Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 23(7): 1076-1085, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35761085

RESUMO

Memory B cells persist for a lifetime and rapidly differentiate into antibody-producing plasmablasts and plasma cells upon antigen re-encounter. The clonal relationship and evolution of memory B cells and circulating plasmablasts is not well understood. Using single-cell sequencing combined with isolation of specific antibodies, we found that in two healthy donors, the memory B cell repertoire was dominated by large IgM, IgA and IgG2 clonal families, whereas IgG1 families, including those specific for recall antigens, were of small size. Analysis of multiyear samples demonstrated stability of memory B cell clonal families and revealed that a large fraction of recently generated plasmablasts was derived from long-term memory B cell families and was found recurrently. Collectively, this study provides a systematic description of the structure, stability and dynamics of the human memory B cell pool and suggests that memory B cells may be active at any time point in the generation of plasmablasts.


Assuntos
Células B de Memória , Plasmócitos , Linfócitos B , Células Cultivadas , Humanos , Imunoglobulina G , Memória Imunológica
2.
Hum Mol Genet ; 30(13): 1175-1187, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33601422

RESUMO

Synaptic dysfunction and cognitive decline in Huntington's disease (HD) involve hyperactive A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). To identify the molecular mechanisms through which ADAM10 is associated with synaptic dysfunction in HD, we performed an immunoaffinity purification-mass spectrometry (IP-MS) study of endogenous ADAM10 in the brains of wild-type and HD mice. We found that proteins implicated in synapse organization, synaptic plasticity, and vesicle and organelles trafficking interact with ADAM10, suggesting that it may act as hub protein at the excitatory synapse. Importantly, the ADAM10 interactome is enriched in presynaptic proteins and ADAM10 co-immunoprecipitates with piccolo (PCLO), a key player in the recycling and maintenance of synaptic vesicles. In contrast, reduced ADAM10/PCLO immunoprecipitation occurs in the HD brain, with decreased density of synaptic vesicles in the reserve and docked pools at the HD presynaptic terminal. Conditional heterozygous deletion of ADAM10 in the forebrain of HD mice reduces active ADAM10 to wild-type level and normalizes ADAM10/PCLO complex formation and synaptic vesicle density and distribution. The results indicate that presynaptic ADAM10 and PCLO are a relevant component of HD pathogenesis.


Assuntos
Proteína ADAM10/metabolismo , Proteínas do Citoesqueleto/metabolismo , Doença de Huntington/metabolismo , Neuropeptídeos/metabolismo , Vesículas Sinápticas/metabolismo , Proteína ADAM10/genética , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Humanos , Doença de Huntington/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Terminações Pré-Sinápticas/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas/genética , Proteômica/métodos , Vesículas Sinápticas/ultraestrutura , Sinaptossomos/metabolismo , Espectrometria de Massas em Tandem/métodos
3.
Nat Immunol ; 12(8): 796-803, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706005

RESUMO

MicroRNAs are small noncoding RNAs that regulate gene expression post-transcriptionally. Here we applied microRNA profiling to 17 human lymphocyte subsets to identify microRNA signatures that were distinct among various subsets and different from those of mouse lymphocytes. One of the signature microRNAs of naive CD4+ T cells, miR-125b, regulated the expression of genes encoding molecules involved in T cell differentiation, including IFNG, IL2RB, IL10RA and PRDM1. The expression of synthetic miR-125b and lentiviral vectors encoding the precursor to miR-125b in naive lymphocytes inhibited differentiation to effector cells. Our data provide an 'atlas' of microRNA expression in human lymphocytes, define subset-specific signatures and their target genes and indicate that the naive state of T cells is enforced by microRNA.


Assuntos
Linfócitos T CD4-Positivos/imunologia , MicroRNAs/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Biologia Computacional/métodos , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Hematol Oncol ; 40(4): 541-553, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35451108

RESUMO

The spatial architecture of the lymphoid tissue in follicular lymphoma (FL) presents unique challenges to studying its immune microenvironment. We investigated the spatial interplay of T cells, macrophages, myeloid cells and natural killer T cells using multispectral immunofluorescence images of diagnostic biopsies of 32 patients. A deep learning-based image analysis pipeline was tailored to the needs of follicular lymphoma spatial histology research, enabling the identification of different immune cells within and outside neoplastic follicles. We analyzed the density and spatial co-localization of immune cells in the inter-follicular and intra-follicular regions of follicular lymphoma. Low inter-follicular density of CD8+FOXP3+ cells and co-localization of CD8+FOXP3+ with CD4+CD8+ cells were significantly associated with relapse (p = 0.0057 and p = 0.0019, respectively) and shorter time to progression after first-line treatment (Logrank p = 0.0097 and log-rank p = 0.0093, respectively). A low inter-follicular density of CD8+FOXP3+ cells is associated with increased risk of relapse independent of follicular lymphoma international prognostic index (FLIPI) (p = 0.038, Hazard ratio (HR) = 0.42 [0.19, 0.95], but not independent of co-localization of CD8+FOXP3+ with CD4+CD8+ cells (p = 0.43). Co-localization of CD8+FOXP3+ with CD4+CD8+ cells is predictors of time to relapse independent of the FLIPI score and density of CD8+FOXP3+ cells (p = 0.027, HR = 0.0019 [7.19 × 10-6 , 0.49], This suggests a potential role of inter-follicular CD8+FOXP3+ and CD4+CD8+ cells in the disease progression of FL, warranting further validation on larger patient cohorts.


Assuntos
Linfoma Folicular , Linfócitos T CD8-Positivos , Fatores de Transcrição Forkhead , Humanos , Linfoma Folicular/patologia , Recidiva Local de Neoplasia , Prognóstico , Microambiente Tumoral
5.
Nucleic Acids Res ; 47(10): 5325-5340, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30937446

RESUMO

Stem cell identity and plasticity are controlled by master regulatory genes and complex circuits also involving non-coding RNAs. Circular RNAs (circRNAs) are a class of RNAs generated from protein-coding genes by backsplicing, resulting in stable RNA structures devoid of free 5' and 3' ends. Little is known of the mechanisms of action of circRNAs, let alone in stem cell biology. In this study, for the first time, we determined that a circRNA controls mesenchymal stem cell (MSC) identity and differentiation. High-throughput MSC expression profiling from different tissues revealed a large number of expressed circRNAs. Among those, circFOXP1 was enriched in MSCs compared to differentiated mesodermal derivatives. Silencing of circFOXP1 dramatically impaired MSC differentiation in culture and in vivo. Furthermore, we demonstrated a direct interaction between circFOXP1 and miR-17-3p/miR-127-5p, which results in the modulation of non-canonical Wnt and EGFR pathways. Finally, we addressed the interplay between canonical and non-canonical Wnt pathways. Reprogramming to pluripotency of MSCs reduced circFOXP1 and non-canonical Wnt, whereas canonical Wnt was boosted. The opposing effect was observed during generation of MSCs from human pluripotent stem cells. Our results provide unprecedented evidence for a regulatory role for circFOXP1 as a gatekeeper of pivotal stem cell molecular networks.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , MicroRNAs/metabolismo , RNA , Proteínas Repressoras/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Proliferação de Células , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Exorribonucleases/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Inativação Gênica , Células HEK293 , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/citologia , Mesoderma/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/citologia , RNA Circular , RNA Interferente Pequeno/metabolismo , Análise de Sequência de RNA , Células-Tronco/citologia , Proteínas Wnt/metabolismo
6.
Neurobiol Dis ; 146: 105140, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33065279

RESUMO

RUES2 cell lines represent the first collection of isogenic human embryonic stem cells (hESCs) carrying different pathological CAG lengths in the HTT gene. However, their neuronal differentiation potential has yet to be thoroughly evaluated. Here, we report that RUES2 during ventral telencephalic differentiation is biased towards medial ganglionic eminence (MGE). We also show that HD-RUES2 cells exhibit an altered MGE transcriptional signature in addition to recapitulating known HD phenotypes, with reduced expression of the neurodevelopmental regulators NEUROD1 and BDNF and increased cleavage of synaptically enriched N-cadherin. Finally, we identified the transcription factor SP1 as a common potential detrimental co-partner of muHTT by de novo motif discovery analysis on the LGE, MGE, and cortical genes differentially expressed in HD human pluripotent stem cells in our and additional datasets. Taken together, these observations suggest a broad deleterious effect of muHTT in the early phases of neuronal development that may unfold through its altered interaction with SP1.


Assuntos
Biomarcadores Tumorais/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes/citologia , Receptores Imunológicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/patologia , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo
7.
Eur J Immunol ; 49(1): 96-111, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30431161

RESUMO

Whether human IL-10-producing regulatory T cells ("Tr1") represent a distinct differentiation lineage or an unstable activation stage remains a key unsolved issue. Here, we report that Eomesodermin (Eomes) acted as a lineage-defining transcription factor in human IFN-γ/IL-10 coproducing Tr1-like cells. In vivo occurring Tr1-like cells expressed Eomes, and were clearly distinct from all other CD4+ T-cell subsets, including conventional cytotoxic CD4+ T cells. They expressed Granzyme (Gzm) K, but had lost CD40L and IL-7R expression. Eomes antagonized the Th17 fate, and directly controlled IFN-γ and GzmK expression. However, Eomes binding to the IL-10 promoter was not detectable in human CD4+ T cells, presumably because critical Tbox binding sites of the mouse were not conserved. A precommitment to a Tr1-like fate, i.e. concominant induction of Eomes, GzmK, and IFN-γ, was promoted by IL-4 and IL-12-secreting myeloid dendritic cells. Consistently, Th1 effector memory cells contained precommitted Eomes+ GzmK+ T cells. Stimulation with T-cell receptor (TCR) agonists and IL-27 promoted the generation of Tr1-like effector cells by inducing switching from CD40L to IL-10. Importantly, CD4+ Eomes+ T-cell subsets were present in lymphoid and nonlymphoid tissues, and their frequencies varied systemically in patients with inflammatory bowel disease and graft-versus-host disease. We propose that Eomes+ Tr1-like cells are effector cells of a unique GzmK-expressing CD4+ T-cell subset.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Doenças Inflamatórias Intestinais/imunologia , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Regulação da Expressão Gênica , Granzimas/metabolismo , Humanos , Memória Imunológica , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos , Proteínas com Domínio T/genética
9.
Cereb Cortex ; 28(5): 1846-1856, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351579

RESUMO

During differentiation, neurons progressively restrict their fate repressing the expression of specific genes. Here we describe the involvement in such developmental steps of the methyl-CpG binding protein 2 (MeCP2), an epigenetic factor that participates to chromatin folding and transcriptional regulation. We previously reported that, due to transcriptional impairments, the maturation of Mecp2 null neurons is delayed. To evaluate whether this could stem from altered progenitors proliferation and differentiation, we investigated whether lack of Mecp2 affects these features both in vitro and in vivo. We show that in Mecp2 null embryonic cortexes the expression of genes defining the identity of proliferating neuroprogenitors is enriched and that their permanence in the G1 phase is prolonged. Moreover, the number of cells transitioning from a stage of maturation to a more mature one is increased in Mecp2 null embryonic cortices, in line with the central role of G1 for cell identity refinement. We thus suggest that, possibly due to the lack of proper transcriptional control normally exerted by Mecp2, fate refinement is impaired in developing null cells. We propose that the maturation delay affecting the developing Mecp2 null cortex originates, at least in part, from deranged mechanisms of cell fate refinement.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína 2 de Ligação a Metil-CpG/deficiência , Neurônios/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina , Células Cultivadas , Ciclina D1/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidade , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Feminino , Fatores de Crescimento de Fibroblastos/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro
10.
J Biol Chem ; 292(7): 2903-2915, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28077577

RESUMO

Upon T cell receptor stimulation, CD4+ T helper (Th) lymphocytes release extracellular vesicles (EVs) containing microRNAs. However, no data are available on whether human CD4+ T cell subsets release EVs containing different pattern of microRNAs. The present work aimed at filling this gap by assessing the microRNA content in EVs released upon in vitro T cell receptor stimulation of Th1, Th17, and T regulatory (Treg) cells. Our results indicate that EVs released by Treg cells are significantly different compared with those released by the other subsets. In particular, miR-146a-5p, miR-150-5p, and miR-21-5p are enriched, whereas miR-106a-5p, miR-155-5p, and miR-19a-3p are depleted in Treg-derived EVs. The in vitro identified EV-associated microRNA signature was increased in serum of autoimmune patients with psoriasis and returned to healthy levels upon effective treatment with etanercept, a biological drug targeting the TNF pathway and suppressing inflammation. Moreover, Gene Set Enrichment Analysis showed an over-representation of genes relevant for T cell activation, such as CD40L, IRAK1, IRAK2, STAT1, and c-Myb in the list of validated targets of Treg-derived EV miRNAs. At functional level, Treg-derived (but not Th1/Th17-derived) EVs inhibited CD4+ T cell proliferation and suppressed two relevant targets of miR-146a-5p: STAT1 and IRAK2. In conclusion, our work identified the miRNAs specifically released by different human CD4+ T cell subsets and started to unveil the potential use of their quantity in human serum to mark the pathological elicitation of these cells in vivo and their biological effect in cell to cell communication during the adaptive immune response.


Assuntos
Doenças Autoimunes/genética , Linfócitos T CD4-Positivos/citologia , Vesículas Extracelulares/metabolismo , MicroRNAs/genética , Subpopulações de Linfócitos T , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Etanercepte/uso terapêutico , Humanos , MicroRNAs/sangue , Psoríase/sangue , Psoríase/tratamento farmacológico , Psoríase/genética
11.
Brief Bioinform ; 17(2): 204-12, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26238539

RESUMO

The high-throughput analysis of microRNAs (miRNAs) circulating within the blood of healthy and diseased individuals is an active area of biomarker research. Whereas quantitative real-time reverse transcription polymerase chain reaction (qPCR)-based methods are widely used, it is yet unresolved how the data should be normalized. Here, we show that a combination of different algorithms results in the identification of candidate reference miRNAs that can be exploited as normalizers, in both discovery and validation phases. Using the methodology considered here, we identify normalizers that are able to reduce nonbiological variation in the data and we present several case studies, to illustrate the relevance in the context of physiological or pathological scenarios. In conclusion, the discovery of stable reference miRNAs from high-throughput studies allows appropriate normalization of focused qPCR assays.


Assuntos
Algoritmos , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , MicroRNAs/sangue , MicroRNAs/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Biomarcadores/sangue , Perfilação da Expressão Gênica/normas , Sequenciamento de Nucleotídeos em Larga Escala/normas , Humanos , MicroRNAs/normas , Reação em Cadeia da Polimerase em Tempo Real/normas , Valores de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
12.
Nucleic Acids Res ; 43(W1): W487-92, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25897123

RESUMO

The miRBase is the official miRNA repository which keeps the annotation updated on newly discovered miRNAs: it is also used as a reference for the design of miRNA profiling platforms. Nomenclature ambiguities generated by loosely updated platforms and design errors lead to incompatibilities among platforms, even from the same vendor. Published miRNA lists are thus generated with different profiling platforms that refer to diverse and not updated annotations. This greatly compromises searches, comparisons and analyses that rely on miRNA names only without taking into account the mature sequences, which is particularly critic when such analyses are carried over automatically. In this paper we introduce miRiadne, a web tool to harmonize miRNA nomenclature, which takes into account the original miRBase versions from 10 up to 21, and annotations of 40 common profiling platforms from nine brands that we manually curated. miRiadne uses the miRNA mature sequence to link miRBase versions and/or platforms to prevent nomenclature ambiguities. miRiadne was designed to simplify and support biologists and bioinformaticians in re-annotating their own miRNA lists and/or data sets. As Ariadne helped Theseus in escaping the mythological maze, miRiadne will help the miRNA researcher in escaping the nomenclature maze. miRiadne is freely accessible from the URL http://www.miriadne.org.


Assuntos
MicroRNAs/química , Anotação de Sequência Molecular , Software , Terminologia como Assunto , Internet , MicroRNAs/metabolismo
13.
Immunol Rev ; 253(1): 82-96, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23550640

RESUMO

CD4(+) T lymphocytes orchestrate adaptive immune responses by differentiating into various subsets of effector T cells such as T-helper 1 (Th1), Th2, Th17, and regulatory T cells. These subsets have been generally described by master transcription factors that dictate the expression of cytokines and receptors, which ultimately define lymphocyte effector functions. However, the view of T-lymphocyte subsets as stable and terminally differentiated lineages has been challenged by increasing evidence of functional plasticity within CD4(+) T-cell subsets, which implies flexible programming of effector functions depending on time and space of T-cell activation. An outstanding question with broad basic and traslational implications relates to the mechanisms, besides transcriptional regulation, which define the plasticity of effector functions. In this study, we discuss the emerging role of regulatory non-coding RNAs in T-cell differentiation and plasticity. Not only microRNAs have been proven to be important for CD4(+) T-cell differentiation, but it is also likely that the overall T-cell functioning is the result of a multilayered network composed by coding RNAs as well as by short and long non-coding RNAs. The integrated study of all the nodes of this network will provide a comprehensive view of the molecular mechanisms underlying T-cell functions in health and disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , MicroRNAs/imunologia , RNA Longo não Codificante/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa , Animais , Comunicação Celular , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Humanos , Imunomodulação , Ativação Linfocitária/genética , Equilíbrio Th1-Th2 , Células Th17/imunologia
14.
Am J Pathol ; 183(2): 413-21, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23731727

RESUMO

Despite the evidence that tumor necrosis factor (TNF) inhibitors block TNF and the downstream inflammatory cascade, their primary mechanism of action in inhibiting the self-sustaining pathogenic cycle in psoriasis is not completely understood. This study has the aim to identify early critical events for the resolution of inflammation in skin lesions using anti-TNF therapy. We used a translational approach that correlates gene expression fold change in lesional skin with the Psoriasis Area and Severity Index score decrease induced by TNF blockade after 4 weeks of treatment. Data were validated by immunofluorescence microscopy on skin biopsy specimens. We found that the anti-TNF-modulated genes that mostly associated with the clinical amelioration were Ccr7, its ligand, Ccl19, and dendritic cell maturation genes. Decreased expression of T-cell activation genes and Vegf also associated with the clinical response. More important, the down-regulation of Ccr7 observed at 4 weeks significantly correlated with the clinical remission occurring at later time points. Immunofluorescence microscopy on skin biopsy specimens showed that reduction of CCR7(+) cells and chemokine ligand (CCL) 19 was paralleled by disaggregation of the dermal lymphoid-like tissue. These data show that an early critical event for the clinical remission of psoriasis in response to TNF inhibitors is the inhibition of the CCR7/CCL19 axis and support its role in psoriasis pathogenesis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Quimiocina CCL19/antagonistas & inibidores , Psoríase/tratamento farmacológico , Receptores CCR7/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adalimumab , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos CD4/metabolismo , Senescência Celular/efeitos dos fármacos , Quimiocina CCL19/genética , Etanercepte , Feminino , Humanos , Imunoglobulina G/uso terapêutico , Infliximab , Células de Langerhans/efeitos dos fármacos , Ativação Linfocitária/genética , Masculino , Pessoa de Meia-Idade , Psoríase/genética , Receptores CCR7/genética , Receptores do Fator de Necrose Tumoral/uso terapêutico , Indução de Remissão , Pele/metabolismo , Linfócitos T/efeitos dos fármacos
15.
PLoS Biol ; 9(6): e1001086, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21738444

RESUMO

The basic unit of genome packaging is the nucleosome, and nucleosomes have long been proposed to restrict DNA accessibility both to damage and to transcription. Nucleosome number in cells was considered fixed, but recently aging yeast and mammalian cells were shown to contain fewer nucleosomes. We show here that mammalian cells lacking High Mobility Group Box 1 protein (HMGB1) contain a reduced amount of core, linker, and variant histones, and a correspondingly reduced number of nucleosomes, possibly because HMGB1 facilitates nucleosome assembly. Yeast nhp6 mutants lacking Nhp6a and -b proteins, which are related to HMGB1, also have a reduced amount of histones and fewer nucleosomes. Nucleosome limitation in both mammalian and yeast cells increases the sensitivity of DNA to damage, increases transcription globally, and affects the relative expression of about 10% of genes. In yeast nhp6 cells the loss of more than one nucleosome in four does not affect the location of nucleosomes and their spacing, but nucleosomal occupancy. The decrease in nucleosomal occupancy is non-uniform and can be modelled assuming that different nucleosomal sites compete for available histones. Sites with a high propensity to occupation are almost always packaged into nucleosomes both in wild type and nucleosome-depleted cells; nucleosomes on sites with low propensity to occupation are disproportionately lost in nucleosome-depleted cells. We suggest that variation in nucleosome number, by affecting nucleosomal occupancy both genomewide and gene-specifically, constitutes a novel layer of epigenetic regulation.


Assuntos
Genoma , Proteína HMGB1/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Transcrição Gênica , Animais , DNA/genética , DNA/metabolismo , Dano ao DNA , Epigênese Genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteína HMGB1/genética , Células HeLa , Histonas/genética , Humanos , Camundongos , Modelos Teóricos , RNA/genética , RNA/metabolismo , Leveduras/genética , Leveduras/metabolismo
16.
Blood Adv ; 8(11): 2880-2889, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38593222

RESUMO

ABSTRACT: Inhibitor development is the most severe complication of hemophilia A (HA) care and is associated with increased morbidity and mortality. This study aimed to use a novel immunoglobulin G epitope mapping method to explore the factor VIII (FVIII)-specific epitope profile in the SIPPET cohort population and to develop an epitope mapping-based inhibitor prediction model. The population consisted of 122 previously untreated patients with severe HA who were followed up for 50 days of exposure to FVIII or 3 years, whichever occurred first. Sampling was performed before FVIII treatment and at the end of the follow-up. The outcome was inhibitor development. The FVIII epitope repertoire was assessed by means of a novel random peptide phage-display assay. A least absolute shrinkage and selection operator (LASSO) regression model and a random forest model were fitted on posttreatment sample data and validated in pretreatment sample data. The predictive performance of these models was assessed by the C-statistic and a calibration plot. We identified 27 775 peptides putatively directed against FVIII, which were used as input for the statistical models. The C-statistic of the LASSO and random forest models were good at 0.78 (95% confidence interval [CI], 0.69-0.86) and 0.80 (95% CI, 0.72-0.89). Model calibration of both models was moderately good. Two statistical models, developed on data from a novel random peptide phage display assay, were used to predict inhibitor development before exposure to exogenous FVIII. These models can be used to set up diagnostic tests that predict the risk of inhibitor development before starting treatment with FVIII.


Assuntos
Fator VIII , Hemofilia A , Biblioteca de Peptídeos , Humanos , Fator VIII/imunologia , Mapeamento de Epitopos , Masculino , Estudos de Coortes , Epitopos/imunologia
17.
Sci Rep ; 11(1): 18043, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34508139

RESUMO

Interferons (IFNs) are key cytokines involved in alerting the immune system to viral infection. After IFN stimulation, cellular transcriptional profile critically changes, leading to the expression of several IFN stimulated genes (ISGs) that exert a wide variety of antiviral activities. Despite many ISGs have been already identified, a comprehensive network of coding and non-coding genes with a central role in IFN-response still needs to be elucidated. We performed a global RNA-Seq transcriptome profile of the HCV permissive human hepatoma cell line Huh7.5 and its parental cell line Huh7, upon IFN treatment, to define a network of genes whose coordinated modulation plays a central role in IFN-response. Our study adds molecular actors, coding and non-coding genes, to the complex molecular network underlying IFN-response and shows how systems biology approaches, such as correlation networks, network's topology and gene ontology analyses can be leveraged to this aim.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética , Interferons/metabolismo , Biologia de Sistemas/métodos , Transcriptoma , Sítios de Ligação , Linhagem Celular Tumoral , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Fatores Reguladores de Interferon/metabolismo , Interferons/farmacologia , Neoplasias Hepáticas , Motivos de Nucleotídeos , Ligação Proteica
18.
Cancer Res ; 80(21): 4693-4706, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32963011

RESUMO

Multiple myeloma is a plasma cell neoplasm characterized by the production of unfolded immunoglobulins, which cause endoplasmic reticulum (ER) stress and sensitivity to proteasome inhibition. The genomic landscape of multiple myeloma is characterized by the loss of several genes rarely mutated in other cancers that may underline specific weaknesses of multiple myeloma cells. One of these is FAM46C that is lost in more than 10% of patients with multiple myeloma. We show here that FAM46C is part of a new complex containing the ER-associated protein FNDC3A, which regulates trafficking and secretion and, by impairing autophagy, exacerbates proteostatic stress. Reconstitution of FAM46C in multiple myeloma cells that had lost it induced apoptosis and ER stress. Apoptosis was preceded by an increase of intracellular aggregates, which was not linked to increased translation of IgG mRNA, but rather to impairment of autophagy. Biochemical analysis showed that FAM46C requires interaction with ER bound protein FNDC3A to reside in the cytoplasmic side of the ER. FNDC3A was lost in some multiple myeloma cell lines. Importantly, depletion of FNDC3A increased the fitness of FAM46C-expressing cells and expression of FNDC3A in cells that had lost it recapitulated the effects of FAM46C, inducing aggregates and apoptosis. FAM46C and FNDC3A formed a complex that modulates secretion routes, increasing lysosome exocytosis. The cellular landscape generated by FAM46C/FNDC3A expression predicted sensitivity to sphingosine kinase inhibition. These results suggest that multiple myeloma cells remodel their trafficking machinery to cope with ER stress. SIGNIFICANCE: This study identifies a new multiple myeloma-specific tumor suppressor complex that regulates autophagy and unconventional secretion, highlighting the sensitivity of multiple myeloma cells to the accumulation of protein aggregates.


Assuntos
Fibronectinas/metabolismo , Mieloma Múltiplo/patologia , Nucleotidiltransferases/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Autofagia/fisiologia , Genes Supressores de Tumor , Xenoenxertos , Humanos , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Nucleotidiltransferases/genética , Agregados Proteicos/fisiologia , Transporte Proteico/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
19.
J Cell Biol ; 167(3): 433-43, 2004 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-15520229

RESUMO

Saccharomyces cerevisiae must reach a carbon source-modulated critical cell size, protein content per cell at the onset of DNA replication (Ps), in order to enter S phase. Cells grown in glucose are larger than cells grown in ethanol. Here, we show that an increased level of the cyclin-dependent inhibitor Far1 increases cell size, whereas far1 Delta cells start bud emergence and DNA replication at a smaller size than wild type. Cln3 Delta, far1 Delta, and strains overexpressing Far1 do not delay budding during an ethanol glucose shift-up as wild type does. Together, these findings indicate that Cln3 has to overcome Far1 to trigger Cln-Cdc28 activation, which then turns on SBF- and MBF-dependent transcription. We show that a second threshold is required together with the Cln3/Far1 threshold for carbon source modulation of Ps. A new molecular network accounting for the setting of Ps is proposed.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclinas/fisiologia , Mitose , Proteínas Repressoras/fisiologia , Fase S , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomycetales/citologia , Proteína Quinase CDC28 de Saccharomyces cerevisiae/metabolismo , Tamanho Celular , Proteínas Inibidoras de Quinase Dependente de Ciclina , Replicação do DNA , Etanol/metabolismo , Glucose/metabolismo , Saccharomycetales/química , Saccharomycetales/metabolismo , Transcrição Gênica
20.
Methods Mol Biol ; 1959: 247-259, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30852827

RESUMO

The diagnostic accuracy of biomarker-based approaches can be considerably improved by combining multiple markers. A biomarker's capacity to identify specific subjects is usually assessed using receiver operating characteristic (ROC) curves. Multimarker signatures are complicated to select as data signatures must be integrated using sophisticated statistical methods. CombiROC, developed as a user-friendly web tool, helps researchers to accurately determine optimal combinations of markers identified by a range of omics methods. With CombiROC, data of different types, such as proteomics and transcriptomics, can be analyzed using Sensitivity/Specificity filters: the number of candidate marker panels arising from combinatorial analysis is easily optimized bypassing limitations imposed by the nature of different experimental approaches. Users have full control over initial selection stringency, then CombiROC computes sensitivity and specificity for all marker combinations, determines performance for the best combinations, and produces ROC curves for automatic comparisons. All steps can be visualized in a graphic interface. CombiROC is designed without hard-coded thresholds, to allow customized fitting of each specific dataset: this approach dramatically reduces computational burden and false-negative rates compared to fixed thresholds. CombiROC can be accessed at www.combiroc.eu .


Assuntos
Biomarcadores , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Proteômica , Software , Interpretação Estatística de Dados , Perfilação da Expressão Gênica/métodos , Humanos , Proteoma , Proteômica/métodos , Curva ROC , Transcriptoma , Interface Usuário-Computador
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA