Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell ; 182(4): 1009-1026.e29, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32730809

RESUMO

Electrophilic compounds originating from nature or chemical synthesis have profound effects on immune cells. These compounds are thought to act by cysteine modification to alter the functions of immune-relevant proteins; however, our understanding of electrophile-sensitive cysteines in the human immune proteome remains limited. Here, we present a global map of cysteines in primary human T cells that are susceptible to covalent modification by electrophilic small molecules. More than 3,000 covalently liganded cysteines were found on functionally and structurally diverse proteins, including many that play fundamental roles in immunology. We further show that electrophilic compounds can impair T cell activation by distinct mechanisms involving the direct functional perturbation and/or degradation of proteins. Our findings reveal a rich content of ligandable cysteines in human T cells and point to electrophilic small molecules as a fertile source for chemical probes and ultimately therapeutics that modulate immunological processes and their associated disorders.


Assuntos
Cisteína/metabolismo , Ligantes , Linfócitos T/metabolismo , Acetamidas/química , Acetamidas/farmacologia , Acrilamidas/química , Acrilamidas/farmacologia , Células Cultivadas , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteólise/efeitos dos fármacos , Proteoma/química , Proteoma/metabolismo , Estereoisomerismo , Linfócitos T/citologia , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/metabolismo
2.
Nat Immunol ; 23(7): 1086-1097, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35739197

RESUMO

Maintenance of tissue homeostasis is dependent on the communication between stem cells and supporting cells in the same niche. Regulatory T cells (Treg cells) are emerging as a critical component of the stem-cell niche for supporting their differentiation. How Treg cells sense dynamic signals in this microenvironment and communicate with stem cells is mostly unknown. In the present study, by using hair follicles (HFs) to study Treg cell-stem cell crosstalk, we show an unrecognized function of the steroid hormone glucocorticoid in instructing skin-resident Treg cells to facilitate HF stem-cell (HFSC) activation and HF regeneration. Ablation of the glucocorticoid receptor (GR) in Treg cells blocks hair regeneration without affecting immune homeostasis. Mechanistically, GR and Foxp3 cooperate in Treg cells to induce transforming growth factor ß3 (TGF-ß3), which activates Smad2/3 in HFSCs and facilitates HFSC proliferation. The present study identifies crosstalk between Treg cells and HFSCs mediated by the GR-TGF-ß3 axis, highlighting a possible means of manipulating Treg cells to support tissue regeneration.


Assuntos
Glucocorticoides , Folículo Piloso , Glucocorticoides/metabolismo , Cabelo/metabolismo , Folículo Piloso/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta3/metabolismo
3.
Cell ; 173(3): 665-676.e14, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29551272

RESUMO

Class 2 CRISPR-Cas systems endow microbes with diverse mechanisms for adaptive immunity. Here, we analyzed prokaryotic genome and metagenome sequences to identify an uncharacterized family of RNA-guided, RNA-targeting CRISPR systems that we classify as type VI-D. Biochemical characterization and protein engineering of seven distinct orthologs generated a ribonuclease effector derived from Ruminococcus flavefaciens XPD3002 (CasRx) with robust activity in human cells. CasRx-mediated knockdown exhibits high efficiency and specificity relative to RNA interference across diverse endogenous transcripts. As one of the most compact single-effector Cas enzymes, CasRx can also be flexibly packaged into adeno-associated virus. We target virally encoded, catalytically inactive CasRx to cis elements of pre-mRNA to manipulate alternative splicing, alleviating dysregulated tau isoform ratios in a neuronal model of frontotemporal dementia. Our results present CasRx as a programmable RNA-binding module for efficient targeting of cellular RNA, enabling a general platform for transcriptome engineering and future therapeutic development.


Assuntos
Sistemas CRISPR-Cas , Biologia Computacional/métodos , Engenharia Genética/métodos , Engenharia de Proteínas/métodos , RNA/análise , Processamento Alternativo , Animais , Proteínas de Bactérias/metabolismo , Diferenciação Celular , Escherichia coli/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Lentivirus/genética , Camundongos , Interferência de RNA , RNA Guia de Cinetoplastídeos/genética , Ruminococcus , Análise de Sequência de RNA , Transcriptoma
4.
Cell ; 175(1): 146-158.e15, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30100182

RESUMO

Pathogen virulence exists on a continuum. The strategies that drive symptomatic or asymptomatic infections remain largely unknown. We took advantage of the concept of lethal dose 50 (LD50) to ask which component of individual non-genetic variation between hosts defines whether they survive or succumb to infection. Using the enteric pathogen Citrobacter, we found no difference in pathogen burdens between healthy and symptomatic populations. Iron metabolism-related genes were induced in asymptomatic hosts compared to symptomatic or naive mice. Dietary iron conferred complete protection without influencing pathogen burdens, even at 1000× the lethal dose of Citrobacter. Dietary iron induced insulin resistance, increasing glucose levels in the intestine that were necessary and sufficient to suppress pathogen virulence. A short course of dietary iron drove the selection of attenuated Citrobacter strains that can transmit and asymptomatically colonize naive hosts, demonstrating that environmental factors and cooperative metabolic strategies can drive conversion of pathogens toward commensalism.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Ferro/metabolismo , Virulência/fisiologia , Animais , Infecções Assintomáticas , Citrobacter rodentium/metabolismo , Citrobacter rodentium/patogenicidade , Colite/tratamento farmacológico , Colite/metabolismo , Colo/microbiologia , Suplementos Nutricionais , Infecções por Enterobacteriaceae/tratamento farmacológico , Feminino , Resistência à Insulina/fisiologia , Intestino Delgado/microbiologia , Ferro/farmacologia , Dose Letal Mediana , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos DBA
5.
Immunity ; 54(7): 1561-1577.e7, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34102100

RESUMO

A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.


Assuntos
Antígenos CD36/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Peroxidação de Lipídeos/fisiologia , Lipoproteínas LDL/metabolismo , Neoplasias/metabolismo , Receptores Depuradores/metabolismo , Animais , Transporte Biológico/fisiologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Leucócitos Mononucleares/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microambiente Tumoral/fisiologia
6.
Nature ; 614(7949): 767-773, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36755096

RESUMO

Cancers arise through the accumulation of genetic and epigenetic alterations that enable cells to evade telomere-based proliferative barriers and achieve immortality. One such barrier is replicative crisis-an autophagy-dependent program that eliminates checkpoint-deficient cells with unstable telomeres and other cancer-relevant chromosomal aberrations1,2. However, little is known about the molecular events that regulate the onset of this important tumour-suppressive barrier. Here we identified the innate immune sensor Z-DNA binding protein 1 (ZBP1) as a regulator of the crisis program. A crisis-associated isoform of ZBP1 is induced by the cGAS-STING DNA-sensing pathway, but reaches full activation only when associated with telomeric-repeat-containing RNA (TERRA) transcripts that are synthesized from dysfunctional telomeres. TERRA-bound ZBP1 oligomerizes into filaments on the outer mitochondrial membrane of a subset of mitochondria, where it activates the innate immune adapter protein mitochondrial antiviral-signalling protein (MAVS). We propose that these oligomerization properties of ZBP1 serve as a signal amplification mechanism, where few TERRA-ZBP1 interactions are sufficient to launch a detrimental MAVS-dependent interferon response. Our study reveals a mechanism for telomere-mediated tumour suppression, whereby dysfunctional telomeres activate innate immune responses through mitochondrial TERRA-ZBP1 complexes to eliminate cells destined for neoplastic transformation.


Assuntos
Replicação do DNA , Mitocôndrias , Transdução de Sinais , Telômero , Humanos , DNA/biossíntese , DNA/genética , DNA/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias/genética , Neoplasias/patologia , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Telômero/genética , Telômero/metabolismo , Interferons , Imunidade Inata , Autofagia
7.
Genes Dev ; 34(13-14): 913-930, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32499403

RESUMO

During mitosis, transcription of genomic DNA is dramatically reduced, before it is reactivated during nuclear reformation in anaphase/telophase. Many aspects of the underlying principles that mediate transcriptional memory and reactivation in the daughter cells remain unclear. Here, we used ChIP-seq on synchronized cells at different stages after mitosis to generate genome-wide maps of histone modifications. Combined with EU-RNA-seq and Hi-C analyses, we found that during prometaphase, promoters, enhancers, and insulators retain H3K4me3 and H3K4me1, while losing H3K27ac. Enhancers globally retaining mitotic H3K4me1 or locally retaining mitotic H3K27ac are associated with cell type-specific genes and their transcription factors for rapid transcriptional activation. As cells exit mitosis, promoters regain H3K27ac, which correlates with transcriptional reactivation. Insulators also gain H3K27ac and CCCTC-binding factor (CTCF) in anaphase/telophase. This increase of H3K27ac in anaphase/telophase is required for posttranscriptional activation and may play a role in the establishment of topologically associating domains (TADs). Together, our results suggest that the genome is reorganized in a sequential order, in which histone methylations occur first in prometaphase, histone acetylation, and CTCF in anaphase/telophase, transcription in cytokinesis, and long-range chromatin interactions in early G1. We thus provide insights into the histone modification landscape that allows faithful reestablishment of the transcriptional program and TADs during cell division.


Assuntos
Cromatina/metabolismo , Código das Histonas/genética , Histonas/metabolismo , Mitose/genética , Processamento de Proteína Pós-Traducional/genética , Ativação Transcricional/genética , Animais , Pontos de Checagem do Ciclo Celular/genética , Cromossomos/genética , Cromossomos/metabolismo , Elementos Facilitadores Genéticos , Genoma/genética , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Tempo
8.
Genes Dev ; 34(19-20): 1330-1344, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32912901

RESUMO

Despite being the frontline therapy for type 2 diabetes, the mechanisms of action of the biguanide drug metformin are still being discovered. In particular, the detailed molecular interplays between the AMPK and the mTORC1 pathway in the hepatic benefits of metformin are still ill defined. Metformin-dependent activation of AMPK classically inhibits mTORC1 via TSC/RHEB, but several lines of evidence suggest additional mechanisms at play in metformin inhibition of mTORC1. Here we investigated the role of direct AMPK-mediated serine phosphorylation of RAPTOR in a new RaptorAA mouse model, in which AMPK phospho-serine sites Ser722 and Ser792 of RAPTOR were mutated to alanine. Metformin treatment of primary hepatocytes and intact murine liver requires AMPK regulation of both RAPTOR and TSC2 to fully inhibit mTORC1, and this regulation is critical for both the translational and transcriptional response to metformin. Transcriptionally, AMPK and mTORC1 were both important for regulation of anabolic metabolism and inflammatory programs triggered by metformin treatment. The hepatic transcriptional response in mice on high-fat diet treated with metformin was largely ablated by AMPK deficiency under the conditions examined, indicating the essential role of this kinase and its targets in metformin action in vivo.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Metformina/farmacologia , Proteína Regulatória Associada a mTOR/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Genótipo , Hipoglicemiantes/farmacologia , Inflamação , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metabolismo/efeitos dos fármacos , Metformina/uso terapêutico , Camundongos , Fosforilação/efeitos dos fármacos , Proteína Regulatória Associada a mTOR/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
9.
Cell ; 149(3): 630-41, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22541433

RESUMO

In female mouse embryos, somatic cells undergo a random form of X chromosome inactivation (XCI), whereas extraembryonic trophoblast cells in the placenta undergo imprinted XCI, silencing exclusively the paternal X chromosome. Initiation of imprinted XCI requires a functional maternal allele of the X-linked gene Rnf12, which encodes the ubiquitin ligase Rnf12/RLIM. We find that knockout (KO) of Rnf12 in female mammary glands inhibits alveolar differentiation and milk production upon pregnancy, with alveolar cells that lack RLIM undergoing apoptosis as they begin to differentiate. Genetic analyses demonstrate that these functions are mediated primarily by the paternal Rnf12 allele due to nonrandom maternal XCI in mammary epithelial cells. These results identify paternal Rnf12/RLIM as a critical survival factor for milk-producing alveolar cells and, together with population models, reveal implications of transgenerational epigenetic inheritance.


Assuntos
Sobrevivência Celular , Glândulas Mamárias Animais/citologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Epigênese Genética , Feminino , Impressão Genômica , Masculino , Glândulas Mamárias Animais/fisiologia , Camundongos , Gravidez , Ubiquitina-Proteína Ligases/genética , Inativação do Cromossomo X
10.
Mol Cell ; 75(5): 891-904.e7, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31375262

RESUMO

Mammalian SWI/SNF complexes are multi-subunit chromatin remodeling complexes associated with an ATPase (either SMARCA4 or SMARCA2). Heterozygous mutations in the SMARCA2 ATPase cause Nicolaides-Baraitser syndrome (NCBRS), an intellectual disability syndrome associated with delayed speech onset. We engineered human embryonic stem cells (hESCs) to carry NCBRS-associated heterozygous SMARCA2 K755R or R1159Q mutations. While SMARCA2 mutant hESCs were phenotypically normal, differentiation to neural progenitors cells (NPCs) was severely impaired. We find that SMARCA2 mutations cause enhancer reorganization with loss of SOX3-dependent neural enhancers and prominent emergence of astrocyte-specific de novo enhancers. Changes in chromatin accessibility at enhancers were associated with an increase in SMARCA2 binding and retargeting of SMARCA4. We show that the AP-1 family member FRA2 is aberrantly overexpressed in SMARCA2 mutant NPCs, where it functions as a pioneer factor at de novo enhancers. Together, our results demonstrate that SMARCA2 mutations cause impaired differentiation through enhancer reprogramming via inappropriate targeting of SMARCA4.


Assuntos
DNA Helicases/metabolismo , Elementos Facilitadores Genéticos , Heterozigoto , Células-Tronco Embrionárias Humanas/metabolismo , Mutação de Sentido Incorreto , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Substituição de Aminoácidos , Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , DNA Helicases/genética , Fácies , Deformidades Congênitas do Pé/genética , Deformidades Congênitas do Pé/metabolismo , Deformidades Congênitas do Pé/patologia , Antígeno 2 Relacionado a Fos/biossíntese , Antígeno 2 Relacionado a Fos/genética , Células HEK293 , Células-Tronco Embrionárias Humanas/patologia , Humanos , Hipotricose/genética , Hipotricose/metabolismo , Hipotricose/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Proteínas Nucleares/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/genética
11.
Genes Dev ; 33(7-8): 418-435, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30819820

RESUMO

The RNA polymerase II (RNAPII) C-terminal domain kinase, CDK12, regulates genome stability, expression of DNA repair genes, and cancer cell resistance to chemotherapy and immunotherapy. In addition to its role in mRNA biosynthesis of DNA repair genes, we show here that CDK12 phosphorylates the mRNA 5' cap-binding repressor, 4E-BP1, to promote translation of mTORC1-dependent mRNAs. In particular, we found that phosphorylation of 4E-BP1 by mTORC1 (T37 and T46) facilitates subsequent CDK12 phosphorylation at two Ser-Pro sites (S65 and T70) that control the exchange of 4E-BP1 with eIF4G at the 5' cap of CHK1 and other target mRNAs. RNA immunoprecipitation coupled with deep sequencing (RIP-seq) revealed that CDK12 regulates release of 4E-BP1, and binding of eIF4G, to many mTORC1 target mRNAs, including those needed for MYC transformation. Genome-wide ribosome profiling (Ribo-seq) further identified specific CDK12 "translation-only" target mRNAs, including many mTORC1 target mRNAs as well as many subunits of mitotic and centromere/centrosome complexes. Accordingly, confocal imaging analyses revealed severe chromosome misalignment, bridging, and segregation defects in cells deprived of CDK12 or CCNK. We conclude that the nuclear RNAPII-CTD kinase CDK12 cooperates with mTORC1, and controls a specialized translation network that is essential for mitotic chromosome stability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quinase 1 do Ponto de Checagem/genética , Quinases Ciclina-Dependentes/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Instabilidade Genômica/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/genética , Ciclinas/genética , Ciclinas/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Humanos , Mitose/genética , Fosforilação/genética , Ligação Proteica/genética
12.
PLoS Genet ; 17(1): e1009277, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411704

RESUMO

The nuclear protein CCCTC-binding factor (CTCF) has diverse roles in chromatin architecture and gene regulation. Functionally, CTCF associates with thousands of genomic sites and interacts with proteins, such as cohesin, or non-coding RNAs to facilitate specific transcriptional programming. In this study, we examined CTCF during the cellular stress response in human primary cells using immune-blotting, quantitative real time-PCR, chromatin immunoprecipitation-sequence (ChIP-seq) analysis, mass spectrometry, RNA immunoprecipitation-sequence analysis (RIP-seq), and Airyscan confocal microscopy. Unexpectedly, we found that CTCF is exquisitely sensitive to diverse forms of stress in normal patient-derived human mammary epithelial cells (HMECs). In HMECs, a subset of CTCF protein forms complexes that localize to Serine/arginine-rich splicing factor (SC-35)-containing nuclear speckles. Upon stress, this species of CTCF protein is rapidly downregulated by changes in protein stability, resulting in loss of CTCF from SC-35 nuclear speckles and changes in CTCF-RNA interactions. Our ChIP-seq analysis indicated that CTCF binding to genomic DNA is largely unchanged. Restoration of the stress-sensitive pool of CTCF protein abundance and re-localization to nuclear speckles can be achieved by inhibition of proteasome-mediated degradation. Surprisingly, we observed the same characteristics of the stress response during neuronal differentiation of human pluripotent stem cells (hPSCs). CTCF forms stress-sensitive complexes that localize to SC-35 nuclear speckles during a specific stage of neuronal commitment/development but not in differentiated neurons. We speculate that these particular CTCF complexes serve a role in RNA processing that may be intimately linked with specific genes in the vicinity of nuclear speckles, potentially to maintain cells in a certain differentiation state, that is dynamically regulated by environmental signals. The stress-regulated activity of CTCF is uncoupled in persistently stressed, epigenetically re-programmed "variant" HMECs and certain cancer cell lines. These results reveal new insights into CTCF function in cell differentiation and the stress-response with implications for oxidative damage-induced cancer initiation and neuro-degenerative diseases.


Assuntos
Fator de Ligação a CCCTC/genética , Proteínas de Ligação a DNA/genética , Neoplasias/genética , Doenças Neurodegenerativas/genética , Fatores de Processamento de Serina-Arginina/genética , Sítios de Ligação , Diferenciação Celular , Linhagem Celular Tumoral , Cromatina , Cromossomos , Epigênese Genética/genética , Regulação da Expressão Gênica , Genômica , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/genética , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Ligação Proteica , Processamento Pós-Transcricional do RNA/genética , Estresse Fisiológico/genética
13.
Proc Natl Acad Sci U S A ; 117(6): 2906-2913, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31988127

RESUMO

The RNA-based organisms from which modern life is thought to have descended would have depended on an RNA polymerase ribozyme to copy functional RNA molecules, including copying the polymerase itself. Such a polymerase must have been capable of copying structured RNAs with high efficiency and high fidelity to maintain genetic information across successive generations. Here the class I RNA polymerase ribozyme was evolved in vitro for the ability to synthesize functional ribozymes, resulting in the markedly improved ability to synthesize complex RNAs using nucleoside 5'-triphosphate (NTP) substrates. The polymerase is descended from the class I ligase, which contains the same catalytic core as the polymerase. The class I ligase can be synthesized by the improved polymerase as three separate RNA strands that assemble to form a functional ligase. The polymerase also can synthesize the complement of each of these three strands. Despite this remarkable level of activity, only a very small fraction of the assembled ligases retain catalytic activity due to the presence of disabling mutations. Thus, the fidelity of RNA polymerization should be considered a major impediment to the construction of a self-sustained, RNA-based evolving system. The propagation of heritable information requires both efficient and accurate synthesis of genetic molecules, a requirement relevant to both laboratory systems and the early history of life on Earth.


Assuntos
RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Catalítico/genética , RNA Catalítico/metabolismo , Sequência de Bases , RNA Polimerases Dirigidas por DNA/química , Evolução Molecular Direcionada , Conformação de Ácido Nucleico , Nucleotídeos , Polimerização , RNA/genética , RNA/metabolismo , RNA Catalítico/química
14.
Nat Chem Biol ; 16(4): 458-468, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31819274

RESUMO

Functional protein-coding small open reading frames (smORFs) are emerging as an important class of genes. However, the number of translated smORFs in the human genome is unclear because proteogenomic methods are not sensitive enough, and, as we show, Ribo-seq strategies require additional measures to ensure comprehensive and accurate smORF annotation. Here, we integrate de novo transcriptome assembly and Ribo-seq into an improved workflow that overcomes obstacles with previous methods, to more confidently annotate thousands of smORFs. Evolutionary conservation analyses suggest that hundreds of smORF-encoded microproteins are likely functional. Additionally, many smORFs are regulated during fundamental biological processes, such as cell stress. Peptides derived from smORFs are also detectable on human leukocyte antigen complexes, revealing smORFs as a source of antigens. Thus, by including additional validation into our smORF annotation workflow, we accurately identify thousands of unannotated translated smORFs that will provide a rich pool of unexplored, functional human genes.


Assuntos
Anotação de Sequência Molecular/métodos , Fases de Leitura Aberta/genética , Análise de Sequência de DNA/métodos , Genoma Humano , Humanos , Peptídeos/química , Transcriptoma/genética
15.
Mol Psychiatry ; 26(6): 2440-2456, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33398088

RESUMO

Bipolar disorder (BD) is a psychiatric condition characterized by depressive and manic episodes that affect 2% of the world population. The first-line long-term treatment for mood stabilization is lithium (Li). Induced pluripotent stem cell modeling of BD using hippocampal dentate gyrus-like neurons derived from Li-responsive (LR) and Li-non-responsive (NR) patients previously showed neuronal hyperexcitability. Li treatment reversed hyperexcitability only on the LR neurons. In this study we searched for specific targets of Li resistance in NR neurons and found that the activity of Wnt/ß-catenin signaling pathway was severely affected, with a significant decrease in expression of LEF1. Li targets the Wnt/ß-catenin signaling pathway by inhibiting GSK-3ß and releasing ß-catenin that forms a nuclear complex with TCF/LEF1, activating the Wnt/ß-catenin transcription program. Therefore, we propose that downregulation of LEF1 may account for Li resistance in NR neurons. Our results show that valproic acid (VPA), a drug used to treat NR patients that also acts downstream of GSK-3ß, upregulated LEF1 and Wnt/ß-catenin gene targets, increased transcriptional activity of complex ß-catenin/TCF/LEF1, and reduced excitability in NR neurons. In addition, decreasing LEF1 expression in control neurons using shLEF1 caused hyperexcitability, confirming that the impact of VPA on excitability in NR neurons was connected to changes in LEF1 and in the Wnt/ß-catenin pathway. Our results suggest that LEF1 may be a useful target for the discovery of new drugs for BD treatment.


Assuntos
Transtorno Bipolar , Lítio , Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/genética , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Lítio/farmacologia , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neurônios/metabolismo , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
16.
Metabolomics ; 16(12): 126, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33294943

RESUMO

INTRODUCTION: Cellular metabolites are generated by a complex network of biochemical reactions. This makes interpreting changes in metabolites exceptionally challenging. OBJECTIVES: To develop a computational tool that integrates multiomics data at the level of reactions. METHODS: Changes in metabolic reactions are modeled with input from transcriptomics/proteomics measurements of enzymes and metabolomic measurements of metabolites. RESULTS: We developed SUMMER, which identified more relevant signals, key metabolic reactions, and relevant underlying biological pathways in a real-world case study. CONCLUSION: SUMMER performs integrative analysis for data interpretation and exploration. SUMMER is freely accessible at http://summer.salk.edu and the code is available at https://bitbucket.org/salkigc/summer .


Assuntos
Biologia Computacional/métodos , Metabolômica , Software , Algoritmos , Animais , Análise de Dados , Perfilação da Expressão Gênica/métodos , Humanos , Metabolômica/métodos , Camundongos , Proteômica/métodos , Curva ROC , Navegador , Fluxo de Trabalho
17.
BMC Bioinformatics ; 19(1): 296, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30089462

RESUMO

BACKGROUND: Microarray experiments comprise more than half of all series in the Gene Expression Omnibus (GEO). However, downloading and analyzing raw or semi-processed microarray data from GEO is not intuitive and requires manual error-prone analysis and a bioinformatics background. This is due to a lack of standardization in array platform fabrication as well as the lack of a simple interactive tool for clustering, plotting, differential expression testing, and testing for functional enrichment. RESULTS: We introduce the Bioinformatics Array Research Tool (BART), an R Shiny web application that automates the microarray download and analysis process across diverse microarray platforms. It provides an intuitive interface, automatically downloads and parses data from GEO, suggests groupings of samples for differential expression testing, performs batch effect correction, outputs quality control plots, converts probe IDs, generates full lists of differentially expressed genes, and performs functional enrichment analysis. We show that BART enables a more comprehensive analysis of a wider range of microarray datasets on GEO by comparing it to four leading online microarray analysis tools. CONCLUSIONS: BART allows a scientist with no bioinformatics background to extract knowledge from their own microarray data or microarray experiments available from GEO. BART is functional on more microarray experiments and provides more comprehensive analyses than extant microarray analysis tools. BART is hosted on bart.salk.edu , includes a user tutorial, and is available for download from https://bitbucket.org/Luisa_amaral/bart .


Assuntos
Biologia Computacional/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Neoplasias da Mama/genética , Bases de Dados Genéticas , Feminino , Humanos , Análise de Componente Principal , RNA/isolamento & purificação , Software
18.
Nucleic Acids Res ; 43(2): 699-707, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25541195

RESUMO

When messenger RNA splicing occurs co-transcriptionally, the potential for kinetic control based on transcription dynamics is widely recognized. Indeed, perturbation studies have reported that when transcription kinetics are perturbed genetically or pharmacologically splice patterns may change. However, whether kinetic control is contributing to the control of splicing within the normal range of physiological conditions remains unknown. We examined if the kinetic determinants for co-transcriptional splicing (CTS) might be reflected in the structure and expression patterns of the genome and epigenome. To identify and then quantitatively relate multiple, simultaneous CTS determinants, we constructed a scalable mathematical model of the kinetic interplay of RNA synthesis and CTS and parameterized it with diverse next generation sequencing (NGS) data. We thus found a variety of CTS determinants encoded in vertebrate genomes and epigenomes, and that these combine variously for different groups of genes such as housekeeping versus regulated genes. Together, our findings indicate that the kinetic basis of splicing is functionally and physiologically relevant, and may meaningfully inform the analysis of genomic and epigenomic data to provide insights that are missed when relying on statistical approaches alone.


Assuntos
Modelos Genéticos , Splicing de RNA , Transcrição Gênica , Animais , Simulação por Computador , Epigênese Genética , Genes Essenciais , Genômica , Humanos , Cinética , Camundongos , RNA Polimerase II/metabolismo , RNA Mensageiro/biossíntese , Análise de Sequência de RNA
19.
Mol Syst Biol ; 11(1): 783, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25680807

RESUMO

Understanding the functions of multi-cellular organs in terms of the molecular networks within each cell is an important step in the quest to predict phenotype from genotype. B-lymphocyte population dynamics, which are predictive of immune response and vaccine effectiveness, are determined by individual cells undergoing division or death seemingly stochastically. Based on tracking single-cell time-lapse trajectories of hundreds of B cells, single-cell transcriptome, and immunofluorescence analyses, we constructed an agent-based multi-modular computational model to simulate lymphocyte population dynamics in terms of the molecular networks that control NF-κB signaling, the cell cycle, and apoptosis. Combining modeling and experimentation, we found that NF-κB cRel enforces the execution of a cellular decision between mutually exclusive fates by promoting survival in growing cells. But as cRel deficiency causes growing B cells to die at similar rates to non-growing cells, our analysis reveals that the phenomenological decision model of wild-type cells is rooted in a biased race of cell fates. We show that a multi-scale modeling approach allows for the prediction of dynamic organ-level physiology in terms of intra-cellular molecular networks.


Assuntos
Linfócitos B/citologia , Divisão Celular , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Animais , Apoptose , Linfócitos B/metabolismo , Proliferação de Células , Camundongos , Modelos Moleculares , Análise de Sequência de RNA , Transdução de Sinais , Baço/citologia
20.
J Immunol ; 192(7): 3121-32, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24591377

RESUMO

The transcription factor NF-κB is a regulator of inflammatory and adaptive immune responses, yet only IκBα was shown to limit NF-κB activation and inflammatory responses. We investigated another negative feedback regulator, IκBε, in the regulation of B cell proliferation and survival. Loss of IκBε resulted in increased B cell proliferation and survival in response to both antigenic and innate stimulation. NF-κB activity was elevated during late-phase activation, but the dimer composition was stimulus specific. In response to IgM, cRel dimers were elevated in IκBε-deficient cells, yet in response to LPS, RelA dimers also were elevated. The corresponding dimer-specific sequences were found in the promoters of hyperactivated genes. Using a mathematical model of the NF-κB-signaling system in B cells, we demonstrated that kinetic considerations of IκB kinase-signaling input and IκBε's interactions with RelA- and cRel-specific dimers could account for this stimulus specificity. cRel is known to be the key regulator of B cell expansion. We found that the RelA-specific phenotype in LPS-stimulated cells was physiologically relevant: unbiased transcriptome profiling revealed that the inflammatory cytokine IL-6 was hyperactivated in IκBε(-/-) B cells. When IL-6R was blocked, LPS-responsive IκBε(-/-) B cell proliferation was reduced to near wild-type levels. Our results provide novel evidence for a critical role for immune-response functions of IκBε in B cells; it regulates proliferative capacity via at least two mechanisms involving cRel- and RelA-containing NF-κB dimers. This study illustrates the importance of kinetic considerations in understanding the functional specificity of negative-feedback regulators.


Assuntos
Linfócitos B/imunologia , Proliferação de Células , Quinase I-kappa B/imunologia , Proteínas Proto-Oncogênicas c-rel/imunologia , Fator de Transcrição RelA/imunologia , Algoritmos , Animais , Linfócitos B/metabolismo , Western Blotting , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Retroalimentação Fisiológica/efeitos dos fármacos , Citometria de Fluxo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-6/metabolismo , Cinética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Multimerização Proteica/imunologia , Proteínas Proto-Oncogênicas c-rel/química , Proteínas Proto-Oncogênicas c-rel/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA