Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 6540, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095402

RÉSUMÉ

Foam cells in atheroma are engorged with lipid droplets (LDs) that contain esters of regulatory lipids whose metabolism remains poorly understood. LD-associated hydrolase (LDAH) has a lipase structure and high affinity for LDs of foam cells. Using knockout and transgenic mice of both sexes, here we show that LDAH inhibits atherosclerosis development and promotes stable lesion architectures. Broad and targeted lipidomic analyzes of primary macrophages and comparative lipid profiling of atheroma identified a broad impact of LDAH on esterified sterols, including natural liver X receptor (LXR) sterol ligands. Transcriptomic analyzes coupled with rescue experiments show that LDAH modulates the expression of prototypical LXR targets and leads macrophages to a less inflammatory phenotype with a profibrotic gene signature. These studies underscore the role of LDs as reservoirs and metabolic hubs of bioactive lipids, and suggest that LDAH favorably modulates macrophage activation and protects against atherosclerosis via lipolytic mobilization of regulatory sterols.


Sujet(s)
Athérosclérose , Gouttelettes lipidiques , Récepteurs hépatiques X , Macrophages , Souris knockout , Animaux , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/prévention et contrôle , Athérosclérose/anatomopathologie , Récepteurs hépatiques X/métabolisme , Récepteurs hépatiques X/génétique , Souris , Mâle , Ligands , Femelle , Gouttelettes lipidiques/métabolisme , Macrophages/métabolisme , Stérols/métabolisme , Cellules spumeuses/métabolisme , Souris transgéniques , Souris de lignée C57BL , Humains , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Activation des macrophages , Sterol Esterase
2.
Heliyon ; 10(12): e32546, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38975228

RÉSUMÉ

Understanding the molecular and physical complexity of the tissue microenvironment (TiME) in the context of its spatiotemporal organization has remained an enduring challenge. Recent advances in engineering and data science are now promising the ability to study the structure, functions, and dynamics of the TiME in unprecedented detail; however, many advances still occur in silos that rarely integrate information to study the TiME in its full detail. This review provides an integrative overview of the engineering principles underlying chemical, optical, electrical, mechanical, and computational science to probe, sense, model, and fabricate the TiME. In individual sections, we first summarize the underlying principles, capabilities, and scope of emerging technologies, the breakthrough discoveries enabled by each technology and recent, promising innovations. We provide perspectives on the potential of these advances in answering critical questions about the TiME and its role in various disease and developmental processes. Finally, we present an integrative view that appreciates the major scientific and educational aspects in the study of the TiME.

3.
bioRxiv ; 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-39026848

RÉSUMÉ

Hepatocyte polyploidy and maturity are critical to acquiring specialized liver functions. Multiple intra- and extracellular factors influence ploidy, but how they cooperate temporally to steer liver polyploidization and maturation or how post-transcriptional mechanisms integrate into these paradigms is unknown. Here, we identified an important regulatory hierarchy in which postnatal activation of Epithelial-Splicing-Regulatory-Protein-2 (ESRP2) stimulates biogenesis of liver-specific microRNA (miR-122), thereby facilitating polyploidization, maturation, and functional competence of hepatocytes. By determining transcriptome-wide protein-RNA interactions in vivo and integrating them with single-cell and bulk hepatocyte RNA-seq datasets, we delineate an ESRP2-driven RNA processing program that drives sequential replacement of fetal-to-adult transcript isoforms. Specifically, ESRP2 binds the primary miR-122 host gene transcript to promote its processing/biogenesis. Combining constitutive and inducible ESRP2 gain- and loss-of-function mice models with miR-122 rescue experiments, we demonstrate that timed activation of ESRP2 augments miR-122-driven program of cytokinesis failure, ensuring proper onset and extent of hepatocyte polyploidization.

4.
Dev Cell ; 59(5): 676-691.e5, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38290519

RÉSUMÉ

Regeneration involves gene expression changes explained in part by context-dependent recruitment of transcriptional activators to distal enhancers. Silencers that engage repressive transcriptional complexes are less studied than enhancers and more technically challenging to validate, but they potentially have profound biological importance for regeneration. Here, we identified candidate silencers through a screening process that examined the ability of DNA sequences to limit injury-induced gene expression in larval zebrafish after fin amputation. A short sequence (s1) on chromosome 5 near several genes that reduce expression during adult fin regeneration could suppress promoter activity in stable transgenic lines and diminish nearby gene expression in knockin lines. High-resolution analysis of chromatin organization identified physical associations of s1 with gene promoters occurring preferentially during fin regeneration, and genomic deletion of s1 elevated the expression of these genes after fin amputation. Our study provides methods to identify "tissue regeneration silencer elements" (TRSEs) with the potential to reduce unnecessary or deleterious gene expression during regeneration.


Sujet(s)
Éléments silenceurs transcriptionnels , Danio zébré , Animaux , Danio zébré/génétique , Animal génétiquement modifié , Régions promotrices (génétique)
5.
J Exp Med ; 220(11)2023 11 06.
Article de Anglais | MEDLINE | ID: mdl-37698554

RÉSUMÉ

Natural killer (NK) cells are lymphocytes capable of controlling tumors and virus infections through direct lysis and cytokine production. While both T and NK cells expand and accumulate in affected tissues, the role of NK cell expansion in tumor and viral control is not well understood. Here, we show that posttranscriptional regulation by the RNA-binding protein HuR is essential for NK cell expansion without negatively affecting effector functions. HuR-deficient NK cells displayed defects in the metaphase of the cell cycle, including decreased expression and alternative splicing of Ska2, a component of the spindle and kinetochore complex. HuR-dependent NK cell expansion contributed to long-term cytomegalovirus control and facilitated control of subcutaneous tumors but not tumor metastases in two independent tumor models. These results show that posttranscriptional regulation by HuR specifically affects NK cell expansion, which is required for the control of long-term virus infection and solid tumors, but not acute infection or tumor metastases, highlighting fundamental differences with antigen-specific T cell control.


Sujet(s)
Protéine-1 similaire à ELAV , Cellules tueuses naturelles , Tumeurs , Maladies virales , Humains , Cycle cellulaire , Division cellulaire , Prolifération cellulaire , Protéine-1 similaire à ELAV/métabolisme , Protéines chromosomiques nonhistones/métabolisme
6.
ACS Nano ; 17(9): 8465-8482, 2023 05 09.
Article de Anglais | MEDLINE | ID: mdl-37126072

RÉSUMÉ

Real-time guidance through fluorescence imaging improves the surgical outcomes of tumor resections, reducing the chances of leaving positive margins behind. As tumors are heterogeneous, it is imperative to interrogate multiple overexpressed cancer biomarkers with high sensitivity and specificity to improve surgical outcomes. However, for accurate tumor delineation and ratiometric detection of tumor biomarkers, current methods require multiple excitation wavelengths to image multiple biomarkers, which is impractical in a clinical setting. Here, we have developed a biomimetic platform comprising near-infrared fluorescent semiconducting polymer nanoparticles (SPNs) with red blood cell membrane (RBC) coating, capable of targeting two representative cell-surface biomarkers (folate, αυß3 integrins) using a single excitation wavelength for tumor delineation during surgical interventions. We evaluate our single excitation ratiometric nanoparticles in in vitro tumor cells, ex vivo tumor-mimicking phantoms, and in vivo mouse xenograft tumor models. Favorable biological properties (improved biocompatibility, prolonged blood circulation, reduced liver uptake) are complemented by superior spectral features: (i) specific fluorescence enhancement in tumor regions with high tumor-to-normal tissue (T/NT) ratios in ex vivo samples and (ii) estimation of cell-surface tumor biomarkers with single wavelength excitation providing insights about cancer progression (metastases). Our single excitation, dual output approach has the potential to differentiate between the tumor and healthy regions and simultaneously provide a qualitative indicator of cancer progression, thereby guiding surgeons in the operating room with the resection process.


Sujet(s)
Nanoparticules , Tumeurs , Humains , Animaux , Souris , Marqueurs biologiques tumoraux , Tumeurs/imagerie diagnostique , Membrane érythrocytaire , Imagerie optique
7.
Nat Commun ; 14(1): 551, 2023 02 09.
Article de Anglais | MEDLINE | ID: mdl-36759613

RÉSUMÉ

Regulation of RNA processing contributes profoundly to tissue development and physiology. Here, we report that serine-arginine-rich splicing factor 1 (SRSF1) is essential for hepatocyte function and survival. Although SRSF1 is mainly known for its many roles in mRNA metabolism, it is also crucial for maintaining genome stability. We show that acute liver damage in the setting of targeted SRSF1 deletion in mice is associated with the excessive formation of deleterious RNA-DNA hybrids (R-loops), which induce DNA damage. Combining hepatocyte-specific transcriptome, proteome, and RNA binding analyses, we demonstrate that widespread genotoxic stress following SRSF1 depletion results in global inhibition of mRNA transcription and protein synthesis, leading to impaired metabolism and trafficking of lipids. Lipid accumulation in SRSF1-deficient hepatocytes is followed by necroptotic cell death, inflammation, and fibrosis, resulting in NASH-like liver pathology. Importantly, SRSF1-depleted human liver cancer cells recapitulate this pathogenesis, illustrating a conserved and fundamental role for SRSF1 in preserving genome integrity and tissue homeostasis. Thus, our study uncovers how the accumulation of detrimental R-loops impedes hepatocellular gene expression, triggering metabolic derangements and liver damage.


Sujet(s)
Stéatose hépatique non alcoolique , Humains , Animaux , Souris , Facteurs d'épissage des ARN/métabolisme , Stéatose hépatique non alcoolique/génétique , ARN/métabolisme , Facteurs d'épissage riches en sérine-arginine/génétique , Facteurs d'épissage riches en sérine-arginine/métabolisme , ARN messager/métabolisme , Épissage alternatif
8.
Nucleic Acids Res ; 51(5): 2397-2414, 2023 03 21.
Article de Anglais | MEDLINE | ID: mdl-36744439

RÉSUMÉ

The intestinal epithelial regeneration is driven by intestinal stem cells under homeostatic conditions. Differentiated intestinal epithelial cells, such as Paneth cells, are capable of acquiring multipotency and contributing to regeneration upon the loss of intestinal stem cells. Paneth cells also support intestinal stem cell survival and regeneration. We report here that depletion of an RNA-binding protein named polypyrimidine tract binding protein 1 (PTBP1) in mouse intestinal epithelial cells causes intestinal stem cell death and epithelial regeneration failure. Mechanistically, we show that PTBP1 inhibits neuronal-like splicing programs in intestinal crypt cells, which is critical for maintaining intestinal stem cell stemness. This function is achieved at least in part through promoting the non-productive splicing of its paralog PTBP2. Moreover, PTBP1 inhibits the expression of an AKT inhibitor PHLDA3 in Paneth cells and permits AKT activation, which presumably maintains Paneth cell plasticity and function in supporting intestinal stem cell niche. We show that PTBP1 directly binds to a CU-rich region in the 3' UTR of Phlda3, which we demonstrate to be critical for downregulating the mRNA and protein levels of Phlda3. Our results thus reveal the multifaceted in vivo regulation of intestinal epithelial regeneration by PTBP1 at the post-transcriptional level.


Sujet(s)
Régulation de l'expression des gènes , Ribonucléoprotéines nucléaires hétérogènes , Protéines proto-oncogènes c-akt , Animaux , Souris , Différenciation cellulaire , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Régénération/génétique , Épissage des ARN
9.
Dev Biol ; 491: 13-30, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36049533

RÉSUMÉ

Corneal Epithelial Stem Cells (CESCs) and their proliferative progeny, the Transit Amplifying Cells (TACs), are responsible for homeostasis and maintaining corneal transparency. Owing to our limited knowledge of cell fates and gene activity within the cornea, the search for unique markers to identify and isolate these cells remains crucial for ocular surface reconstruction. We performed single-cell RNA sequencing of corneal cells from larval and adult stages of Xenopus. Our results indicate that as the cornea develops and matures, there is an increase in cellular diversity, which is accompanied by a substantial shift in transcriptional profile, gene regulatory network and cell-cell communication dynamics. Our data also reveals several novel genes expressed in corneal cells and changes in gene expression during corneal differentiation at both developmental time-points. Importantly, we identify specific basal cell clusters in both the larval and adult cornea that comprise a relatively undifferentiated cell type and express distinct stem cell markers, which we propose are the putative larval and adult CESCs, respectively. This study offers a detailed atlas of single-cell transcriptomes in the frog cornea. In the future, this work will be useful to elucidate the function of novel genes in corneal epithelial homeostasis, wound healing and regeneration.


Sujet(s)
Épithélium antérieur de la cornée , Animaux , Cornée , Épithélium antérieur de la cornée/métabolisme , Larve/génétique , Larve/métabolisme , Cellules souches/métabolisme , Xenopus laevis/génétique , Xenopus laevis/métabolisme
10.
Commun Biol ; 5(1): 351, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35414121

RÉSUMÉ

Single-cell RNA-Sequencing has the potential to provide deep biological insights by revealing complex regulatory interactions across diverse cell phenotypes at single-cell resolution. However, current single-cell gene regulatory network inference methods produce a single regulatory network per input dataset, limiting their capability to uncover complex regulatory relationships across related cell phenotypes. We present SimiC, a single-cell gene regulatory inference framework that overcomes this limitation by jointly inferring distinct, but related, gene regulatory dynamics per phenotype. We show that SimiC uncovers key regulatory dynamics missed by previously proposed methods across a range of systems, both model and non-model alike. In particular, SimiC was able to uncover CAR T cell dynamics after tumor recognition and key regulatory patterns on a regenerating liver, and was able to implicate glial cells in the generation of distinct behavioral states in honeybees. SimiC hence establishes a new approach to quantitating regulatory architectures between distinct cellular phenotypes, with far-reaching implications for systems biology.


Sujet(s)
Réseaux de régulation génique , Tumeurs , Animaux , Abeilles , Régulation de l'expression des gènes , Phénotype , Biologie des systèmes
11.
Genome Res ; 31(4): 576-591, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33649154

RÉSUMÉ

The adult liver has an exceptional ability to regenerate, but how it maintains its specialized functions during regeneration is unclear. Here, we used partial hepatectomy (PHx) in tandem with single-cell transcriptomics to track cellular transitions and heterogeneities of ∼22,000 liver cells through the initiation, progression, and termination phases of mouse liver regeneration. Our results uncovered that, following PHx, a subset of hepatocytes transiently reactivates an early-postnatal-like gene expression program to proliferate, while a distinct population of metabolically hyperactive cells appears to compensate for any temporary deficits in liver function. Cumulative EdU labeling and immunostaining of metabolic, portal, and central vein-specific markers revealed that hepatocyte proliferation after PHx initiates in the midlobular region before proceeding toward the periportal and pericentral areas. We further demonstrate that portal and central vein proximal hepatocytes retain their metabolically active state to preserve essential liver functions while midlobular cells proliferate nearby. Through combined analysis of gene regulatory networks and cell-cell interaction maps, we found that regenerating hepatocytes redeploy key developmental regulons, which are guided by extensive ligand-receptor-mediated signaling events between hepatocytes and nonparenchymal cells. Altogether, our study offers a detailed blueprint of the intercellular crosstalk and cellular reprogramming that balances the metabolic and proliferative requirements of a regenerating liver.


Sujet(s)
Plasticité cellulaire , Régénération hépatique , Foie/cytologie , Foie/métabolisme , Animaux , Prolifération cellulaire , Hépatectomie , Hépatocytes/cytologie , Hépatocytes/métabolisme , Souris , Analyse sur cellule unique , Transcriptome
12.
Biosci Rep ; 40(9)2020 09 30.
Article de Anglais | MEDLINE | ID: mdl-32880391

RÉSUMÉ

Helicobacter pylori is a carcinogenic bacterium that is responsible for 5.5% of all human gastric cancers. H. pylori codes for an unusually large number of restriction-modification (R-M) systems and several of them are strain-specific and phase-variable. HpyAII is a novel Type IIs phase-variable restriction endonuclease present in 26695 strain of H. pylori. We show that HpyAII prefers two-site substrates over one-site substrates for maximal cleavage activity. HpyAII is less stringent in metal ion requirement and shows higher cleavage activity with Ni2+ over Mg2+. Mutational analysis of the putative residues of the HNH motif of HpyAII confirms that the protein has an active HNH site for the cleavage of DNA. However, mutation of the first Histidine residue of the HNH motif to Alanine does not abolish the enzymatic activity, but instead causes loss of fidelity compared with wildtype HpyAII. Previous studies have shown that mutation of the first Histidine residue of the HNH motif of all other known HNH motif motif-containing enzymes completely abolishes enzymatic activity. We found, in the case of HpyAII, mutation of an active site residue leads to the loss of endonuclease fidelity. The present study provides further insights into the evolution of restriction enzymes.


Sujet(s)
Protéines bactériennes/métabolisme , Type II site-specific deoxyribonuclease/métabolisme , Helicobacter pylori/enzymologie , Protéines bactériennes/génétique , Sites de fixation , Domaine catalytique/génétique , Cations divalents/métabolisme , Coenzymes/métabolisme , Clivage de l'ADN , Type II site-specific deoxyribonuclease/génétique , Helicobacter pylori/génétique , Magnésium/métabolisme , Mutagenèse dirigée , Nickel/métabolisme , Spécificité du substrat
13.
RNA ; 26(11): 1603-1620, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32675111

RÉSUMÉ

Cellular quiescence and cell cycle reentry regulate vital biological processes such as cellular development and tissue homeostasis and are controlled by precise regulation of gene expression. The roles of long noncoding RNAs (lncRNAs) during these processes remain to be elucidated. By performing genome-wide transcriptome analyses, we identify differential expression of several hundreds of lncRNAs, including a significant number of the less-characterized class of microRNA-host-gene (MIRHG) lncRNAs or lnc-MIRHGs, during cellular quiescence and cell cycle reentry in human diploid fibroblasts. We observe that MIR222HG lncRNA displays serum-stimulated RNA processing due to enhanced splicing of the host nascent pri-MIR222HG transcript. The pre-mRNA splicing factor SRSF1 negatively regulates the microprocessor-catalyzed cleavage of pri-miR-222, thereby increasing the cellular pool of the mature MIR222HG Association of SRSF1 to pri-MIR222HG, including to a mini-exon, which partially overlaps with the primary miR-222 precursor, promotes serum-stimulated splicing over microRNA processing of MIR222HG Further, we observe that the increased levels of spliced MIR222HG in serum-stimulated cells promote the cell cycle reentry post quiescence in a microRNA-independent manner. MIR222HG interacts with DNM3OS, another lncRNA whose expression is elevated upon serum-stimulation, and promotes cell cycle reentry. The double-stranded RNA binding protein ILF3/2 complex facilitates MIR222HG:DNM3OS RNP complex assembly, thereby promoting DNM3OS RNA stability. Our study identifies a novel mechanism whereby competition between the splicing and microprocessor machinery modulates the serum-induced RNA processing of MIR222HG, which dictates cell cycle reentry.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Poumon/cytologie , ARN long non codant/génétique , Facteurs d'épissage riches en sérine-arginine/métabolisme , Sérum/composition chimique , Cycle cellulaire , Lignée cellulaire , Fibroblastes/composition chimique , Fibroblastes/cytologie , Cellules HEK293 , Humains , Poumon/composition chimique , Facteur nucléaire-45/métabolisme , Facteurs nucléaires-90/métabolisme , Maturation post-transcriptionnelle des ARN , Épissage des ARN , Analyse de séquence d'ARN , Imagerie de molécules uniques , Régulation positive ,
14.
ACS Appl Mater Interfaces ; 12(14): 16137-16149, 2020 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-32182420

RÉSUMÉ

By using complementary DNA sequences as surface ligands, we selectively allow two individual diffusing "dual-color" carbon dots to interact in situ and in vitro. Spontaneous nanoscale oxidation of surface-abundant nitroso-/nitro-functionalities leads to two distinctly colored carbon dots (CD) which are isolated by polarity driven chromatographic separation. Green- and red-emitting carbon dots (gCD and rCD) were decorated by complementary single-stranded DNAs which produce a marked increase in the fluorescence emission of the respective carbon dots. Mutual colloidal interactions are achieved through hybridization of complementary DNA base pairs attached to the respective particles, resulting in quenching of their photoluminescence. The observed post-hybridization quenching is presumably due to a combined effect from an aggregation of CDs post duplex DNA formation and close proximity of multicolored CDs, having overlapped spectral regions leading to a nonradiative energy transfer process possibly released as heat. This strategy may contribute to the rational design of mutually interacting carbon dots for a better control over the resulting assembly structure for studying different biological phenomenon including molecular cytogenetics. One of the newly synthesized CDs was successfully used to image intracellular location of GAPDH mRNA using an event of change in fluorescence intensity (FI) of CDs. This selectivity was introduced by conjugating an oligonucleotide harboring complementary sequence to GAPDH mRNA. FI of this conjugated carbon dot, rCD-GAPDH, was also found to decrease in the presence of Ca2+, varied in relation to H+ concentrations, and could serve as a tool to quantify the intracellular concentrations of Ca2+ and pH value (H+) which can give important information about cell survival. Therefore, CD-oligonucleotide conjugates could serve as efficient probes for cellular events and interventions.


Sujet(s)
Techniques de biocapteur , Glyceraldehyde 3-phosphate dehydrogenase (phosphorylating)/isolement et purification , Hybridation d'acides nucléiques , Oligonucléotides/composition chimique , Carbone/composition chimique , ADN simple brin/composition chimique , Glyceraldehyde 3-phosphate dehydrogenase (phosphorylating)/composition chimique , Humains , Boîtes quantiques/composition chimique , ARN messager/composition chimique , ARN messager/isolement et purification
15.
Dev Cell ; 52(6): 748-763.e6, 2020 03 23.
Article de Anglais | MEDLINE | ID: mdl-32109384

RÉSUMÉ

Myotonic dystrophy type 1 (DM1) is a multisystemic genetic disorder caused by the CTG repeat expansion in the 3'-untranslated region of DMPK gene. Heart dysfunctions occur in ∼80% of DM1 patients and are the second leading cause of DM1-related deaths. Herein, we report that upregulation of a non-muscle splice isoform of RNA-binding protein RBFOX2 in DM1 heart tissue-due to altered splicing factor and microRNA activities-induces cardiac conduction defects in DM1 individuals. Mice engineered to express the non-muscle RBFOX240 isoform in heart via tetracycline-inducible transgenesis, or CRISPR/Cas9-mediated genome editing, reproduced DM1-related cardiac conduction delay and spontaneous episodes of arrhythmia. Further, by integrating RNA binding with cardiac transcriptome datasets from DM1 patients and mice expressing the non-muscle RBFOX2 isoform, we identified RBFOX240-driven splicing defects in voltage-gated sodium and potassium channels, which alter their electrophysiological properties. Thus, our results uncover a trans-dominant role for an aberrantly expressed RBFOX240 isoform in DM1 cardiac pathogenesis.


Sujet(s)
Potentiels d'action , Rythme cardiaque , Dystrophie myotonique/génétique , Facteurs d'épissage des ARN/génétique , Épissage des ARN , Protéines de répression/génétique , Adulte , Animaux , Cellules cultivées , Femelle , Humains , Mâle , Souris de lignée C57BL , Souris de lignée ICR , microARN/génétique , microARN/métabolisme , Adulte d'âge moyen , Myocytes cardiaques/métabolisme , Myocytes cardiaques/physiologie , Dystrophie myotonique/métabolisme , Dystrophie myotonique/physiopathologie , Canaux potassiques voltage-dépendants/génétique , Canaux potassiques voltage-dépendants/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Facteurs d'épissage des ARN/métabolisme , Protéines de répression/métabolisme , Canaux sodiques voltage-dépendants/génétique , Canaux sodiques voltage-dépendants/métabolisme
16.
Semin Cell Dev Biol ; 100: 74-87, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31980376

RÉSUMÉ

Recent advances in genetics and genomics have reinvigorated the field of liver regeneration. It is now possible to combine lineage-tracing with genome-wide studies to genetically mark individual liver cells and their progenies and detect precise changes in their genome, transcriptome, and proteome under normal versus regenerative settings. The recent use of single-cell RNA sequencing methodologies in model organisms has, in some ways, transformed our understanding of the cellular and molecular biology of liver regeneration. Here, we review the latest strides in our knowledge of general principles that coordinate regeneration of the liver and reflect on some conflicting evidence and controversies surrounding this topic. We consider the prominent mechanisms that stimulate homeostasis-related vis-à-vis injury-driven regenerative responses, highlight the likely cellular sources/depots that reconstitute the liver following various injuries and discuss the extrinsic and intrinsic signals that direct liver cells to proliferate, de-differentiate, or trans-differentiate while the tissue recovers from acute or chronic damage.


Sujet(s)
Régénération hépatique , Foie/cytologie , Foie/métabolisme , Animaux , Différenciation cellulaire , Prolifération cellulaire , Humains
17.
J Clin Invest ; 130(4): 2129-2145, 2020 04 01.
Article de Anglais | MEDLINE | ID: mdl-31945016

RÉSUMÉ

Severe alcoholic hepatitis (SAH) is a deadly liver disease without an effective medical therapy. Although SAH mortality is known to correlate with hepatic accumulation of immature liver cells, why this occurs and how it causes death are unclear. Here, we demonstrate that expression of epithelial splicing regulatory protein 2 (ESRP2), an RNA-splicing factor that maintains the nonproliferative, mature phenotype of adult hepatocytes, was suppressed in both human SAH and various mouse models of SAH in parallel with the severity of alcohol consumption and liver damage. Inflammatory cytokines released by excessive alcohol ingestion reprogrammed adult hepatocytes into proliferative, fetal-like cells by suppressing ESRP2. Sustained loss of ESRP2 permitted reemergence of a fetal RNA-splicing program that attenuates the Hippo signaling pathway and thus allows fetal transcriptional regulators to accumulate in adult liver. We further showed that depleting ESRP2 in mice exacerbated alcohol-induced steatohepatitis, enabling surviving hepatocytes to shed adult hepatocyte functions and become more regenerative, but threatening overall survival by populating the liver with functionally immature hepatocytes. Our findings revealed a mechanism that explains why liver failure develops in patients with the clinical syndrome of SAH, suggesting that recovery from SAH might be improved by limiting adult-to-fetal reprogramming in hepatocytes.


Sujet(s)
Épissage alternatif , Reprogrammation cellulaire , Hépatite alcoolique/métabolisme , Hépatocytes/métabolisme , Protéines de liaison à l'ARN/métabolisme , Transduction du signal , Animaux , Lignée cellulaire , Survie cellulaire , Modèles animaux de maladie humaine , Femelle , Hépatite alcoolique/génétique , Hépatite alcoolique/anatomopathologie , Hépatocytes/anatomopathologie , Humains , Mâle , Souris , Souris knockout , Protéines de liaison à l'ARN/génétique , Indice de gravité de la maladie
18.
Methods ; 155: 131-139, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30500367

RÉSUMÉ

The regulation of gene expression occurs through complex relationships between transcription, processing, turnover, and translation, which are only beginning to be elucidated. We know that at least for certain messenger (m) RNAs, processing, modifications, and sequence elements can greatly influence their translational output through recognition by translation and turn-over machinery. Recently, we and others have combined high-throughput sequencing technologies with traditional biochemical methods of studying translation to extend our understanding of these relationships. Additionally, there is growing importance given to how these processes may be regulated across varied cell types as a means to achieve tissue-specific expression of proteins. Here, we provide an in-depth methodology for polysome profiling to dissect the composition of mRNAs and proteins that make up the translatome from both whole tissues and a specific cell type isolated from mammalian tissue. Also, we provide a detailed computational workflow for the analysis of the next-generation sequencing data generated from these experiments.


Sujet(s)
Biologie informatique/méthodes , Polyribosomes/génétique , Biosynthèse des protéines , ARN messager/génétique , Analyse de séquence d'ARN/statistiques et données numériques , Animaux , Encéphale/cytologie , Encéphale/métabolisme , Fractionnement cellulaire/méthodes , Centrifugation en gradient de densité/méthodes , Gene Ontology , Réseaux de régulation génique , Hépatocytes/cytologie , Hépatocytes/métabolisme , Séquençage nucléotidique à haut débit , Foie/cytologie , Foie/métabolisme , Souris , Annotation de séquence moléculaire , Myocarde/cytologie , Myocarde/métabolisme , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Neurones/cytologie , Neurones/métabolisme , Spécificité d'organe , Polyribosomes/classification , Polyribosomes/métabolisme , ARN messager/métabolisme
19.
Nat Struct Mol Biol ; 25(10): 928-939, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30250226

RÉSUMÉ

During liver regeneration, most new hepatocytes arise via self-duplication; yet, the underlying mechanisms that drive hepatocyte proliferation following injury remain poorly defined. By combining high-resolution transcriptome and polysome profiling of hepatocytes purified from quiescent and toxin-injured mouse livers, we uncover pervasive alterations in messenger RNA translation of metabolic and RNA-processing factors, which modulate the protein levels of a set of splicing regulators. Specifically, downregulation of the splicing regulator ESRP2 activates a neonatal alternative splicing program that rewires the Hippo signaling pathway in regenerating hepatocytes. We show that production of neonatal splice isoforms attenuates Hippo signaling, enables greater transcriptional activation of downstream target genes, and facilitates liver regeneration. We further demonstrate that ESRP2 deletion in mice causes excessive hepatocyte proliferation upon injury, whereas forced expression of ESRP2 inhibits proliferation by suppressing the expression of neonatal Hippo pathway isoforms. Thus, our findings reveal an alternative splicing axis that supports regeneration following chronic liver injury.


Sujet(s)
Épissage alternatif , Régénération hépatique/génétique , Protein-Serine-Threonine Kinases/physiologie , Animaux , Prolifération cellulaire/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Hépatocytes/cytologie , Hépatocytes/métabolisme , Hépatocytes/physiologie , Voie de signalisation Hippo , Souris , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , ARN messager/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/physiologie , Transduction du signal/génétique
20.
F1000Res ; 5: 2668, 2016.
Article de Anglais | MEDLINE | ID: mdl-27909578

RÉSUMÉ

Alternative splicing, polyadenylation, and chemical modifications of RNA generate astonishing complexity within eukaryotic transcriptomes. The last decade has brought numerous advances in sequencing technologies that allow biologists to investigate these phenomena with greater depth and accuracy while reducing time and cost. A commensurate development in biochemical techniques for the enrichment and analysis of different RNA variants has accompanied the advancement of global sequencing analysis platforms. Here, we present a detailed overview of the latest biochemical methods, along with bioinformatics pipelines that have aided in identifying different RNA variants. We also highlight the ongoing developments and challenges associated with RNA variant detection and quantification, including sample heterogeneity and isolation, as well as 'Omics' big data handling.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE