Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Nat Immunol ; 20(4): 471-481, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30778241

RESUMO

Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.


Assuntos
Imunoglobulina A/biossíntese , Intestinos/imunologia , Microbiota , Proteínas Proto-Oncogênicas c-maf/fisiologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Células Cultivadas , Colite/imunologia , Citocinas/metabolismo , Disbiose , Regulação da Expressão Gênica , Homeostase , Interleucina-10/biossíntese , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-maf/genética , Proteínas Proto-Oncogênicas c-maf/metabolismo , Linfócitos T Reguladores/enzimologia
3.
Immunity ; 48(3): 570-583.e8, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29562203

RESUMO

Polymorphisms in NFKB1 that diminish its expression have been linked to human inflammatory diseases and increased risk for epithelial cancers. The underlying mechanisms are unknown, and the link is perplexing given that NF-κB signaling reportedly typically exerts pro-tumorigenic activity. Here we have shown that NF-κB1 deficiency, even loss of a single allele, resulted in spontaneous invasive gastric cancer (GC) in mice that mirrored the histopathological progression of human intestinal-type gastric adenocarcinoma. Bone marrow chimeras revealed that NF-κB1 exerted tumor suppressive functions in both epithelial and hematopoietic cells. RNA-seq analysis showed that NF-κB1 deficiency resulted in aberrant JAK-STAT signaling, which dysregulated expression of effectors of inflammation, antigen presentation, and immune checkpoints. Concomitant loss of STAT1 prevented these immune abnormalities and GC development. These findings provide mechanistic insight into how polymorphisms that attenuate NFKB1 expression predispose humans to epithelial cancers, highlighting the pro-tumorigenic activity of STAT1 and identifying targetable vulnerabilities in GC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , NF-kappa B/deficiência , Fator de Transcrição STAT1/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animais , Apresentação de Antígeno/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Redes Reguladoras de Genes , Humanos , Inflamação/patologia , Camundongos , Camundongos Knockout , Fator de Transcrição STAT1/deficiência , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia
4.
Clin Immunol ; 259: 109871, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38101498

RESUMO

To clarify the role of the C5a/C5aR (C5a receptor) and C5b-9 pathways in macrovascular thrombosis (MAT) and renal microthrombosis (MIT), 73 renal biopsy-proven complement-mediated thrombotic microangiopathy (C-TMA) patients were enrolled; 9 patients with pure MAT and 13 patients with pure MIT were selected for further study. Twenty-five external C-TMA patients were selected as the validation cohort. Plasma C5a and sC5b-9 (soluble C5b-9) levels were significantly higher in patients with MAT than in those with MIT (P = 0.008, P = 0.041, respectively). The mean optical density of C5aR1 in the kidney was significantly higher in MAT patients than in those with MIT (P < 0.001). Both urinary sC5b-9 levels (MIT: P < 0.001, MAT: P = 0.004) and renal deposition of C5b-9 (MIT: P < 0.001, MAT: P = 0.001) were significantly higher in C-TMA patients compared to normal control, but were similar between MAT and MIT groups. In the correlation analysis within 22C-TMA patients, urinary sC5b-9 levels and renal deposition of C5b-9 were positively correlated to renal MIT formation (P = 0.009 and P = 0.031, respectively). Furthermore, the renal citrullinated histone H3 (CitH3)- and neutrophil elastase (NE)-positive area ratios were both significantly higher in the MAT group than in the MIT group (P = 0.006 and P = 0.020, respectively). Therefore, the local C5b-9 and C5a/C5aR1 pathways might have differential contributions to MIT and MAT formation in the disease.


Assuntos
Trombose , Microangiopatias Trombóticas , Humanos , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento , Receptor da Anafilatoxina C5a , Complemento C5b
5.
Twin Res Hum Genet ; 27(2): 80-84, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38634508

RESUMO

Gestational diabetes mellitus (GDM) is a frequent complication of pregnancy. The specific mechanisms underlying GDM have not yet been fully elucidated. Contemporary research indicates a potential association between liver enzyme irregularities and an increased risk of metabolic disorders, including diabetes. The alanine aminotransferase (ALT) level is recognized as a sensitive marker of liver injury. An increase in ALT levels is hypothesized to be linked to the pathogenesis of insulin resistance and diabetes. Nonetheless, the definitive causal link between ALT levels and GDM still needs to be determined. This investigation utilized two-sample Mendelian randomization (MR) to examine the genetic causation between alanine aminotransferase (ALT) and GDM. We acquired alanine aminotransferase (ALT)-related GWAS summary data from the UK Biobank, Million Veteran Program, Rotterdam Study, and Lifeline Study. Gestational diabetes data were obtained from the FinnGen Consortium. We employed various MR analysis techniques, including inverse-variance weighted (IVW), MR Egger, weighted median, simple, and weighted weighting. In addition to MR-Egger intercepts, Cochrane's Q test was also used to assess heterogeneity in the MR data, and the MR-PRESSO test was used to assess horizontal pleiotropy. To assess the association's sensitivity, a leave-one-out approach was employed. The IVW results confirmed the independent risk factor for GDM development, as indicated by the ALT level (p = .011). As shown by leave-one-out analysis, horizontal pleiotrophy did not significantly skew the causative link (p > .05). Our dual-sample MR analysis provides substantiated evidence of a genetic causal relationship between alanine aminotransferase (ALT) levels and gestational diabetes.


Assuntos
Alanina Transaminase , Diabetes Gestacional , Análise da Randomização Mendeliana , Humanos , Feminino , Diabetes Gestacional/genética , Diabetes Gestacional/sangue , Diabetes Gestacional/epidemiologia , Alanina Transaminase/sangue , Alanina Transaminase/genética , Gravidez , Estudo de Associação Genômica Ampla , Fatores de Risco , Adulto , Polimorfismo de Nucleotídeo Único , Predisposição Genética para Doença
6.
Artigo em Inglês | MEDLINE | ID: mdl-38923718

RESUMO

BACKGROUND: Gestational Diabetes Mellitus (GDM) poses significant risks to maternal and fetal health. Current diagnostic methods based on glucose tolerance tests have limitations for early detection. tRNA-derived small RNAs (tsRNAs) have emerged as potential molecular regulators in various diseases, including metabolic disorders. However, the diagnostic value of tsRNAs in plasma for early GDM or postpartum remains unclear. METHODS: This longitudinal study profiled the expression of tsRNAs across different gestational stages and postpartum in women with GDM (n = 40) and healthy control gestational women (HCs, n = 40). High-throughput small RNA sequencing identified candidate tsRNAs, which were then validated and correlated with clinical biochemical markers such as fasting blood glucose (FBG), HOMA-IR, and GHbA1c. RESULTS: tRF-1:32-Val-AAC-1-M6, tRF-1:31-Glu-CTC-1-M2, and tRF-1:30-Gly-CCC-1-M4 were consistently upregulated in the GDM group compared to HCs during the second trimester (p < 0.05). Only tRF-1:31-Glu-CTC-1-M2 was highly expressed during the first trimester, and tRF-1:30-Gly-CCC-1-M4 increased during postpartum. tRF-1:31-Glu-CTC-1-M2 showed a significant correlation with FBG levels in the first trimester (R = 0.317, p = 0.047). The expression of tRF-1:30-Gly-CCC-1-M4 was significantly correlated with HOMA-IR (r = 0.65, p < 0.001) and GHBA1c (r = 0.33, p = 0.037) during postpartum. A joint diagnostic model incorporating tsRNAs expression and clinical markers demonstrated enhanced predictive power for GDM (ROC AUC = 0.768). CONCLUSION: Our results revealed distinct expression patterns of specific tsRNAs in GDM, showcasing their correlation with key metabolic parameters. This underscores their promising role as biomarkers for early prediction and diagnosis of GDM. The integration of tRFs into a composite biomarker panel holds the potential to improve clinical outcomes by enabling personalized risk assessment and targeted interventions.

7.
Ren Fail ; 46(1): 2332491, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38584145

RESUMO

OBJECTIVE: Lipoprotein glomerulopathy (LPG) is a rare disorder characterized by the development of glomerular lipoprotein thrombosis. LPG exhibits familial aggregation, with mutations in the apolipoprotein E (APOE) gene identified as the leading cause of this disease. This study aimed to investigate APOE gene mutations and the clinicopathological features in eleven LPG patients. METHODS: Clinicopathological and follow-up data were obtained by extracting DNA, followed by APOE coding region sequencing analysis. This study analyzed clinical and pathological manifestations, gene mutations, treatment and prognosis. RESULTS: The mean age of the eleven patients was 33.82 years. Among them, five had a positive family history for LPG, ten presented with proteinuria, four exhibited nephrotic syndrome, and six presented with microscopic hematuria. Dyslipidemia was identified in ten patients. In all renal specimens, there was evident dilation of glomerular capillary lumens containing lipoprotein thrombi, and positive oil red O staining was observed in frozen sections of all samples. APOE gene testing revealed that one patient had no mutations, while the remaining ten patients exhibited mutations in the APOE gene, with three patients presenting with multiple mutations simultaneously. Following the confirmation of LPG diagnosis, treatment with angiotensin-converting enzyme inhibitor (ACEI)/angiotensin receptor blocker (ARB) was initiated, and the disease progressed slowly. CONCLUSION: LPG is histologically characterized by lamellated lipoprotein thrombi in glomeruli, and kidney biopsy is essential for diagnosis. Mutations in the APOE gene are the leading cause of LPG. This study revealed clinicopathological characteristics and APOE gene mutations in patients with LPG, which helps us better understand the disease.


Assuntos
Antagonistas de Receptores de Angiotensina , Nefropatias , Humanos , Adulto , Inibidores da Enzima Conversora de Angiotensina , Nefropatias/patologia , Mutação , Apolipoproteínas E/genética
8.
Genes Dev ; 28(12): 1337-50, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24939936

RESUMO

Loss-of-function mutations in hematopoietic transcription factors including PAX5 occur in most cases of B-progenitor acute lymphoblastic leukemia (B-ALL), a disease characterized by the accumulation of undifferentiated lymphoblasts. Although PAX5 mutation is a critical driver of B-ALL development in mice and humans, it remains unclear how its loss contributes to leukemogenesis and whether ongoing PAX5 deficiency is required for B-ALL maintenance. Here we used transgenic RNAi to reversibly suppress endogenous Pax5 expression in the hematopoietic compartment of mice, which cooperates with activated signal transducer and activator of transcription 5 (STAT5) to induce B-ALL. In this model, restoring endogenous Pax5 expression in established B-ALL triggers immunophenotypic maturation and durable disease remission by engaging a transcriptional program reminiscent of normal B-cell differentiation. Notably, even brief Pax5 restoration in B-ALL cells causes rapid cell cycle exit and disables their leukemia-initiating capacity. These and similar findings in human B-ALL cell lines establish that Pax5 hypomorphism promotes B-ALL self-renewal by impairing a differentiation program that can be re-engaged despite the presence of additional oncogenic lesions. Our results establish a causal relationship between the hallmark genetic and phenotypic features of B-ALL and suggest that engaging the latent differentiation potential of B-ALL cells may provide new therapeutic entry points.


Assuntos
Diferenciação Celular/genética , Fator de Transcrição PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Células Precursoras de Linfócitos B/citologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes myc/genética , Humanos , Camundongos , Camundongos Transgênicos , Fator de Transcrição PAX5/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
9.
Blood ; 133(16): 1729-1741, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30755422

RESUMO

Somatically acquired mutations in PHF6 (plant homeodomain finger 6) frequently occur in hematopoietic malignancies and often coincide with ectopic expression of TLX3. However, there is no functional evidence to demonstrate whether these mutations contribute to tumorigenesis. Similarly, the role of PHF6 in hematopoiesis is unknown. We report here that Phf6 deletion in mice resulted in a reduced number of hematopoietic stem cells (HSCs), an increased number of hematopoietic progenitor cells, and an increased proportion of cycling stem and progenitor cells. Loss of PHF6 caused increased and sustained hematopoietic reconstitution in serial transplantation experiments. Interferon-stimulated gene expression was upregulated in the absence of PHF6 in hematopoietic stem and progenitor cells. The numbers of hematopoietic progenitor cells and cycling hematopoietic stem and progenitor cells were restored to normal by combined loss of PHF6 and the interferon α and ß receptor subunit 1. Ectopic expression of TLX3 alone caused partially penetrant leukemia. TLX3 expression and loss of PHF6 combined caused fully penetrant early-onset leukemia. Our data suggest that PHF6 is a hematopoietic tumor suppressor and is important for fine-tuning hematopoietic stem and progenitor cell homeostasis.


Assuntos
Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Leucemia/etiologia , Proteínas Repressoras/fisiologia , Animais , Carcinogênese , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Receptores de Interferon , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor
10.
PLoS Biol ; 16(8): e2004986, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30080881

RESUMO

Distinct transcriptional states are maintained through organization of chromatin, resulting from the sum of numerous repressive and active histone modifications, into tightly packaged heterochromatin versus more accessible euchromatin. Polycomb repressive complex 2 (PRC2) is the main mammalian complex responsible for histone 3 lysine 27 trimethylation (H3K27me3) and is integral to chromatin organization. Using in vitro and in vivo studies, we show that deletion of Suz12, a core component of all PRC2 complexes, results in loss of H3K27me3 and H3K27 dimethylation (H3K27me2), completely blocks normal mammary gland development, and profoundly curtails progenitor activity in 3D organoid cultures. Through the application of mammary organoids to bypass the severe phenotype associated with Suz12 loss in vivo, we have explored gene expression and chromatin structure in wild-type and Suz12-deleted basal-derived organoids. Analysis of organoids led to the identification of lineage-specific changes in gene expression and chromatin structure, inferring cell type-specific PRC2-mediated gene silencing of the chromatin state. These expression changes were accompanied by cell cycle arrest but not lineage infidelity. Together, these data indicate that canonical PRC2 function is essential for development of the mammary gland through the repression of alternate transcription programs and maintenance of chromatin states.


Assuntos
Glândulas Mamárias Animais/embriologia , Complexo Repressor Polycomb 2/fisiologia , Animais , Cromatina/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Feminino , Heterocromatina/metabolismo , Código das Histonas , Histonas/metabolismo , Lisina/metabolismo , Glândulas Mamárias Animais/metabolismo , Metilação , Camundongos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb , Cultura Primária de Células , Processamento de Proteína Pós-Traducional
11.
J Immunol ; 202(11): 3282-3296, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31004011

RESUMO

The adult heart contains macrophages derived from both embryonic and adult bone marrow (BM)-derived precursors. This population diversity prompted us to explore how distinct macrophage subsets localize within the heart, and their relative contributions in cardiac disease. In this study, using the reciprocal expression of Lyve-1 and Ccr2 to distinguish macrophages with distinct origins, we show that, in the steady state, both embryonic (Lyvepos) and BM-derived (Ccr2pos) macrophages populate the major vessels of the heart in mice and humans. However, cardiac macrophage populations are markedly perturbed by inflammation. In a mouse model of Kawasaki disease, BM-derived macrophages preferentially increase during acute cardiac inflammation and selectively accumulate around major cardiac vessels. The accumulation of BM-derived macrophages coincides with the loss of their embryonic counterparts and is an initiating, essential step in the emergence of subsequent cardiac vasculitis in this experimental model. Finally, we demonstrate that the accumulation of Ccr2pos macrophages (and the development of vasculitis) occurs in close proximity to a population of Ccr2 chemokine ligand-producing epicardial cells, suggesting that the epicardium may be involved in localizing inflammation to cardiac vessels. Collectively, our findings identify the perivascular accumulation of BM-derived macrophages as pivotal in the pathogenesis of cardiac vasculitis and provide evidence about the mechanisms governing their recruitment to the heart.


Assuntos
Células-Tronco Embrionárias/citologia , Macrófagos/imunologia , Síndrome de Linfonodos Mucocutâneos/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Pericárdio/imunologia , Vasculite/imunologia , Animais , Movimento Celular , Proliferação de Células , Vasos Coronários/patologia , Modelos Animais de Doenças , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Receptores CCR2/metabolismo
12.
PLoS Genet ; 14(11): e1007735, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30399141

RESUMO

Diet may be modified seasonally or by biogeographic, demographic or cultural shifts. It can differentially influence mitochondrial bioenergetics, retrograde signalling to the nuclear genome, and anterograde signalling to mitochondria. All these interactions have the potential to alter the frequencies of mtDNA haplotypes (mitotypes) in nature and may impact human health. In a model laboratory system, we fed four diets varying in Protein: Carbohydrate (P:C) ratio (1:2, 1:4, 1:8 and 1:16 P:C) to four homoplasmic Drosophila melanogaster mitotypes (nuclear genome standardised) and assayed their frequency in population cages. When fed a high protein 1:2 P:C diet, the frequency of flies harbouring Alstonville mtDNA increased. In contrast, when fed the high carbohydrate 1:16 P:C food the incidence of flies harbouring Dahomey mtDNA increased. This result, driven by differences in larval development, was generalisable to the replacement of the laboratory diet with fruits having high and low P:C ratios, perturbation of the nuclear genome and changes to the microbiome. Structural modelling and cellular assays suggested a V161L mutation in the ND4 subunit of complex I of Dahomey mtDNA was mildly deleterious, reduced mitochondrial functions, increased oxidative stress and resulted in an increase in larval development time on the 1:2 P:C diet. The 1:16 P:C diet triggered a cascade of changes in both mitotypes. In Dahomey larvae, increased feeding fuelled increased ß-oxidation and the partial bypass of the complex I mutation. Conversely, Alstonville larvae upregulated genes involved with oxidative phosphorylation, increased glycogen metabolism and they were more physically active. We hypothesise that the increased physical activity diverted energy from growth and cell division and thereby slowed development. These data further question the use of mtDNA as an assumed neutral marker in evolutionary and population genetic studies. Moreover, if humans respond similarly, we posit that individuals with specific mtDNA variations may differentially metabolise carbohydrates, which has implications for a variety of diseases including cardiovascular disease, obesity, and perhaps Parkinson's Disease.


Assuntos
Estudos de Associação Genética , Genótipo , Fenótipo , Animais , DNA Mitocondrial , Dieta , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Metabolismo Energético , Aptidão Genética , Haplótipos , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metaboloma , Mitocôndrias/genética , Mitocôndrias/metabolismo , Modelos Biológicos , Modelos Moleculares , Mutação , Conformação Proteica , Reprodutibilidade dos Testes , Transcriptoma
13.
Am J Nephrol ; 51(9): 752-760, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32862175

RESUMO

BACKGROUND: The current study aimed to evaluate the associations between podocyte injury and clinicopathological features in renal thrombotic microangiopathy (TMA) based on a Chinese cohort, which might be underscored in this disease. METHODS: The clinical, laboratory, and renal histopathological data of patients with renal biopsy-proven TMA from 2000 to 2015 in our institute were collected. Foot process effacement (FPE) was quantified by foot process width (FPW) by electron microscopy. Podocytes in the renal specimens were also detected by stainings for podocyte-specific markers, including Wilms tumor 1 (WT-1), synaptopodin, and podocalyxin. The associations between FPW and clinico-histopathological data were further analyzed. A composite end-point was defined by all-cause death or end-stage renal disease to address the predictive value of FPW. RESULTS: Sixty-three patients with renal biopsy-proven TMA were enrolled. The FPW of renal TMA patients was 1,090 ± 637 nm (range, 572-4,748 nm), which was significantly higher than the normal range in our center (p = 0.005). By immunohistochemistry and immunofluorescence assays, we found decreased expressions of synaptopodin, podocalyxin, and WT-1 and continued stainings of WT-1 in some podocytes without detectable synaptopodin stainings in the areas of sclerotic tufts and cellular crescents. The FPW value was correlated with the serum albumin concentration (rs = -0.281, p = 0.026), proteinuria amount (rs = 0.255, p = 0.047), serum creatinine levels (rs = 0.339, p = 0.007), and eGFR (rs = -0.335, p = 0.007). According to ROC curve analysis, the optimal cutoff level of FPW for predicting the composite end-point was 869 nm. In patients with FPW ≥ 869 nm, FPW levels were further correlated with the severity of mesangiolysis (rs = 0.351, p = 0.033) and glomerulosclerosis (rs = 0.369, p = 0.025) in pathological evaluations. Patients without clinical remission also had higher FPW than those with remission (1,240 ± 793 vs. 925 ± 344 nm, p = 0.013). The multivariate Cox hazard model showed that FPW ≥ 869 nm was an independent risk factor for the composite end-point (hazard ratio: 3.64, 95% CI: 1.37-9.66, p = 0.009). CONCLUSION: The podocyte injury was prevalent and the FPW levels were closely associated with clinicopathological features, especially prognosis, in renal TMA patients.


Assuntos
Falência Renal Crônica/epidemiologia , Glomérulos Renais/patologia , Microangiopatias Trombóticas/complicações , Adulto , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Biomarcadores/análise , Biomarcadores/metabolismo , Biópsia , Causas de Morte , Estudos de Coortes , Feminino , Seguimentos , Humanos , Imunossupressores/uso terapêutico , Falência Renal Crônica/etiologia , Falência Renal Crônica/patologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/ultraestrutura , Masculino , Proteínas dos Microfilamentos/análise , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica , Pessoa de Meia-Idade , Troca Plasmática , Prognóstico , Medição de Risco/métodos , Sialoglicoproteínas/análise , Sialoglicoproteínas/metabolismo , Microangiopatias Trombóticas/mortalidade , Microangiopatias Trombóticas/patologia , Microangiopatias Trombóticas/terapia , Proteínas WT1/análise , Proteínas WT1/metabolismo , Adulto Jovem
14.
PLoS Biol ; 15(1): e2000731, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28125611

RESUMO

Lung squamous cell carcinoma (SqCC), the second most common subtype of lung cancer, is strongly associated with tobacco smoking and exhibits genomic instability. The cellular origins and molecular processes that contribute to SqCC formation are largely unexplored. Here we show that human basal stem cells (BSCs) isolated from heavy smokers proliferate extensively, whereas their alveolar progenitor cell counterparts have limited colony-forming capacity. We demonstrate that this difference arises in part because of the ability of BSCs to repair their DNA more efficiently than alveolar cells following ionizing radiation or chemical-induced DNA damage. Analysis of mice harbouring a mutation in the DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a key enzyme in DNA damage repair by nonhomologous end joining (NHEJ), indicated that BSCs preferentially repair their DNA by this error-prone process. Interestingly, polyploidy, a phenomenon associated with genetically unstable cells, was only observed in the human BSC subset. Expression signature analysis indicated that BSCs are the likely cells of origin of human SqCC and that high levels of NHEJ genes in SqCC are correlated with increasing genomic instability. Hence, our results favour a model in which heavy smoking promotes proliferation of BSCs, and their predilection for error-prone NHEJ could lead to the high mutagenic burden that culminates in SqCC. Targeting DNA repair processes may therefore have a role in the prevention and therapy of SqCC.


Assuntos
Dano ao DNA , Reparo do DNA por Junção de Extremidades , Pulmão/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Morte Celular , Separação Celular , Quebras de DNA de Cadeia Dupla , Células Epiteliais/citologia , Células Epiteliais/ultraestrutura , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Neoplasias de Células Escamosas/patologia , Alvéolos Pulmonares/citologia , Fumar/efeitos adversos , Traqueia/citologia
15.
Physiol Genomics ; 50(3): 190-196, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29341866

RESUMO

Endomyocardial biopsy (EMB) remains the gold standard for detecting rejection after heart transplantation but is costly and invasive. This study aims to distinguish no rejection (0R) from low-grade rejection (1R/2R) after heart transplantation in children by using global gene expression profiling in blood. A total of 106 blood samples with corresponding EMB from 18 children who underwent heart transplantation from 2011 to 2014 were analyzed (18 baseline/pretransplantation samples, 88 EMB samples). Corresponding rejection grades for each blood sample were 0R in 39% (34/88), 1R in 51% (45/88), and 2R in 10% (9/88). mRNA from each sample was sequenced. Differential expression analysis was performed at the gene level. A k-nearest neighbor (kNN) analysis was applied to the most differentially expressed (DE) genes to identify rejection after transplantation. Mean age at transplantation was 10.0 ± 5.4 yr. Expression of B cell and T cell receptor sequences was used to measure the effect of posttransplantation immunosuppression. Follow-up samples had lower levels of immunoglobulin gene families compared with pretransplantation ( P < 3E-5) (lower numbers of activated B cells). T cell receptor alpha and beta gene families had decreased expression in 0R samples compared with pretransplantation ( P < 4E-5) but recovered to near baseline levels in 1R/2R samples. kNN using the most DE gene (MKS1) and k = 9 nearest neighbors correctly identified 83% (73/88) of 1R/2R compared with 0R by leave-one-out cross validation. Using a genomic approach we can distinguish low-grade cellular allograft rejection (1R/2R) from no rejection (0R) after heart transplantation in children despite a wide age range.


Assuntos
Perfilação da Expressão Gênica , Rejeição de Enxerto/genética , Transplante de Coração , Adolescente , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Humanos , Lactente , Masculino
16.
Immunol Cell Biol ; 96(10): 1083-1094, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29870118

RESUMO

Plasmacytoid dendritic cells (pDCs) play a critical role in bridging the innate and adaptive immune systems. pDCs are specialized type I interferon (IFN) producers, which has implicated them as initiators of autoimmune pathogenesis. However, little is known about the downstream effectors of type I IFN signaling that amplify autoimmune responses. Here, we have used a chemokine reporter mouse to determine the CXCR3 ligand responses in DCs subsets. Following TLR7 stimulation, conventional type 1 and type 2 DCs (cDC1 and cDC2, respectively) uniformly upregulate CXCL10. By contrast, the proportion of chemokine positive pDCs was significantly less, and stable CXCL10+ and CXCL10- populations could be distinguished. CXCL9 expression was induced in all cDC1s, in half of the cDC2 but not by pDCs. The requirement for IFNAR signaling for chemokine reporter expression was interrogated by receptor blocking and deficiency and shown to be critical for CXCR3 ligand expression in Flt3-ligand-derived DCs. Chemokine-producing potential was not concordant with the previously identified markers of pDC heterogeneity. Finally, we show that CXCL10+ and CXCL10- populations are transcriptionally distinct, expressing unique transcriptional regulators, IFN signaling molecules, chemokines, cytokines, and cell surface markers. This work highlights CXCL10 as a downstream effector of type I IFN signaling and suggests a division of labor in pDCs subtypes that likely impacts their function as effectors of viral responses and as drivers of inflammation.


Assuntos
Quimiocina CXCL10/genética , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Receptor 7 Toll-Like/agonistas , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Células Cultivadas , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , Perfilação da Expressão Gênica , Imunofenotipagem , Interferon Tipo I/metabolismo , Camundongos , Receptores CXCR3/metabolismo , Transdução de Sinais
17.
PLoS Genet ; 11(5): e1005211, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25973911

RESUMO

Down syndrome (DS), with trisomy of chromosome 21 (HSA21), is the commonest human aneuploidy. Pre-leukemic myeloproliferative changes in DS foetal livers precede the acquisition of GATA1 mutations, transient myeloproliferative disorder (DS-TMD) and acute megakaryocytic leukemia (DS-AMKL). Trisomy of the Erg gene is required for myeloproliferation in the Ts(1716)65Dn DS mouse model. We demonstrate here that genetic changes specifically attributable to trisomy of Erg lead to lineage priming of primitive and early multipotential progenitor cells in Ts(1716)65Dn mice, excess megakaryocyte-erythroid progenitors, and malignant myeloproliferation. Gene expression changes dependent on trisomy of Erg in Ts(1716)65Dn multilineage progenitor cells were correlated with those associated with trisomy of HSA21 in human DS hematopoietic stem and primitive progenitor cells. These data suggest a role for ERG as a regulator of hematopoietic lineage potential, and that trisomy of ERG in the context of DS foetal liver hemopoiesis drives the pre-leukemic changes that predispose to subsequent DS-TMD and DS-AMKL.


Assuntos
Cromossomos Humanos Par 21/genética , Síndrome de Down/genética , Proteínas Oncogênicas/genética , Células-Tronco/citologia , Transativadores/genética , Fatores de Transcrição/genética , Trissomia , ADP-Ribosil Ciclase 1/metabolismo , Alelos , Animais , Antígenos CD34/metabolismo , Linhagem da Célula , Proliferação de Células , Modelos Animais de Doenças , Células Eritroides/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genótipo , Hematopoese/genética , Sistema Hematopoético/citologia , Sistema Hematopoético/metabolismo , Humanos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Análise em Microsséries , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Análise de Sequência de RNA , Células-Tronco/metabolismo , Regulador Transcricional ERG , Transcriptoma
18.
Eur Arch Otorhinolaryngol ; 275(8): 2127-2134, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29951933

RESUMO

OBJECTIVE: The study aimed to explore some novel diagnostic biomarkers for papillary thyroid carcinoma (PTC) by identifying the different expression of TROP-2, SLP-2 and CD56 in benign and malignant thyroid lesions. METHODS: We evaluated the mRNA expressions of TROP-2 and SLP-2 in fine needle aspirates (FNAs) which contained 10 PTCs and 10 benign follicular adenomas (FAs) using quantitative real-time PCR (qRT-PCR). Immunohistochemical (IHC) staining of TROP-2, SLP-2 and CD56 was also performed on postoperative samples of 30 PTCs and 29 FAs. Membranous or cytoplasmic staining in > 10% of cells was considered as positive. Diagnostic sensitivity, specificity, positive predictive value, negative predictive value (NPV) and diagnostic accuracy of these three biomarkers were carried out. We further analyzed the associations between the clinical features and the expressions of markers in PTCs. RESULTS: The mRNA expressions of both TROP-2 and SLP-2 were increased substantially in PTCs in comparison with those in FAs (P < 0.05). Similarly, IHC for these two proteins demonstrated higher positive staining in PTCs than in FAs (96.5% vs. 12.5% for TROP-2, 83.3% vs. 20.7% for SLP-2, P < 0.05). Conversely, CD56 expression was lost with 86.7% of PTCs. In identifying malignancy, TROP-2 was the most sensitive marker and CD56 was the most specific one. When the markers were combined, the sensitivity and NPV increased to 100% and had better diagnostic accuracy. However, no association was found between biomarker expressions and clinicopathological factors in PTCs. CONCLUSIONS: We found that TROP-2, SLP-2 and CD56 were effective diagnostic markers for PTC, especially when they were combined to use.


Assuntos
Antígenos de Neoplasias/genética , Proteínas Sanguíneas/genética , Antígeno CD56/genética , Carcinoma Papilar/diagnóstico , Moléculas de Adesão Celular/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/genética , RNA Neoplásico/genética , Neoplasias da Glândula Tireoide/diagnóstico , Adulto , Antígenos de Neoplasias/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Biópsia por Agulha Fina , Proteínas Sanguíneas/biossíntese , Antígeno CD56/biossíntese , Carcinoma Papilar/metabolismo , Moléculas de Adesão Celular/biossíntese , Contagem de Células , Feminino , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/metabolismo
19.
J Biol Chem ; 291(41): 21703-21716, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27539849

RESUMO

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are IL-6 family members with a wide range of biological functions. Human OSM (hOSM) and murine LIF (mLIF) act in mouse cells via a LIF receptor (LIFR)-glycoprotein 130 (gp130) heterodimer. In contrast, murine OSM (mOSM) signals mainly via an OSM receptor (OSMR)-gp130 heterodimer and binds with only very low affinity to mLIFR. hOSM and mLIF stimulate bone remodeling by both reducing osteocytic sclerostin and up-regulating the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) in osteoblasts. In the absence of OSMR, mOSM still strongly suppressed sclerostin and stimulated bone formation but did not induce RANKL, suggesting that intracellular signaling activated by the low affinity interaction of mOSM with mLIFR is different from the downstream effects when mLIF or hOSM interacts with the same receptor. Both STAT1 and STAT3 were activated by mOSM in wild type cells or by mLIF/hOSM in wild type and Osmr-/- cells. In contrast, in Osmr-/- primary osteocyte-like cells stimulated with mOSM (therefore acting through mLIFR), microarray expression profiling and Western blotting analysis identified preferential phosphorylation of STAT3 and induction of its target genes but not of STAT1 and its target genes; this correlated with reduced phosphorylation of both gp130 and LIFR. In a mouse model of spontaneous osteopenia caused by hyperactivation of STAT1/3 signaling downstream of gp130 (gp130Y757F/Y757F), STAT1 deletion rescued the osteopenic phenotype, indicating a beneficial effect of promoting STAT3 signaling over STAT1 downstream of gp130 in this low bone mass condition, and this may have therapeutic value.


Assuntos
Doenças Ósseas Metabólicas/metabolismo , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/metabolismo , Oncostatina M/metabolismo , Osteócitos/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/patologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Modelos Animais de Doenças , Humanos , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/genética , Camundongos , Oncostatina M/genética , Subunidade beta de Receptor de Oncostatina M/genética , Subunidade beta de Receptor de Oncostatina M/metabolismo , Tamanho do Órgão , Osteócitos/patologia , Fosforilação/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/genética
20.
Blood ; 125(12): 1910-21, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25605372

RESUMO

The histone acetyltransferase MOZ (MYST3, KAT6A) is the target of recurrent chromosomal translocations fusing the MOZ gene to CBP, p300, NCOA3, or TIF2 in particularly aggressive cases of acute myeloid leukemia. In this study, we report the role of wild-type MOZ in regulating B-cell progenitor proliferation and hematopoietic malignancy. In the Eµ-Myc model of aggressive pre-B/B-cell lymphoma, the loss of just one allele of Moz increased the median survival of mice by 3.9-fold. MOZ was required to maintain the proliferative capacity of B-cell progenitors, even in the presence of c-MYC overexpression, by directly maintaining the transcriptional activity of genes required for normal B-cell development. Hence, B-cell progenitor numbers were significantly reduced in Moz haploinsufficient animals. Interestingly, we find a significant overlap in genes regulated by MOZ, mixed lineage leukemia 1, and mixed lineage leukemia 1 cofactor menin. This includes Meis1, a TALE class homeobox transcription factor required for B-cell development, characteristically upregulated as a result of MLL1 translocations in leukemia. We demonstrate that MOZ localizes to the Meis1 locus in pre-B-cells and maintains Meis1 expression. Our results suggest that even partial inhibition of MOZ may reduce the proliferative capacity of MEIS1, and HOX-driven lymphoma and leukemia cells.


Assuntos
Linfócitos B/citologia , Regulação Neoplásica da Expressão Gênica , Histona Acetiltransferases/genética , Linfoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Células-Tronco/citologia , Alelos , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Senescência Celular , Feminino , Haploinsuficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência de RNA , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA