Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.485
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 186(19): 4172-4188.e18, 2023 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-37633267

RESUMO

Selective clearance of organelles, including endoplasmic reticulum (ER) and mitochondria, by autophagy plays an important role in cell health. Here, we describe a developmentally programmed selective ER clearance by autophagy. We show that Parkinson's disease-associated PINK1, as well as Atl, Rtnl1, and Trp1 receptors, regulate ER clearance by autophagy. The E3 ubiquitin ligase Parkin functions downstream of PINK1 and is required for mitochondrial clearance while having the opposite function in ER clearance. By contrast, Keap1 and the E3 ubiquitin ligase Cullin3 function downstream of PINK1 to regulate ER clearance by influencing Rtnl1 and Atl. PINK1 regulates a change in Keap1 localization and Keap1-dependent ubiquitylation of the ER-phagy receptor Rtnl1 to facilitate ER clearance. Thus, PINK1 regulates the selective clearance of ER and mitochondria by influencing the balance of Keap1- and Parkin-dependent ubiquitylation of substrates that determine which organelle is removed by autophagy.


Assuntos
Retículo Endoplasmático , Fator 2 Relacionado a NF-E2 , Retículo Endoplasmático/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Proteínas Quinases , Ubiquitina-Proteína Ligases , Drosophila melanogaster , Animais
2.
Cell ; 185(6): 1025-1040.e14, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35148837

RESUMO

During the SARS-CoV-2 pandemic, novel and traditional vaccine strategies have been deployed globally. We investigated whether antibodies stimulated by mRNA vaccination (BNT162b2), including third-dose boosting, differ from those generated by infection or adenoviral (ChAdOx1-S and Gam-COVID-Vac) or inactivated viral (BBIBP-CorV) vaccines. We analyzed human lymph nodes after infection or mRNA vaccination for correlates of serological differences. Antibody breadth against viral variants is lower after infection compared with all vaccines evaluated but improves over several months. Viral variant infection elicits variant-specific antibodies, but prior mRNA vaccination imprints serological responses toward Wuhan-Hu-1 rather than variant antigens. In contrast to disrupted germinal centers (GCs) in lymph nodes during infection, mRNA vaccination stimulates robust GCs containing vaccine mRNA and spike antigen up to 8 weeks postvaccination in some cases. SARS-CoV-2 antibody specificity, breadth, and maturation are affected by imprinting from exposure history and distinct histological and antigenic contexts in infection compared with vaccination.


Assuntos
Anticorpos Antivirais , Vacina BNT162 , COVID-19 , Centro Germinativo , Antígenos Virais , COVID-19/prevenção & controle , Humanos , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus , Vacinação
3.
Cell ; 184(3): 596-614.e14, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33508232

RESUMO

Checkpoint inhibitors (CPIs) augment adaptive immunity. Systematic pan-tumor analyses may reveal the relative importance of tumor-cell-intrinsic and microenvironmental features underpinning CPI sensitization. Here, we collated whole-exome and transcriptomic data for >1,000 CPI-treated patients across seven tumor types, utilizing standardized bioinformatics workflows and clinical outcome criteria to validate multivariable predictors of CPI sensitization. Clonal tumor mutation burden (TMB) was the strongest predictor of CPI response, followed by total TMB and CXCL9 expression. Subclonal TMB, somatic copy alteration burden, and histocompatibility leukocyte antigen (HLA) evolutionary divergence failed to attain pan-cancer significance. Dinucleotide variants were identified as a source of immunogenic epitopes associated with radical amino acid substitutions and enhanced peptide hydrophobicity/immunogenicity. Copy-number analysis revealed two additional determinants of CPI outcome supported by prior functional evidence: 9q34 (TRAF2) loss associated with response and CCND1 amplification associated with resistance. Finally, single-cell RNA sequencing (RNA-seq) of clonal neoantigen-reactive CD8 tumor-infiltrating lymphocytes (TILs), combined with bulk RNA-seq analysis of CPI-responding tumors, identified CCR5 and CXCL13 as T-cell-intrinsic markers of CPI sensitivity.


Assuntos
Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias/imunologia , Linfócitos T/imunologia , Biomarcadores Tumorais/metabolismo , Antígenos CD8/metabolismo , Quimiocina CXCL13/metabolismo , Cromossomos Humanos Par 9/genética , Estudos de Coortes , Ciclina D1/genética , Variações do Número de Cópias de DNA/genética , Exoma/genética , Amplificação de Genes , Humanos , Evasão da Resposta Imune/efeitos dos fármacos , Análise Multivariada , Mutação/genética , Neoplasias/patologia , Polimorfismo de Nucleotídeo Único/genética , Receptores CCR5/metabolismo , Linfócitos T/efeitos dos fármacos , Carga Tumoral/genética
4.
Nat Immunol ; 24(10): 1762-1777, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37653247

RESUMO

Multivalent viral epitopes induce rapid, robust and T cell-independent humoral immune responses, but the biochemical basis for such potency remains incompletely understood. We take advantage of a set of liposomes of viral size engineered to display affinity mutants of the model antigen (Ag) hen egg lysozyme. Particulate Ag induces potent 'all-or-none' B cell responses that are density dependent but affinity independent. Unlike soluble Ag, particulate Ag induces signal amplification downstream of the B cell receptor by selectively evading LYN-dependent inhibitory pathways and maximally activates NF-κB in a manner that mimics T cell help. Such signaling induces MYC expression and enables even low doses of particulate Ag to trigger robust B cell proliferation in vivo in the absence of adjuvant. We uncover a molecular basis for highly sensitive B cell responses to viral Ag display that is independent of encapsulated nucleic acids and is not merely accounted for by avidity and B cell receptor cross-linking.


Assuntos
Antígenos , Linfócitos B , Receptores de Antígenos de Linfócitos B/metabolismo , Ativação Linfocitária , Epitopos/metabolismo
5.
Nat Immunol ; 21(10): 1267-1279, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32868928

RESUMO

Antigen stimulation (signal 1) triggers B cell proliferation and primes B cells to recruit, engage and respond to T cell help (signal 2). Failure to receive signal 2 within a defined time window results in B cell apoptosis, yet the mechanisms that enforce dependence on co-stimulation are incompletely understood. Nr4a1-3 encode a small family of orphan nuclear receptors that are rapidly induced by B cell antigen receptor stimulation. Here, we show that Nr4a1 and Nr4a3 play partially redundant roles to restrain B cell responses to antigen in the absence of co-stimulation and do so, in part, by repressing the expression of BATF and, consequently, MYC. The NR4A family also restrains B cell access to T cell help by repressing expression of the T cell chemokines CCL3 and CCL4, as well as CD86 and ICAM1. Such NR4A-mediated regulation plays a role specifically under conditions of competition for limiting T cell help.


Assuntos
Linfócitos B/imunologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Comunicação Celular , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Imunidade Humoral , Imunomodulação , Ativação Linfocitária , Camundongos , Camundongos Knockout , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/genética , Receptores dos Hormônios Tireóideos/metabolismo , Transdução de Sinais
6.
Immunity ; 56(11): 2469-2471, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37967529

RESUMO

Neutrophils have remained understudied in malignant brain tumors. In a recent issue of Cell, Maas et al. analyze brain tumor-patient samples and demonstrate that the brain microenvironment reprograms infiltrating neutrophils to enhance their longevity and increase their immune-suppressive and pro-angiogenic properties.


