Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
bioRxiv ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38645116

RESUMEN

Arising as co-products of canonical gene expression, transcription-associated lincRNAs, such as promoter upstream transcripts (PROMPTs), enhancer RNAs (eRNAs), and readthrough (RT) transcripts, are often regarded as byproducts of transcription, although they may be important for the expression of nearby genes. We identified regions of nascent expression of these lincRNA in 16 human cell lines using Bru-seq techniques, and found distinctly regulated patterns of PROMPT, eRNA, and RT transcription using the diverse biochemical approaches in the ENCODE4 deeply profiled cell lines collection. Transcription of these lincRNAs was influenced by sequence-specific features and the local or 3D chromatin landscape. However, these sequence and chromatin features do not describe the full spectrum of lincRNA expression variability we identify, highlighting the complexity of their regulation. This may suggest that transcription-associated lincRNAs are not merely byproducts, but rather that the transcript itself, or the act of its transcription, is important for genomic function.

2.
Geriatrics (Basel) ; 8(6)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38132486

RESUMEN

PURPOSE: The COVID-19 pandemic limited access to community fall prevention programs, thus establishing the need for virtual interventions. Herein, we describe the feasibility, effectiveness, and acceptability of a virtual, multicomponent fall prevention program (MOVing FREEly). METHODS: A team of clinical falls prevention experts developed a six-week multicomponent fall prevention exercise and education class for older community-dwelling adults at risk of falling. Feasibility was measured through class attendance; effectiveness was measured through changes in performance measures, self-report of falling risk, and concern about falling; acceptability was assessed through questionnaires completed immediately upon program completion and at a three-month follow up. RESULTS: A total of 32 patients participated in the MOVing FREEly program. Attendance for education and exercise classes on average was greater than 80% with little attrition. Patient reported reduced concern of falling, improvement in the falls efficacy scale-international (FES-I) short form, and had statistically significant improvement in 30 s sit-to-stand and single-leg balance tests. The program was well received by participants, saving them significant time and costs of travel. CONCLUSIONS: A virtual, multicomponent fall prevention program is feasible and acceptable and effective as reducing falling risk. Future studies can explore the ability of this program to reduce falling incident and injury.

3.
Int J Cancer ; 153(3): 552-570, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37140208

RESUMEN

Although KMT2D, also known as MLL2, is known to play an essential role in development, differentiation, and tumor suppression, its role in pancreatic cancer development is not well understood. Here, we discovered a novel signaling axis mediated by KMT2D, which links TGF-ß to the activin A pathway. We found that TGF-ß upregulates a microRNA, miR-147b, which in turn leads to post-transcriptional silencing of KMT2D. Loss of KMT2D induces the expression and secretion of activin A, which activates a noncanonical p38 MAPK-mediated pathway to modulate cancer cell plasticity, promote a mesenchymal phenotype, and enhance tumor invasion and metastasis in mice. We observed a decreased KMT2D expression in human primary and metastatic pancreatic cancer. Furthermore, inhibition or knockdown of activin A reversed the protumoral role of KMT2D loss. These findings support a tumor-suppressive role of KMT2D in pancreatic cancer and identify miR-147b and activin A as novel therapeutic targets.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Plasticidad de la Célula , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Pancreáticas/patología , Factor de Crecimiento Transformador beta/metabolismo , Activinas/genética , Neoplasias Pancreáticas
4.
Cell Commun Signal ; 21(1): 15, 2023 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-36691073

RESUMEN

Grainyhead like 2 (GRHL2) is an essential transcription factor for development and function of epithelial tissues. It has dual roles in cancer by supporting tumor growth while suppressing epithelial to mesenchymal transitions (EMT). GRHL2 cooperates with androgen and estrogen receptors (ER) to regulate gene expression. We explore genome wide GRHL2 binding sites conserved in three ER⍺/GRHL2 positive luminal breast cancer cell lines by ChIP-Seq. Interaction with the ER⍺/FOXA1/GATA3 complex is observed, however, only for a minor fraction of conserved GRHL2 peaks. We determine genome wide transcriptional dynamics in response to loss of GRHL2 by nascent RNA Bru-seq using an MCF7 conditional knockout model. Integration of ChIP- and Bru-seq pinpoints candidate direct GRHL2 target genes in luminal breast cancer. Multiple connections between GRHL2 and proliferation are uncovered, including transcriptional activation of ETS and E2F transcription factors. Among EMT-related genes, direct regulation of CLDN4 is corroborated but several targets identified in other cells (including CDH1 and ZEB1) are ruled out by both ChIP- and Bru-seq as being directly controlled by GRHL2 in luminal breast cancer cells. Gene clusters correlating positively (including known GRHL2 targets such as ErbB3, CLDN4/7) or negatively (including TGFB1 and TGFBR2) with GRHL2 in the MCF7 knockout model, display similar correlation with GRHL2 in ER positive as well as ER negative breast cancer patients. Altogether, this study uncovers gene sets regulated directly or indirectly by GRHL2 in luminal breast cancer, identifies novel GRHL2-regulated genes, and points to distinct GRHL2 regulation of EMT in luminal breast cancer cells. Video Abstract.


Asunto(s)
Neoplasias de la Mama , Proteínas de Unión al ADN , Humanos , Femenino , Proteínas de Unión al ADN/metabolismo , Neoplasias de la Mama/patología , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
5.
Hum Mol Genet ; 32(6): 934-947, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36219176

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease. Its complex pathogenesis and phenotypic heterogeneity hinder therapeutic development and early diagnosis. Altered RNA metabolism is a recurrent pathophysiologic theme, including distinct microRNA (miRNA) profiles in ALS tissues. We profiled miRNAs in accessible biosamples, including skin fibroblasts and whole blood and compared them in age- and sex-matched healthy controls versus ALS participants with and without repeat expansions to chromosome 9 open reading frame 72 (C9orf72; C9-ALS and nonC9-ALS), the most frequent ALS mutation. We identified unique and shared profiles of differential miRNA (DmiRNA) levels in each C9-ALS and nonC9-ALS tissues versus controls. Fibroblast DmiRNAs were validated by quantitative real-time PCR and their target mRNAs by 5-bromouridine and 5-bromouridine-chase sequencing. We also performed pathway analysis to infer biological meaning, revealing anticipated, tissue-specific pathways and pathways previously linked to ALS, as well as novel pathways that could inform future research directions. Overall, we report a comprehensive study of a miRNA profile dataset from C9-ALS and nonC9-ALS participants across two accessible biosamples, providing evidence of dysregulated miRNAs in ALS and possible targets of interest. Distinct miRNA patterns in accessible tissues may also be leveraged to distinguish ALS participants from healthy controls for earlier diagnosis. Future directions may look at potential correlations of miRNA profiles with clinical parameters.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , MicroARNs , Enfermedades Neurodegenerativas , Humanos , Esclerosis Amiotrófica Lateral/patología , MicroARNs/genética , MicroARNs/metabolismo , Demencia Frontotemporal/genética , Mutación
6.
Cells ; 11(23)2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36497055

RESUMEN

Cancer risk after ionizing radiation (IR) is assumed to be linear with the dose; however, for low doses, definite evidence is lacking. Here, using temporal multi-omic systems analyses after a low (LD; 0.1 Gy) or a high (HD; 1 Gy) dose of X-rays, we show that, although the DNA damage response (DDR) displayed dose proportionality, many other molecular and cellular responses did not. Phosphoproteomics uncovered a novel mode of phospho-signaling via S12-PPP1R7, and large-scale dephosphorylation events that regulate mitotic exit control in undamaged cells and the G2/M checkpoint upon IR in a dose-dependent manner. The phosphoproteomics of irradiated DNA double-strand breaks (DSBs) repair-deficient cells unveiled extended phospho-signaling duration in either a dose-dependent (DDR signaling) or independent (mTOR-ERK-MAPK signaling) manner without affecting signal magnitude. Nascent transcriptomics revealed the transcriptional activation of genes involved in NRF2-regulated antioxidant defense, redox-sensitive ERK-MAPK signaling, glycolysis and mitochondrial function after LD, suggesting a prominent role for reactive oxygen species (ROS) in molecular and cellular responses to LD exposure, whereas DDR genes were prominently activated after HD. However, how and to what extent the observed dose-dependent differences in molecular and cellular responses may impact cancer development remain unclear, as the induction of chromosomal damage was found to be dose-proportional (10-200 mGy).


Asunto(s)
Roturas del ADN de Doble Cadena , Radiación Ionizante , Puntos de Control de la Fase G2 del Ciclo Celular , Especies Reactivas de Oxígeno , Transducción de Señal
7.
iScience ; 25(9): 105030, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36111258

RESUMEN

The cyclin-dependent kinase CDK12 has garnered interest as a cancer therapeutic target as DNA damage response genes are particularly suppressed by loss of CDK12 activity. In this study, we assessed the acute effects of CDK12 inhibition on transcription and RNA processing using nascent RNA Bru-seq and BruChase-seq. Acute transcriptional changes were overall small after CDK12 inhibition but over 600 genes showed intragenic premature termination, including DNA repair and cell cycle genes. Furthermore, many genes showed reduced transcriptional readthrough past the end of genes in the absence of CDK12 activity. RNA turnover was dramatically affected by CDK12 inhibition and importantly, caused increased degradation of many transcripts from DNA damage response genes. We also show that co-transcriptional splicing was suppressed by CDK12 inhibition. Taken together, these studies reveal the roles of CDK12 in regulating transcription elongation, transcription termination, co-transcriptional splicing, and RNA turnover.

8.
Sci Immunol ; 7(74): eabn8144, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36026441

RESUMEN

FOXN1 is a transcription factor critical for the development of both thymic epithelial cell (TEC) and hair follicle cell (HFC) compartments. However, mechanisms controlling its expression remain poorly understood. To address this question, we performed thorough analyses of the evolutionary conservation and chromatin status of the Foxn1 locus in different tissues and states and identified several putative cis-regulatory regions unique to TECs versus HFCs. Furthermore, experiments using genetically modified mice with specific deletions in the Foxn1 locus and additional bioinformatic analyses helped us identify key regions and transcription factors involved in either positive or negative regulation of Foxn1 in both TECs and HFCs. Specifically, we identified SIX1 and FOXN1 itself as key factors inducing Foxn1 expression in embryonic and neonatal TECs. Together, our data provide important mechanistic insights into the transcriptional regulation of the Foxn1 gene in TEC versus HFC and highlight the role of FOXN1 in its autoregulation.


Asunto(s)
Células Epiteliales , Regulación de la Expresión Génica , Animales , Ratones , Proteínas de Unión al ARN , Timo
9.
Curr Biol ; 32(12): 2581-2595.e6, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35580604

RESUMEN

Autophagy is a conserved, multi-step process of capturing proteolytic cargo in autophagosomes for lysosome degradation. The capacity to remove toxic proteins that accumulate in neurodegenerative disorders attests to the disease-modifying potential of the autophagy pathway. However, neurons respond only marginally to conventional methods for inducing autophagy, limiting efforts to develop therapeutic autophagy modulators for neurodegenerative diseases. The determinants underlying poor autophagy induction in neurons and the degree to which neurons and other cell types are differentially sensitive to autophagy stimuli are incompletely defined. Accordingly, we sampled nascent transcript synthesis and stabilities in fibroblasts, induced pluripotent stem cells (iPSCs), and iPSC-derived neurons (iNeurons), thereby uncovering a neuron-specific stability of transcripts encoding myotubularin-related phosphatase 5 (MTMR5). MTMR5 is an autophagy suppressor that acts with its binding partner, MTMR2, to dephosphorylate phosphoinositides critical for autophagy initiation and autophagosome maturation. We found that MTMR5 is necessary and sufficient to suppress autophagy in iNeurons and undifferentiated iPSCs. Using optical pulse labeling to visualize the turnover of endogenously encoded proteins in live cells, we observed that knockdown of MTMR5 or MTMR2, but not the unrelated phosphatase MTMR9, significantly enhances neuronal degradation of TDP-43, an autophagy substrate implicated in several neurodegenerative diseases. Our findings thus establish a regulatory mechanism of autophagy intrinsic to neurons and targetable for clearing disease-related proteins in a cell-type-specific manner. In so doing, our results not only unravel novel aspects of neuronal biology and proteostasis but also elucidate a strategy for modulating neuronal autophagy that could be of high therapeutic potential for multiple neurodegenerative diseases.


Asunto(s)
Autofagosomas , Proteínas Tirosina Fosfatasas no Receptoras , Autofagosomas/metabolismo , Autofagia/genética , Neuronas/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo
10.
J Med Chem ; 65(8): 6133-6156, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35439009

RESUMEN

Using a cytotoxicity-based phenotypic screen of a highly diverse library of 20,000 small-molecule compounds, we identified a quinolin-8-yl-nicotinamide, QN519, as a promising lead. QN519 represents a novel scaffold with drug-like properties, showing potent in vitro cytotoxicity in a panel of 12 cancer cell lines. Subsequently, lead optimization campaign generated compounds with IC50 values < 1 µM. An optimized compound, QN523, shows significant in vivo efficacy in a pancreatic cancer xenograft model. QN523 treatment significantly increased the expression of HSPA5, DDIT3, TRIB3, and ATF3 genes, suggesting activation of the stress response pathway. We also observed a significant increase in the expression of WIPI1, HERPUD1, GABARAPL1, and MAP1LC3B, implicating autophagy as a major mechanism of action. Due to the lack of effective treatments for pancreatic cancer, discovery of novel agents such as the QN series of compounds with unique mechanism of action has the potential to fulfill a clear unmet medical need.


Asunto(s)
Antineoplásicos , Neoplasias Pancreáticas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia , Línea Celular Tumoral , Humanos , Niacinamida/farmacología , Niacinamida/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
11.
PLoS Pathog ; 18(2): e1010272, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108347

RESUMEN

Respiratory syncytial virus (RSV) can cause bronchiolitis and viral pneumonia in young children and the elderly. Lack of vaccines and recurrence of RSV infection indicate the difficulty in eliciting protective memory immune responses. Tissue resident memory T cells (TRM) can confer protection from pathogen re-infection and, in human experimental RSV infection, the presence of lung CD8+ TRM cells correlates with a better outcome. However, the requirements for generating and maintaining lung TRM cells during RSV infection are not fully understood. Here, we use mouse models to assess the impact of innate immune response determinants in the generation and subsequent expansion of the TRM cell pool during RSV infection. We show that CD8+ TRM cells expand independently from systemic CD8+ T cells after RSV re-infection. Re-infected MAVS and MyD88/TRIF deficient mice, lacking key components involved in innate immune recognition of RSV and induction of type I interferons (IFN-α/ß), display impaired expansion of CD8+ TRM cells and reduction in antigen specific production of granzyme B and IFN-γ. IFN-α treatment of MAVS deficient mice during primary RSV infection restored TRM cell expansion upon re-challenge but failed to recover TRM cell functionality. Our data reveal how innate immunity, including the axis controlling type I IFN induction, instructs and regulates CD8+ TRM cell responses to RSV infection, suggesting possible mechanisms for therapeutic intervention.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón Tipo I/inmunología , Células T de Memoria/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Granzimas/inmunología , Granzimas/metabolismo , Inmunidad Innata , Memoria Inmunológica , Interferón Tipo I/metabolismo , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Virus Sincitial Respiratorio/virología , Transducción de Señal
12.
Cell Mol Gastroenterol Hepatol ; 13(2): 643-667, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34583087

RESUMEN

BACKGROUND & AIMS: Inactivating mutations of KDM6A, a histone demethylase, were frequently found in pancreatic ductal adenocarcinoma (PDAC). We investigated the role of KDM6A (lysine demethylase 6A) in PDAC development. METHODS: We performed a pancreatic tissue microarray analysis of KDM6A protein levels. We used human PDAC cell lines for KDM6A knockout and knockdown experiments. We performed bromouridine sequencing analysis to elucidate the effects of KDM6A loss on global transcription. We performed studies with Ptf1aCre; LSL-KrasG12D; Trp53R172H/+; Kdm6afl/fl or fl/Y, Ptf1aCre; Kdm6afl/fl or fl/Y, and orthotopic xenograft mice to investigate the impacts of Kdm6a deficiency on pancreatic tumorigenesis and pancreatitis. RESULTS: Loss of KDM6A was associated with metastasis in PDAC patients. Bromouridine sequencing analysis showed up-regulation of the epithelial-mesenchymal transition pathway in PDAC cells deficient in KDM6A. Loss of KDM6A promoted mesenchymal morphology, migration, and invasion in PDAC cells in vitro. Mechanistically, activin A and subsequent p38 activation likely mediated the role of KDM6A loss. Inhibiting either activin A or p38 reversed the effect. Pancreas-specific Kdm6a-knockout mice pancreata showed accelerated PDAC progression, developed a more aggressive undifferentiated type of PDAC, and increased metastases in the background of Kras and p53 mutations. Kdm6a-deficient pancreata in a pancreatitis model had a delayed recovery with increased PDAC precursor lesions compared with wild-type pancreata. CONCLUSIONS: Loss of KDM6A accelerates PDAC progression and metastasis, most likely by a noncanonical p38-dependent activin A pathway. KDM6A also promotes pancreatic tissue recovery from pancreatitis. Activin A might be used as a therapeutic target for KDM6A-deficient PDACs.


Asunto(s)
Plasticidad de la Célula , Neoplasias Pancreáticas , Activinas/metabolismo , Animales , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Ratones , Páncreas/patología , Neoplasias Pancreáticas/patología
13.
Nat Commun ; 12(1): 6520, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764283

RESUMEN

Lung diseases, such as cystic fibrosis and COPD, are characterized by mucus obstruction and chronic airway inflammation, but their mechanistic link remains poorly understood. Here, we focus on the function of the mucostatic airway microenvironment on epigenetic reprogramming of airway macrophages (AM) and resulting transcriptomic and phenotypical changes. Using a mouse model of muco-obstructive lung disease (Scnn1b-transgenic), we identify epigenetically controlled, differentially regulated pathways and transcription factors involved in inflammatory responses and macrophage polarization. Functionally, AMs from Scnn1b-transgenic mice have reduced efferocytosis and phagocytosis, and excessive inflammatory responses upon lipopolysaccharide challenge, mediated through enhanced Irf1 function and expression. Ex vivo stimulation of wild-type AMs with native mucus impairs efferocytosis and phagocytosis capacities. In addition, mucus induces gene expression changes, comparable with those observed in AMs from Scnn1b-transgenic mice. Our data show that mucostasis induces epigenetic reprogramming of AMs, leading to changes favoring tissue damage and disease progression. Targeting these altered AMs may support therapeutic approaches in patients with muco-obstructive lung diseases.


Asunto(s)
Fibrosis Quística/metabolismo , Epigenómica/métodos , Macrófagos Alveolares/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Animales , Fibrosis Quística/genética , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Fagocitosis/genética , Fagocitosis/fisiología , Enfermedad Pulmonar Obstructiva Crónica/genética
14.
Nat Commun ; 12(1): 3835, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158510

RESUMEN

Transcription restart after a genotoxic challenge is a fundamental yet poorly understood process. Here, we dissect the interplay between transcription and chromatin restoration after DNA damage by focusing on the human histone chaperone complex HIRA, which is required for transcription recovery post UV. We demonstrate that HIRA is recruited to UV-damaged chromatin via the ubiquitin-dependent segregase VCP to deposit new H3.3 histones. However, this local activity of HIRA is dispensable for transcription recovery. Instead, we reveal a genome-wide function of HIRA in transcription restart that is independent of new H3.3 and not restricted to UV-damaged loci. HIRA coordinates with ASF1B to control transcription restart by two independent pathways: by stabilising the associated subunit UBN2 and by reducing the expression of the transcription repressor ATF3. Thus, HIRA primes UV-damaged chromatin for transcription restart at least in part by relieving transcription inhibition rather than by depositing new H3.3 as an activating bookmark.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN , Chaperonas de Histonas/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Transcripción Genética , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Cromatina/efectos de la radiación , Reparación del ADN , Células HeLa , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Humanos , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Proteína que Contiene Valosina/genética , Proteína que Contiene Valosina/metabolismo
15.
RNA ; 2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-33975916

RESUMEN

Pre-mRNA splicing is carried out by the spliceosome and involves splice site recognition, removal of introns, and ligation of exons. Components of the spliceosome have been shown to interact with the elongating RNA polymerase II (RNAPII) which is thought to allow splicing to occur concurrently with transcription. However, little is known about the regulation and efficiency of co-transcriptional splicing in human cells. In this study, we used Bru-seq and BruChase-seq to determine the co-transcriptional splicing efficiencies of 17,000 introns expressed across 6 human cell lines. We found that less than half of all introns across these 6 cell lines were co-transcriptionally spliced. Splicing efficiencies for individual introns showed variations across cell lines, suggesting that splicing may be regulated in a cell-type specific manner. Moreover, the splicing efficiency of introns varied within genes. The efficiency of co-transcriptional splicing did not correlate with gene length, intron position, splice site strengths, or the intron/neighboring exons GC content. However, we identified binding signals from multiple RNA binding proteins (RBPs) that correlated with splicing efficiency, including core spliceosomal machinery components-such as SF3B4, U2AF1 and U2AF2 showing higher binding signals in poorly spliced introns. In addition, multiple RBPs, such as BUD13, PUM1 and SND1, showed preferential binding in exons that flank introns with high splicing efficiencies. The nascent RNA splicing patterns presented here across multiple cell types add to our understanding of the complexity in RNA splicing, wherein RNA-binding proteins may play important roles in determining splicing outcomes in a cell type- and intron-specific manner.

16.
Nat Commun ; 12(1): 1342, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637760

RESUMEN

Bulky DNA lesions in transcribed strands block RNA polymerase II (RNAPII) elongation and induce a genome-wide transcriptional arrest. The transcription-coupled repair (TCR) pathway efficiently removes transcription-blocking DNA lesions, but how transcription is restored in the genome following DNA repair remains unresolved. Here, we find that the TCR-specific CSB protein loads the PAF1 complex (PAF1C) onto RNAPII in promoter-proximal regions in response to DNA damage. Although dispensable for TCR-mediated repair, PAF1C is essential for transcription recovery after UV irradiation. We find that PAF1C promotes RNAPII pause release in promoter-proximal regions and subsequently acts as a processivity factor that stimulates transcription elongation throughout genes. Our findings expose the molecular basis for a non-canonical PAF1C-dependent pathway that restores transcription throughout the human genome after genotoxic stress.


Asunto(s)
Daño del ADN/fisiología , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN/fisiología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Factores de Transcripción/metabolismo , Núcleo Celular , ADN/efectos de la radiación , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/genética , Mapas de Interacción de Proteínas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Rayos Ultravioleta
17.
J Pharm Pract ; 34(6): 943-951, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32938309

RESUMEN

PURPOSE: Current evidence regarding efficacy and safety of human papillomavirus 9-valent (9vHPV), recombinant zoster (RZV), and CpG-adjuvanted recombinant hepatitis B (HepB-CpG) vaccines in adults with human immunodeficiency virus, inflammatory bowel disease, solid organ transplant, and allogeneic hematopoietic stem cell transplant is reviewed. SUMMARY: Patients immunocompromised due to underlying disease or treatment are at increased risk for infections; however, insufficient understanding of various vaccines' efficacy, safety, indications, and contraindications in this population has led to suboptimal vaccination rates. The Infectious Disease Society of America (IDSA) published guidelines on vaccines in immunocompromised populations in 2013. Since then, several advances have been made including an expanded indication with 9vHPV for use in males and females 9 to 45 years old, and the introduction of new vaccines for herpes zoster (RZV) and hepatitis B (HepB-CpG). Pharmacists are instrumental to vaccination efforts and may benefit from a review of recent vaccine updates. CONCLUSION: The 9vHPV can be used in men and women ages 9 to 45 years old regardless of immune status. RZV safety and efficacy in several immunocompromised populations has been demonstrated; however, manufacturers and major societies have yet to update their recommendations. HepB-CpG may be used in most immunocompromised patients yet remains under-utilized.


Asunto(s)
Alphapapillomavirus , Hepatitis B , Herpes Zóster , Adolescente , Adulto , Niño , Femenino , Hepatitis B/prevención & control , Herpes Zóster/epidemiología , Herpes Zóster/prevención & control , Humanos , Huésped Inmunocomprometido , Masculino , Persona de Mediana Edad , Papillomaviridae , Farmacéuticos , Vacunación , Adulto Joven
19.
Front Immunol ; 11: 572747, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123150

RESUMEN

Infections with respiratory syncytial virus (RSV) occurs repeatedly throughout life because sustained, protective memory responses fail to develop. Why this occurs is not known. During RSV infection the recognition of the virus via the cytosolic RIG-I like receptors and signaling via the adaptor protein MAVS is crucial for mounting an innate immune response. However, if this signaling pathway is important for T cell responses during primary infection and during re-infection is not fully elucidated. We describe a second peak of pro-inflammatory mediators during the primary immune response to RSV that coincides with the arrival of T cells into the lung. This second peak of cytokines/chemokines is regulated differently than the early peak and is largely independent of signaling via MAVS. This was concurrent with Mavs-/- mice mounting a strong T cell response to primary RSV infection, with robust IFN-γ; and Granzyme B production. However, after RSV re-infection, Mavs-/- mice showed fewer CD4+ and CD8+ short term memory T cells and their capacity to produce IFN-γ; and Granzyme B, was decreased. In sum, cytosolic recognition of RSV is important not only for initiating innate anti-viral responses but also for generating or maintaining efficient, short term T cell memory responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Mucosa Respiratoria/fisiología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Granzimas/metabolismo , Humanos , Inmunidad Innata , Memoria Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Noqueados , Transducción de Señal
20.
Nat Commun ; 11(1): 4755, 2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32958772

RESUMEN

We hereby provide the initial portrait of lincNORS, a spliced lincRNA generated by the MIR193BHG locus, entirely distinct from the previously described miR-193b-365a tandem. While inducible by low O2 in a variety of cells and associated with hypoxia in vivo, our studies show that lincNORS is subject to multiple regulatory inputs, including estrogen signals. Biochemically, this lincRNA fine-tunes cellular sterol/steroid biosynthesis by repressing the expression of multiple pathway components. Mechanistically, the function of lincNORS requires the presence of RALY, an RNA-binding protein recently found to be implicated in cholesterol homeostasis. We also noticed the proximity between this locus and naturally occurring genetic variations highly significant for sterol/steroid-related phenotypes, in particular the age of sexual maturation. An integrative analysis of these variants provided a more formal link between these phenotypes and lincNORS, further strengthening the case for its biological relevance.


Asunto(s)
Homeostasis , Oxígeno/metabolismo , ARN Largo no Codificante/fisiología , Esteroles/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Núcleo Celular/metabolismo , Colesterol/metabolismo , Estrógenos/metabolismo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Humanos , Células MCF-7 , Fenotipo , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...