Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Ther ; 32(6): 1875-1894, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38549378

RESUMO

Chimeric antigen receptor-T (CAR-T) cell has been developed as a promising agent for patients with refractory or relapsed lymphoma and leukemia, but not all the recipients could achieve a long-lasting remission. The limited capacity of in vivo expansion and memory differentiation post activation is one of the major reasons for suboptimal CAR-T therapeutic efficiency. Nitric oxide (NO) plays multifaceted roles in mitochondrial dynamics and T cell activation, but its function on CAR-T cell persistence and anti-tumor efficacy remains unknown. Herein, we found the continuous signaling from CAR not only promotes excessive NO production, but also suppressed S-nitrosoglutathione reductase (GSNOR) expression in T cells, which collectively led to increased protein S-nitrosylation, resulting in impaired mitochondrial fitness and deficiency of T cell stemness. Intriguingly, enforced expression of GSNOR promoted memory differentiation of CAR-T cell after immune activation, rendered CAR-T better resistance to mitochondrial dysfunction, further enhanced CAR-T cell expansion and anti-tumor capacity in vitro and in a mouse tumor model. Thus, we revealed a critical role of NO in restricting CAR-T cell persistence and functionality, and defined that GSNOR overexpression may provide a solution to combat NO stress and render patients with more durable protection from CAR-T therapy.


Assuntos
Imunoterapia Adotiva , Mitocôndrias , Receptores de Antígenos Quiméricos , Animais , Camundongos , Mitocôndrias/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Antígenos Quiméricos/genética , Humanos , Imunoterapia Adotiva/métodos , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Linfócitos T/metabolismo , Linfócitos T/imunologia , Óxido Nítrico/metabolismo , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Ativação Linfocitária , Transdução de Sinais
2.
Cell Mol Life Sci ; 80(3): 63, 2023 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-36781473

RESUMO

Adipose tissue CD11c+ myeloid cell is an independent risk factor associated with obesity and metabolic disorders. However, the underlying molecular basis remains elusive. Here, we demonstrated that liver kinase B1 (Lkb1), a key bioenergetic sensor, is involved in CD11c+ cell-mediated immune responses in diet-induced obesity. Loss of Lkb1 in CD11c+ cells results in obesity resistance but lower glucose tolerance, which accompanies tissue-specific immune abnormalities. The accumulation and CD80's expression of Lkb1 deficient adipose-tissue specific dendritic cells but not macrophages is restrained. Additionally, the balance of IL-17A and IFN-γ remarkably tips towards the latter in fat T cells and CD11c- macrophages. Mechanistically, IFN-γ promotes apoptosis of preadipocytes and inhibits their adipogenesis while IL-17A promotes the adipogenesis in vitro, which might account in part for the fat gain resistant phenotype. In summary, these findings reveal that Lkb1 is essential for fat CD11c+ dendritic cells responding to HFD exposure and provides new insights into the IL-17A/IFN-γ balance in HFD-induced obesity.


Assuntos
Proteínas Quinases Ativadas por AMP , Intolerância à Glucose , Resistência à Insulina , Obesidade , Animais , Camundongos , Tecido Adiposo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/metabolismo , Inflamação/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/complicações , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Interferon gama/metabolismo
3.
Lancet Oncol ; 24(11): 1229-1241, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37863088

RESUMO

BACKGROUND: Relapses frequently occur following CD19-directed chimeric antigen receptor (CAR) T-cell treatment for relapsed or refractory B-cell acute lymphocytic leukaemia in children. We aimed to assess the activity and safety of sequential CD19-directed and CD22-directed CAR T-cell treatments. METHODS: This single-centre, single-arm, phase 2 trial, done at Beijing GoBroad Boren Hospital, Beijing, China, included patients aged 1-18 years who had relapsed or refractory B-cell acute lymphocytic leukaemia with CD19 and CD22 positivity greater than 95% and an Eastern Cooperative Oncology Group performance status of 0-2. Patients were initially infused with CD19-directed CAR T cells intravenously, followed by CD22-directed CAR T-cell infusion after minimal residual disease-negative complete remission (or complete remission with incomplete haematological recovery) was reached and all adverse events (except haematological adverse events) were grade 2 or better. The target dose for each infusion was 0·5 × 106 to 5·0 × 106 cells per kg. The primary endpoint was objective response rate at 3 months after the first infusion. Secondary endpoints were duration of remission, event-free survival, disease-free survival, overall survival, safety, pharmacokinetics, and B-cell quantification. The prespecified activity analysis included patients who received the target dose and the safety analysis included all treated patients. This study is registered with ClinicalTrials.gov, NCT04340154, and enrolment has ended. FINDINGS: Between May 28, 2020, and Aug 16, 2022, 81 participants were enrolled, of whom 31 (38%) were female and 50 (62%) were male. Median age was 8 years (IQR 6-10), all patients were Asian. All 81 patients received the first infusion and 79 (98%) patients received sequential infusions, CD19-directed CAR T cells at a median dose of 2·7 × 106 per kg (IQR 1·1 × 106 to 3·7 × 106) and CD22-directed CAR T cells at a median dose of 2·2 × 106 per kg (1·1 × 106 to 3·7 × 106), with a median interval of 39 days (37-41) between the two infusions. 62 (77%) patients received the target dose, including two patients who did not receive CD22 CAR T cells. At 3 months, 60 (97%, 95% CI 89-100) of the 62 patients who received the target dose had an objective response. Median follow-up was 17·7 months (IQR 11·4-20·9). 18-month event-free survival for patients who received the target dose was 79% (95% CI 66-91), duration of remission was 80% (68-92), and disease-free survival was 80% (68-92) with transplantation censoring; overall survival was 96% (91-100). Common adverse events of grade 3 or 4 between CD19-directed CAR T-cell infusion and 30 days after CD22-directed CAR T-cell infusion included cytopenias (64 [79%] of 81 patients), cytokine release syndrome (15 [19%]), neurotoxicity (four [5%]), and infections (five [6%]). Non-haematological adverse events of grade 3 or worse more than 30 days after CD22-directed CAR T-cell infusion occurred in six (8%) of 79 patients. No treatment-related deaths occurred. CAR T-cell expansion was observed in all patients, with a median peak at 9 days (IQR 7-14) after CD19-directed and 12 days (10-15) after CD22-directed CAR T-cell infusion. At data cutoff, 35 (45%) of 77 evaluable patients had CAR transgenes and 59 (77%) had B-cell aplasia. INTERPRETATION: This sequential strategy induced deep and sustained responses with an acceptable toxicity profile, and thus potentially provides long-term benefits for children with this condition. FUNDING: The National Key Research & Development Program of China, the CAMS Innovation Fund for Medical Sciences (CIFMS), and the Non-Profit Central Research Institute Fund of Chinese Academy of Medical Sciences. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Assuntos
Leucemia Linfocítica Crônica de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Humanos , Masculino , Criança , Feminino , Receptores de Antígenos Quiméricos/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Imunoterapia Adotiva/efeitos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Terapia Baseada em Transplante de Células e Tecidos , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/uso terapêutico
4.
Exp Cell Res ; 392(2): 112003, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32278689

RESUMO

Dendritic cells (DCs) play a central role in autoimmunity, immune homeostasis, and presentation of tumor antigens to T cells in order to prime antitumor responses. The number of tumor-infiltrating DCs is associated with survival and prognosis in cancer. Twist1 is a well-known regulator of tumor initiation and promotion, but whether and how DC-derived Twist1 regulates antitumor responses remains poorly understood. Here, we generated a mouse line with Twist1 conditionally depleted in DCs and found that Twist1-deficiency in DCs did not affect the DCs and T cell homeostasis under steady-state conditions; however, in melanoma models, the proportion of conventional DCs (cDCs) in draining lymph nodes (DLNs) was significantly decreased. Accordingly, a decreased ratio and number of tumor-infiltrating cDCs were observed, which reduced the recruitment of tumor-infiltrating T cells. Furthermore, production of IFN-γ, a crucial antitumor factor, by T cells, was dramatically decreased, which can further dampen the T cell antitumor functions. Collectively, our data indicate that Twist1 in DCs regulates antitumor functions by maintain the number of tumor-infiltrating DCs and T cells, and their antitumor activity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Imunidade Celular/imunologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Antígenos de Neoplasias/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout
5.
Exp Cell Res ; 363(1): 73-83, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29294307

RESUMO

Dendritic cells (DCs) are pivotal to initiating adaptive immune response. Emerging evidence highlights important roles of tuberous sclerosis complex 1 (Tsc1) in DC development and activation. Our previous study also showed that Tsc1 expression in DCs was required to promote T-cell homeostasis and response partially through inhibiting mammalian target of rapamycin complex1 (mTORC1). However, the molecular mechanism of transcriptional regulation by which Tsc1 control DC homeostasis and function remains largely unknown. Here we globally identified the Tsc1-regulated genes by comparing the transcriptional profiling of Tsc1-deficient DCs with wild-type DCs. It showed that Tsc1 specifically regulated the expression of groups of gene sets critically involved in DC survival, proliferation, metabolism and antigen presentation. The impacts of Tsc1 on DC gene expression were partially dependent on inhibition of mTORC1 signal. Our study thus provides a comprehensive molecular basis for understanding how Tsc1 programs the homeostasis and function of DCs through transcriptional regulation.


Assuntos
Células Dendríticas/citologia , Homeostase/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo , Animais , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Camundongos Transgênicos , Complexos Multiproteicos/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
6.
J Craniofac Surg ; 28(6): 1615-1619, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28863113

RESUMO

The authors aim to track the distribution of human umbilical cord mesenchymal stem cells (MSCs) in large blood vessel of traumatic brain injury -rats through immunohistochemical method and small animal imaging system. After green fluorescent protein (GFP) gene was transfected into 293T cell, virus was packaged and MSCs were transfected. Mesenchymal stem cells containing GFP were transplanted into brain ventricle of rats when the infection rate reaches 95%. The immunohistochemical and small animal imaging system was used to detect the distribution of MSCs in large blood vessels of rats. Mesenchymal stem cells could be observed in large vessels with positive GFP expression 10 days after transplantation, while control groups (normal group and traumatic brain injury group) have negative GFP expression. The vascular endothelial growth factor in transplantation group was higher than that in control groups. The in vivo imaging showed obvious distribution of MSCs in the blood vessels of rats, while no MSCs could be seen in control groups. The intravascular migration and homing of MSCs could be seen in rats received MSCs transplantation, and new angiogenesis could be seen in MSCs-transplanted blood vessels.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Cordão Umbilical/citologia , Animais , Vasos Sanguíneos/química , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Ratos , Projetos de Pesquisa
7.
Cell Physiol Biochem ; 36(4): 1406-18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26159807

RESUMO

BACKGROUND/AIMS: Tuberous sclerosis complex 1 (Tsc1) has been shown to regulate M1/M2 polarization of macrophages, but the precise roles of Tsc1 in the function and stability of macrophages are not fully understood. Here we show that Tsc1 is required for regulating the survival, migration and phagocytosis of macrophages. METHODS: Mice with Tsc1 homozygous deletion in myeloid cells (LysMCreTsc1(flox/flox); Tsc1 KO) were obtained by crossing Tsc1(flox/flox) mice with mice expressing Cre recombinase under the control of Lysozyme promoter (LysMCre). The apoptosis and growth of macrophages were determined by flow cytometry and Real-time PCR (RT-PCR). The phagocytosis was determined using a Vybrant™ phagocytosis assay kit. The migration of macrophages was determined using transwell migration assay. RESULTS: Peritoneal macrophages of Tsc1 KO mice exhibited increased apoptosis and enlarged cell size. Both M1 and M2 phenotypes in Tsc1-deficient macrophages were elevated in steady-state as well as in inflammatory conditions. Tsc1-deficient macrophages demonstrated impaired migration and reduced expression of chemokine receptors including CCR2 and CCR5. Phagocytosis activity and ROS production were enhanced in Tsc1-deficient macrophages. Furthermore, pharmacological inhibition of the mammalian target of rapamycin complex 1 (mTORC1) partially reversed the aberrance of Tsc1-deficient macrophages. CONCLUSION: Tsc1 plays a critical role in regulating macrophage survival, function and polarization via inhibition of mTORC1 activity.


Assuntos
Deleção de Genes , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Quimiocinas/genética , Feminino , Regulação da Expressão Gênica , Ativação Linfocitária , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Células Mieloides/citologia , Células Mieloides/metabolismo , Fagocitose , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa
8.
Acta Biochim Biophys Sin (Shanghai) ; 47(8): 597-603, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26094142

RESUMO

Interleukin 37b (IL-37b) plays a key role in suppressing immune responses, partially by modulating the function of dendritic cells (DCs). However, the precise mechanisms are still largely unknown. Here, we investigated the effects of IL-37b on DC maturation and T cell responses induced by DCs, and explored the involved signaling pathways. It was found that IL-37b down-regulated the expressions of co-stimulatory molecules CD80 and CD86 on DCs in vitro. At the same time, the expressions of pro-inflammatory cytokines, such as TNF-α and IL-6, were suppressed, while the expression of the T cell inhibitory cytokine TGF-ß was increased in IL-37b-treated DCs. In addition, the activation effect of DCs on T cells was impaired by IL-37b. We further revealed that extracellular single-regulated kinase (ERK), nuclear factor-κB (NF-κB), and mTOR-S6K signaling pathways were involved in the inhibition of DCs induced by IL-37b. This was confirmed by the similarly suppressive effect of chemical inhibitors against NF-κB, ERK, and S6K on the expressions of IL-6 and TNF-α in DCs. In conclusion, these results demonstrated that IL-37b suppressed DC maturation and immunostimulatory capacity in T cell priming by involving in ERK, NF-κB, and S6K-based inhibitory signaling pathways.


Assuntos
Apresentação Cruzada , Citocinas/biossíntese , Células Dendríticas/metabolismo , NF-kappa B/fisiologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Células Dendríticas/imunologia , Interleucinas/genética , Interleucinas/imunologia , Interleucinas/farmacologia , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , NF-kappa B/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/imunologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fatores Supressores Imunológicos/imunologia
9.
Hepatology ; 57(2): 667-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22996586

RESUMO

UNLABELLED: MicroRNAs (miRNAs) have been reported to be associated with the development of cancers. However, the function of miRNAs in human hepatocellular carcinoma (HCC) remains largely undefined. Here we found that overexpression of miR-10a promoted the migration and invasion of QGY-7703 and HepG2 cells in vitro but suppressed metastasis in vivo. Cell adhesion assays showed that miR-10a suppressed HCC cell-matrix adhesion, which could explain the results of the in vivo animal experiments. The Eph tyrosine kinase receptor, EphA4, was identified as the direct and functional target gene of miR-10a. Knockdown of EphA4 phenocopied the effect of miR-10a and ectopic expression of EphA4 restored the effect of miR-10a on migration, invasion, and adhesion in HCC cells. We further demonstrated that miR-10a and EphA4 regulated the epithelial-mesenchymal transition process and the ß1-integrin pathway to affect cell invasion and adhesion. CONCLUSION: Our findings highlight the importance of miR-10a in regulating the metastatic properties of HCC by directly targeting EphA4 and may provide new insights into the pathogenesis of HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/secundário , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Hepáticas/patologia , MicroRNAs/fisiologia , Metástase Neoplásica/fisiopatologia , Receptor EphA4/metabolismo , Linhagem Celular Tumoral , Junções Célula-Matriz/metabolismo , Feminino , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Invasividade Neoplásica , Receptor EphA4/biossíntese
11.
Acta Biochim Biophys Sin (Shanghai) ; 45(7): 561-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23676772

RESUMO

The binding of the stromal cell-derived factor-1α (SDF-1α) to the cysteine (C)-X-C motif chemokine receptor 4 (CXCR4) has emerged as a key signal for stem and progenitor cells trafficking to the circulation from the bone marrow. Our aim was to investigate the role of daily intermittent administration of AMD3100 (a specific reversible CXCR4 receptor antagonist) during the healing process after myocardial infarction (MI). Wistar rats were subjected to MI and AMD3100 was injected intraperitoneally after surgery. SDF-1α mRNA expression was measured by real-time polymerase chain reaction. Histology changes were analyzed with immunofluorescence, Masson's trichrome staining, and wheat germ agglutinin. The number of leukocytes in peripheral blood was measured by complete blood cell count analysis. The activities of matrix metalloproteinase-2/9 (MMP-2/9) were determined by gelatin zymography. The expression level of SDF-1α mRNA in the infarcted tissue was enhanced rapidly (6 h), peaked at 24 h, and then declined to the normal level at 7 days post-MI. AMD3100 further enhanced the increase of SDF-1α in infarct area. Increased leukocytes were observed in AMD3100-treated groups. The mobilization of c-kit(+) stem/progenitor cells and enhanced neovascularization were augmented by AMD3100. Additionally, AMD3100 improved ventricular remodeling, which was revealed by the decrease of infarct size, viable cardiomyocyte cross-sectional area and left ventricle (LV) expansion index, and the increase of LV free wall thickness. The activities of MMP-2/9 were up-regulated by AMD3100. In conclusion, short-term intermittent administration of AMD3100 could accelerate the wound healing process in experimental MI and be a potential therapy for the treatment of MI.


Assuntos
Compostos Heterocíclicos/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Animais , Sequência de Bases , Benzilaminas , Ciclamos , Primers do DNA , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos/uso terapêutico , Masculino , Infarto do Miocárdio/patologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 23(12): 731-3, 2011 Dec.
Artigo em Zh | MEDLINE | ID: mdl-22153010

RESUMO

OBJECTIVE: To examine the level of endothelin-1 (ET-1) in serum and its expression in myocardium tissue during the development of acute myocardial ischemia/reperfusion (I/R) injury in rat and the effects of L-arginine (L-Arg) administration on these indexes. METHODS: One hundred and ten Wistar rats were randomly divided into nine groups to receive: (1) sham surgery, (2)ischemia (I, by ligation of anterior descending coronary artery for 30 minutes), (3) I+reperfusion (R, by the removal of the ligature) for 0.5 hour, (4) I+R for 1 hour, (5) I+R for 2 hours; group (6) ~ (9) also received I/R treatment as in group 2 ~ 5 respectively but with L-Arg pretreatment. Blood and myocardium tissue samples were collected by the end of the experiment for the analysis of: serum level of creatine kinase (CK), lactate dehydrogenase [LDH, by enzyme linked immunosorbent assay (ELISA)], ET-1 (radioimmunoassay), and the tissue content of ET-1 mRNA/peptide [by reverse-transcription polymerase chain-reaction (RT-PCR) and Western blotting]. RESULTS: In comparison with the sham treated control animals, the serum levels of CK, LDH, and ET-1 were all significantly higher in the groups treated with I/R (particularly those exposed to reperfusion). The myocardial tissue content of ET-1 mRNA/peptide were also significantly increased in I/R treated groups (particularly the I+R 2 hours group) as compared to control (ET-1 mRNA: 0.775 ± 0.029 vs. 0.310 ± 0.076; ET-1 peptide: 0.773 ± 0.055 vs. 0.340 ± 0.099, both P < 0.05). The i.v. administration of L-Arg significantly suppressed the up-regulation of tissue content of ET-1 mRNA /peptide in I/R treated animals (ET-1 mRNA: 0.340 ± 0.049 vs. 0.775 ± 0.029; ET-1 peptide: 0.390 ± 0.094 vs. 0.773 ± 0.055, both P < 0.05). CONCLUSION: L-Arg may be tested during certain stage of I/R injury as a therapeutic intervention for the suppression of ET-1 up-regulation.


Assuntos
Arginina/farmacologia , Endotelina-1/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Animais , Creatina Quinase/sangue , Ratos , Ratos Wistar
13.
Nat Commun ; 12(1): 3913, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162888

RESUMO

Human FOXP3+ regulatory T (Treg) cells are central to immune tolerance. However, their heterogeneity and differentiation remain incompletely understood. Here we use single-cell RNA and T cell receptor sequencing to resolve Treg cells from healthy individuals and patients with or without acute graft-versus-host disease (aGVHD) who undergo stem cell transplantation. These analyses, combined with functional assays, separate Treg cells into naïve, activated, and effector stages, and resolve the HLA-DRhi, LIMS1hi, highly suppressive FOXP3hi, and highly proliferative MKI67hi effector subsets. Trajectory analysis assembles Treg subsets into two differentiation paths (I/II) with distinctive phenotypic and functional programs, ending with the FOXP3hi and MKI67hi subsets, respectively. Transcription factors FOXP3 and SUB1 contribute to some Path I and Path II phenotypes, respectively. These FOXP3hi and MKI67hi subsets and two differentiation pathways are conserved in transplanted patients, despite having functional and migratory impairments under aGVHD. These findings expand the understanding of Treg cell heterogeneity and differentiation and provide a single-cell atlas for the dissection of Treg complexity in health and disease.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição Forkhead/imunologia , Transdução de Sinais/genética , Análise de Célula Única/métodos , Linfócitos T Reguladores/imunologia , Transcriptoma/genética , Western Blotting , Ensaio de Imunoadsorção Enzimática , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , RNA-Seq/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo
14.
Cell Mol Immunol ; 17(5): 483-495, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31664223

RESUMO

Accumulating evidence suggests that a reduction in the number of Foxp3+ regulatory T cells (Tregs) contributes to the pathogenesis of acute graft-versus-host disease (aGVHD), which is a major adverse complication that can occur after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the precise features and mechanism underlying the defects in Tregs remain largely unknown. In this study, we demonstrated that Tregs were more dramatically decreased in bone marrow compared with those in peripheral blood from aGVHD patients and that bone marrow Treg defects were negatively associated with hematopoietic reconstitution. Tregs from aGVHD patients exhibited multiple defects, including the instability of Foxp3 expression, especially in response to IL-12, impaired suppressor function, decreased migratory capacity, and increased apoptosis. Transcriptional profiling revealed the downregulation of Lkb1, a previously identified critical regulator of murine Treg identity and metabolism, and murine Lkb1-regulated genes in Tregs from aGVHD patients. Foxp3 expression in human Tregs could be decreased and increased by the knockdown and overexpression of the Lkb1 gene, respectively. Furthermore, a loss-of-function assay in an aGVHD murine model confirmed that Lkb1 deficiency could impair Tregs and aggravate disease severity. These findings reveal that Lkb1 downregulation contributes to multiple defects in Tregs in human aGVHD and highlight the Lkb1-related pathways that could serve as therapeutic targets that may potentially be manipulated to mitigate aGVHD.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T Reguladores/imunologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Adolescente , Adulto , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Criança , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Fatores de Transcrição Forkhead/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Doença Enxerto-Hospedeiro/sangue , Humanos , Masculino , Metilação/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Linfócitos T Reguladores/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
15.
Biomed Pharmacother ; 120: 109352, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31586905

RESUMO

Inflammatory monocyte and macrophage subset accumulation during the inflammatory response that drives atherosclerosis can exacerbate the extent of atherosclerosis. It has been demonstrated that voltage-gated sodium channels (VGSCs) can regulate cell bioactivities in monocytes/macrophages. We hypothesized that blockade of mononuclear phagocyte VGSCs was atheroprotective through monocyte/macrophage subset modulation and macrophage proliferation suppression in atherosclerotic lesions. In this experimental study, when VGSCs were knocked down with RNA interference plasmid transfection in mouse peripheral blood monocytes and monocyte-macrophage lineage RAW264.7 cells in vitro, the biological characteristics of proliferation, phagocytosis, and migration in RAW264.7 cells declined. In addition, suppression of LPS-induced M1 polarization and facilitation of IL-4-induced M2 polarization were also observed. In an in vivo study, ApoE knockout (ApoE-/-) mice were fed a standard chow diet (CD) or a western diet (WD). After feeding with phenytoin (PHT), no significant differences were detected in plasma lipids, and the anti-inflammatory phenotypes of both monocytes and macrophages were elevated and proinflammatory phenotypes declined. The local proliferation of macrophages was also distinctly suppressed, along with a significant reduction in atheromatous plaques. In conclusion, blockade of VGSCs in the mononuclear phagocyte system reduced atherosclerotic lesions, which may occur through altering monocyte/macrophage subsets and suppressing macrophage proliferation in atherosclerotic plaques. Blockage of VGSCs may play an important role in cardiovascular protection.


Assuntos
Aterosclerose/prevenção & controle , Ativação de Macrófagos , Macrófagos/metabolismo , Monócitos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Proliferação de Células , Modelos Animais de Doenças , Regulação para Baixo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Monócitos/patologia , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Fagocitose , Placa Aterosclerótica , Células RAW 264.7 , Interferência de RNA , Transdução de Sinais
16.
Am J Transl Res ; 10(3): 901-906, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636880

RESUMO

This study reports a case of a 4-year-old boy patient with abnormalities of muscle tone, movement and motor skills, as well as unstable gait leading to frequent falls. The results of the electroencephalogram (EEG) indicate moderately abnormal EEG, accompanied by irregular seizures. Based on these clinical characteristics, the patient was diagnosed with cerebral palsy (CP) in our hospital. In this study, the patient was treated with umbilical cord mesenchymal stem cell (UC-MSC) transplantation therapy. This patient received UC-MSC transplantation 3 times (5.3*107) in total. After three successive cell transplantations, the patient recovered well and showed obvious improvements in EEG and limb strength, motor function, and language expression. However, the improvement in intelligence quotient (IQ) was less obvious. These results indicate that UC-MSC transplantation is a promising treatment for cerebral palsy.

17.
Nat Commun ; 9(1): 5298, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30546010

RESUMO

To balance immunity and tolerance, the endogenous pool of Foxp3+ regulatory T (Treg) cells is tightly controlled, but the underlying mechanisms of this control remain poorly understood. Here we show that the number of Treg cells is negatively regulated by the kinase Lkb1 in dendritic cells (DCs). Conditional knockout of the Lkb1 gene in DCs leads to excessive Treg cell expansion in multiple organs and dampens antigen-specific T cell immunity. Lkb1-deficient DCs are capable of enhancing, compared with wild-type DCs, Treg cell proliferation via cell-cell contact involving the IKK/IKBα-independent activation of the NF-κB/OX40L pathway. Intriguingly, treating wild-type mice with lipopolysaccharide selectively depletes Lkb1 protein in DCs, resulting in Treg cell expansion and suppressed inflammatory injury upon subsequent challenge. Loss of Lkb1 does not obviously upregulate proinflammatory molecules expression on DCs. We thus identify Lkb1 as a regulatory switch in DCs for controlling Treg cell homeostasis, immune response and tolerance.


Assuntos
Proliferação de Células/genética , Células Dendríticas/imunologia , Proteínas Serina-Treonina Quinases/genética , Linfócitos T Reguladores/imunologia , Proteínas Quinases Ativadas por AMP , Animais , Apoptose/imunologia , Técnicas de Inativação de Genes , Homeostase/fisiologia , Quinase I-kappa B/metabolismo , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Ligante OX40 , Receptores OX40/metabolismo , Fatores de Necrose Tumoral/metabolismo
18.
Cell Death Dis ; 8(1): e2553, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28079897

RESUMO

Dendritic cells (DCs) are pivotal to the induction of adaptive T-cell immune responses. Recent evidence highlights a critical role of tuberous sclerosis complex 1 (Tsc1), a primarily upstream negative regulator of mammalian target of rapamycin (mTOR), in DC development, but whether and how Tsc1 directly regulate mature DC function in vivo remains elusive. Here we show that selective disruption of Tsc1 in DCs results in a lymphoproliferative disorder with the spontaneous activation of T cells. Tsc1 deficiency results in the activation of mTORC1-PPARγ pathway, which leads to the upregulation of neuropilin-1 (Nrp1) expression on DCs to stimulate naive T-cell proliferation. However, Tsc1-deficient DCs have defects in the ability to induce antigen-specific T-cell responses in vitro and in vivo owing to impaired survival during antigen transportation and presentation. Indeed, Tsc1 promotes DC survival through restraining independent mTORC1 and ROS-Bim pathways. Our study identifies Tsc1 as a crucial signaling checkpoint in DCs essential for preserving T-cell homeostasis and response.


Assuntos
Células Dendríticas/metabolismo , Transtornos Linfoproliferativos/genética , Linfócitos T/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Animais , Diferenciação Celular/genética , Células Dendríticas/imunologia , Células Dendríticas/patologia , Homeostase , Humanos , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/genética , Transdução de Sinais/genética , Linfócitos T/imunologia , Linfócitos T/patologia , Serina-Treonina Quinases TOR/biossíntese , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
19.
Nat Commun ; 8: 15876, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28621313

RESUMO

Regulatory T (Treg) cells are a distinct T-cell lineage characterized by sustained Foxp3 expression and potent suppressor function, but the upstream dominant factors that preserve Treg lineage-specific features are mostly unknown. Here, we show that Lkb1 maintains Treg cell lineage identity by stabilizing Foxp3 expression and enforcing suppressor function. Upon T-cell receptor (TCR) stimulation Lkb1 protein expression is upregulated in Treg cells but not in conventional T cells. Mice with Treg cell-specific deletion of Lkb1 develop a fatal early-onset autoimmune disease, with no Foxp3 expression in most Treg cells. Lkb1 stabilizes Foxp3 expression by preventing STAT4-mediated methylation of the conserved noncoding sequence 2 (CNS2) in the Foxp3 locus. Independent of maintaining Foxp3 expression, Lkb1 programs the expression of a wide spectrum of immunosuppressive genes, through mechanisms involving the augmentation of TGF-ß signalling. These findings identify a critical function of Lkb1 in maintaining Treg cell lineage identity.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T Reguladores/citologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Doenças Autoimunes/genética , Linhagem da Célula , Metilação de DNA , Feminino , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Transcrição STAT4/genética , Fator de Transcrição STAT4/metabolismo , Linfócitos T Reguladores/fisiologia
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(3): 289-93, 2016 Mar.
Artigo em Zh | MEDLINE | ID: mdl-26927543

RESUMO

OBJECTIVE: To investigate the effect of blocking p38 mitogen-activated protein kinase (p38MAPK) pathway on the proliferation of OT-II cells mediated by splenic dendritic cells (DCs). METHODS: Splenic DCs of C57BL/6 mice were purified with anti-CD11c immunomagnetic beads, and OT-II cells were isolated from the splenic of CD4(+)-ovalbumin transgenic mice (OT-IItransgenic mice) by mouse CD4 T cell isolation kits. After being pretreated with SB203580, an inhibitor of p38MAPK, DCs were stimulated with lipopolysaccharides (LPS). Then the expression levels of co-stimulatory molecules (CD80, CD86) and MHCII in DCs, and antigen-presenting ability of DCs treated with the 52-68 fragment of the E alpha-chain of I-E class II molecules (Eα52-68 peptide) were detected by flow cytometry. The protein levels of tumor necrosis factor α (TNF-α), interleukin 1α (IL-1α), interleukin 6 (IL-6) and transforming growth factor ß (TGF-ß) in the culture supernatants were measured by ELISA. The proliferation of OT-II cells which were co-cultured with OVA323-339-treated DCs was analyzed by flow cytometry. RESULTS: The purity of both DCs and OT-II cells reached over 90% after isolation. SB203580 downregulated the expressions of CD80, CD86 and MHC II, and suppressed the antigen-presenting ability of DCs. The expressions of TNF-α, IL-1α and IL-6 were downregulated, while the expression of TGF-ß was raised. Finally, SB203580 inhibited DCs-mediated proliferation of OT-II cells. CONCLUSION: Blocking p38MAPK pathway with SB203580 could inhibit DCs-mediated proliferation of OT-II cells, which might be involved in modulating the expressions of CD80, CD86 and MHC II, the antigen-presenting ability, as well as the expressions of pro-inflammatory and anti-inflammatory cytokines.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células/fisiologia , Células Dendríticas/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/imunologia , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Imidazóis/farmacologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/farmacologia , Piridinas/farmacologia , Receptores de Antígenos de Linfócitos T , Baço/citologia , Baço/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA