Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 208(7): 1554-1565, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35321879

RESUMO

Type 1 diabetes (T1D) is characterized by the loss of immune self-tolerance, resulting in an aberrant immune responses against self-tissue. A few therapeutics have been partially successful in reverting or slowing down T1D progression in patients, and the infusion of autologous hematopoietic stem cells (HSCs) is emerging as an option to be explored. In this study, we proposed to pharmacologically enhance by ex vivo modulation with small molecules the immunoregulatory and trafficking properties of HSCs to provide a safer and more efficacious treatment option for patients with T1D and other autoimmune disorders. A high-throughput targeted RNA sequencing screening strategy was used to identify a combination of small molecules (16,16-dimethyl PGE2 and dexamethasone), which significantly upregulate key genes involved in trafficking (e.g., CXCR4) and immunoregulation (e.g., programmed death ligand 1). The pharmacologically enhanced, ex vivo-modulated HSCs (regulatory HSCs [HSC.Regs]) have strong trafficking properties to sites of inflammation in a mouse model of T1D, reverted autoimmune diabetes in NOD mice, and delayed experimental multiple sclerosis and rheumatoid arthritis in preclinical models. Mechanistically, HSC.Regs reduced lymphocytic infiltration of pancreatic ß cells and inhibited the activity of autoreactive T cells. Moreover, when tested in clinically relevant in vitro autoimmune assays, HSC.Regs abrogated the autoimmune response. Ex vivo pharmacological modulation enhances the immunoregulatory and trafficking properties of HSCs, thus generating HSC.Regs, which mitigated autoimmune diabetes and other autoimmune disorders.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1 , Transplante de Células-Tronco Hematopoéticas , Animais , Doenças Autoimunes/terapia , Diabetes Mellitus Tipo 1/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos NOD
3.
J Exp Med ; 213(9): 1901-19, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27481129

RESUMO

MicroRNA (miRNA) deficiency impairs the generation of T follicular helper (Tfh) cells, but the contribution of individual miRNAs to this phenotype remains poorly understood. In this study, we performed deep sequencing analysis of miRNAs expressed in Tfh cells and identified a five-miRNA signature. Analyses of mutant mice deficient of these miRNAs revealed that miR-22 and miR-183/96/182 are dispensable, but miR-155 is essential for the generation and function of Tfh cells. miR-155 deficiency led to decreased proliferation specifically at the late stage of Tfh cell differentiation and reduced CD40 ligand (CD40L) expression on antigen-specific CD4(+) T cells. Mechanistically, miR-155 repressed the expression of Peli1, a ubiquitin ligase that promotes the degradation of the NF-κB family transcription factor c-Rel, which controls cellular proliferation and CD40L expression. Therefore, our study identifies a novel miR-155-Peli1-c-Rel pathway that specifically regulates Tfh cell generation and function.


Assuntos
MicroRNAs/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas c-rel/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T Auxiliares-Indutores/imunologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Ligante de CD40/análise , Diferenciação Celular , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Linfócitos T Citotóxicos/imunologia
4.
Nat Chem Biol ; 12(10): 802-809, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27501396

RESUMO

Guanine-nucleotide dissociation inhibitors (GDIs) are negative regulators of Rho family GTPases that sequester the GTPases away from the membrane. Here we ask how GDI-Cdc42 interaction regulates localized Cdc42 activation for cell motility. The sensitivity of cells to overexpression of Rho family pathway components led us to a new biosensor, GDI.Cdc42 FLARE, in which Cdc42 is modified with a fluorescence resonance energy transfer (FRET) 'binding antenna' that selectively reports Cdc42 binding to endogenous GDIs. Similar antennae could also report GDI-Rac1 and GDI-RhoA interaction. Through computational multiplexing and simultaneous imaging, we determined the spatiotemporal dynamics of GDI-Cdc42 interaction and Cdc42 activation during cell protrusion and retraction. This revealed remarkably tight coordination of GTPase release and activation on a time scale of 10 s, suggesting that GDI-Cdc42 interactions are a critical component of the spatiotemporal regulation of Cdc42 activity, and not merely a mechanism for global sequestration of an inactivated pool of signaling molecules.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Inibidores de Dissociação do Nucleotídeo Guanina/química , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteína cdc42 de Ligação ao GTP/química , Proteína cdc42 de Ligação ao GTP/metabolismo , Sítios de Ligação , Células HEK293 , Humanos , Análise Espaço-Temporal
5.
Immunity ; 44(6): 1284-98, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27332731

RESUMO

T helper 17 (Th17) cells are key players in autoimmune diseases. However, the roles of non-coding RNAs in Th17 cell development and function are largely unknown. We found that deletion of the endoribonuclease-encoding Dicer1 specifically in Th17 cells protected mice from experimental autoimmune encephalomyelitis. We found that the Dicer1-regulated microRNA (miR)-183-96-182 cluster (miR-183C) was highly expressed in Th17 cells and was induced by cytokine IL-6-STAT3 signaling. miR-183C expression enhanced pathogenic cytokine production from Th17 cells during their development and promoted autoimmunity. Mechanistically, miR-183C in Th17 cells directly repressed expression of the transcription factor Foxo1. Foxo1 negatively regulated the pathogenicity of Th17 cells in part by inhibiting expression of cytokine receptor IL-1R1. These findings indicate that the miR-183C drives Th17 pathogenicity in autoimmune diseases via inhibition of Foxo1 and present promising therapeutic targets.


Assuntos
RNA Helicases DEAD-box/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Proteína Forkhead Box O1/metabolismo , MicroRNAs/genética , Esclerose Múltipla/imunologia , Ribonuclease III/metabolismo , Células Th17/fisiologia , Animais , Células Cultivadas , RNA Helicases DEAD-box/genética , Proteína Forkhead Box O1/genética , Humanos , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Interleucina-1/metabolismo , Ribonuclease III/genética , Fator de Transcrição STAT3/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(16): E2316-25, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27044095

RESUMO

To study the development and function of "natural-arising" T regulatory (nTreg) cells, we developed a novel nTreg model on pure nonobese diabetic background using epigenetic reprogramming via somatic cell nuclear transfer. On RAG1-deficient background, we found that monoclonal FoxP3(+)CD4(+)Treg cells developed in the thymus in the absence of other T cells. Adoptive transfer experiments revealed that the thymic niche is not a limiting factor in nTreg development. In addition, we showed that the T-cell receptor (TCR) ß-chain of our nTreg model was not only sufficient to bias T-cell development toward the CD4 lineage, but we also demonstrated that this TCR ß-chain was able to provide stronger TCR signals. This TCR-ß-driven mechanism would thus unify former per se contradicting hypotheses of TCR-dependent and -independent nTreg development. Strikingly, peripheral FoxP3(-)CD4(+)T cells expressing the same TCR as this somatic cell nuclear transfer nTreg model had a reduced capability to differentiate into Th1 cells but were poised to differentiate better into induced nTreg cells, both in vitro and in vivo, representing a novel peripheral precursor subset of nTreg cells to which we refer to as pre-nTreg cells.


Assuntos
Diferenciação Celular/imunologia , Modelos Imunológicos , Técnicas de Transferência Nuclear , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Camundongos , Camundongos Knockout , Linfócitos T Reguladores/citologia
7.
Front Genet ; 6: 340, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26697058

RESUMO

Transient transfection of chemically synthesized microRNA (miRNA) mimics is being used extensively to study the functions and mechanisms of endogenous miRNAs. However, it remains unclear whether transfected miRNAs behave similarly to endogenous miRNAs. Here we show that transient transfection of miRNA mimics into HeLa cells by a commonly used method led to the accumulation of high molecular weight RNA species and a few hundred fold increase in mature miRNA levels. In contrast, expression of the same miRNAs through lentiviral infection or plasmid transfection of HeLa cells, transgenic expression in primary lymphocytes, and endogenous overexpression in lymphoma and leukemia cell lines did not lead to the appearance of high molecular weight RNA species. The increase of mature miRNA levels in these cells was below 10-fold, which was sufficient to suppress target gene expression and to drive lymphoma development in mice. Moreover, transient transfection of miRNA mimics at high concentrations caused non-specific alterations in gene expression, while at low concentrations achieved expression levels comparable to other methods but failed to efficiently suppress target gene expression. Small RNA deep sequencing analysis revealed that the guide strands of miRNA mimics were frequently mutated, while unnatural passenger strands of some miRNA mimics accumulated to high levels. The high molecular weight RNA species were a heterogeneous mixture of several classes of RNA species generated by concatemerization, 5'- and 3'-end tailing of miRNA mimics. We speculate that the supraphysiological levels of mature miRNAs and these artifactual RNA species led to non-specific changes in gene expression. Our results have important implications for the design and interpretation of experiments primarily employing transient transfection of miRNA mimics.

8.
Phys Biol ; 11(1): 016002, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24406475

RESUMO

Elevated levels of circulating endothelial cells (CECs) occur in response to various pathological conditions including myocardial infarction (MI). Here, we adapted a fluid phase biopsy technology platform that successfully detects circulating tumor cells in the blood of cancer patients (HD-CTC assay), to create a high-definition circulating endothelial cell (HD-CEC) assay for the detection and characterization of CECs. Peripheral blood samples were collected from 79 MI patients, 25 healthy controls and six patients undergoing vascular surgery (VS). CECs were defined by positive staining for DAPI, CD146 and von Willebrand Factor and negative staining for CD45. In addition, CECs exhibited distinct morphological features that enable differentiation from surrounding white blood cells. CECs were found both as individual cells and as aggregates. CEC numbers were higher in MI patients compared with healthy controls. VS patients had lower CEC counts when compared with MI patients but were not different from healthy controls. Both HD-CEC and CellSearch® assays could discriminate MI patients from healthy controls with comparable accuracy but the HD-CEC assay exhibited higher specificity while maintaining high sensitivity. Our HD-CEC assay may be used as a robust diagnostic biomarker in MI patients.


Assuntos
Biópsia/métodos , Células Endoteliais/patologia , Infarto do Miocárdio/sangue , Infarto do Miocárdio/patologia , Contagem de Células , Humanos , Infarto do Miocárdio/diagnóstico , Sensibilidade e Especificidade
9.
PLoS One ; 8(11): e79877, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24224016

RESUMO

RhoA and RhoC GTPases share 92% amino acid sequence identity, yet play different roles in regulating cell motility and morphology. To understand these differences, we developed and validated a biosensor of RhoC activation (RhoC FLARE). This was used together with a RhoA biosensor to compare the spatio-temporal dynamics of RhoA and RhoC activity during cell protrusion/retraction and macropinocytosis. Both GTPases were activated similarly at the cell edge, but in regions more distal from the edge RhoC showed higher activation during protrusion. The two isoforms differed markedly in the kinetics of activation. RhoC was activated concomitantly with RhoA at the cell edge, but distally, RhoC activation preceded RhoA activation, occurring before edge protrusion. During macropinocytosis, differences were observed during vesicle closure and in the area surrounding vesicle formation.


Assuntos
Técnicas Biossensoriais/métodos , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Humanos , Cinética , Camundongos
10.
EMBO J ; 32(17): 2377-91, 2013 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-23921550

RESUMO

MicroRNAs (miRNAs) have been broadly implicated in cancer, but their exact function and mechanism in carcinogenesis remain poorly understood. Elevated miR-17~92 expression is frequently found in human cancers, mainly due to gene amplification and Myc-mediated transcriptional upregulation. Here we show that B cell-specific miR-17~92 transgenic mice developed lymphomas with high penetrance and that, conversely, Myc-driven lymphomagenesis stringently requires two intact alleles of miR-17~92. We experimentally identified miR-17~92 target genes by PAR-CLIP and validated select target genes in miR-17~92 transgenic mice. These analyses demonstrate that miR-17~92 drives lymphomagenesis by suppressing the expression of multiple negative regulators of the PI3K and NFκB pathways and by inhibiting the mitochondrial apoptosis pathway. Accordingly, miR-17~92-driven lymphoma cells exhibited constitutive activation of the PI3K and NFκB pathways and chemical inhibition of either pathway reduced tumour size and prolonged the survival of lymphoma-bearing mice. These findings establish miR-17~92 as a powerful cancer driver that coordinates the activation of multiple oncogenic pathways, and demonstrate for the first time that chemical inhibition of miRNA downstream pathways has therapeutic value in treating cancers caused by miRNA dysregulation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Linfoma/genética , MicroRNAs/fisiologia , Animais , Linfócitos B/patologia , Linfócitos B/fisiologia , Linfoma de Burkitt/genética , Linfoma de Burkitt/patologia , Proliferação de Células , Sobrevivência Celular/genética , Proteínas de Homeodomínio/genética , Humanos , Imidazóis/farmacologia , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/administração & dosagem , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Quinoxalinas/farmacologia , RNA Longo não Codificante , Reprodutibilidade dos Testes
11.
Nat Cell Biol ; 13(6): 660-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21572420

RESUMO

The cyclical protrusion and retraction of the leading edge is a hallmark of many migrating cells involved in processes such as development, inflammation and tumorigenesis. The molecular identity of the signalling mechanisms that control these cycles has remained unknown. Here, we used live-cell imaging of biosensors to monitor spontaneous morphodynamic and signalling activities, and employed correlative image analysis to examine the role of cyclic-AMP-activated protein kinase A (PKA) in protrusion regulation. PKA activity at the leading edge is closely synchronized with rapid protrusion and with the activity of RhoA. Ensuing PKA phosphorylation of RhoA and the resulting increased interaction between RhoA and RhoGDI (Rho GDP-dissociation inhibitor) establish a negative feedback mechanism that controls the cycling of RhoA activity at the leading edge. Thus, cooperation between PKA, RhoA and RhoGDI forms a pacemaker that governs the morphodynamic behaviour of migrating cells.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Movimento Celular , Dipodomys , Feminino , Rim/citologia , Rim/enzimologia , Ratos , Transdução de Sinais , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
12.
Exp Cell Res ; 316(13): 2027-41, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20406634

RESUMO

Directed cell migration requires continuous cycles of protrusion of the leading edge and contraction to pull up the cell rear. How these spatially distributed processes are coordinated to maintain a state of persistent protrusion remains unknown. During wound healing responses of epithelial sheets, cells along the wound edge display two distinct morphologies: 'leader cells' exhibit persistent edge protrusions, while the greater majority of 'follower cells' randomly cycle between protrusion and retraction. Here, we exploit the heterogeneity in cell morphodynamic behaviors to deduce the requirements in terms of cytoskeleton dynamics for persistent and sporadic protrusion events. We used quantitative Fluorescent Speckle Microscopy (qFSM) to compare rates of F-actin assembly and flow relative to the local protrusion and retraction dynamics of the leading edge. Persistently protruding cells are characterized by contractile actomyosin structures that align with the direction of migration, with converging F-actin flows interpenetrating over a wide band in the lamella. Conversely, non-persistent protruders have their actomyosin structures aligned perpendicular to the axis of migration, and are characterized by prominent F-actin retrograde flows that end into transverse arcs. Analysis of F-actin kinetics in the lamellipodia showed that leader cells have three-fold higher assembly rates when compared to followers. To further investigate a putative relationship between actomyosin contraction and F-actin assembly, myosin II was inhibited by blebbistatin. Treated cells at the wound edge adopted a homogeneously persistent protrusion behavior, with rates matching those of leader cells. Surprisingly, we found that disintegration of actomyosin structures led to a significant decrease in F-actin assembly. Our data suggests that persistent protrusion in these cells is achieved by a reduction in overall F-actin retrograde flow, with lower assembly rates now sufficient to propel forward the leading edge. Based on our data we propose that differences in the protrusion persistence of leaders and followers originate in the distinct actomyosin contraction modules that differentially regulate leading edge protrusion-promoting F-actin assembly, and retraction-promoting retrograde flow.


Assuntos
Actinas/fisiologia , Movimento Celular , Citoesqueleto/metabolismo , Epitélio/ultraestrutura , Pseudópodes/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Células Cultivadas , Epitélio/metabolismo , Cinética , Microscopia de Fluorescência
13.
Biophys J ; 98(8): 1571-81, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20409477

RESUMO

We propose a mathematical model for simulating the leading-edge dynamics of a migrating cell from the interplay among elastic properties, architecture of the actin cytoskeleton, and the mechanics of the membrane. Our approach is based on the description of the length and attachment dynamics of actin filaments in the lamellipodium network. It is used to determine the total force exerted on the membrane at each position along the leading edge and at each time step. The model reproduces the marked state switches in protrusion morphodynamics found experimentally between epithelial cells in control conditions and cells expressing constitutively active Rac, a signaling molecule involved in the regulation of lamellipodium network assembly. The model also suggests a mechanistic explanation of experimental distortions in protrusion morphodynamics induced by deregulation of Arp2/3 and cofilin activity.


Assuntos
Actinas/metabolismo , Membrana Celular/metabolismo , Modelos Biológicos , Pseudópodes/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Simulação por Computador , Fenótipo
14.
Curr Opin Cell Biol ; 20(5): 541-50, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18586481

RESUMO

Understanding the structural adaptation and signaling of adhesion sites in response to mechanical stimuli requires in situ characterization of the dynamic activation of a large number of adhesion components. Here, we review high-resolution live cell imaging approaches to measure forces, assembly, and interaction of adhesion components, and the activation of adhesion-mediated signals. We conclude by outlining computational multiplexing as a framework for the integration of these data into comprehensive models of adhesion signaling pathways.


Assuntos
Técnicas Biossensoriais , Moléculas de Adesão Celular/metabolismo , Adesão Celular/fisiologia , Mecanotransdução Celular/fisiologia , Microscopia de Fluorescência/métodos , Análise Espectral/métodos , Animais , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Estresse Mecânico
15.
J Theor Biol ; 250(1): 209-18, 2008 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-17950756

RESUMO

A protein interaction network (PIN) is a set of proteins that modulate one another's activities by regulated synthesis and degradation, by reversible binding to form complexes, and by catalytic reactions (e.g., phosphorylation and dephosphorylation). Most PINs are so complex that their dynamical characteristics cannot be deduced accurately by intuitive reasoning alone. To predict the properties of such networks, many research groups have turned to mathematical models (differential equations based on standard biochemical rate laws, e.g., mass-action, Michaelis-Menten, Hill). When using Michaelis-Menten rate expressions to model PINs, care must be exercised to avoid making inconsistent assumptions about enzyme-substrate complexes. We show that an appealingly simple model of a PIN that functions as a bistable switch is compromised by neglecting enzyme-substrate intermediates. When the neglected intermediates are put back into the model, bistability of the switch is lost. The theory of chemical reaction networks predicts that bistability can be recovered by adding specific reaction channels to the molecular mechanism. We explore two very different routes to recover bistability. In both cases, we show how to convert the original 'phenomenological' model into a consistent set of mass-action rate laws that retains the desired bistability properties. Once an equivalent model is formulated in terms of elementary chemical reactions, it can be simulated accurately either by deterministic differential equations or by Gillespie's stochastic simulation algorithm.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclo Celular/fisiologia , Modelos Químicos , Mapeamento de Interação de Proteínas/métodos , Algoritmos , Senescência Celular/fisiologia , Ligação Proteica/fisiologia , Proteínas/antagonistas & inibidores , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Processos Estocásticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA