Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 2024 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-39471815

RESUMO

Fructose is associated with colorectal cancer tumorigenesis and metastasis through ketohexokinase-mediated metabolism in the colorectal epithelium, yet its role in the tumor immune microenvironment remains largely unknown. Here, we show that a modest amount of fructose, without affecting obesity and associated complications, promotes colorectal cancer tumorigenesis and growth by suppressing the polarization of M1-like macrophages. Fructose inhibits M1-like macrophage polarization independently of fructose-mediated metabolism. Instead, it serves as a signal molecule to promote the interaction between hexokinase 2 and inositol 1,4,5-trisphophate receptor type 3, the predominant Ca2+ channel on the endoplasmic reticulum. The interaction reduces Ca2+ levels in cytosol and mitochondria, thereby suppressing the activation of mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 1 (STAT1) as well as NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Consequently, this impedes M1-like macrophage polarization. Our study highlights the critical role of fructose as a signaling molecule that impairs the polarization of M1-like macrophages for tumor growth.

2.
BMJ Open ; 13(10): e077219, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37879700

RESUMO

INTRODUCTION: Conventional interventional modalities for preserving or improving cognitive function in patients with brain tumour undergoing radiotherapy usually involve pharmacological and/or cognitive rehabilitation therapy administered at fixed doses or intensities, often resulting in suboptimal or no response, due to the dynamically evolving patient state over the course of disease. The personalisation of interventions may result in more effective results for this population. We have developed the CURATE.AI COR-Tx platform, which combines a previously validated, artificial intelligence-derived personalised dosing technology with digital cognitive training. METHODS AND ANALYSIS: This is a prospective, single-centre, single-arm, mixed-methods feasibility clinical trial with the primary objective of testing the feasibility of the CURATE.AI COR-Tx platform intervention as both a digital intervention and digital diagnostic for cognitive function. Fifteen patient participants diagnosed with a brain tumour requiring radiotherapy will be recruited. Participants will undergo a remote, home-based 10-week personalised digital intervention using the CURATE.AI COR-Tx platform three times a week. Cognitive function will be assessed via a combined non-digital cognitive evaluation and a digital diagnostic session at five time points: preradiotherapy, preintervention and postintervention and 16-weeks and 32-weeks postintervention. Feasibility outcomes relating to acceptability, demand, implementation, practicality and limited efficacy testing as well as usability and user experience will be assessed at the end of the intervention through semistructured patient interviews and a study team focus group discussion at study completion. All outcomes will be analysed quantitatively and qualitatively. ETHICS AND DISSEMINATION: This study has been approved by the National Healthcare Group (NHG) DSRB (DSRB2020/00249). We will report our findings at scientific conferences and/or in peer-reviewed journals. TRIAL REGISTRATION NUMBER: NCT04848935.


Assuntos
Inteligência Artificial , Neoplasias Encefálicas , Humanos , Neoplasias Encefálicas/radioterapia , Cognição , Estudos de Viabilidade , Estudos Prospectivos
3.
Mol Cell Biol ; 42(2): e0028221, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34842443

RESUMO

Glycemic variability has been considered one of the predictors of diabetes complications in patients with diabetes mellitus (DM). In this work, we evaluated whether glycemic variability induces cardiac fibrosis through regulating cardiac fibroblast activation and macrophage polarization. Moreover, we determined whether glucose transporter sodium-glucose cotransporter 1 (SGLT1) plays an important role in this process. Glycemic variability-induced mice were established using DM mice (GVDM mice), and intermittent high-glucose (IHG) treatment was used to simulate glycemic variability in RAW264.7 macrophages and cardiac fibroblasts. The short hairpin RNA for SGLT1 was used to knock down SGLT1. The results showed that glycemic variability aggravated the cardiac fibrosis in GVDM mice. Additionally, glycemic variability promoted the expression of fibrogenic cytokine and the extracellular matrix proteins in left ventricular tissues and cardiac fibroblasts. GVDM mice showed a higher incidence of macrophage infiltration and M1 polarization in left ventricular tissues. Moreover, IHG-promoted RAW264.7 macrophages tended to differentiate to M1 phenotype. SGLT1 knockdown alleviated cardiac fibrosis in GVDM mice and inhibited activations of cardiac fibroblast and macrophage M1 polarization. Our results indicated that glycemic variability aggravates cardiac fibrosis through activating cardiac fibroblast and macrophage M1 polarization, which could be partially inhibited by SGLT1 knockdown.


Assuntos
Glicemia/metabolismo , Fibroblastos/metabolismo , Ativação de Macrófagos/fisiologia , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Animais , Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamento de Genes/métodos , Glucose/metabolismo , Coração/fisiopatologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo
4.
Polymers (Basel) ; 15(1)2022 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36616565

RESUMO

Lactoferrin (LF) is an iron-binding glycoprotein with various biological activities that has been extensively used in food and medical applications. Several methods for detecting LF have been reported, but they still face challenges in terms of sensitivity and simplicity of detection. To achieve an accurate and efficient detection of LF, we developed a method for the determination of LF in lactoferrin supplements using carbon dots (CDs) fluorescent probes. The N, S-doped PPI carbon dots (N, S-PPI-CDs) were prepared using a protein (peanut protein isolate) and cysteamine as precursors. The prepared N, S-PPI-CDs exhibited intense blue fluorescence and good biocompatibility, while the fluorescence intensity of the N, S-PPI-CDs showed a good linear relationship with Fe2+/Fe3+ concentration (0-2 µM). The N, S-PPI-CDs exhibited a high potential ability to rapidly detect Fe2+/Fe3+ within 30 s, with a limit of detection (LoD) of 0.21 µM/0.17 µM. Due to the reversible binding of LF to Fe, the N, S-PPI-CDs showed a high sensitivity and selectivity for LF, with a limit of detection (LoD) of 1.92 µg/mL. In addition, LF was quantified in real sample LF supplements and showed a fluctuation in recovery of less than 2.48%, further demonstrating the effectiveness of the fluorescent N, S-PPI-CDs sensor.

5.
Front Immunol ; 12: 707404, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276703

RESUMO

Thymic blood vessels at the perivascular space (PVS) are the critical site for both homing of hematopoietic progenitor cells (HPCs) and egress of mature thymocytes. It has been intriguing how different opposite migrations can happen in the same place. A subset of specialized thymic portal endothelial cells (TPECs) associated with PVS has been identified to function as the entry site for HPCs. However, the cellular basis and mechanism underlying egress of mature thymocytes has not been well defined. In this study, using various conventional and conditional gene-deficient mouse models, we first confirmed the role of endothelial lymphotoxin beta receptor (LTßR) for thymic egress and ruled out the role of LTßR from epithelial cells or dendritic cells. In addition, we found that T cell-derived ligands lymphotoxin (LT) and LIGHT are required for thymic egress, suggesting a crosstalk between T cells and endothelial cells (ECs) for thymic egress control. Furthermore, immunofluorescence staining analysis interestingly showed that TPECs are also the exit site for mature thymocytes. Single-cell transcriptomic analysis of thymic endothelial cells suggested that TPECs are heterogeneous and can be further divided into two subsets depending on BST-1 expression level. Importantly, BST-1hi population is associated with thymic egressing thymocytes while BST-1lo/- population is associated with HPC settling. Thus, we have defined a LT/LIGHT-LTßR signaling-mediated cellular crosstalk regulating thymic egress and uncovered distinct subsets of TPECs controlling thymic homing and egress, respectively.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/metabolismo , Receptor beta de Linfotoxina/metabolismo , Timócitos/metabolismo , Timo/metabolismo , Animais , Linfotoxina-alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Timo/citologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
6.
J Immunol ; 205(3): 853-863, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32591396

RESUMO

As a specialized subset of intestinal epithelial cells (IECs), goblet cells (GCs) play an important role during the antibacterial response via mucin production. However, the regulatory mechanisms involved in GC differentiation and function during infection, particularly the role of immune cell-IEC cross-talk, remain largely unknown. In this study, using Villin∆Ltbr conditional knockout mice, we demonstrate that LTßR, expressed on IECs, is required for GC hyperplasia and mucin 2 (MUC2) expression during Listeria infection for host defense but not homeostatic maintenance in the naive state. Analysis of single gene-deficient mice revealed that the ligand lymphotoxin (LT), but not LIGHT, and type 3 innate lymphoid cells (ILC3s), but not conventional T cells, are required for MUC2-dependent Listeria control. Conditional deficiency of LT in ILC3s further confirmed the importance of LT signals derived from ILC3s. Lack of ILC3-derived LT or IEC-derived LTßR resulted in the defective expression of genes related to GC differentiation but was not correlated with IEC proliferation and cell death, which were found to be normal by Ki-67 and Annexin V staining. In addition, the alternative NF-κB signaling pathway (involving RelB) in IECs was found to be required for the expression of GC differentiation-related genes and Muc2 and required for the anti-Listeria response. Therefore, our data together suggest a previously unrecognized ILC3-IEC interaction and LT-LTßR-RelB signaling axis governing GC differentiation and function during Listeria infection for host defense.


Assuntos
Diferenciação Celular/imunologia , Células Caliciformes/imunologia , Listeria/imunologia , Listeriose/imunologia , Linfócitos/imunologia , Linfotoxina-alfa/imunologia , Transdução de Sinais/imunologia , Animais , Diferenciação Celular/genética , Células Caliciformes/patologia , Listeriose/genética , Listeriose/patologia , Linfócitos/patologia , Receptor beta de Linfotoxina , Linfotoxina-alfa/genética , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais/genética
7.
J Immunol ; 202(10): 2999-3007, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30952816

RESUMO

The lymphatic vasculature is an important route for dendritic cell (DC) or tumor cell migration from peripheral tissues to draining lymph nodes (DLNs). However, the underlying molecular and cellular mechanisms remain poorly understood. In this study, using conventional bone marrow chimeric mice and additional UVB radiation, we found that deficiency of LIGHT but not lymphotoxin (LT) α1ß2, likely on radioresistant Langerhans cells (LCs), resulted in impaired skin DC migration to DLNs during LPS-induced inflammation. In addition, LT ß receptor (LTßR), but not herpes virus entry mediator, was found to be the receptor of LIGHT controlling DC migration. Furthermore, conditional deficiency of LTßR in Tie2 cre or Lyve1 cre mice, but not in LTßR-deficient bone marrow chimeric mice, impaired DC migration, suggesting an important role of LTßR in radioresistant lymphatic endothelial cells (LECs), although the role of LTßR in blood endothelial cells remains intriguing. Mechanistically, the gene expression of both CCL21 and CCL19 was found to be reduced in skin LECs isolated from LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice compared with their controls upon LPS stimulation. Soluble recombinant LIGHT was able to upregulate CCL21 and CCL19 gene expression on SVEC4-10 endothelial cells. Doxycycline, an inhibitor of soluble LIGHT release in the inflamed skin, impaired skin CCL21 and CCL19 expression and DC migration. In addition, melanoma cell metastasis to DLNs was also inhibited in LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice. Together, our data suggest, to our knowledge, a previously unrecognized scenario in which LCs activate LECs via the LIGHT-LTßR signaling axis to promote DC migration or tumor cell metastasis.


Assuntos
Células Endoteliais/imunologia , Células de Langerhans/imunologia , Vasos Linfáticos/imunologia , Receptor beta de Linfotoxina/imunologia , Transdução de Sinais/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Células Endoteliais/patologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Células de Langerhans/patologia , Lipopolissacarídeos/toxicidade , Vasos Linfáticos/patologia , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
8.
J Immunol ; 201(1): 69-76, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29760194

RESUMO

Cellular cross-talk mediated by lymphotoxin αß-lymphotoxin ß receptor (LTßR) signaling plays a critical role in lymph node (LN) development. Although the major role of LTßR signaling has long been considered to occur in mesenchymal lymphoid tissue organizer cells, a recent study using a VE-cadherincreLtbrfl/fl mouse model suggested that endothelial LTßR signaling contributes to the formation of LNs. However, the detailed roles of LTßR in different endothelial cells (ECs) in LN development remain unknown. Using various cre transgenic mouse models (Tekcre , a strain targeting ECs, and Lyve1cre , mainly targeting lymphatic ECs), we observed that specific LTßR ablation in Tekcre+ or Lyve1cre+ cells is not required for LN formation. Moreover, double-cre-mediated LTßR depletion does not interrupt LN formation. Nevertheless, TekcreLtbrfl/fl mice exhibit reduced lymphoid tissue inducer cell accumulation at the LN anlagen and impaired LN maturation. Interestingly, a subset of ECs (VE-cadherin+Tekcre-low/neg ECs) was found to be enriched in transcripts related to hematopoietic cell recruitment and transendothelial migration, resembling LN high ECs in adult animals. Furthermore, endothelial Tek was observed to negatively regulate hematopoietic cell transmigration. Taken together, our data suggest that although Tekcre+ endothelial LTßR is required for the accumulation of hematopoietic cells and full LN maturation, LTßR in VE-cadherin+Tekcre-low/neg ECs in embryos might represent a critical portal-determining factor for LN formation.


Assuntos
Células Endoteliais/metabolismo , Linfonodos/embriologia , Linfonodos/crescimento & desenvolvimento , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Receptor beta de Linfotoxina/metabolismo , Receptor TIE-2/metabolismo , Animais , Linhagem Celular , Movimento Celular/fisiologia , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organogênese/fisiologia , Transdução de Sinais , Migração Transendotelial e Transepitelial/fisiologia
9.
J Clin Invest ; 127(4): 1574-1588, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28319044

RESUMO

Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Linfonodos/patologia , Receptores Notch/fisiologia , Baço/patologia , Linfócitos T/imunologia , Aloenxertos , Animais , Transplante de Medula Óssea , Proteínas de Ligação ao Cálcio , Células Cultivadas , Feminino , Fibroblastos/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/patologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Ligantes , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Baço/imunologia , Linfócitos T/metabolismo
10.
Sci Rep ; 7: 44481, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28290551

RESUMO

The establishment of T cell central tolerance critically relies on the development and maintenance of the medullary thymic epithelial cells (mTECs). Disrupted signaling of lymphotoxin beta receptor (LTßR) results in dramatically reduced mTEC population. However, whether LTßR directly or indirectly control mTECs remains undetermined; how LTßR controls this process also remain unclear. In this study, by utilizing K14-Cre × Ltbrfl/fl conditional knockout (cKO) mice, we show that epithelial intrinsic LTßR was essential for the mTEC development postnatally. Mechanistically, LTßR did not directly impact the proliferation or survival of mTECs; the maturation of mTECs from MHC-IIlo to MHC-IIhi stage was also unaltered in the absence of LTßR; interestingly, the number of mTEC progenitors (Cld3,4hiSSEA-1+) was found significantly reduced in LTßR cKO mice at the neonatal stage, but not at E18.5. Consequently, epithelial deficiency of LTßR resulted in significant defect of thymic negative selection as demonstrated using OT-I and RIP-OVA transgenic mouse system. In summary, our study clarifies the epithelial intrinsic role of LTßR on mTEC development and function; more importantly, it reveals a previously unrecognized function of LTßR on the control of the size of mTEC progenitor population.


Assuntos
Células Epiteliais/citologia , Receptor beta de Linfotoxina/genética , Células-Tronco/metabolismo , Timo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos/genética , Linhagem da Célula/genética , Proliferação de Células/genética , Células Epiteliais/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Células-Tronco/citologia , Linfócitos T/metabolismo , Timo/metabolismo
11.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27798617

RESUMO

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Coronavirus/imunologia , Infecções por Enterobacteriaceae/imunologia , Fibroblastos/imunologia , Interleucina-15/metabolismo , Linfócitos/imunologia , Vírus da Hepatite Murina/imunologia , Animais , Células Cultivadas , Imunidade Inata , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Nódulos Linfáticos Agregados/patologia , Células Th1/imunologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo
12.
Trials ; 17(1): 512, 2016 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-27769284

RESUMO

BACKGROUND: Although Traditional Chinese Medicine (TCM) has been widely used in clinical settings, a major challenge that remains in TCM is to evaluate its efficacy scientifically. This randomized controlled trial aims to evaluate the efficacy and safety of berberine in the treatment of patients with polycystic ovary syndrome. In order to improve the transparency and research quality of this clinical trial, we prepared this statistical analysis plan (SAP). METHODS: The trial design, primary and secondary outcomes, and safety outcomes were declared to reduce selection biases in data analysis and result reporting. We specified detailed methods for data management and statistical analyses. Statistics in corresponding tables, listings, and graphs were outlined. DISCUSSION: The SAP provided more detailed information than trial protocol on data management and statistical analysis methods. Any post hoc analyses could be identified via referring to this SAP, and the possible selection bias and performance bias will be reduced in the trial. TRIAL REGISTRATION: This study is registered at ClinicalTrials.gov, NCT01138930 , registered on 7 June 2010.


Assuntos
Berberina/uso terapêutico , Interpretação Estatística de Dados , Medicamentos de Ervas Chinesas/uso terapêutico , Resistência à Insulina , Síndrome do Ovário Policístico/tratamento farmacológico , Adolescente , Adulto , Berberina/efeitos adversos , Protocolos Clínicos , Medicamentos de Ervas Chinesas/efeitos adversos , Feminino , Humanos , Modelos Estatísticos , Síndrome do Ovário Policístico/diagnóstico , Projetos de Pesquisa , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
13.
Nat Commun ; 7: 12369, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27493002

RESUMO

Continuous thymic homing of haematopoietic progenitor cells (HPCs) via the blood is critical for normal T-cell development. However, the nature and the differentiation programme of specialized thymic endothelial cells (ECs) controlling this process remain poorly understood. Here using conditional gene-deficient mice, we find that lymphotoxin beta receptor (LTßR) directly controls thymic ECs to guide HPC homing. Interestingly, T-cell deficiency or conditional ablation of T-cell-engaged LTßR signalling results in a defect in thymic HPC homing, suggesting the feedback regulation of thymic progenitor homing by thymic products. Furthermore, we identify and characterize a special thymic portal EC population with features that guide HPC homing. LTßR is essential for the differentiation and homeostasis of these thymic portal ECs. Finally, we show that LTßR is required for T-cell regeneration on irradiation-induced thymic injury. Together, these results uncover a cellular and molecular pathway that governs thymic EC differentiation for HPC homing.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia , Animais , Homeostase , Camundongos Endogâmicos C57BL , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
14.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-25973789

RESUMO

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Glicoproteínas de Membrana/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/citologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , Transdução de Sinais/genética , Células-Tronco/citologia , Timo/citologia
15.
J Exp Med ; 211(11): 2265-79, 2014 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-25311507

RESUMO

Fibroblast-like cells of secondary lymphoid organs (SLO) are important for tissue architecture. In addition, they regulate lymphocyte compartmentalization through the secretion of chemokines, and participate in the orchestration of appropriate cell-cell interactions required for adaptive immunity. Here, we provide data demonstrating the functional importance of SLO fibroblasts during Notch-mediated lineage specification and immune response. Genetic ablation of the Notch ligand Delta-like (DL)1 identified splenic fibroblasts rather than hematopoietic or endothelial cells as niche cells, allowing Notch 2-driven differentiation of marginal zone B cells and of Esam(+) dendritic cells. Moreover, conditional inactivation of DL4 in lymph node fibroblasts resulted in impaired follicular helper T cell differentiation and, consequently, in reduced numbers of germinal center B cells and absence of high-affinity antibodies. Our data demonstrate previously unknown roles for DL ligand-expressing fibroblasts in SLO niches as drivers of multiple Notch-mediated immune differentiation processes.


Assuntos
Microambiente Celular/imunologia , Fibroblastos/metabolismo , Imunidade , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores/metabolismo , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Quimiocina CCL19/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imunofenotipagem , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Baço/imunologia , Baço/metabolismo , Células Estromais/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
16.
PLoS One ; 9(2): e89143, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551234

RESUMO

AcrB is a multidrug transporter in the inner membrane of Escherichia coli. It is an obligate homotrimer and forms a tripartite efflux complex with AcrA and TolC. AcrB is the engine of the efflux machinery and determines substrate specificity. Active efflux depends on several functional features including proton translocation across the inner membrane through a proton relay pathway in the transmembrane domain of AcrB; substrate binding and migration through the substrate translocation pathway; the interaction of AcrB with AcrA and TolC; and the formation of AcrB homotrimer. Here we investigated two aspects of the inter-correlation between these functional features, the dependence of AcrA-AcrB interaction on AcrB trimerization, and the reliance of substrate binding and penetration on protein-protein interaction. Interaction between AcrA and AcrB was investigated through chemical crosslinking, and a previously established in vivo fluorescent labeling method was used to probe substrate binding. Our data suggested that dissociation of the AcrB trimer drastically decreased its interaction with AcrA. In addition, while substrate binding with AcrB seemed to be irrelevant to the presence or absence of AcrA and TolC, the capability of trimerization and conduction of proton influx did affect substrate binding at selected sites along the substrate translocation pathway in AcrB.


Assuntos
Proteínas de Escherichia coli/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Multimerização Proteica , Aminoácidos/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , Cisteína/genética , Proteínas de Escherichia coli/química , Técnicas de Inativação de Genes , Proteínas de Membrana Transportadoras/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Proteínas Associadas à Resistência a Múltiplos Medicamentos/química , Proteínas Mutantes/metabolismo , Mutação/genética , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Especificidade por Substrato/efeitos dos fármacos
17.
Immunity ; 38(5): 1013-24, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23623380

RESUMO

The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-ß receptor (LTßR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTßR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTßR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection.


Assuntos
Infecções por Coronavirus/imunologia , Linfonodos/citologia , Linfonodos/metabolismo , Miofibroblastos/fisiologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/imunologia , Interleucina-7/biossíntese , Linfonodos/imunologia , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-beta/biossíntese , Linfotoxina-beta/metabolismo , Glicoproteínas de Membrana/biossíntese , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vírus da Hepatite Murina/imunologia , Miofibroblastos/citologia , Transdução de Sinais
18.
J Exp Med ; 210(3): 465-73, 2013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-23420877

RESUMO

The development of lymph nodes (LNs) and formation of LN stromal cell microenvironments is dependent on lymphotoxin-ß receptor (LTßR) signaling. In particular, the LTßR-dependent crosstalk between mesenchymal lymphoid tissue organizer and hematopoietic lymphoid tissue inducer cells has been regarded as critical for these processes. Here, we assessed whether endothelial cell (EC)-restricted LTßR signaling impacts on LN development and the vascular LN microenvironment. Using EC-specific ablation of LTßR in mice, we found that conditionally LTßR-deficient animals failed to develop a significant proportion of their peripheral LNs. However, remnant LNs showed impaired formation of high endothelial venules (HEVs). Venules had lost their cuboidal shape, showed reduced segment length and branching points, and reduced adhesion molecule and constitutive chemokine expression. Due to the altered EC-lymphocyte interaction, homing of lymphocytes to peripheral LNs was significantly impaired. Thus, this study identifies ECs as an important LTßR-dependent lymphoid tissue organizer cell population and indicates that continuous triggering of the LTßR on LN ECs is critical for lymphocyte homeostasis.


Assuntos
Células Endoteliais/fisiologia , Linfonodos/fisiologia , Receptor beta de Linfotoxina/fisiologia , Transdução de Sinais/fisiologia , Vênulas/fisiologia , Animais , Caderinas/fisiologia , Homeostase , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA