Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Med ; 21(1): 256, 2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37452335

RESUMO

BACKGROUND: Genome-wide association studies (GWAS) have reported single-nucleotide polymorphisms (SNPs) in the VRK serine/threonine kinase 2 gene (VRK2) showing genome-wide significant associations with major depression, but the regulation effect of the risk SNPs on VRK2 as well as their roles in the illness are yet to be elucidated. METHODS: Based on the summary statistics of major depression GWAS, we conducted population genetic analyses, epigenome bioinformatics analyses, dual luciferase reporter assays, and expression quantitative trait loci (eQTL) analyses to identify the functional SNPs regulating VRK2; we also carried out behavioral assessments, dendritic spine morphological analyses, and phosphorylated 4D-label-free quantitative proteomics analyses in mice with Vrk2 repression. RESULTS: We identified a SNP rs2678907 located in the 5' upstream of VRK2 gene exhibiting large spatial overlap with enhancer regulatory marks in human neural cells and brain tissues. Using luciferase reporter gene assays and eQTL analyses, the depression risk allele of rs2678907 decreased enhancer activities and predicted lower VRK2 mRNA expression, which is consistent with the observations of reduced VRK2 level in the patients with major depression compared with controls. Notably, Vrk2-/- mice exhibited depressive-like behaviors compared to Vrk2+/+ mice and specifically repressing Vrk2 in the ventral hippocampus using adeno-associated virus (AAV) lead to consistent and even stronger depressive-like behaviors in mice. Compared with Vrk2+/+ mice, the density of mushroom and thin spines in the ventral hippocampus was significantly altered in Vrk2-/- mice, which is in line with the phosphoproteomic analyses showing dysregulated synapse-associated proteins and pathways in Vrk2-/- mice. CONCLUSIONS: Vrk2 deficiency mice showed behavioral abnormalities that mimic human depressive phenotypes, which may serve as a useful murine model for studying the pathophysiology of depression.


Assuntos
Estudo de Associação Genômica Ampla , Leucemia Mieloide Aguda , Humanos , Camundongos , Animais , Depressão/genética , Polimorfismo de Nucleotídeo Único , Proteínas Serina-Treonina Quinases/metabolismo
2.
Front Physiol ; 13: 1046497, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36589424

RESUMO

Non-alcoholic Fatty Liver Disease (NAFLD) is a chronic liver disease that is strongly related to insulin resistance and metabolic syndrome, and it has become the most common liver disorder in developed countries. NAFLD embraces the full pathological process of three conditions: steatosis, non-alcoholic steatohepatitis, and finally, cirrhosis. As NAFLD progresses, symptoms will become increasingly severe as fibrosis develops. Therefore, evaluating the fibrosis stage is crucial for patients with NAFLD. A liver biopsy is currently considered the gold standard for staging fibrosis. However, due to the limitations of liver biopsy, non-invasive alternatives were extensively studied and validated in patients with NAFLD. The advantages of non-invasive methods include their high safety and convenience compared with other invasive approaches. This review introduces the non-invasive methods, summarizes their benefits and limitations, and assesses their diagnostic performance for NAFLD-induced fibrosis.

3.
Cytotherapy ; 23(12): 1085-1096, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34593327

RESUMO

BACKGROUND AIMS: Despite the impressive efficacy of chimeric antigen receptor (CAR) T-cell therapy, adverse effects, including cytokine release syndrome and neurotoxicity, impede its therapeutic application, thus making the modulation of CAR T-cell activity a priority. The destabilizing domain mutated from Escherichia coli dihydrofolate reductase (DHFR) is inherently unstable and degraded by proteasomes unless it is stabilized by its chemical ligand trimethoprim (TMP), a Food and Drug Administration-approved drug. Here the authors reveal a strategy to modulate CAR T-cell activity at the protein level by employing DHFR and TMP as a chemical switch system. METHODS: First, the system was demonstrated to work in human primary T cells. To introduce the system to CAR T cells, DHFR was genetically fused to the carboxyl terminal of a third-generation CAR molecule targeting CD19 (CD19-CAR), constructing the CD19-CAR-DHFR fusion. RESULTS: The CD19-CAR-DHFR molecule level was shown to be modulated by TMP. Importantly, the incorporation of DHFR had no impact on the recognition specificity and normal function of the CAR molecule. Little adverse effect on cell proliferation and apoptosis was detected. It was proved that TMP could regulate cytokine secretion and the in vitro cytotoxicity of CD19-CAR-DHFR T cells. Furthermore, the in vivo anti-tumor efficacy was demonstrated to be controllable through the manipulation of TMP administration. The approach to control CD19-CAR also succeeded in 19-BBZ(71), another CD19-targeting CAR with a different structure. CONCLUSIONS: The proposed approach based on DHFR and TMP provides a facile strategy to bring CAR T-cell therapy under conditional user control, and the strategy may have the potential to be transplantable.


Assuntos
Receptores de Antígenos Quiméricos , Antígenos CD19/genética , Humanos , Imunoterapia Adotiva , Ligantes , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T
4.
Am J Med Genet B Neuropsychiatr Genet ; 186(1): 28-39, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33522098

RESUMO

Schizophrenia is a severe mental disease characterized with positive symptoms, negative symptoms, and cognitive impairments. Although recent genome-wide association studies (GWASs) have identified over 145 risk loci for schizophrenia, pinpointing the causal variants and genes at the reported loci and elucidating their roles in schizophrenia remain major challenges. Here we identify a functional single-nucleotide polymorphism (SNP; rs213237) in ZNF323 promoter by using functional fine-mapping. We found that allelic differences at rs213237 affected the ZNF323 promoter activity significantly. Consistently, expression quantitative trait loci (eQTL) analysis showed that rs213237 was significantly associated with ZNF323 expression in diverse human brain tissues, suggesting that rs213237 may contribute to schizophrenia risk through regulating ZNF323 expression. Interestingly, we found that ZNF323 protein was localized in the nucleus and knockdown of ZNF323 in macaque neural stem cells (mNSCs) significantly impaired proliferation and survival of mNSCs. We further showed that stable knockdown of ZNF323 in SH-SY5Y cells resulted in significant decrease of the tyrosine hydroxylase (TH) protein expression. Finally, transcriptome analysis revealed that ZNF323 may regulate pivotal schizophrenia risk genes (including VIPR2 and NPY) and schizophrenia-associated pathways (including PI3K-AKT and NOTCH signaling pathways), suggesting that ZNF323 may be a major regulator of schizophrenia risk genes. Our study reveals how a genetic variant in ZNF323 promoter contributes to schizophrenia risk through regulating ZNF323 expression. More importantly, our findings demonstrate that ZNF323 may have a pivotal role in schizophrenia pathogenesis through regulating schizophrenia risk genes and schizophrenia-associated biological processes (including neurodevelopment, PI3K-AKT, and NOTCH signaling pathways).


Assuntos
Proteínas de Ligação a DNA/metabolismo , Estudo de Associação Genômica Ampla , Células-Tronco Neurais/patologia , Neuroblastoma/patologia , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Esquizofrenia/patologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Predisposição Genética para Doença , Humanos , Macaca , Células-Tronco Neurais/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Fenótipo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Fatores de Transcrição/genética , Células Tumorais Cultivadas
5.
J Genet Genomics ; 47(5): 233-248, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32712163

RESUMO

The Psychiatric Genomics Consortium (PGC) has recently identified 10 potential functional coding variants for schizophrenia. However, how these coding variants confer schizophrenia risk remains largely unknown. Here, we investigate the associations between eight potential functional coding variants identified by PGC and schizophrenia in a large Han Chinese sample (n = 4022 cases and 9270 controls). Among the eight tested single nucelotide polymorphisms (SNPs), rs3617 (a missense variant, p.K315Q in the ITIH3 gene) showed genome-wide significant association with schizophrenia in the Han Chinese population (P = 8.36 × 10-16), with the same risk allele as in PGC. Interestingly, rs3617 is located in a genomic region that is highly evolutionarily conserved, and its schizophrenia risk allele (C allele) was associated with lower ITIH3 mRNA and protein expression. Intriguingly, mouse neural stem cells stably overexpressing ITIH3 with different alleles of rs3617 exhibited significant differences in proliferation, migration, and differentiation, suggesting the impact of rs3617 on neurodevelopment. Subsequent transcriptome analysis found that the differentially expressed genes in neural stem cells stably overexpressing different alleles of rs3617 were significantly enriched in schizophrenia-related pathways, including cell adhesion, synapse assembly, MAPK and PI3K-AKT pathways. Our study provides convergent lines of evidence suggesting that rs3617 in ITIH3 likely affects protein function and neurodevelopment and thereby confers risk of schizophrenia.


Assuntos
alfa-Globulinas/genética , Predisposição Genética para Doença , Transtornos do Neurodesenvolvimento/genética , Esquizofrenia/genética , Adolescente , Adulto , Povo Asiático , Proliferação de Células , China/epidemiologia , Feminino , Humanos , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Mutação de Sentido Incorreto/genética , Transtornos do Neurodesenvolvimento/patologia , Fosfatidilinositol 3-Quinases/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Risco , Esquizofrenia/patologia , Adulto Jovem
6.
ACS Appl Mater Interfaces ; 10(17): 14410-14417, 2018 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-29671572

RESUMO

The fluorescent probe for the detection of calcium ions is an indispensable tool in the biomedical field. The millimolar order of Ca(II) ions is associated with many physiological processes and diseases, such as hypercalcemia, soft tissue calcification, and bone microcracks. However, the conventional fluorescent probes are only suitable for imaging Ca(II) ions in the nanomolar to micromolar range, which can be because of their high affinities toward Ca(II) ions and aggregation-caused quenching drawbacks. To tackle this challenge, we herein develop an aggregation-induced emission (AIE) probe SA-4CO2Na for selective and light-up detection of Ca(II) ions in the millimolar range (0.6-3.0 mM), which can efficiently distinguish between hypercalcemic (1.4-3.0 mM) and normal (1.0-1.4 mM) Ca2+ ion levels. The formation of fibrillar aggregates between SA-4CO2Na and Ca(II) ions was clearly verified by fluorescence, scanning electron microscopy, and transmission electron analysis. Moreover, this AIE-active probe can be used for wash-free and light-up imaging of a high concentration of Ca(II) ions even in the solid analytes, including calcium deposits in psammomatous meningioma slice, microcracks on bovine bone surface, and microdefects on hydroxyapatite-based scaffold. It is thus expected that this AIE-active probe would have broad biomedical applications through light-up imaging and sensing of Ca(II) ions at the millimolar level.


Assuntos
Cálcio/química , Animais , Cátions Bivalentes , Bovinos , Fluorescência , Corantes Fluorescentes
7.
Onco Targets Ther ; 11: 1767-1776, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29662316

RESUMO

BACKGROUND: CD19-chimericantigen receptor (CAR) modified T cells (CD19-CAR T cells) have been well documented to possess potent anti-tumor properties against CD19-expressingleukemia cells. As a traditional medicine, metformin has been widely used to treat type II diabetes mellitus and more recently has become a candidate for the treatment of cancer. However, no report has revealed the direct effect of metformin on CD19-CAR T cell biological function and its underling mechanisms. PURPOSE: The purpose of this research was to explore the effect of metformin on CD19-CAR T cell biological function and the mechanisms involved. METHODS: CD19-CAR T cells proliferation, apoptosis and cytotoxicity were mainly tested by CCK-8 assay, flow cytometry and ELISA. The detection of mechanism primarily used western blot. Bioluminescence imaging is the main application technology of animal studies. RESULTS: In the current study, it was found that metformin inhibited CD19-CAR T cell proliferation and cytotoxicity and induced apoptosis. Furthermore, our study revealed that metformin activated AMPK and suppressed mTOR and HIF1α expression. By using an AMPK inhibitor, compound C, we demonstrated the crucial roles of AMPK in CD19-CAR T cells when they were treated with metformin. Finally, we verified that metformin suppressed the cytotoxicity of CD19-CAR T cell in vivo. CONCLUSION: Taken together, these results indicated that metformin may play an important role in modulating CD19-CAR T cell biological functions in an AMPK-dependent and mTOR/HIF1α-independent manner.

8.
Proc Natl Acad Sci U S A ; 114(34): 8980-8985, 2017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28784797

RESUMO

Glycans possess significant chemical diversity; glycan binding proteins (GBPs) recognize specific glycans to translate their structures to functions in various physiological and pathological processes. Therefore, the discovery and characterization of novel GBPs and characterization of glycan-GBP interactions are significant to provide potential targets for therapeutic intervention of many diseases. Here, we report the biochemical, functional, and structural characterization of a 130-amino-acid protein, Y3, from the mushroom Coprinus comatus Biochemical studies of recombinant Y3 from a yeast expression system demonstrated the protein is a unique GBP. Additionally, we show that Y3 exhibits selective and potent cytotoxicity toward human T-cell leukemia Jurkat cells compared with a panel of cancer cell lines via inducing caspase-dependent apoptosis. Screening of a glycan array demonstrated GalNAcß1-4(Fucα1-3)GlcNAc (LDNF) as a specific Y3-binding ligand. To provide a structural basis for function, the crystal structure was solved to a resolution of 1.2 Å, revealing a single-domain αßα-sandwich motif. Two monomers were dimerized to form a large 10-stranded, antiparallel ß-sheet flanked by α-helices on each side, representing a unique oligomerization mode among GBPs. A large glycan binding pocket extends into the dimeric interface, and docking of LDNF identified key residues for glycan interactions. Disruption of residues predicted to be involved in LDNF/Y3 interactions resulted in the significant loss of binding to Jurkat T-cells and severely impaired their cytotoxicity. Collectively, these results demonstrate Y3 to be a GBP with selective cytotoxicity toward human T-cell leukemia cells and indicate its potential use in cancer diagnosis and treatment.


Assuntos
Agaricales/metabolismo , Coprinus/metabolismo , Proteínas Fúngicas/metabolismo , Polissacarídeos/metabolismo , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Proteínas Fúngicas/química , Proteínas Fúngicas/farmacologia , Células HEK293 , Humanos , Células Jurkat , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Homologia de Sequência de Aminoácidos
9.
ACS Appl Mater Interfaces ; 8(28): 17878-84, 2016 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-27400339

RESUMO

Bone marrow-derived mesenchymal stem cells (BMSCs) have shown great potential for bone repair due to their strong proliferation ability and osteogenic capacity. To evaluate and improve the stem cell-based therapy, long-term tracking of stem cell differentiation into bone-forming osteoblasts is required. However, conventional fluorescent trackers such as fluorescent proteins, quantum dots, and fluorophores with aggregation-caused quenching (ACQ) characteristics have intrinsic limitations of possible interference with stem cell differentiation, heavy metal cytotoxicity, and self-quenching at a high labeling intensity. Herein, we developed aggregation-induced emission nanoparticles decorated with the Tat peptide (AIE-Tat NPs) for long-term tracking of the osteogenic differentiation of mouse BMSCs without interference of cell viability and differentiation ability. Compared with the ability of the commercial Qtracker 655 for tracking of only 6 passages of mouse BMSCs, AIE-Tat NPs have shown a much superior performance in long-term tracking for over 12 passages. Moreover, long-term tracking of the osteogenic differentiation process of mouse BMSCs was successfully conducted on the biocompatible hydroxyapatite scaffold, which is widely used in bone tissue engineering. Thus, AIE-Tat NPs have promising applications in tracking stem cell fate for bone repair.


Assuntos
Células da Medula Óssea/citologia , Corantes Fluorescentes/análise , Células-Tronco Mesenquimais/citologia , Nanopartículas/análise , Osteogênese/fisiologia , Fragmentos de Peptídeos/análise , Produtos do Gene tat do Vírus da Imunodeficiência Humana/análise , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Corantes Fluorescentes/química , Camundongos , Nanopartículas/química , Fragmentos de Peptídeos/química , Espectrofotometria Ultravioleta , Engenharia Tecidual , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química
10.
Nanoscale ; 8(32): 15027-32, 2016 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-27468980

RESUMO

To improve the treatment efficiency and reduce side effects in cancer therapy, accurate diagnosis of cancer cell types at a molecular level is highly desirable. Fluorescent nanoparticles (NPs) are especially suitable for detecting molecular biomarkers of cancer with advantages of superior brightness, easy decoration and high resolution. However, the conventional organic fluorophores, conjugated polymers, and inorganic quantum dots suffer from the drawbacks of aggregation-caused quenching (ACQ), low photostability, and heavy metal toxicity, respectively, which severely restrict their applications in NPs-based fluorescence imaging. To overcome these limitations, herein, we have developed fluorescent nanoparticles based on a t-BuPITBT-TPE fluorophore derived from aggregation-induced emission (AIE)-active tetraphenylethene. Through encapsulating t-BuPITBT-TPE within biocompatible DSPE-PEG and further decorating with a monoclonal antibody cetuximab (C225), the obtained t-BuPITBT-TPE-C225 NPs can be used for targeted imaging of non-small cell lung cancer cells with an overexpressed epidermal growth factor receptor (EGFR). The specific targeting ability of t-BuPITBT-TPE-C225 NPs has been well verified by confocal microscopy and flow cytometry experiments. The t-BuPITBT-TPE-C225 NPs have shown significant advantages in terms of highly efficient red emission, good bio-compatibility, and excellent photostability. This work provides a promising method for precise diagnosis of cancer cells by antibody-functionalized fluorescent NPs with high brightness.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Cetuximab/administração & dosagem , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Microscopia de Fluorescência , Nanopartículas , Linhagem Celular Tumoral , Corantes Fluorescentes , Humanos
11.
J Immunol ; 197(2): 458-69, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27296664

RESUMO

Pbx1 controls chromatin accessibility to a large number of genes and is entirely conserved between mice and humans. The Pbx1-d dominant-negative isoform is more frequent in CD4(+) T cells from lupus patients than from healthy controls. Pbx1-d is associated with the production of autoreactive T cells in mice carrying the Sle1a1 lupus-susceptibility locus. Transgenic (Tg) expression of Pbx1-d in CD4(+) T cells reproduced the phenotypes of Sle1a1 mice, with increased inflammatory functions of CD4(+) T cells and impaired Foxp3(+) regulatory T cell (Treg) homeostasis. Pbx1-d-Tg expression also expanded the number of follicular helper T cells (TFHs) in a cell-intrinsic and Ag-specific manner, which was enhanced in recall responses and resulted in Th1-biased Abs. Moreover, Pbx1-d-Tg CD4(+) T cells upregulated the expression of miR-10a, miR-21, and miR-155, which were implicated in Treg and follicular helper T cell homeostasis. Our results suggest that Pbx1-d impacts lupus development by regulating effector T cell differentiation and promoting TFHs at the expense of Tregs. In addition, our results identify Pbx1 as a novel regulator of CD4(+) T cell effector function.


Assuntos
Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas Proto-Oncogênicas/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Animais , Citometria de Fluxo , Proteínas de Homeodomínio/imunologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Fator de Transcrição 1 de Leucemia de Células Pré-B , Fatores de Transcrição/imunologia
12.
Cell Transplant ; 25(7): 1319-29, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26771084

RESUMO

Toll-like receptor 4 (TLR4) activation in pancreatic ß cells activates aberrant islet graft cellular pathways and contributes to immune rejection in allogeneic islet transplantation. As an approach to overcoming this problem, we determined the capacity of a 33-amino acid peptide consisting of a protein transduction domain (PTD) from the Hph-1 virus and a fragment of the intracellular domain of TLR4 from the C3H mice (PTD-dnTLR4) to block TLR4 signaling and improve allogeneic islet survival in vitro and after transplantation. The efficacy of PTD-dnTLR4 in blocking TLR4 signaling was assessed in the Raw264.7 macrophage line, in the islets, and the ßTC3 cell line. In Raw264.7 cells, preculture with the peptide reduced LPS-induced NF-κB activation and production of proinflammatory cytokines (IL-1ß, TNF-α, iNOS, and IL-6). In islets and ß cells, preincubation with PTD-dnTLR4 suppressed LPS-induced TNF-α expression via inhibition of NF-κB activation and protected them from stress-induced cell death. In vivo, preincubation of BALB/c (H-2(d)) islets with PTD-dnTLR4 resulted in significantly longer survival than control islets in a streptozotocin-induced diabetes model (two of seven grafts survived long term >100 days). PTD-dnTLR4-treated grafts exhibited reduced expression of TNF-α and iNOS and reduced macrophage infiltration posttransplant. The data indicate that PTD-dnTLR4 blocked TLR4 signaling in both macrophages and ß cells, and prolonged allograft survival at least in part by suppressing inflammation and macrophage infiltration. This strategy for blocking TLR4 activity has potential utilization in the treatment of diseases where excessive TLR4 activation contributes to the pathologic cellular pathways such as islet transplantation.


Assuntos
Aloenxertos/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Citoproteção/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Antígenos de Histocompatibilidade/metabolismo , Inflamação/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Domínios Proteicos , Células RAW 264.7
13.
Biochim Biophys Acta ; 1861(2): 130-137, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26569053

RESUMO

AIM: Atypical angiopoietin-like 8 (ANGPTL8), also known as betatrophin, is known to regulate lipid metabolism. However, its mechanism of action remains elusive. METHODS: HepG2, 3T3-L1, and NIT-1 cells were cultured in amino acid-complete MEM or histidine-free MEM to detect ANGPTL8 expression. The three cell types were treated with or without recombinant ANGPTL8 to investigate its role in lipid metabolism. Hydrodynamic tail vein gene delivery was also used to examine the role of ANGPTL8 in mice. RESULTS: ANGPTL8 is significantly up-regulated in amino acid-deprived cultured cells in vitro. The activation of ANGPTL8 gene transcription was mediated through the RAS/c-RAF/MAPK signaling pathway rather than the general GCN2/ATF4 pathways. ANGPTL8 activated the ERK signal transduction pathway in hepatocytes, adipocytes, and pancreatic ß-cells, up-regulating early growth response transcription factor (Egr1) and down-regulating adipose triglyceride lipase (ATGL). CONCLUSION: ANGPTL8 is a stress-response protein that regulates fat metabolism by suppressing ATGL expression, revealing a mechanistic connection between ANGPTL8 and lipid homeostasis in mammalian cells.


Assuntos
Adipócitos/metabolismo , Angiopoietinas/genética , Lipase/genética , Triglicerídeos/metabolismo , Células 3T3-L1 , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/metabolismo , Angiopoietinas/farmacologia , Animais , Diferenciação Celular , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Meios de Cultura/química , Meios de Cultura/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Glicerol/metabolismo , Células Hep G2 , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Lipase/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Proteínas ras/genética , Proteínas ras/metabolismo
14.
Biochem Biophys Res Commun ; 466(4): 656-63, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26403969

RESUMO

Type 1 diabetes is a T cell-mediated organ-specific autoimmune disease. Antigen-specific immune intervention allows the selective targeting of autoreactive T cell, while leaving the remainder of the immune system intact. However, immune intervention for type 1 diabetes has not yielded perfect results clinically. In our paper published previously, we asked whether pancreatic duodenal home box 1 (PDX1) is a target of anti-islet autoimmunity in type 1 diabetes. In this experiment, we assessed the therapeutic effect of oral administration of PDX1 on diabetes development of 4-week-old non-obese diabetic (NOD) mice. The results indicate that PDX1 immunization is an effective intervention strategy for delaying the onset of diabetes in NOD mice in association with: 1) reduced insulitis; 2) suppression of destructive autoreactive T cells; 3) augmentation of regulatory T cells; 4) a shift in cytokine production. The present observations suggest that immunization with PDX1 modulates immune cell responses in NOD mice, raising the possibility that it is beneficial in ameliorating autoimmune destruction of beta-cells and delaying type 1 diabetes development clinically.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Proteínas de Homeodomínio/administração & dosagem , Proteínas de Homeodomínio/imunologia , Transativadores/administração & dosagem , Transativadores/imunologia , Administração Oral , Transferência Adotiva , Animais , Autoimunidade , Citocinas/biossíntese , Citocinas/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Imunização , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Transcriptoma
15.
PLoS One ; 9(10): e110273, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25338001

RESUMO

Irisin is a newly discovered myokine that links exercise with metabolic homeostasis. It is involved in modest weight loss and improves glucose intolerance. However, the direct effects and mechanisms of irisin on vascular endothelial cells (ECs) are not fully understood. In the current study, we demonstrated that irisin promoted Human Umbilical Vein Endothelial Cell (HUVEC) proliferation. It was further demonstrated that this pro-proliferation effect was mediated by irisin-induced activation of extracellular signal-related kinase (ERK) signaling pathways. Inhibition of ERK signaling with U0126 decreased the pro-proliferation effect of irisin on HUVECs. It was also demonstrated that irisin reduced high glucose-induced apoptosis by up-regulating Bcl-2 expression and down-regulating Bax, Caspase-9 and Caspase-3 expression. In summary, these results suggested that irisin plays a novel role in sustaining endothelial homeostasis by promoting HUVEC proliferation via the ERK signaling pathway and protects the cell from high glucose-induced apoptosis by regulating Bcl-2,Bax and Caspase expression.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/genética , Fibronectinas/farmacologia , Glucose/antagonistas & inibidores , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Proliferação de Células/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
PLoS One ; 9(5): e97694, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24835010

RESUMO

Chronic myelogenous leukemia patients treated with tyrosine kinase inhibitor, Imatinib, were shown to have increased serum levels of C-peptide. Imatinib specifically inhibits the tyrosine kinase, c-Abl. However, the mechanism of how Imatinib treatment can lead to increased insulin level is unclear. Specifically, there is little investigation into whether Imatinib directly affects ß cells to promote insulin production. In this study, we showed that Imatinib significantly induced insulin expression in both glucose-stimulated and resting ß cells. In line with this finding, c-Abl knockdown by siRNA and overexpression of c-Abl markedly enhanced and inhibited insulin expression in ß cells, respectively. Unexpectedly, high concentrations of glucose significantly induced c-Abl expression, suggesting c-Abl may play a role in balancing insulin production during glucose stimulation. Further studies demonstrated that c-Abl inhibition did not affect the major insulin gene transcription factor, pancreatic and duodenal homeobox-1 (PDX-1) expression. Of interest, inhibition of c-Abl enhanced NKx2.2 and overexpression of c-Abl in ß cells markedly down-regulated NKx2.2, which is a positive regulator for insulin gene expression. Additionally, we found that c-Abl inhibition significantly enhanced the expression of glucose transporter GLUT2 on ß cells. Our study demonstrates a previously unrecognized mechanism that controls insulin expression through c-Abl-regulated NKx2.2 and GLUT2. Therapeutic targeting ß cell c-Abl could be employed in the treatment of diabetes or ß cell tumor, insulinoma.


Assuntos
Benzamidas/farmacologia , Transportador de Glucose Tipo 2/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/biossíntese , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirimidinas/farmacologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Glucose/metabolismo , Transportador de Glucose Tipo 2/genética , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/genética , Mesilato de Imatinib , Insulina/genética , Leucemia Mieloide/metabolismo , Camundongos , Biossíntese Peptídica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/fisiologia , RNA Interferente Pequeno/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra
17.
J Cell Sci ; 126(Pt 16): 3638-48, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23750005

RESUMO

Induced pluripotent stem cells (iPSCs) hold great promise for cell therapy. However, their low efficiency of lineage-specific differentiation and tumorigenesis severely hinder clinical translation. We hypothesized that reprogramming of somatic cells into lineage-specific progenitor cells might allow for large-scale expansion, avoiding the tumorigenesis inherent with iPSCs and simultaneously facilitating lineage-specific differentiation. Here we aimed at reprogramming rat hepatic WB cells, using four Yamanaka factors, into pancreatic progenitor cells (PPCs) or intermediate (IM) cells that have characteristics of PPCs. IM clones were selected based on their specific morphology and alkaline phosphatase activity and stably passaged under defined culture conditions. IM cells did not have iPSC properties, could be stably expanded in large quantity, and expressed all 14 genes that are used to define the PPC developmental stage. Directed differentiation of IM and WB cells by Pdx1-Ngn3-MafA (PNM) into pancreatic beta-like cells revealed that the IM cells are more susceptible to directed beta cell differentiation because of their open chromatin configuration, as demonstrated by expression of key pancreatic beta cell genes, secretion of insulin in response to glucose stimulation, and easy access to exogenous PNM proteins at the rat insulin 1 and Pdx1 promoters. This notion that IM cells are superior to their parental cells is further supported by the epigenetic demonstration of accessibility of Pdx1 and insulin 1 promoters. In conclusion, we have developed a strategy to derive and expand PPC cells from hepatic WB cells using conventional cell reprogramming. This proof-of-principal study may offer a novel, safe and effective way to generate autologous pancreatic beta cells for cell therapy of diabetes.


Assuntos
Hepatócitos/citologia , Células Secretoras de Insulina/citologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Metilação de DNA , Hepatócitos/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Regiões Promotoras Genéticas , Ratos , Células-Tronco/citologia , Células-Tronco/metabolismo , Transativadores/genética
18.
Am J Transl Res ; 5(2): 184-99, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23573363

RESUMO

Many previous studies demonstrate that hepatocytes can be reprogrammed into insulin-producing cells (IPCs) utilizing viral vector-mediated delivery of pancreatic transcription factors (PTFs). However, whether these liver-derived IPCs are susceptible to autoimmune attack in animal models of type 1 diabetes remains unclear, in part due to the immunogenicity of the viral vectors used to introduce PTF genes. Adeno-associated virus serotype 2 vector-expressing Pdx1-VP16 (Pdx1) and Ngn3 were prepared and injected into the portal vein of streptozotocin (Stz)/diabetic NOD/SCID mice. The presence of glucose-responsive liver-IPCs and their susceptibility to anti-beta cell autoimmunity were assessed by blood glucose levels, insulin content, IPC cell distribution, and intraperitoneal glucose tolerance test following subtotal pancreatectomy (Px) and passive transfer of diabetogenic splenocytes isolated from diabetic female NOD mice. A combination of two PTF genes (Pdx1/Ngn3) effectively reprogrammed liver cells into glucose-responsive IPCs. These IPCs corrected hyperglycemia in Stz/diabetic NOD/SCID mice and maintained normoglycemia following subtotal Px, indicating that liver-derived IPCs could maintain glucose homeostasis. Importantly, we also demonstrated that the glucose-responsive liver-derived IPCs were susceptible to autoimmune destruction by diabetogenic splenocytes, as indicated by progressive elevation in blood glucose levels as well as mixed T-, and B-lymphocytic infiltrates surrounding liver-IPCs 2~3 weeks following transferring of diabetogenic splenocytes into NOD/SCID mice, and confirmed by immunohistochemical studies. In conclusion, genetically reprogrammed liver-IPCs, like pancreatic islet beta-cells, are susceptible to autoimmune attack, suggesting that for cell-replacement therapy of treating type 1 diabetes, beta-cell surrogates may require concomitant immunotherapy to avoid autoimmune destruction.

19.
J Immunol ; 188(8): 4113-21, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22422888

RESUMO

Genetic polymorphisms of IFN regulatory factor 5 (IRF5) are associated with an increased risk of lupus in humans. In this study, we examined the role of IRF5 in the pathogenesis of pristane-induced lupus in mice. The pathological response to pristane in IRF5(-/-) mice shared many features with type I IFN receptor (IFNAR)(-/-) and TLR7(-/-) mice: production of anti-Sm/RNP autoantibodies, glomerulonephritis, generation of Ly6C(hi) monocytes, and IFN-I production all were greatly attenuated. Lymphocyte activation following pristane injection was greatly diminished in IRF5(-/-) mice, and Th cell differentiation was deviated from Th1 in wild-type mice toward Th2 in IRF5(-/-) mice. Th cell development was skewed similarly in TLR7(-/-) or IFNAR(-/-) mice, suggesting that IRF5 alters T cell activation and differentiation by affecting cytokine production. Indeed, production of IFN-I, IL-12, and IL-23 in response to pristane was markedly decreased, whereas IL-4 increased. Unexpectedly, plasmacytoid dendritic cells (pDC) were not recruited to the site of inflammation in IRF5(-/-) or MyD88(-/-) mice, but were recruited normally in IFNAR(-/-) and TLR7(-/-) mice. In striking contrast to wild-type mice, pristane did not stimulate local expression of CCL19 and CCL21 in IRF5(-/-) mice, suggesting that IRF5 regulates chemokine-mediated pDC migration independently of its effects on IFN-I. Collectively, these data indicate that altered production of IFN-I and other cytokines in IRF5(-/-) mice prevents pristane from inducing lupus pathology by broadly affecting T and B lymphocyte activation/differentiation. Additionally, we uncovered a new, IFN-I-independent role of IRF5 in regulating chemokines involved in the homing of pDCs and certain lymphocyte subsets.


Assuntos
Fatores Reguladores de Interferon/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Carcinógenos , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Humanos , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/genética , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Terpenos , Equilíbrio Th1-Th2
20.
Planta Med ; 78(2): 172-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22083900

RESUMO

Two new lactones, 1 and 2, together with five known compounds, 3-7, were isolated from the marine mangrove fungus Xylaria sp. BL321. Their structures were determined by comprehensive analysis of their MS and NMR spectroscopic data. The absolute configurations of 1 and 2 were established on the basis of electronic circular dichroism calculations. It was found that the exocyclic double bond of 1 rearranged into a cyclic double bond to form a new crystal compound (1a) in diluted NaOH solution. Compound 3 was isolated for the first time as a natural product; its absolute configuration was determined by single-crystal X-ray crystallography. Compounds 4-7 displayed cytotoxicity against human breast cancer cell lines MCF-7 and MDA-MB-435, while compounds 1- 3 were inactive (IC(50) > 50 µM).


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Ascomicetos/química , Benzofuranos/uso terapêutico , Produtos Biológicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Cromanos/uso terapêutico , Citocalasinas/uso terapêutico , Lactonas/uso terapêutico , Naftalenos/uso terapêutico , Sesquiterpenos/uso terapêutico , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Benzofuranos/isolamento & purificação , Benzofuranos/farmacologia , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Linhagem Celular Tumoral , China , Cromanos/isolamento & purificação , Cromanos/farmacologia , Dicroísmo Circular , Cristalografia por Raios X , Citocalasinas/isolamento & purificação , Citocalasinas/farmacologia , Feminino , Humanos , Concentração Inibidora 50 , Lactonas/isolamento & purificação , Lactonas/farmacologia , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Naftalenos/isolamento & purificação , Naftalenos/farmacologia , Sesquiterpenos/isolamento & purificação , Sesquiterpenos/farmacologia , Árvores/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA