Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 112022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35511221

RESUMO

Thymic homing of hematopoietic progenitor cells (HPCs) is tightly regulated for proper T cell development. Previously we have identified a subset of specialized thymic portal endothelial cells (TPECs), which is important for thymic HPC homing. However, the underlying molecular mechanism still remains unknown. Here, we found that signal regulatory protein alpha (SIRPα) is preferentially expressed on TPECs. Disruption of CD47-SIRPα signaling in mice resulted in reduced number of thymic early T cell progenitors (ETPs), impaired thymic HPC homing, and altered early development of thymocytes. Mechanistically, Sirpa-deficient ECs and Cd47-deficient bone marrow progenitor cells or T lymphocytes demonstrated impaired transendothelial migration (TEM). Specifically, SIRPα intracellular ITIM motif-initiated downstream signaling in ECs was found to be required for TEM in an SHP2- and Src-dependent manner. Furthermore, CD47 signaling from migrating cells and SIRPα intracellular signaling were found to be required for VE-cadherin endocytosis in ECs. Thus, our study reveals a novel role of endothelial SIRPα signaling for thymic HPC homing for T cell development.


Assuntos
Antígeno CD47 , Células Endoteliais , Animais , Antígenos CD , Antígeno CD47/genética , Caderinas , Endocitose , Células Endoteliais/metabolismo , Camundongos , Receptores Imunológicos , Timócitos/metabolismo
2.
Mol Biol Rep ; 49(3): 2531-2542, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35031926

RESUMO

Non-small cell lung cancer (NSCLC) poses a serious threat to public health due to its significant morbidity and mortality rates. The processes of NSCLC formation and development are quite complex and involve numerous regulatory biomolecules. Long non-coding RNAs (lncRNAs) have attracted attention since they have been found to play critical roles in the tumorigenesis of various human malignancies. Recently, double homeobox A pseudogene 8 (DUXAP8) was identified as an oncogenic lncRNA that is overexpressed in different tumor types. In NSCLC, high expression of DUXAP8 is associated with poor prognosis in patients. The regulatory mechanism underlying the oncogenic effects of DUXAP8 can be divided into transcriptional level and post-transcriptional level. DUXAP8 promotes proliferation, epithelial-mesenchymal transition, and aerobic glycolysis in NSCLC cells. Moreover, DUXAP8 shows potential for the diagnosis and treatment of NSCLC. Herein, we review the molecular mechanisms underlying the DUXAP8-mediated phenotypes of NSCLC as well as its potential clinical applications.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes Homeobox , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Pseudogenes , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
3.
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
4.
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
5.
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
6.
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
7.
J Immunol ; 186(12): 7156-63, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21572030

RESUMO

Lymph node (LN) hypertrophy, the increased cellularity of LNs, is the major indication of the initiation and expansion of the immune response against infection, vaccination, cancer, or autoimmunity. The mechanisms underlying LN hypertrophy remain poorly defined. In this article, we demonstrate that LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by lymphocytes) (TNFSF14) is a novel factor essential for LN hypertrophy after CFA immunization. Mechanistically, LIGHT is required for the influx of lymphocytes into but not egress out of LNs. In addition, LIGHT is required for dendritic cell migration from the skin to draining LNs. Compared with wild type mice, LIGHT(-)(/)(-) mice express lower levels of chemokines in skin and addressins in LN vascular endothelial cells after CFA immunization. We unexpectedly observed that LIGHT from radioresistant rather than radiosensitive cells, likely Langerhans cells, is required for LN hypertrophy. Importantly, Ag-specific T cell responses were impaired in draining LNs of LIGHT(-)(/)(-) mice, suggesting the importance of LIGHT regulation of LN hypertrophy in the generation of an adaptive immune response. Collectively, our data reveal a novel cellular and molecular mechanism for the regulation of LN hypertrophy and its potential impact on the generation of an optimal adaptive immune response.


Assuntos
Hipertrofia/patologia , Inflamação/patologia , Linfonodos/patologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Imunidade Adaptativa , Animais , Quimiocinas/metabolismo , Endotélio Vascular/metabolismo , Imunização , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Linfócitos T/patologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 18(1): 85-9, 2010 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-20137124

RESUMO

This study was aimed to construct the shRNA eukaryotic expression vectors of M2-pyruvate kinase gene (pkm2) and to investigate the effects of pkm2 gene interference on the drug resistance of acute promyelocytic leukemia (APL) cells in vitro. Three specific shRNAs of pkm2 gene were designed and cloned into PBSU6 vector containing a U6 promotor. The constructed plasmids were identified and proved by the restriction sequence analysis. Then the effect of pkm2-shRNA on the protein expression of endogenous PKM2 was detected in NB4R2 cells, a drug resistant cell line of APL by Western blot. The alteration of NB4R2 cell differentiation with the interference of pkm2 gene was also validated by nitroblue tetrazolium (NBT) reduction test. The results showed that three specific shRNA eukaryotic expression vectors targeting pkm2 were successfully constructed. The efficiency of pkm2 gene silence was proved at protein level. The interference of pkm2 gene could significantly enhance the cell differentiation in the drug resistant NB4R2 cell line. It is concluded that the DNA vector containing pkm2 targeting shRNA remarkably promotes the differentiation of NB4R2 cells, showing the prospects of developing the gene target drug.


Assuntos
Proteínas de Bactérias/genética , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia Promielocítica Aguda/genética , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Plasmídeos , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA