Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Biol Psychiatry Glob Open Sci ; 3(4): 1104-1115, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37881572

RESUMO

Background: A salient effect of addictive drugs is to hijack the dopamine reward system, an evolutionarily conserved driver of goal-directed behavior and learning. Reduced dopamine type 2 receptor availability in the striatum is an important pathophysiological mechanism for addiction that is both consequential and causal for other molecular, cellular, and neuronal network differences etiologic for this disorder. Here, we sought to identify gene expression changes attributable to innate low expression of the Drd2 gene in the striatum and specific to striatal indirect medium spiny neurons (iMSNs). Methods: Cre-conditional, translating ribosome affinity purification (TRAP) was used to purify and analyze the translatome (ribosome-bound messenger RNA) of iMSNs from mice with low/heterozygous or wild-type Drd2 expression in iMSNs. Complementary electrophysiological recordings and gene expression analysis of postmortem brain tissue from human cocaine users were performed. Results: Innate low expression of Drd2 in iMSNs led to differential expression of genes involved in GABA (gamma-aminobutyric acid) and cAMP (cyclic adenosine monophosphate) signaling, neural growth, lipid metabolism, neural excitability, and inflammation. Creb1 was identified as a likely upstream regulator, among others. In human brain, expression of FXYD2, a modulatory subunit of the Na/K pump, was negatively correlated with DRD2 messenger RNA expression. In iMSN-TRAP-Drd2HET mice, increased Cartpt and reduced S100a10 (p11) expression recapitulated previous observations in cocaine paradigms. Electrophysiology experiments supported a higher GABA tone in iMSN-Drd2HET mice. Conclusions: This study provides strong molecular evidence that, in addiction, inhibition by the indirect pathway is constitutively enhanced through neural growth and increased GABA signaling.

3.
Commun Biol ; 4(1): 1201, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34671088

RESUMO

To capture the global gene network regulating the differentiation of immature T cells in an unbiased manner, large-scale forward genetic screens in zebrafish were conducted and combined with genetic interaction analysis. After ENU mutagenesis, genetic lesions associated with failure of T cell development were identified by meiotic recombination mapping, positional cloning, and whole genome sequencing. Recessive genetic variants in 33 genes were identified and confirmed as causative by additional experiments. The mutations affected T cell development but did not perturb the development of an unrelated cell type, growth hormone-expressing somatotrophs, providing an important measure of cell-type specificity of the genetic variants. The structure of the genetic network encompassing the identified components was established by a subsequent genetic interaction analysis, which identified many instances of positive (alleviating) and negative (synthetic) genetic interactions. Several examples of synthetic lethality were subsequently phenocopied using combinations of small molecule inhibitors. These drugs not only interfered with normal T cell development, but also elicited remission in a model of T cell acute lymphoblastic leukaemia. Our findings illustrate how genetic interaction data obtained in the context of entire organisms can be exploited for targeted interference with specific cell types and their malignant derivatives.


Assuntos
Redes Reguladoras de Genes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Mutações Sintéticas Letais , Linfócitos T/metabolismo , Animais , Modelos Animais de Doenças , Epistasia Genética , Fenótipo , Peixe-Zebra
4.
J Clin Invest ; 131(9)2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33724957

RESUMO

Cholangiopathies caused by biliary epithelial cell (BEC) injury represent a leading cause of liver failure. No effective pharmacologic therapies exist, and the underlying mechanisms remain obscure. We aimed to explore the mechanisms of bile duct repair after targeted BEC injury. Injection of intermedilysin into BEC-specific human CD59 (hCD59) transgenic mice induced acute and specific BEC death, representing a model to study the early signals that drive bile duct repair. Acute BEC injury induced cholestasis followed by CCR2+ monocyte recruitment and BEC proliferation. Using microdissection and next-generation RNA-Seq, we identified 5 genes, including Mapk8ip2, Cdkn1a, Itgb6, Rgs4, and Ccl2, that were most upregulated in proliferating BECs after acute injury. Immunohistochemical analyses confirmed robust upregulation of integrin αvß6 (ITGß6) expression in this BEC injury model, after bile duct ligation, and in patients with chronic cholangiopathies. Deletion of the Itgb6 gene attenuated BEC proliferation after acute bile duct injury. Macrophage depletion or Ccr2 deficiency impaired ITGß6 expression and BEC proliferation. In vitro experiments revealed that bile acid-activated monocytes promoted BEC proliferation through ITGß6. Our data suggest that BEC injury induces cholestasis, monocyte recruitment, and induction of ITGß6, which work together to promote BEC proliferation and therefore represent potential therapeutic targets for cholangiopathies.


Assuntos
Antígenos de Neoplasias/biossíntese , Ácidos e Sais Biliares/metabolismo , Sistema Biliar/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Integrinas/biossíntese , Ativação de Macrófagos , Macrófagos/metabolismo , Regulação para Cima , Animais , Antígenos de Neoplasias/genética , Ácidos e Sais Biliares/genética , Feminino , Humanos , Integrinas/genética , Masculino , Camundongos , Camundongos Transgênicos , RNA-Seq
5.
Front Neurorobot ; 14: 6, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116636

RESUMO

Understanding the brain is a fascinating challenge, captivating the scientific community and the public alike. The lack of effective treatment for most brain disorders makes the training of the next generation of neuroscientists, engineers and physicians a key concern. Over the past decade there has been a growing effort to introduce neuroscience in primary and secondary schools, however, hands-on laboratories have been limited to anatomical or electrophysiological activities. Modern neuroscience research labs are increasingly using computational tools to model circuits of the brain to understand information processing. Here we introduce the use of neurorobots - robots controlled by computer models of biological brains - as an introduction to computational neuroscience in the classroom. Neurorobotics has enormous potential as an education technology because it combines multiple activities with clear educational benefits including neuroscience, active learning, and robotics. We describe a 1-week introductory neurorobot workshop that teaches high school students how to use neurorobots to investigate key concepts in neuroscience, including spiking neural networks, synaptic plasticity, and adaptive action selection. Our do-it-yourself (DIY) neurorobot uses wheels, a camera, a speaker, and a distance sensor to interact with its environment, and can be built from generic parts costing about $170 in under 4 h. Our Neurorobot App visualizes the neurorobot's visual input and brain activity in real-time, and enables students to design new brains and deliver dopamine-like reward signals to reinforce chosen behaviors. We ran the neurorobot workshop at two high schools (n = 295 students total) and found significant improvement in students' understanding of key neuroscience concepts and in students' confidence in neuroscience, as assessed by a pre/post workshop survey. Here we provide DIY hardware assembly instructions, discuss our open-source Neurorobot App and demonstrate how to teach the Neurorobot Workshop. By doing this we hope to accelerate research in educational neurorobotics and promote the use of neurorobots to teach computational neuroscience in high school.

6.
Cell Rep ; 17(9): 2259-2270, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27880902

RESUMO

Lymphocytes represent basic components of vertebrate adaptive immune systems, suggesting the utility of non-mammalian models to define the molecular basis of their development and differentiation. Our forward genetic screens in zebrafish for recessive mutations affecting early T cell development revealed several major genetic pathways. The identification of lineage-specific transcription factors and specific components of cytokine signaling and DNA replication and/or repair pathways known from studies of immunocompromised mammals provided an evolutionary cross-validation of the screen design. Unexpectedly, however, genes encoding proteins required for pre-mRNA processing were enriched in the collection of mutants identified here. In both zebrafish and mice, deficiency of the splice regulator TNPO3 impairs intrathymic T cell differentiation, illustrating the evolutionarily conserved and cell-type-specific functions of certain pre-mRNA-processing factors for T cell development.


Assuntos
Testes Genéticos , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA/genética , Linfócitos T/citologia , Linfócitos T/metabolismo , Peixe-Zebra/genética , Processamento Alternativo/genética , Animais , Epistasia Genética , Regulação da Expressão Gênica no Desenvolvimento , Larva/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Especificidade de Órgãos/genética , Precursores de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Transcriptoma/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , beta Carioferinas/deficiência , beta Carioferinas/metabolismo
7.
J Immunol ; 191(12): 6002-9, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24244014

RESUMO

NKT and mucosal-associated invariant T (MAIT) cells express semi-invariant TCR and restriction by nonclassical MHC class Ib molecules. Despite common features, the respective development of NKT and MAIT subsets is distinct. NKTs proliferate extensively and acquire effector properties prior to thymic export. MAIT cells exit the thymus as naive cells and acquire an effector/memory phenotype in a process requiring both commensal flora and B cells. During thymic development, NKTs are selected by CD1d-expressing cortical thymocytes; however, the hematopoietic cell type responsible for MAIT cell selection remains unresolved. Using reaggregated thymic organ culture and bone marrow chimeras, we demonstrate that positive selection of mouse iVα19 transgenic and Vß6 transgenic MAIT cell progenitors requires MHC-related 1-expressing CD4(+)CD8(+) double positive thymocytes, whereas thymic B cells, macrophages, and dendritic cell subsets are dispensable. Preincubation of double positive thymocytes with exogenous bacterial ligand increases MHC-related 1 surface expression and enhances mature MAIT cell activation in the in vitro cocultures. The revelation of a common cell type for the selection of both NKT and MAIT subsets raises questions about the mechanisms underlying acquisition of their specific features.


Assuntos
Antígenos CD4/análise , Antígenos CD8/análise , Seleção Clonal Mediada por Antígeno , Antígenos de Histocompatibilidade Classe I/imunologia , Linfopoese/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Antígenos de Diferenciação de Linfócitos T/análise , Linhagem da Célula , Células Cultivadas , Técnicas de Cocultura , Escherichia coli/imunologia , Feminino , Genes de Imunoglobulinas , Células-Tronco Hematopoéticas/classificação , Células-Tronco Hematopoéticas/citologia , Antígenos de Histocompatibilidade Classe I/genética , Região Variável de Imunoglobulina/genética , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Técnicas de Cultura de Órgãos , Quimera por Radiação , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Organismos Livres de Patógenos Específicos , Células Estromais/fisiologia , Subpopulações de Linfócitos T/química , Timo/citologia , Timo/imunologia
8.
J Immunol ; 190(5): 2102-10, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23365074

RESUMO

Tolerance to self-Ags is generated in the thymus. Both epithelial and hematopoietic thymic stromal cells play an active and essential role in this process. However, the role of each of the various stromal cell types remains unresolved. To our knowledge, we describe the first comparative analysis of several types of thymic hematopoietic stromal cells (THSCs) for their ability to induce CD4 tolerance to self, in parallel with the thymic epithelium. The THSCs--two types of conventional dendritic cells (cDCs), plasmacytoid dendritic cells, macrophages (MΦs), B lymphocytes, and eosinophils--were first characterized and quantified in adult mouse thymus. They were then examined in reaggregated thymic organ cultures containing mixtures of monoclonal and polyclonal thymocytes. This thymocyte mixture allows for the analysis of Ag-specific events while avoiding the extreme skewing frequently seen in purely monoclonal systems. Our data indicate that thymic epithelium alone is capable of promoting self-tolerance by eliminating autoreactive CD4 single-positive thymocytes and by supporting regulatory T cell (Treg) development. We also show that both non-Treg CD4 single-positive thymocytes and Tregs are efficiently deleted by the two populations of cDCs present in the thymus, as well as to a lesser extent by MΦs. Plasmacytoid dendritic cells, B lymphocytes, and eosinophils were not able to do so. Finally, cDCs were also the most efficient THSCs at supporting Treg development in the thymus, suggesting that although they may share some characteristics required for negative selection with MΦs, they do not share those required for the support of Treg development, making cDCs a unique cell subset in the thymus.


Assuntos
Antígenos CD/imunologia , Células Dendríticas/citologia , Células Estromais/citologia , Linfócitos T Reguladores/citologia , Timócitos/citologia , Timo/citologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Técnicas de Cultura de Células , Células Dendríticas/classificação , Células Dendríticas/imunologia , Eosinófilos/citologia , Eosinófilos/imunologia , Epitélio/imunologia , Feminino , Feto , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Tolerância a Antígenos Próprios , Células Estromais/classificação , Células Estromais/imunologia , Linfócitos T Reguladores/imunologia , Timócitos/imunologia , Timo/imunologia
9.
PLoS One ; 7(12): e52591, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300712

RESUMO

The thymic medulla is dedicated for purging the T-cell receptor (TCR) repertoire of self-reactive specificities. Medullary thymic epithelial cells (mTECs) play a pivotal role in this process because they express numerous peripheral tissue-restricted self-antigens. Although it is well known that medulla formation depends on the development of single-positive (SP) thymocytes, the mechanisms underlying this requirement are incompletely understood. We demonstrate here that conventional SP CD4⁺ thymocytes bearing autoreactive TCRs drive a homeostatic process that fine-tunes medullary plasticity in adult mice by governing the expansion and patterning of the medulla. This process exhibits strict dependence on TCR-reactivity with self-antigens expressed by mTECs, as well as engagement of the CD28-CD80/CD86 costimulatory axis. These interactions induce the expression of lymphotoxin α in autoreactive CD4⁺ thymocytes and RANK in mTECs. Lymphotoxin in turn drives mTEC development in synergy with RANKL and CD40L. Our results show that Ag-dependent interactions between autoreactive CD4⁺ thymocytes and mTECs fine-tune homeostasis of the medulla by completing the signaling axes implicated in mTEC expansion and medullary organization.


Assuntos
Autoantígenos/imunologia , Antígenos CD4/metabolismo , Homeostase , Timócitos/fisiologia , Timo/crescimento & desenvolvimento , Animais , Padronização Corporal , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Expressão Gênica , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Timócitos/imunologia , Timócitos/metabolismo , Timo/citologia , Timo/imunologia , Técnicas de Cultura de Tecidos
10.
Trends Immunol ; 32(5): 212-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21459674

RESUMO

Mucosal-associated invariant T (MAIT) cells are a population of T cells that display a semi-invariant T cell receptor (TCR) and are restricted by the evolutionarily conserved major histocompatibility complex related molecule, MR1. Here, we review recent knowledge of this T cell population. MAIT cells are abundant in human blood, gut and liver, and display an effector phenotype. They follow an atypical pathway of development and preferentially locate to peripheral tissues. Human and mouse MAIT cells react to bacterially infected cells in an MR1-dependent manner. They migrate to the infection site and can be protective in experimental infection models. MAIT cells secrete interferon-γ, and interleukin-17 under certain conditions. The species conservation, as well as the wide microbial reactivity, infer an important role for this cell population in immunity.


Assuntos
Infecções Bacterianas/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Mucosa/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Interferon gama/metabolismo , Interleucina-17/metabolismo , Ativação Linfocitária , Camundongos , Antígenos de Histocompatibilidade Menor , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/metabolismo
11.
PLoS Biol ; 7(3): e54, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19278296

RESUMO

Mucosal-associated invariant T (MAIT) cells display two evolutionarily conserved features: an invariant T cell receptor (TCR)alpha (iTCRalpha) chain and restriction by the nonpolymorphic class Ib major histocompatibility complex (MHC) molecule, MHC-related molecule 1 (MR1). MR1 expression on thymus epithelial cells is not necessary for MAIT cell development but their accumulation in the gut requires MR1 expressing B cells and commensal flora. MAIT cell development is poorly known, as these cells have not been found in the thymus so far. Herein, complementary human and mouse experiments using an anti-humanValpha7.2 antibody and MAIT cell-specific iTCRalpha and TCRbeta transgenic mice in different genetic backgrounds show that MAIT cell development is a stepwise process, with an intra-thymic selection followed by peripheral expansion. Mouse MAIT cells are selected in an MR1-dependent manner both in fetal thymic organ culture and in double iTCRalpha and TCRbeta transgenic RAG knockout mice. In the latter mice, MAIT cells do not expand in the periphery unless B cells are added back by adoptive transfer, showing that B cells are not required for the initial thymic selection step but for the peripheral accumulation. In humans, contrary to natural killer T (NKT) cells, MAIT cells display a naïve phenotype in the thymus as well as in cord blood where they are in low numbers. After birth, MAIT cells acquire a memory phenotype and expand dramatically, up to 1%-4% of blood T cells. Finally, in contrast with NKT cells, human MAIT cell development is independent of the molecular adaptor SAP. Interestingly, mouse MAIT cells display a naïve phenotype and do not express the ZBTB16 transcription factor, which, in contrast, is expressed by NKT cells and the memory human MAIT cells found in the periphery after birth. In conclusion, MAIT cells are selected by MR1 in the thymus on a non-B non-T hematopoietic cell, and acquire a memory phenotype and expand in the periphery in a process dependent both upon B cells and the bacterial flora. Thus, their development follows a unique pattern at the crossroad of NKT and gammadelta T cells.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade nas Mucosas/imunologia , Células T Matadoras Naturais/fisiologia , Subpopulações de Linfócitos T/fisiologia , Timo/imunologia , Animais , Linfócitos B/fisiologia , Criança , Sangue Fetal/imunologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor , Proteína com Dedos de Zinco da Leucemia Promielocítica , Receptores de Antígenos de Linfócitos T alfa-beta , Linfócitos T , Timo/citologia
12.
Oncol Rep ; 11(2): 523-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14719094

RESUMO

The aim of this study was to determine the presence of angiostatin in ascitic and pleural effusions from cancer patients, as well as of metalloproteinases (MMPs) and urokinase-type plasminogen activator (uPA), both involved in angiostatin generation in in vitro models. Ascitic fluids, pleural exudates, and sera from 21 cancer patients were analyzed for the presence of angiostatin by western blot, whereas gelatinases MMP-2 and MMP-9, and uPA were evaluated by zymography. Our study revealed elevated levels of angiostatin in effusions of cancer patients, contrasting with mostly intermediate levels in less than half of their sera, and undetectable levels in normal sera. Despite the observation of enhanced levels of HMW-uPA and MMP-2 in malignant effusions from cancer patients, their analysis in individual samples showed no association between angiostatin presence and the enzymes, suggesting that the latter would not play an unimportant role, if any, in in vivo generation of angiostatin.


Assuntos
Angiostatinas/metabolismo , Ascite/fisiopatologia , Neoplasias/metabolismo , Derrame Pleural/química , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/análise , Feminino , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Pessoa de Meia-Idade , Valores de Referência , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...