Assuntos
Neoplasias , Neutrófilos , Humanos , Encéfalo , Neoplasias/patologia , Microambiente Tumoral
7.
Cell ; 165(3): 742-53, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27040499

RESUMO

RNA transcripts are bound and regulated by RNA-binding proteins (RBPs). Current methods for identifying in vivo targets of an RBP are imperfect and not amenable to examining small numbers of cells. To address these issues, we developed TRIBE (targets of RNA-binding proteins identified by editing), a technique that couples an RBP to the catalytic domain of the Drosophila RNA-editing enzyme ADAR and expresses the fusion protein in vivo. RBP targets are marked with novel RNA editing events and identified by sequencing RNA. We have used TRIBE to identify the targets of three RBPs (Hrp48, dFMR1, and NonA). TRIBE compares favorably to other methods, including CLIP, and we have identified RBP targets from as little as 150 specific fly neurons. TRIBE can be performed without an antibody and in small numbers of specific cells.


Assuntos
Adenosina Desaminase/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Técnicas Genéticas , Edição de RNA , Regiões 3' não Traduzidas , Animais , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteínas de Ligação a RNA
8.
Genes Dev ; 37(21-24): 968-983, 2023 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-37977822

RESUMO

The spliceosomal gene SF3B1 is frequently mutated in cancer. While it is known that SF3B1 hotspot mutations lead to loss of splicing factor SUGP1 from spliceosomes, the cancer-relevant SF3B1-SUGP1 interaction has not been characterized. To address this issue, we show by structural modeling that two regions flanking the SUGP1 G-patch make numerous contacts with the region of SF3B1 harboring hotspot mutations. Experiments confirmed that all the cancer-associated mutations in these regions, as well as mutations affecting other residues in the SF3B1-SUGP1 interface, not only weaken or disrupt the interaction but also alter splicing similarly to SF3B1 cancer mutations. Finally, structural modeling of a trimeric protein complex reveals that the SF3B1-SUGP1 interaction "loops out" the G-patch for interaction with the helicase DHX15. Our study thus provides an unprecedented molecular view of a protein complex essential for accurate splicing and also reveals that numerous cancer-associated mutations disrupt the critical SF3B1-SUGP1 interaction.


Assuntos
Neoplasias , Spliceossomos , Humanos , RNA Mensageiro/metabolismo , Spliceossomos/genética , Spliceossomos/metabolismo , Fatores de Processamento de RNA/química , Splicing de RNA/genética , Neoplasias/genética , Neoplasias/metabolismo , Mutação , Fosfoproteínas/metabolismo
9.
Cell ; 161(7): 1619-32, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26091039

RESUMO

The existence of extracellular phosphoproteins has been acknowledged for over a century. However, research in this area has been undeveloped largely because the kinases that phosphorylate secreted proteins have escaped identification. Fam20C is a kinase that phosphorylates S-x-E/pS motifs on proteins in milk and in the extracellular matrix of bones and teeth. Here, we show that Fam20C generates the majority of the extracellular phosphoproteome. Using CRISPR/Cas9 genome editing, mass spectrometry, and biochemistry, we identify more than 100 secreted phosphoproteins as genuine Fam20C substrates. Further, we show that Fam20C exhibits broader substrate specificity than previously appreciated. Functional annotations of Fam20C substrates suggest roles for the kinase beyond biomineralization, including lipid homeostasis, wound healing, and cell migration and adhesion. Our results establish Fam20C as the major secretory pathway protein kinase and serve as a foundation for new areas of investigation into the role of secreted protein phosphorylation in human biology and disease.


Assuntos
Caseína Quinase I/química , Caseína Quinase I/metabolismo , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Sequência de Aminoácidos , Proteínas Sanguíneas/metabolismo , Caseína Quinase I/genética , Adesão Celular , Movimento Celular , Proteínas do Líquido Cefalorraquidiano/metabolismo , Proteínas da Matriz Extracelular/genética , Técnicas de Inativação de Genes , Ontologia Genética , Humanos , Dados de Sequência Molecular , Fosfoproteínas/análise , Via Secretória , Especificidade por Substrato
10.
Cell ; 161(7): 1516-26, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26091036

RESUMO

The 2013-2015 Ebola virus disease (EVD) epidemic is caused by the Makona variant of Ebola virus (EBOV). Early in the epidemic, genome sequencing provided insights into virus evolution and transmission and offered important information for outbreak response. Here, we analyze sequences from 232 patients sampled over 7 months in Sierra Leone, along with 86 previously released genomes from earlier in the epidemic. We confirm sustained human-to-human transmission within Sierra Leone and find no evidence for import or export of EBOV across national borders after its initial introduction. Using high-depth replicate sequencing, we observe both host-to-host transmission and recurrent emergence of intrahost genetic variants. We trace the increasing impact of purifying selection in suppressing the accumulation of nonsynonymous mutations over time. Finally, we note changes in the mucin-like domain of EBOV glycoprotein that merit further investigation. These findings clarify the movement of EBOV within the region and describe viral evolution during prolonged human-to-human transmission.


Assuntos
Ebolavirus/genética , Ebolavirus/isolamento & purificação , Genoma Viral , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/virologia , Mutação , Evolução Biológica , Surtos de Doenças , Ebolavirus/classificação , Doença pelo Vírus Ebola/transmissão , Humanos , Serra Leoa/epidemiologia , Manejo de Espécimes
11.
Nature ; 629(8010): 219-227, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38570683

RESUMO

The Integrator complex can terminate RNA polymerase II (Pol II) in the promoter-proximal region of genes. Previous work has shed light on how Integrator binds to the paused elongation complex consisting of Pol II, the DRB sensitivity-inducing factor (DSIF) and the negative elongation factor (NELF) and how it cleaves the nascent RNA transcript1, but has not explained how Integrator removes Pol II from the DNA template. Here we present three cryo-electron microscopy structures of the complete Integrator-PP2A complex in different functional states. The structure of the pre-termination complex reveals a previously unresolved, scorpion-tail-shaped INTS10-INTS13-INTS14-INTS15 module that may use its 'sting' to open the DSIF DNA clamp and facilitate termination. The structure of the post-termination complex shows that the previously unresolved subunit INTS3 and associated sensor of single-stranded DNA complex (SOSS) factors prevent Pol II rebinding to Integrator after termination. The structure of the free Integrator-PP2A complex in an inactive closed conformation2 reveals that INTS6 blocks the PP2A phosphatase active site. These results lead to a model for how Integrator terminates Pol II transcription in three steps that involve major rearrangements.


Assuntos
Microscopia Crioeletrônica , Modelos Moleculares , Proteína Fosfatase 2 , RNA Polimerase II , RNA Polimerase II/metabolismo , RNA Polimerase II/química , RNA Polimerase II/ultraestrutura , Proteína Fosfatase 2/metabolismo , Proteína Fosfatase 2/química , Proteína Fosfatase 2/ultraestrutura , Terminação da Transcrição Genética , Humanos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/química , Ligação Proteica , Fatores de Elongação da Transcrição/metabolismo , Fatores de Elongação da Transcrição/química , Proteínas Nucleares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestrutura , Subunidades Proteicas/metabolismo , Subunidades Proteicas/química
12.
Genes Dev ; 36(15-16): 876-886, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36207140

RESUMO

Nucleoli are the major cellular compartments for the synthesis of rRNA and assembly of ribosomes, the macromolecular complexes responsible for protein synthesis. Given the abundance of ribosomes, there is a huge demand for rRNA, which indeed constitutes ∼80% of the mass of RNA in the cell. Thus, nucleoli are characterized by extensive transcription of multiple rDNA loci by the dedicated polymerase, RNA polymerase (Pol) I. However, in addition to producing rRNAs, there is considerable additional transcription in nucleoli by RNA Pol II as well as Pol I, producing multiple noncoding (nc) and, in one instance, coding RNAs. In this review, we discuss important features of these transcripts, which often appear species-specific and reflect transcription antisense to pre-rRNA by Pol II and within the intergenic spacer regions on both strands by both Pol I and Pol II. We discuss how expression of these RNAs is regulated, their propensity to form cotranscriptional R loops, and how they modulate rRNA transcription, nucleolar structure, and cellular homeostasis more generally.


Assuntos
RNA Polimerase II , Precursores de RNA , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , DNA Intergênico , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Homeostase/genética , Substâncias Macromoleculares/metabolismo , RNA Polimerase I/genética , RNA Polimerase I/metabolismo , RNA Polimerase II/metabolismo , Precursores de RNA/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Transcrição Gênica
13.
Nat Immunol ; 18(5): 552-562, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28346408

RESUMO

Gut dysbiosis might underlie the pathogenesis of type 1 diabetes. In mice of the non-obese diabetic (NOD) strain, we found that key features of disease correlated inversely with blood and fecal concentrations of the microbial metabolites acetate and butyrate. We therefore fed NOD mice specialized diets designed to release large amounts of acetate or butyrate after bacterial fermentation in the colon. Each diet provided a high degree of protection from diabetes, even when administered after breakdown of immunotolerance. Feeding mice a combined acetate- and butyrate-yielding diet provided complete protection, which suggested that acetate and butyrate might operate through distinct mechanisms. Acetate markedly decreased the frequency of autoreactive T cells in lymphoid tissues, through effects on B cells and their ability to expand populations of autoreactive T cells. A diet containing butyrate boosted the number and function of regulatory T cells, whereas acetate- and butyrate-yielding diets enhanced gut integrity and decreased serum concentration of diabetogenic cytokines such as IL-21. Medicinal foods or metabolites might represent an effective and natural approach for countering the numerous immunological defects that contribute to T cell-dependent autoimmune diseases.


Assuntos
Acetatos/metabolismo , Linfócitos B/imunologia , Butiratos/metabolismo , Colo/metabolismo , Diabetes Mellitus Tipo 1/dietoterapia , Disbiose/dietoterapia , Linfócitos T Reguladores/imunologia , Animais , Autoimunidade , Linfócitos B/microbiologia , Células Cultivadas , Colo/patologia , Dietoterapia , Microbioma Gastrointestinal , Interleucinas/sangue , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T Reguladores/microbiologia
15.
Immunity ; 53(5): 925-933.e4, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33129373

RESUMO

We conducted a serological study to define correlates of immunity against SARS-CoV-2. Compared to those with mild coronavirus disease 2019 (COVID-19) cases, individuals with severe disease exhibited elevated virus-neutralizing titers and antibodies against the nucleocapsid (N) and the receptor binding domain (RBD) of the spike protein. Age and sex played lesser roles. All cases, including asymptomatic individuals, seroconverted by 2 weeks after PCR confirmation. Spike RBD and S2 and neutralizing antibodies remained detectable through 5-7 months after onset, whereas α-N titers diminished. Testing 5,882 members of the local community revealed only 1 sample with seroreactivity to both RBD and S2 that lacked neutralizing antibodies. This fidelity could not be achieved with either RBD or S2 alone. Thus, inclusion of multiple independent assays improved the accuracy of antibody tests in low-seroprevalence communities and revealed differences in antibody kinetics depending on the antigen. We conclude that neutralizing antibodies are stably produced for at least 5-7 months after SARS-CoV-2 infection.


Assuntos
Betacoronavirus/imunologia , Técnicas de Laboratório Clínico/métodos , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Imunidade Humoral , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Arizona/epidemiologia , Betacoronavirus/isolamento & purificação , COVID-19 , Teste para COVID-19 , Infecções por Coronavirus/sangue , Infecções por Coronavirus/diagnóstico , Proteínas do Nucleocapsídeo de Coronavírus , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Nucleocapsídeo/imunologia , Pandemias , Fosfoproteínas , Pneumonia Viral/sangue , Pneumonia Viral/diagnóstico , Prevalência , Domínios e Motivos de Interação entre Proteínas , SARS-CoV-2 , Estudos Soroepidemiológicos , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Adulto Jovem
16.
Nat Rev Mol Cell Biol ; 18(1): 18-30, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27677860

RESUMO

Alternative polyadenylation (APA) is an RNA-processing mechanism that generates distinct 3' termini on mRNAs and other RNA polymerase II transcripts. It is widespread across all eukaryotic species and is recognized as a major mechanism of gene regulation. APA exhibits tissue specificity and is important for cell proliferation and differentiation. In this Review, we discuss the roles of APA in diverse cellular processes, including mRNA metabolism, protein diversification and protein localization, and more generally in gene regulation. We also discuss the molecular mechanisms underlying APA, such as variation in the concentration of core processing factors and RNA-binding proteins, as well as transcription-based regulation.


Assuntos
Regiões 3' não Traduzidas , Poliadenilação , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Transporte Ativo do Núcleo Celular , Éxons , Regulação da Expressão Gênica , Humanos , Precursores de RNA/genética , Estabilidade de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo
17.
Genes Dev ; 35(23-24): 1579-1594, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34819354

RESUMO

The nucleolus is an important cellular compartment in which ribosomal RNAs (rRNAs) are transcribed and where certain stress pathways that are crucial for cell growth are coordinated. Here we report novel functions of the DNA replication and repair factor replication protein A (RPA) in control of nucleolar homeostasis. We show that loss of the DNA:RNA helicase senataxin (SETX) promotes RPA nucleolar localization, and that this relocalization is dependent on the presence of R loops. Notably, this nucleolar RPA phenotype was also observed in the presence of camptothecin (CPT)-induced genotoxic stress, as well as in SETX-deficient AOA2 patient fibroblasts. Extending these results, we found that RPA is recruited to rDNA following CPT treatment, where RPA prevents R-loop-induced DNA double-strand breaks. Furthermore, we show that loss of RPA significantly decreased 47S pre-rRNA levels, which was accompanied by increased expression of both RNAP II-mediated "promoter and pre-rRNA antisense" RNA as well as RNAP I-transcribed intragenic spacer RNAs. Finally, and likely reflecting the above, we found that loss of RPA promoted nucleolar structural disorganization, characterized by the appearance of reduced size nucleoli. Our findings both indicate new roles for RPA in nucleoli through pre-rRNA transcriptional control and also emphasize that RPA function in nucleolar homeostasis is linked to R-loop resolution under both physiological and pathological conditions.


Assuntos
Estruturas R-Loop , Proteína de Replicação A , Nucléolo Celular/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Humanos , Enzimas Multifuncionais , RNA Helicases/metabolismo , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Transcrição Gênica
18.
Nature ; 606(7916): 896-901, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35676485

RESUMO

The observation of the Higgs boson solidified the standard model of particle physics. However, explanations of anomalies (for example, dark matter) rely on further symmetry breaking, calling for an undiscovered axial Higgs mode1. The Higgs mode was also seen in magnetic, superconducting and charge density wave (CDW) systems2,3. Uncovering the vector properties of a low-energy mode is challenging, and requires going beyond typical spectroscopic or scattering techniques. Here we discover an axial Higgs mode in the CDW system RTe3 using the interference of quantum pathways. In RTe3 (R = La, Gd), the electronic ordering couples bands of equal or different angular momenta4-6. As such, the Raman scattering tensor associated with the Higgs mode contains both symmetric and antisymmetric components, which are excited via two distinct but degenerate pathways. This leads to constructive or destructive interference of these pathways, depending on the choice of the incident and Raman-scattered light polarization. The qualitative behaviour of the Raman spectra is well captured by an appropriate tight-binding model, including an axial Higgs mode. Elucidation of the antisymmetric component is direct evidence that the Higgs mode contains an axial vector representation (that is, a pseudo-angular momentum) and hints that the CDW is unconventional. Thus, we provide a means for measuring quantum properties of collective modes without resorting to extreme experimental conditions.

19.
Nature ; 610(7930): 173-181, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36171288

RESUMO

Combination therapy with PD-1 blockade and IL-2 is highly effective during chronic lymphocytic choriomeningitis virus infection1. Here we examine the underlying basis for this synergy. We show that PD-1 + IL-2 combination therapy, in contrast to PD-1 monotherapy, substantially changes the differentiation program of the PD-1+TCF1+ stem-like CD8+ T cells and results in the generation of transcriptionally and epigenetically distinct effector CD8+ T cells that resemble highly functional effector CD8+ T cells seen after an acute viral infection. The generation of these qualitatively superior CD8+ T cells that mediate viral control underlies the synergy between PD-1 and IL-2. Our results show that the PD-1+TCF1+ stem-like CD8+ T cells, also referred to as precursors of exhausted CD8+ T cells, are not fate-locked into the exhaustion program and their differentiation trajectory can be changed by IL-2 signals. These virus-specific effector CD8+ T cells emerging from the stem-like CD8+ T cells after combination therapy expressed increased levels of the high-affinity IL-2 trimeric (CD25-CD122-CD132) receptor. This was not seen after PD-1 blockade alone. Finally, we show that CD25 engagement with IL-2 has an important role in the observed synergy between IL-2 cytokine and PD-1 blockade. Either blocking CD25 with an antibody or using a mutated version of IL-2 that does not bind to CD25 but still binds to CD122 and CD132 almost completely abrogated the synergistic effects observed after PD-1 + IL-2 combination therapy. There is considerable interest in PD-1 + IL-2 combination therapy for patients with cancer2,3, and our fundamental studies defining the underlying mechanisms of how IL-2 synergizes with PD-1 blockade should inform these human translational studies.


Assuntos
Linfócitos T CD8-Positivos , Interleucina-2 , Receptor de Morte Celular Programada 1 , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Quimioterapia Combinada , Humanos , Subunidade gama Comum de Receptores de Interleucina , Interleucina-2/imunologia , Interleucina-2/farmacologia , Interleucina-2/uso terapêutico , Subunidade alfa de Receptor de Interleucina-2 , Subunidade beta de Receptor de Interleucina-2 , Coriomeningite Linfocítica/tratamento farmacológico , Coriomeningite Linfocítica/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Fator 1 de Transcrição de Linfócitos T
20.
Genes Dev ; 34(11-12): 785-805, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32381627

RESUMO

Dysregulation of the DNA/RNA-binding protein FUS causes certain subtypes of ALS/FTD by largely unknown mechanisms. Recent evidence has shown that FUS toxic gain of function due either to mutations or to increased expression can disrupt critical cellular processes, including mitochondrial functions. Here, we demonstrate that in human cells overexpressing wild-type FUS or expressing mutant derivatives, the protein associates with multiple mRNAs, and these are enriched in mRNAs encoding mitochondrial respiratory chain components. Notably, this sequestration leads to reduced levels of the encoded proteins, which is sufficient to bring about disorganized mitochondrial networks, reduced aerobic respiration and increased reactive oxygen species. We further show that mutant FUS associates with mitochondria and with mRNAs encoded by the mitochondrial genome. Importantly, similar results were also observed in fibroblasts derived from ALS patients with FUS mutations. Finally, we demonstrate that FUS loss of function does not underlie the observed mitochondrial dysfunction, and also provides a mechanism for the preferential sequestration of the respiratory chain complex mRNAs by FUS that does not involve sequence-specific binding. Together, our data reveal that respiratory chain complex mRNA sequestration underlies the mitochondrial defects characteristic of ALS/FTD and contributes to the FUS toxic gain of function linked to this disease spectrum.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Regulação da Expressão Gênica/genética , Mitocôndrias/patologia , RNA Mensageiro/metabolismo , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Linhagem Celular , Respiração Celular/genética , Células Cultivadas , Transporte de Elétrons/genética , Genoma Mitocondrial , Humanos , Mitocôndrias/genética , Mutação , Agregação Patológica de Proteínas/genética , Ligação Proteica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA