vivo and highlight the importance the SCYL family of proteins in regulating neuronal function and survival. Only male mice were used in this study.SIGNIFICANCE STATEMENT SCYL1 and SCYL2, members of the SCY1-like family of pseudokinases, have well established roles in neuronal function. Herein, we uncover the role of SCYL3 in maintaining motor neuron viability. Although targeted disruption of Scyl3 in mice had little or no effect on embryonic development and postnatal life, it accelerated disease onset associated with the loss of Scyl1, a novel motor neuron disease gene in humans. Scyl1 and Scyl3 double-deficient mice had neuronal defects characteristic of amyotrophic lateral sclerosis, including TDP-43 pathology, at an earlier age than did Scyl1-deficient mice. Thus, we show that SCYL1 and SCYL3 play overlapping roles in maintaining motor neuronal viability in vivo and confirm that SCYL family members are critical regulators of neuronal function and survival.


Assuntos
Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Proteínas de Membrana/fisiologia , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Quinases/genética , Proteínas Adaptadoras de Transporte Vesicular , Animais , Atrofia , Axônios/patologia , Caspases/metabolismo , Proteínas de Ligação a DNA/genética , Fibroblastos/patologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/patologia , Músculo Esquelético/patologia , Degeneração Neural/genética , Proteínas do Tecido Nervoso/genética , Paralisia/genética , Paralisia/patologia
17.
Gastroenterology ; 154(4): 948-964.e8, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29203393

RESUMO

BACKGROUND & AIMS: Inflammatory bowel diseases (IBD) increase risk for colorectal cancer. Mutations in the Mediterranean fever gene (MEFV or pyrin) are associated with hereditary autoinflammatory disease and severe IBD. Expression of MEFV, a sensor protein that the initiates assembly of the inflammasome complex, is increased in colon biopsies from patients with IBD. We investigated the role of pyrin in intestinal homeostasis in mice. METHODS: Mefv-/- mice and C57/BL6 mice (controls) were given azoxymethane followed by multiple rounds of dextran sodium sulfate (DSS) to induce colitis and tumorigenesis. In some experiments, Mefv-/- mice were given injections of recombinant interleukin 18 (rIL18) or saline (control) during DSS administration. Colon tissues were collected at different time points during colitis development and analyzed by histology, immunohistochemistry, immunoblots, or ELISAs (to measure cytokines). Spleen and mesenteric lymph node were collected, processed, and analyzed by flow cytometry. Colon epithelial permeability was measured in mice with colitis by gavage of fluorescent dextran and quantification of serum levels. RESULTS: MEFV was expressed in colons of control mice and expression increased during chronic and acute inflammation; high levels were detected in colon tumor and adjacent non-tumor tissues. Mefv-/- mice developed more severe colitis than control mice, with a greater extent of epithelial hyperplasia and a larger tumor burden. Levels of inflammatory cytokines (IL6) and chemokines were significantly higher in colons of Mefv-/- mice than control mice following colitis induction, whereas the level IL18, which depends on the inflammasome for maturation and release, was significantly lower in colons of Mefv-/- mice. Mefv-/- mice had increased epithelial permeability following administration of DSS than control mice, and loss of the tight junction proteins occludin and claudin-2 from intercellular junctions. STAT3 was activated (phosphorylated) in inflamed colon tissues from Mefv-/-, which also had increased expression of stem cell markers (OLFM4, BMI1, and MSI1) compared with colons from control mice. Administration of rIL18 to Mefv-/- mice reduced epithelial permeability, intestinal inflammation, the severity of colitis, and colon tumorigenesis. CONCLUSIONS: In studies with DSS-induced colitis, we found that pyrin (MEFV) is required for inflammasome activation and IL18 maturation, which promote intestinal barrier integrity and prevent colon inflammation and tumorigenesis. Strategies to increase activity of MEFV or IL18 might be developed for the treatment of IBD and prevention of colitis-associated tumorigenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Colite/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Inflamassomos/metabolismo , Pirina/metabolismo , Junções Íntimas/metabolismo , Animais , Azoximetano , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/imunologia , Colo/patologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Sulfato de Dextrana , Modelos Animais de Doenças , Predisposição Genética para Doença , Inflamassomos/imunologia , Interleucina-18/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Fenótipo , Fosforilação , Pirina/deficiência , Pirina/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Junções Íntimas/imunologia , Junções Íntimas/patologia , Fatores de Tempo , Carga Tumoral , Microambiente Tumoral
18.
J Immunol ; 199(5): 1555-1560, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28733484

RESUMO

For the αß or γδTCR chains to integrate extracellular stimuli into the appropriate intracellular cellular response, they must use the 10 ITAMs found within the CD3 subunits (CD3γε, CD3δε, and ζζ) of the TCR signaling complex. However, it remains unclear whether each specific ITAM sequence of the individual subunit (γεδζ) is required for thymocyte development or whether any particular CD3 ITAM motif is sufficient. In this article, we show that mice utilizing a single ITAM sequence (γ, ε, δ, ζa, ζb, or ζc) at each of the 10 ITAM locations exhibit a substantial reduction in thymic cellularity and limited CD4-CD8- (double-negative) to CD4+CD8+ (double-positive) maturation because of low TCR expression and signaling. Together, the data suggest that ITAM sequence diversity is required for optimal TCR signal transduction and subsequent T cell maturation.


Assuntos
Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Complexos Multiproteicos/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Timo/imunologia , Motivos de Aminoácidos/genética , Animais , Complexo CD3/genética , Diferenciação Celular , Células Cultivadas , Hematopoese , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Transdução de Sinais
19.
J Exp Med ; 214(8): 2217-2229, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28634194

RESUMO

Innate sensing of influenza virus infection induces activation of programmed cell death pathways. We have recently identified Z-DNA-binding protein 1 (ZBP1) as an innate sensor of influenza A virus (IAV). ZBP1-mediated IAV sensing is critical for triggering programmed cell death in the infected lungs. Surprisingly, little is known about the mechanisms regulating ZBP1 activation to induce programmed cell death. Here, we report that the sensing of IAV RNA by retinoic acid inducible gene I (RIG-I) initiates ZBP1-mediated cell death via the RIG-I-MAVS-IFN-ß signaling axis. IAV infection induces ubiquitination of ZBP1, suggesting potential regulation of ZBP1 function through posttranslational modifications. We further demonstrate that ZBP1 senses viral ribonucleoprotein (vRNP) complexes of IAV to trigger cell death. These findings collectively indicate that ZBP1 activation requires RIG-I signaling, ubiquitination, and vRNP sensing to trigger activation of programmed cell death pathways during IAV infection. The mechanism of ZBP1 activation described here may have broader implications in the context of virus-induced cell death.


Assuntos
Morte Celular/fisiologia , Glicoproteínas/fisiologia , Ribonucleoproteínas/metabolismo , Animais , Glicoproteínas/metabolismo , Vírus da Influenza A/fisiologia , Proteínas de Membrana/fisiologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas do Tecido Nervoso/fisiologia , Infecções por Orthomyxoviridae/metabolismo , Proteínas de Ligação a RNA , Receptores de Superfície Celular , Transdução de Sinais/fisiologia , Ubiquitinação
20.
Front Immunol ; 8: 482, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28507546

RESUMO

Defects in cartilage homeostasis can give rise to various skeletal disorders including osteochondromas. Osteochondromas are benign bone tumors caused by excess accumulation of chondrocytes, the main cell type of cartilage. The extracellular signal-regulated kinase (ERK) pathway is a major signaling node that functions within chondrocytes to regulate their growth and differentiation. However, it is not known whether the ERK pathway in other cell types regulates cartilage homeostasis. We show here that mice with a germline deficiency of Erk1 and a conditional deletion of Erk2 in cells that express CD4, or expressed CD4 at one point in development, unexpectedly developed bone deformities. The bone lesions were due to neoplastic outgrowths of chondrocytes and disordered growth plates, similar to tumors observed in the human disease, osteochondromatosis. Chondrocyte accumulation was not due to deletion of Erk2 in the T cells. Rather, CD4cre was expressed in cell types other than T cells, including a small fraction of chondrocytes. Surprisingly, the removal of T cells accelerated osteochondroma formation and enhanced disease severity. These data show for the first time that T cells impact the growth of osteochondromas and describe a novel model to study cartilage homeostasis and osteochondroma formation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...