Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.725
Filtrer
1.
Mol Cancer ; 23(1): 142, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987766

RÉSUMÉ

BACKGROUND: Breast cancer (BC) is the most common cancer in women, with triple negative BC (TNBC) accounting for 20% of cases. While early detection and targeted therapies have improved overall life expectancy, TNBC remains resistant to current treatments. Although parity reduces the lifetime risk of developing BC, pregnancy increases the risk of developing TNBC for years after childbirth. Although numerous gene mutations have been associated with BC, no single gene alteration has been identified as a universal driver. RRAS2 is a RAS-related GTPase rarely found mutated in cancer. METHODS: Conditional knock-in mice were generated to overexpress wild type human RRAS2 in mammary epithelial cells. A human sample cohort was analyzed by RT-qPCR to measure RRAS2 transcriptional expression and to determine the frequency of both a single-nucleotide polymorphism (SNP rs8570) in the 3'UTR region of RRAS2 and of genomic DNA amplification in tumoral and non-tumoral human BC samples. RESULTS: Here we show that overexpression of wild-type RRAS2 in mice is sufficient to develop TNBC in 100% of females in a pregnancy-dependent manner. In human BC, wild-type RRAS2 is overexpressed in 68% of tumors across grade, location, and molecular type, surpassing the prevalence of any previously implicated alteration. Still, RRAS2 overexpression is notably higher and more frequent in TNBC and young parous patients. The increased prevalence of the alternate C allele at the SNP position in tumor samples, along with frequent RRAS2 gene amplification in both tumors and blood of BC patients, suggests a cause-and-effect relationship between RRAS2 overexpression and breast cancer. CONCLUSIONS: Higher than normal expression of RRAS2 not bearing activating mutations is a key driver in the majority of breast cancers, especially those of the triple-negative type and those linked to pregnancy.


Sujet(s)
Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Femelle , Animaux , Humains , Souris , Grossesse , Oncogènes , Polymorphisme de nucléotide simple , Période du postpartum/génétique , Mutation , Régulation de l'expression des gènes tumoraux , Techniques de knock-in de gènes , Protéines G ras/génétique , Protéines G ras/métabolisme , Souris transgéniques , Modèles animaux de maladie humaine , Protéines membranaires , Protéines G monomériques
2.
Molecules ; 29(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38930955

RÉSUMÉ

The CRISPR-Cas9 system has emerged as the most prevalent gene editing technology due to its simplicity, high efficiency, and low cost. However, the homology-directed repair (HDR)-mediated gene knock-in in this system suffers from low efficiency, which limits its application in animal model preparation, gene therapy, and agricultural genetic improvement. Here, we report the design and optimization of a simple and efficient reporter-based assay to visualize and quantify HDR efficiency. Through random screening of a small molecule compound library, two groups of compounds, including the topoisomerase inhibitors and PIM1 kinase inhibitors, have been identified to promote HDR. Two representative compounds, etoposide and quercetagetin, also significantly enhance the efficiency of CRISPR-Cas9 and HDR-mediated gene knock-in in mouse embryos. Our study not only provides an assay to screen compounds that may facilitate HDR but also identifies useful tool compounds to facilitate the construction of genetically modified animal models with the CRISPR-Cas9 system.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Inhibiteurs de protéines kinases , Protéines proto-oncogènes c-pim-1 , Édition de gène/méthodes , Protéines proto-oncogènes c-pim-1/antagonistes et inhibiteurs , Protéines proto-oncogènes c-pim-1/génétique , Protéines proto-oncogènes c-pim-1/métabolisme , Animaux , Souris , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs des topoisomérases/pharmacologie , Humains , Réparation de l'ADN par recombinaison/effets des médicaments et des substances chimiques , Techniques de knock-in de gènes
3.
Nat Commun ; 15(1): 5489, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38942786

RÉSUMÉ

Lipid droplets (LDs) are dynamic lipid storage organelles. They are tightly linked to metabolism and can exert protective functions, making them important players in health and disease. Most LD studies in vivo rely on staining methods, providing only a snapshot. We therefore developed a LD-reporter mouse by labelling the endogenous LD coat protein perilipin 2 (PLIN2) with tdTomato, enabling staining-free fluorescent LD visualisation in living and fixed tissues and cells. Here we validate this model under standard and high-fat diet conditions and demonstrate that LDs are highly abundant in various cell types in the healthy brain, including neurons, astrocytes, ependymal cells, neural stem/progenitor cells and microglia. Furthermore, we also show that LDs are abundant during brain development and can be visualized using live imaging of embryonic slices. Taken together, our tdTom-Plin2 mouse serves as a novel tool to study LDs and their dynamics under both physiological and diseased conditions in all tissues expressing Plin2.


Sujet(s)
Encéphale , Gouttelettes lipidiques , Périlipine-2 , Animaux , Périlipine-2/métabolisme , Périlipine-2/génétique , Gouttelettes lipidiques/métabolisme , Encéphale/métabolisme , Souris , Neurones/métabolisme , Techniques de knock-in de gènes , Souris transgéniques , Femelle , Protéines luminescentes/métabolisme , Protéines luminescentes/génétique , Mâle , Astrocytes/métabolisme , Alimentation riche en graisse , Souris de lignée C57BL , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Microglie/métabolisme
4.
Yi Chuan ; 46(6): 466-477, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38886150

RÉSUMÉ

Gene knock-in in mammalian cells usually uses homology-directed repair (HDR) mechanism to integrate exogenous DNA template into the target genome site. However, HDR efficiency is often low, and the co-localization of exogenous DNA template and target genome site is one of the key limiting factors. To improve the efficiency of HDR mediated by CRISPR/Cas9 system, our team and previous studies fused different adaptor proteins with SpCas9 protein and expressed them. By using their characteristics of binding to specific DNA sequences, many different CRISPR/SpCas9 donor adapter gene editing systems were constructed. In this study, we used them to knock-in eGFP gene at the 3'-end of the terminal exon of GAPDH and ACTB genes in HEK293T cells to facilitate a comparison and optimization of these systems. We utilized an optimized donor DNA template design method, validated the knock-in accuracy via PCR and Sanger sequencing, and assessed the efficiency using flow cytometry. The results showed that the fusion of yGal4BD, hGal4BD, hLacI, hTHAP11 as well as N57 and other adaptor proteins with the C-terminus of SpCas9 protein had no significant effect on its activity. At the GAPDH site, the donor adapter systems of SpCas9 fused with yGal4BD, hGal4BD, hLacI and hTHAP11 significantly improved the knock-in efficiency. At the ACTB site, SpCas9 fused with yGal4BD and hGal4BD significantly improved the knock-in efficiency. Furthermore, increasing the number of BS in the donor DNA template was beneficial to enhance the knock-in efficiency mediated by SpCas9-hTHAP11 system. In conclusion, this study compares and optimizes multiple CRISPR/Cas9 donor adapter gene editing systems, providing valuable insights for future gene editing applications.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Humains , Édition de gène/méthodes , Cellules HEK293 , Techniques de knock-in de gènes/méthodes , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme
5.
Elife ; 122024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38869243

RÉSUMÉ

An expanded CAG repeat in the huntingtin gene (HTT) causes Huntington's disease (HD). Since the length of uninterrupted CAG repeat, not polyglutamine, determines the age-at-onset in HD, base editing strategies to convert CAG to CAA are anticipated to delay onset by shortening the uninterrupted CAG repeat. Here, we developed base editing strategies to convert CAG in the repeat to CAA and determined their molecular outcomes and effects on relevant disease phenotypes. Base editing strategies employing combinations of cytosine base editors and guide RNAs (gRNAs) efficiently converted CAG to CAA at various sites in the CAG repeat without generating significant indels, off-target edits, or transcriptome alterations, demonstrating their feasibility and specificity. Candidate BE strategies converted CAG to CAA on both expanded and non-expanded CAG repeats without altering HTT mRNA and protein levels. In addition, somatic CAG repeat expansion, which is the major disease driver in HD, was significantly decreased in the liver by a candidate BE strategy treatment in HD knock-in mice carrying canonical CAG repeats. Notably, CAG repeat expansion was abolished entirely in HD knock-in mice carrying CAA-interrupted repeats, supporting the therapeutic potential of CAG-to-CAA conversion strategies in HD and potentially other repeat expansion disorders.


Sujet(s)
Édition de gène , Protéine huntingtine , Maladie de Huntington , Expansion de trinucléotide répété , Maladie de Huntington/génétique , Maladie de Huntington/thérapie , Animaux , Édition de gène/méthodes , Souris , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Expansion de trinucléotide répété/génétique , Modèles animaux de maladie humaine , Humains , Mutation , Techniques de knock-in de gènes
6.
Dev Biol ; 514: 78-86, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38880275

RÉSUMÉ

The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.


Sujet(s)
Coeur , Integrases , Facteur-1 liant le domaine de régulation positive I , Animaux , Souris , Coeur/embryologie , Integrases/métabolisme , Integrases/génétique , Facteur-1 liant le domaine de régulation positive I/génétique , Facteur-1 liant le domaine de régulation positive I/métabolisme , Facteurs de transcription MEF2/génétique , Facteurs de transcription MEF2/métabolisme , Souris transgéniques , Régulation de l'expression des gènes au cours du développement/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Techniques de knock-in de gènes
7.
Pestic Biochem Physiol ; 202: 105953, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38879307

RÉSUMÉ

The brown planthopper (Nilaparvata lugens) is a major destructive rice pest in Asia. High levels of insecticide resistance have been frequently reported, and the G932C mutation in the chitin synthase 1 (CHS1) gene has been found to mediate buprofezin resistance. However, there has been no direct evidence to confirm the functional significance of the single G932C substitution mutation leading to buprofezin resistance in N. lugens. Here, we successfully constructed a knock-in homozygous strain (Nl-G932C) of N. lugens using CRISPR/Cas9 coupled with homology-directed repair (HDR). Compared with the background strain susceptible to buprofezin (Nl-SS), the knock-in strain (Nl-G932C) showed a 94.9-fold resistance to buprofezin. Furthermore, resistant strains (Nl-932C) isolated from the field exhibited a 2078.8-fold resistance to buprofezin, indicating that there are other mechanisms contributing to buprofezin resistance in the field. Inheritance analysis showed that the resistance trait is incomplete dominance. In addition, the Nl-G932C strain had a relative fitness of 0.33 with a substantially decreased survival rate, emergence rate, and fecundity. This study provided in vivo functional evidence for the causality of G932C substitution mutation of CHS1 with buprofezin resistance and valuable information for facilitating the development of resistance management strategies in N. lugens. This is the first example of using CRISPR/Cas9 gene-editing technology in a hemipteran insect to directly confirm the role of a candidate target site mutation in insecticide resistance.


Sujet(s)
Systèmes CRISPR-Cas , Chitine synthase , Hemiptera , Résistance aux insecticides , Insecticides , Thiadiazines , Animaux , Hemiptera/génétique , Résistance aux insecticides/génétique , Thiadiazines/pharmacologie , Chitine synthase/génétique , Insecticides/pharmacologie , Mutation , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Techniques de knock-in de gènes , Femelle , Mâle
8.
Platelets ; 35(1): 2369766, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38904212

RÉSUMÉ

Receptor-induced tyrosine phosphorylation of spleen tyrosine kinase (Syk) has been studied extensively in hematopoietic cells. Metabolic mapping and high-resolution mass spectrometry, however, indicate that one of the most frequently detected phosphorylation sites encompassed S297 (S291 in mice) located within the linker B region of Syk. It has been reported that Protein kinase C (PKC) phosphorylates Syk S297, thus influencing Syk activity. However, conflicting studies suggest that this phosphorylation enhances as well as reduces Syk activity. To clarify the function of this site, we generated Syk S291A knock-in mice. We used platelets as a model system as they possess Glycoprotein VI (GPVI), a receptor containing an immunoreceptor tyrosine-based activation motif (ITAM) which transduces signals through Syk. Our analysis of the homozygous mice indicated that the knock-in platelets express only one isoform of Syk, while the wild-type expresses two isoforms at 69 and 66 kDa. When the GPVI receptor was activated with collagen-related peptide (CRP), we observed an increase in functional responses and phosphorylations in Syk S291A platelets. This potentiation did not occur with AYPGKF or 2-MeSADP, although they also activate PKC isoforms. Although there was potentiation of platelet functional responses, there was no difference in tail bleeding times. However, the time to occlusion in the FeCl3 injury model was enhanced. These data indicate that the effects of Syk S291 phosphorylation represent a significant outcome on platelet activation and signaling in vitro but also reveals its multifaceted nature demonstrated by the differential effects on physiological responses in vivo.


What is the context Spleen tyrosine kinase (Syk) is present a number of cells and important in controlling the functions of various cells and organs.Syk is known to exist in two isoforms Syk L (long form or Syk A) and Syk S (short form or Syk B).It is known that phosphorylation events regulate Syk activation and activity.In several inflammatory disease conditions, Syk mutants are known to play a role.Phosphorylation of the Syk residue Serine 291 is known to occur, but its function in the regulation of Syk activation or activity is not known.What is new In this study, we generated a mutant mouse Syk S291A, which cannot be phosphorylated on serine residue. We evaluated the function of platelets isolated from these mice and compared them to platelets isolated from wild type littermates.We observed that the mutation in Syk L unexpectedly caused Syk S to disappear from a number of tissues.Platelet functions are enhanced in mutant mouse platelets compared to those from wild-type mice.What is the impact These studies enhance our understanding of the impact of Serine 291 phosphorylation on the function of Syk in platelets.


Sujet(s)
Plaquettes , Transduction du signal , Syk kinase , Animaux , Syk kinase/métabolisme , Plaquettes/métabolisme , Souris , Phosphorylation , Motif d'activation de l'immunorécepteur dépendant de la tyrosine , Techniques de knock-in de gènes , Humains , Glycoprotéines de membrane plaquettaire/métabolisme , Activation plaquettaire
9.
Nat Commun ; 15(1): 4756, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834544

RÉSUMÉ

Given the absence of approved treatments for pathogenic variants in Peripherin-2 (PRPH2), it is imperative to identify a universally effective therapeutic target for PRPH2 pathogenic variants. To test the hypothesis that formation of the elongated discs in presence of PRPH2 pathogenic variants is due to the presence of the full complement of rhodopsin in absence of the required amounts of functional PRPH2. Here we demonstrate the therapeutic potential of reducing rhodopsin levels in ameliorating disease phenotype in knockin models for p.Lys154del (c.458-460del) and p.Tyr141Cys (c.422 A > G) in PRPH2. Reducing rhodopsin levels improves physiological function, mitigates the severity of disc abnormalities, and decreases retinal gliosis. Additionally, intravitreal injections of a rhodopsin-specific antisense oligonucleotide successfully enhance the physiological function of photoreceptors and improves the ultrastructure of discs in mutant mice. Presented findings shows that reducing rhodopsin levels is an effective therapeutic strategy for the treatment of inherited retinal degeneration associated with PRPH2 pathogenic variants.


Sujet(s)
Périphérines , Rhodopsine , Périphérines/génétique , Périphérines/métabolisme , Animaux , Rhodopsine/génétique , Rhodopsine/métabolisme , Souris , Humains , Modèles animaux de maladie humaine , Régulation négative , Dégénérescence de la rétine/génétique , Dégénérescence de la rétine/métabolisme , Dégénérescence de la rétine/thérapie , Oligonucléotides antisens/génétique , Rétine/métabolisme , Rétine/anatomopathologie , Rétinopathies/génétique , Rétinopathies/métabolisme , Rétinopathies/anatomopathologie , Rétinopathies/thérapie , Souris de lignée C57BL , Mutation , Femelle , Techniques de knock-in de gènes , Mâle
10.
PLoS Genet ; 20(6): e1011298, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38870088

RÉSUMÉ

Tardigrades are small aquatic invertebrates known for their remarkable tolerance to diverse extreme stresses. To elucidate the in vivo mechanisms underlying this extraordinary resilience, methods for genetically manipulating tardigrades have long been desired. Despite our prior success in somatic cell gene editing by microinjecting Cas9 ribonucleoproteins (RNPs) into the body cavity of tardigrades, the generation of gene-edited individuals remained elusive. In this study, employing an extremotolerant parthenogenetic tardigrade species, Ramazzottius varieornatus, we established conditions that led to the generation of gene-edited tardigrade individuals. Drawing inspiration from the direct parental CRISPR (DIPA-CRISPR) technique employed in several insects, we simply injected a concentrated Cas9 RNP solution into the body cavity of parental females shortly before their initial oviposition. This approach yielded gene-edited G0 progeny. Notably, only a single allele was predominantly detected at the target locus for each G0 individual, indicative of homozygous mutations. By co-injecting single-stranded oligodeoxynucleotides (ssODNs) with Cas9 RNPs, we achieved the generation of homozygously knocked-in G0 progeny, and these edited alleles were inherited by G1/G2 progeny. This is the first example of heritable gene editing in the entire phylum of Tardigrada. This establishment of a straightforward method for generating homozygous knockout/knock-in individuals not only facilitates in vivo analyses of the molecular mechanisms underpinning extreme tolerance, but also opens up avenues for exploring various topics, including Evo-Devo, in tardigrades.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Homozygote , Parthénogenèse , Tardigrada , Animaux , Tardigrada/génétique , Édition de gène/méthodes , Parthénogenèse/génétique , Femelle , Techniques de knock-in de gènes/méthodes , Techniques de knock-out de gènes , Allèles
11.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38743626

RÉSUMÉ

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Sujet(s)
Maladies héréditaires de l'oeil , Maladies génétiques liées au chromosome X , Myopie , Héméralopie , Rhodopsine , Animaux , Héméralopie/génétique , Héméralopie/métabolisme , Maladies héréditaires de l'oeil/génétique , Maladies héréditaires de l'oeil/métabolisme , Souris , Rhodopsine/génétique , Rhodopsine/métabolisme , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/métabolisme , Myopie/génétique , Myopie/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/anatomopathologie , Obscurité , Transducine/génétique , Transducine/métabolisme , Techniques de knock-in de gènes , Modèles animaux de maladie humaine
12.
Stem Cell Res ; 78: 103445, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38820864

RÉSUMÉ

Forkhead box protein J1 (FOXJ1), a member of the forkhead family, is an important transcription factor regulating multiciliated cell differentiation and motile ciliogenic program. Here, we established a FOXJ1- EGFP knock-in human embryonic stem cell (hESC) line by inserting a P2A-EGFP gene cassette of FOXJ1 using CRISPR/Cas9 system. The reporter cell line retained a normal karyotype, expressed comparable pluripotent marker genes, and maintained differentiation potential. This reporter cell line enables live identification of multiciliated cells during the general lung differentiation and will be a valuable tool for studying the multiciliated cell differentiation, ciliogenesis and mechanism of related pulmonary diseases.


Sujet(s)
Systèmes CRISPR-Cas , Facteurs de transcription Forkhead , Cellules souches embryonnaires humaines , Humains , Cellules souches embryonnaires humaines/métabolisme , Cellules souches embryonnaires humaines/cytologie , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Lignée cellulaire , Différenciation cellulaire , Techniques de knock-in de gènes/méthodes , Ciblage de gène/méthodes , Gènes rapporteurs
13.
Matrix Biol ; 131: 17-29, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38759902

RÉSUMÉ

Amelogenin (AMELX), the predominant matrix protein in enamel formation, contains a singular phosphorylation site at Serine 16 (S16) that greatly enhances AMELX's capacity to stabilize amorphous calcium phosphate (ACP) and inhibit its transformation to apatitic enamel crystals. To explore the potential role of AMELX phosphorylation in vivo, we developed a knock-in (KI) mouse model in which AMELX phosphorylation is prevented by substituting S16 with Ala (A). As anticipated, AMELXS16A KI mice displayed a severe phenotype characterized by weak hypoplastic enamel, absence of enamel rods, extensive ectopic calcifications, a greater rate of ACP transformation to apatitic crystals, and progressive cell pathology in enamel-forming cells (ameloblasts). In the present investigation, our focus was on understanding the mechanisms of action of phosphorylated AMELX in amelogenesis. We have hypothesized that the absence of AMELX phosphorylation would result in a loss of controlled mineralization during the secretory stage of amelogenesis, leading to an enhanced rate of enamel mineralization that causes enamel acidification due to excessive proton release. To test these hypotheses, we employed microcomputed tomography (µCT), colorimetric pH assessment, and Fourier Transform Infrared (FTIR) microspectroscopy of apical portions of mandibular incisors from 8-week old wildtype (WT) and KI mice. As hypothesized, µCT analyses demonstrated significantly higher rates of enamel mineral densification in KI mice during the secretory stage compared to the WT. Despite a greater rate of enamel densification, maximal KI enamel thickness increased at a significantly lower rate than that of the WT during the secretory stage of amelogenesis, reaching a thickness in mid-maturation that is approximately half that of the WT. pH assessments revealed a lower pH in secretory enamel in KI compared to WT mice, as hypothesized. FTIR findings further demonstrated that KI enamel is comprised of significantly greater amounts of acid phosphate compared to the WT, consistent with our pH assessments. Furthermore, FTIR microspectroscopy indicated a significantly higher mineral-to-organic ratio in KI enamel, as supported by µCT findings. Collectively, our current findings demonstrate that phosphorylated AMELX plays crucial mechanistic roles in regulating the rate of enamel mineral formation, and in maintaining physico-chemical homeostasis and the enamel growth pattern during early stages of amelogenesis.


Sujet(s)
Améloblastes , Amélogenèse , Amélogénine , Émail dentaire , Microtomographie aux rayons X , Animaux , Amélogénine/métabolisme , Amélogénine/génétique , Phosphorylation , Émail dentaire/métabolisme , Émail dentaire/croissance et développement , Souris , Amélogenèse/génétique , Améloblastes/métabolisme , Techniques de knock-in de gènes , Phosphates de calcium/métabolisme , Concentration en ions d'hydrogène
14.
Histochem Cell Biol ; 162(1-2): 41-52, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38762823

RÉSUMÉ

During development and differentiation, histone modifications dynamically change locally and globally, associated with transcriptional regulation, DNA replication and repair, and chromosome condensation. The level of histone H4 Lys20 monomethylation (H4K20me1) increases during the G2 to M phases of the cell cycle and is enriched in facultative heterochromatin, such as inactive X chromosomes in cycling cells. To track the dynamic changes of H4K20me1 in living cells, we have developed a genetically encoded modification-specific intracellular antibody (mintbody) probe that specifically binds to the modification. Here, we report the generation of knock-in mice in which the coding sequence of the mCherry-tagged version of the H4K20me1-mintbody is inserted into the Rosa26 locus. The knock-in mice, which ubiquitously expressed the H4K20me1-mintbody, developed normally and were fertile, indicating that the expression of the probe does not disturb the cell growth, development, or differentiation. Various tissues isolated from the knock-in mice exhibited nuclear fluorescence without the need for fixation. The H4K20me1-mintbody was enriched in inactive X chromosomes in developing embryos and in XY bodies during spermatogenesis. The knock-in mice will be useful for the histochemical analysis of H4K20me1 in any cell types.


Sujet(s)
Techniques de knock-in de gènes , Histone , Protéines luminescentes , Animaux , Souris , Histone/métabolisme , Protéines luminescentes/métabolisme , Protéines luminescentes/génétique , Anticorps/métabolisme , , Mâle , Souris de lignée C57BL , Souris transgéniques
15.
JCI Insight ; 9(13)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38771644

RÉSUMÉ

Hypotrichosis is a genetic disorder characterized by a diffuse and progressive loss of scalp and/or body hair. Nonetheless, the causative genes for several affected individuals remain elusive, and the underlying mechanisms have yet to be fully elucidated. Here, we discovered a dominant variant in a disintegrin and a metalloproteinase domain 17 (ADAM17) gene caused hypotrichosis with woolly hair. Adam17 (p.D647N) knockin mice mimicked the hair abnormality in patients. ADAM17 (p.D647N) mutation led to hair follicle stem cell (HFSC) exhaustion and caused abnormal hair follicles, ultimately resulting in alopecia. Mechanistic studies revealed that ADAM17 binds directly to E3 ubiquitin ligase tripartite motif-containing protein 47 (TRIM47). ADAM17 variant enhanced the association between ADAM17 and TRIM47, leading to an increase in ubiquitination and subsequent degradation of ADAM17 protein. Furthermore, reduced ADAM17 protein expression affected the Notch signaling pathway, impairing the activation, proliferation, and differentiation of HFSCs during hair follicle regeneration. Overexpression of Notch intracellular domain rescued the reduced proliferation ability caused by Adam17 variant in primary fibroblast cells.


Sujet(s)
Protéine ADAM17 , Alopécie , Follicule pileux , Ubiquitin-protein ligases , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Animaux , Alopécie/génétique , Alopécie/métabolisme , Alopécie/anatomopathologie , Souris , Follicule pileux/métabolisme , Follicule pileux/anatomopathologie , Humains , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitination , Mâle , Transduction du signal/génétique , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Femelle , Mutation , Techniques de knock-in de gènes , Prolifération cellulaire/génétique , Différenciation cellulaire/génétique , Protéolyse , Modèles animaux de maladie humaine , Fibroblastes/métabolisme , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique
16.
Nat Immunol ; 25(6): 1083-1096, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38816616

RÉSUMÉ

Current prophylactic human immunodeficiency virus 1 (HIV-1) vaccine research aims to elicit broadly neutralizing antibodies (bnAbs). Membrane-proximal external region (MPER)-targeting bnAbs, such as 10E8, provide exceptionally broad neutralization, but some are autoreactive. Here, we generated humanized B cell antigen receptor knock-in mouse models to test whether a series of germline-targeting immunogens could drive MPER-specific precursors toward bnAbs. We found that recruitment of 10E8 precursors to germinal centers (GCs) required a minimum affinity for germline-targeting immunogens, but the GC residency of MPER precursors was brief due to displacement by higher-affinity endogenous B cell competitors. Higher-affinity germline-targeting immunogens extended the GC residency of MPER precursors, but robust long-term GC residency and maturation were only observed for MPER-HuGL18, an MPER precursor clonotype able to close the affinity gap with endogenous B cell competitors in the GC. Thus, germline-targeting immunogens could induce MPER-targeting antibodies, and B cell residency in the GC may be regulated by a precursor-competitor affinity gap.


Sujet(s)
Affinité des anticorps , Lymphocytes B , Centre germinatif , Anticorps anti-VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Centre germinatif/immunologie , Animaux , Souris , Humains , Lymphocytes B/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Anticorps anti-VIH/immunologie , Affinité des anticorps/immunologie , Anticorps neutralisants/immunologie , Infections à VIH/immunologie , Vaccins contre le SIDA/immunologie , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Récepteurs pour l'antigène des lymphocytes B/immunologie , Techniques de knock-in de gènes , Souris transgéniques , Anticorps neutralisants à large spectre/immunologie , Souris de lignée C57BL
17.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758779

RÉSUMÉ

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Sujet(s)
Vieillissement de la cellule , Mitochondries , Stéatose hépatique non alcoolique , Acyl-(acyl-carrier-protein)desaturase , Animaux , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/anatomopathologie , Souris , Vieillissement de la cellule/génétique , Acétylation , Mitochondries/métabolisme , Acyl-(acyl-carrier-protein)desaturase/métabolisme , Acyl-(acyl-carrier-protein)desaturase/génétique , Mâle , Acetate coA-ligase/métabolisme , Acetate coA-ligase/génétique , Techniques de knock-in de gènes , Foie/métabolisme , Foie/anatomopathologie , Métabolisme lipidique , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Modèles animaux de maladie humaine , Coenzyme A ligases , Fatty acid synthase type I
18.
Genesis ; 62(3): e23601, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38703044

RÉSUMÉ

HAND2 is a basic helix-loop-helix transcription factor with diverse functions during development. To facilitate the investigation of genetic and functional diversity among Hand2-expressing cells in the mouse, we have generated Hand2Dre, a knock-in allele expressing Dre recombinase. To avoid disrupting Hand2 function, the Dre cDNA is inserted at the 3' end of the Hand2 coding sequence following a viral 2A peptide. Hand2Dre homozygotes can therefore be used in complex crosses to increase the proportion of useful genotypes among offspring. Dre expression in mid-gestation Hand2Dre embryos is indistinguishable from wild-type Hand2 expression, and HandDre efficiently recombines rox target sites in vivo. In combination with existing Cre and Flp mouse lines, Hand2Dre will therefore extend the ability to perform genetic intersectional labeling, fate mapping, and functional manipulation of subpopulations of cells characterized by developmental expression of Hand2.


Sujet(s)
Allèles , Facteurs de transcription à motif basique hélice-boucle-hélice , Techniques de knock-in de gènes , Animaux , Femelle , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Techniques de knock-in de gènes/méthodes , Integrases/génétique , Integrases/métabolisme , Mâle
19.
Nat Commun ; 15(1): 4561, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811575

RÉSUMÉ

The mammalian SWI/SNF-like BAF complexes play critical roles during animal development and pathological conditions. Previous gene deletion studies and characterization of human gene mutations implicate that the complexes both repress and activate a large number of genes. However, the direct function of the complexes in cells remains largely unclear due to the relatively long-term nature of gene deletion or natural mutation. Here we generate a mouse line by knocking in the auxin-inducible degron tag (AID) to the Smarca4 gene, which encodes BRG1, the essential ATPase subunit of the BAF complexes. We show that the tagged BRG1 can be efficiently depleted by osTIR1 expression and auxin treatment for 6 to 10 h in CD4 + T cells, hepatocytes, and fibroblasts isolated from the knock-in mice. The acute depletion of BRG1 leads to decreases in nascent RNAs and RNA polymerase II binding at a large number of genes, which are positively correlated with the loss of BRG1. Further, these changes are correlated with diminished accessibility at DNase I Hypersensitive Sites (DHSs) and p300 binding. The acute BRG1 depletion results in three major patterns of nucleosome shifts leading to narrower nucleosome spacing surrounding transcription factor motifs and at enhancers and transcription start sites (TSSs), which are correlated with loss of BRG1, decreased chromatin accessibility and decreased nascent RNAs. Acute depletion of BRG1 severely compromises the Trichostatin A (TSA) -induced histone acetylation, suggesting a substantial interplay between the chromatin remodeling activity of BRG1 and histone acetylation. Our data suggest BRG1 mainly plays a direct positive role in chromatin accessibility, RNAPII binding, and nascent RNA production by regulating nucleosome positioning and facilitating transcription factor binding to their target sites.


Sujet(s)
Helicase , Protéines nucléaires , Facteurs de transcription , Animaux , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Helicase/métabolisme , Helicase/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Souris , Nucléosomes/métabolisme , Nucléosomes/génétique , Acides indolacétiques/métabolisme , RNA polymerase II/métabolisme , Fibroblastes/métabolisme , Techniques de knock-in de gènes , Hépatocytes/métabolisme , Protéine p300-E1A/métabolisme , Protéine p300-E1A/génétique , Activation de la transcription , Transcription génétique , Histone/métabolisme , Deoxyribonuclease I/métabolisme , Chromatine/métabolisme , Humains
20.
Sci Rep ; 14(1): 12393, 2024 05 29.
Article de Anglais | MEDLINE | ID: mdl-38811759

RÉSUMÉ

Parkinson's disease (PD) is a progressive late-onset neurodegenerative disease leading to physical and cognitive decline. Mutations of leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of PD. LRRK2 is a complex scaffolding protein with known regulatory roles in multiple molecular pathways. Two prominent examples of LRRK2-modulated pathways are Wingless/Int (Wnt) and nuclear factor of activated T-cells (NFAT) signaling. Both are well described key regulators of immune and nervous system development as well as maturation. The aim of this study was to establish the physiological and pathogenic role of LRRK2 in Wnt and NFAT signaling in the brain, as well as the potential contribution of the non-canonical Wnt/Calcium pathway. In vivo cerebral Wnt and NFATc1 signaling activity was quantified in LRRK2 G2019S mutant knock-in (KI) and LRRK2 knockout (KO) male and female mice with repeated measures over 28 weeks, employing lentiviral luciferase biosensors, and analyzed using a mixed-effect model. To establish spatial resolution, we investigated tissues, and primary neuronal cell cultures from different brain regions combining luciferase signaling activity, immunohistochemistry, qPCR and western blot assays. Results were analyzed by unpaired t-test with Welch's correction or 2-way ANOVA with post hoc corrections. In vivo Wnt signaling activity in LRRK2 KO and LRRK2 G2019S KI mice was increased significantly ~ threefold, with a more pronounced effect in males (~ fourfold) than females (~ twofold). NFATc1 signaling was reduced ~ 0.5-fold in LRRK2 G2019S KI mice. Brain tissue analysis showed region-specific expression changes in Wnt and NFAT signaling components. These effects were predominantly observed at the protein level in the striatum and cerebral cortex of LRRK2 KI mice. Primary neuronal cell culture analysis showed significant genotype-dependent alterations in Wnt and NFATc1 signaling under basal and stimulated conditions. Wnt and NFATc1 signaling was primarily dysregulated in cortical and hippocampal neurons respectively. Our study further built on knowledge of LRRK2 as a Wnt and NFAT signaling protein. We identified complex changes in neuronal models of LRRK2 PD, suggesting a role for mutant LRRK2 in the dysregulation of NFAT, and canonical and non-canonical Wnt signaling.


Sujet(s)
Modèles animaux de maladie humaine , Leucine-rich repeat serine-threonine protein kinase-2 , Facteurs de transcription NFATC , Maladie de Parkinson , Voie de signalisation Wnt , Animaux , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Leucine-rich repeat serine-threonine protein kinase-2/métabolisme , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Mâle , Souris , Femelle , Techniques de knock-in de gènes , Souris knockout , Neurones/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Mutation , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...