Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Lab Anim (NY) ; 52(12): 324-331, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38017180

ABSTRACT

Adoptive cell transfer between genetically identical hosts relies on the use of a congenic marker to distinguish the donor cells from the host cells. CD45, a glycoprotein expressed by all hematopoietic cells, is one of the main congenic markers used because its two isoforms, CD45.1 and CD45.2, can be discriminated by flow cytometry. As a consequence, C57BL/6J (B6; CD45.2) and B6.SJL-Ptprca Pepcb/BoyJ (B6.SJL; CD45.1) mice are widely used in adoptive cell transfer experiments, under the presumption that they differ only at the CD45 (Ptprc) locus. However, recent studies have identified genetic variations between these congenic strains and have notably highlighted a differential expression of cathepsin E (CTSE). The B6.SJL mouse presents a number of functional differences in hematopoietic stem cell engraftment potential and immune cell numbers compared with the B6 mouse. In this study, we showed that B6 and B6.SJL mice also differ in their CD8+ T cell compartment and CD8+ T cell responses to viral infection. We identified Ctse as the most differentially expressed gene between CD8+ T cells of B6 and B6.SJL and demonstrated that the differences reported between these two mouse strains are not due to CTSE. Finally, using CRISPR-Cas9 genome editing, we generated a CD45.1-expressing B6 mouse by inserting one nucleotide mutation (A904G) leading to an amino acid change (K302E) in the Ptprc gene of the B6 mouse. We showed that this new B6-Ptprcem(K302E)Jmar/J mouse resolves the experimental biases reported between the B6 and B6.SJL mouse lines and should thus represent the new gold standard for adoptive cell transfer experiments in B6.


Subject(s)
CD8-Positive T-Lymphocytes , Hematopoietic Stem Cells , Mice , Animals , Mice, Inbred C57BL , Epitopes , Mice, Inbred Strains , Adoptive Transfer
2.
Nat Commun ; 10(1): 45, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604748

ABSTRACT

Programmable nucleases have enabled rapid and accessible genome engineering in eukaryotic cells and living organisms. However, their delivery into target cells can be technically challenging when working with primary cells or in vivo. Here, we use engineered murine leukemia virus-like particles loaded with Cas9-sgRNA ribonucleoproteins (Nanoblades) to induce efficient genome-editing in cell lines and primary cells including human induced pluripotent stem cells, human hematopoietic stem cells and mouse bone-marrow cells. Transgene-free Nanoblades are also capable of in vivo genome-editing in mouse embryos and in the liver of injected mice. Nanoblades can be complexed with donor DNA for "all-in-one" homology-directed repair or programmed with modified Cas9 variants to mediate transcriptional up-regulation of target genes. Nanoblades preparation process is simple, relatively inexpensive and can be easily implemented in any laboratory equipped for cellular biology.


Subject(s)
CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Genetic Vectors/genetics , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/genetics , Animals , Cell Line, Tumor , DNA Repair/genetics , Embryo, Mammalian , Fibroblasts , Gene Editing/economics , Genome/genetics , HEK293 Cells , Hematopoietic Stem Cells , Humans , Induced Pluripotent Stem Cells , Leukemia Virus, Murine/genetics , Macrophages , Mice , Mice, Inbred C57BL , Primary Cell Culture , Transcriptional Activation/genetics
3.
Sci Rep ; 8(1): 4679, 2018 Mar 13.
Article in English | MEDLINE | ID: mdl-29535400

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

4.
Sci Rep ; 8(1): 474, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323173

ABSTRACT

In this report, we present an improved protocol for CRISPR/Cas9 genome editing in mice. The procedure consists in the electroporation of intact mouse zygotes with ribonucleoprotein complexes prepared in vitro from recombinant Cas9 nuclease and synthetic dual guide RNA. This simple cloning-free method proves to be extremely efficient for the generation of indels and small deletions by non-homologous end joining, and for the generation of specific point mutations by homology-directed repair. The procedure, which avoids DNA construction, in vitro transcription and oocyte microinjection, greatly simplifies genome editing in mice.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , RNA, Guide, Kinetoplastida/genetics , Zygote/metabolism , Animals , DNA End-Joining Repair , Deubiquitinating Enzymes , Electroporation , Endopeptidases/chemistry , Endopeptidases/genetics , Female , Genetic Loci , Genotyping Techniques , INDEL Mutation , Male , Mice , Mice, Inbred C57BL , Mutation, Missense
5.
Thyroid ; 28(1): 139-150, 2018 01.
Article in English | MEDLINE | ID: mdl-29205102

ABSTRACT

BACKGROUND: Resistance to thyroid hormone due to THRA mutations (RTHα) is a recently discovered genetic disease, displaying important variability in its clinical presentation. The mutations alter the function of TRα1, one of the two nuclear receptors for thyroid hormone. METHODS: The aim of this study was to understand the relationship between specific THRA mutations and phenotype. CRISPR/Cas9 genome editing was used to generate five new mouse models of RTHα, with frameshift or missense mutations. RESULTS: Like human patients, mutant mice displayed a hypothyroid-like phenotype, with altered development. Phenotype severity varied between the different mouse models, mainly depending on the ability of the mutant receptor to interact with transcription corepressor in the presence of thyroid hormone. CONCLUSION: The present mutant mice represent highly relevant models for the human genetic disease which will be useful for future investigations.


Subject(s)
Genes, erbA/genetics , Thyroid Hormone Resistance Syndrome/genetics , Animals , CRISPR-Cas Systems , Mice , Mutation , Phenotype
6.
Genesis ; 51(3): 193-200, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281269

ABSTRACT

Immune tolerance to self-antigens is a complex process that utilizes multiple mechanisms working in concert to maintain homeostasis and prevent autoimmunity. Considerable progress in deciphering the mechanisms controlling the activation or deletion of T cells has been made by using T cell receptor (TCR) transgenic mice. One such model is the F5 model in which CD8 T cells express a TCR specific for an epitope derived from the influenza NP68 protein. Our aim was to create transgenic mouse models expressing constitutively the NP68 epitope fused to enhanced green fluorescent protein (EGFP) in order to assess unambiguously the relative levels of NP68 epitope expressed by single cells. We used a lentiviral-based approach to generate two independent transgenic mouse strains expressing the fusion protein EGFP-NP68 under the control of CAG (CMV immediate early enhancer and the chicken ß-actin promoter) or spleen focus-forming virus (SFFV) promoters. Analysis of the pattern of EGFP expression in the hematopoietic compartment showed that CAG and SFFV promoters are differentially regulated during T cell development. However, both promoters drove high EGFP-NP68 expression in dendritic cells (pDCs, CD8α(+) cDCs, and CD8α(-) cDCs) from spleen or generated in vitro following differentiation from bone-marrow progenitors. NP68 epitope was properly processed and successfully presented by dendritic cells (DCs) by direct presentation and cross-presentation to F5 CD8 T cells. The models presented here are valuable tools to investigate the priming of F5 CD8 T cells by different subsets of DCs.


Subject(s)
Epitopes, T-Lymphocyte/genetics , Green Fluorescent Proteins/genetics , Transgenes , Viral Proteins/genetics , Animals , Dendritic Cells/metabolism , Genetic Engineering/methods , Genetic Vectors , Histocompatibility Antigens Class I/metabolism , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , T-Lymphocytes/metabolism
7.
Immunol Res ; 52(3): 284-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22528127

ABSTRACT

Besides the classically described subsets of memory CD8 T cells generated under infectious conditions, are T inflammatory memory cells generated under sterile priming conditions, such as sensitization to allergens. Although not fully differentiated as pathogen-induced memory cells, they display memory properties that distinguish them from naive CD8 T cells. Given these memory cells are generated in an antigen-specific context that is devoid of pathogen-derived danger signals and CD4 T cell help, we herein questioned whether they maintained their activation and differentiation potential, could be recruited in an immune response directed against a pathogen expressing their cognate antigen and further differentiate in fully competent secondary memory cells. We show that T inflammatory memory cells can indeed take part to the immune response triggered by a viral infection, differentiate into secondary effectors and further generate typical central memory CD8 T cells and effector memory CD8 T cells. Furthermore, the secondary memory cells they generate display a functional advantage over primary memory cells in their capacity to produce TNF-α and the XCL1 chemokine. These results suggest that cross-reactive stimulations and differentiation of cells directed against allergens or self into fully competent pathogen-induced memory cells might have incidences in inflammatory immuno-pathologies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, C/immunology , Immunologic Memory , Lymphocyte Activation , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Chemokines, C/biosynthesis , Cross Reactions/genetics , Cross Reactions/immunology , Mice , Mice, Transgenic , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
8.
FEBS Lett ; 585(17): 2703-8, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21820436

ABSTRACT

A strict control of endolymph composition (high potassium, low sodium fluid) and volume is instrumental for a proper functioning of the inner ear. Alteration of endolymph homeostasis is proposed in the pathogenesis of Menière's disease. However, the mechanisms controlling endolymph secretion remain elusive. By using the vestibular EC5v cells, we provide evidence for the presence of vasopressin, catecholamine and purinergic signaling pathways, coupled to adenylate cyclase, phosphoinositidase C and Ca(2+) activation. We demonstrate that vasopressin and catecholamines stimulate while ATP inhibits apical potassium secretion by EC5v cells. These results open new interesting perspectives for the management of inner ear diseases.


Subject(s)
Adenosine Triphosphate/pharmacology , Catecholamines/pharmacology , Ear, Inner/drug effects , Ear, Inner/metabolism , Potassium/metabolism , Vasopressins/pharmacology , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Calcium Signaling/drug effects , Cell Line , Cyclic AMP/metabolism , Ear, Inner/cytology , Mice , Phosphoric Diester Hydrolases/metabolism , Receptors, Purinergic P2Y/metabolism , Receptors, Vasopressin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Endocrinol ; 23(12): 1948-62, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19846540

ABSTRACT

Aldosterone effects are mediated by the mineralocorticoid receptor (MR), a transcription factor highly expressed in the distal nephron. Given that MR expression level constitutes a key element controlling hormone responsiveness, there is much interest in elucidating the molecular mechanisms governing MR expression. To investigate whether hyper- or hypotonicity could affect MR abundance, we established by targeted oncogenesis a novel immortalized cortical collecting duct (CCD) cell line and examined the impact of osmotic stress on MR expression. KC3AC1 cells form domes, exhibit a high transepithelial resistance, express 11beta-hydroxysteroid dehydrogenase 2 and functional endogenous MR, which mediates aldosterone-stimulated Na(+) reabsorption through the epithelial sodium channel activation. MR expression is tightly regulated by osmotic stress. Hypertonic conditions induce expression of tonicity-responsive enhancer binding protein, an osmoregulatory transcription factor capable of binding tonicity-responsive enhancer response elements located in MR regulatory sequences. Surprisingly, hypertonicity leads to a severe reduction in MR transcript and protein levels. This is accompanied by a concomitant tonicity-induced expression of Tis11b, a mRNA-destabilizing protein that, by binding to the AU-rich sequences of the 3'-untranslated region of MR mRNA, may favor hypertonicity-dependent degradation of labile MR transcripts. In sharp contrast, hypotonicity causes a strong increase in MR transcript and protein levels. Collectively, we demonstrate for the first time that optimal adaptation of CCD cells to changes in extracellular fluid composition is accompanied by drastic modification in MR abundance via transcriptional and posttranscriptional mechanisms. Osmotic stress-regulated MR expression may represent an important molecular determinant for cell-specific MR action, most notably in renal failure, hypertension, or mineralocorticoid resistance.


Subject(s)
Aldosterone/pharmacology , Osmosis/drug effects , Osmosis/physiology , Receptors, Mineralocorticoid/metabolism , Animals , Blotting, Western , Cell Line , Electrophoretic Mobility Shift Assay , Gene Expression Regulation/drug effects , Hypertonic Solutions/pharmacology , Hypotonic Solutions/pharmacology , Immunohistochemistry , Ion Transport/drug effects , Kidney Tubules, Collecting/cytology , Male , Mice , Mice, Transgenic , Protein Stability , Receptors, Mineralocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , Transfection
10.
Acta Otolaryngol ; 128(6): 608-12, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18568492

ABSTRACT

CONCLUSION: The permeability of the blood-labyrinth barrier for radioactive mannitol was unchanged after impulse noise trauma. The present findings are contradictory to the theory of an increased permeability in the blood-labyrinth barrier as a result of extensive noise exposure. OBJECTIVE: Noise trauma is reported to cause multiple effects on the cochlea including mechanical and metabolic damage. The aim of the study was to observe the effects of impulse noise on cochlear homeostasis. MATERIALS AND METHODS: A well-established rat model was used for evaluation of the early effects of impulse noise trauma on the integrity of the blood-perilymph barrier. To evaluate whether a blood-perilymph barrier disruption contributes to cochlear injury after impulse noise, the paracellular transport of radioactive mannitol into scala vestibuli perilymph (PLV) and electrolyte concentration in perilymph were estimated. Thirteen animals exposed to synthesized impulses of 160 dB SPL peak value, at a rate of 100 pulses, were designed as the study group and 15 rats not exposed to noise were designed as the control group. After mannitol infusion each ear of the animal in the study group was separately exposed to impulse noise and PLV samples were taken during 2 h post-infusion. In the control group, corresponding PLV samples were taken after mannitol injection. RESULTS: At 2 h after mannitol infusion there was no difference in PLV mannitol concentration in the study group and control group (21.5%+/-2.2 and 20.5%+/-2.1, respectively). Impulse noise had no effect on the electrochemical composition of PLV.


Subject(s)
Blood , Capillary Permeability , Hearing Loss, Noise-Induced/physiopathology , Perilymph/physiology , Scala Vestibuli/blood supply , Animals , Mannitol/pharmacokinetics , Noise , Perilymph/chemistry , Potassium/analysis , Rats , Rats, Long-Evans , Sodium/analysis , Tritium
11.
J Biol Chem ; 281(15): 10496-507, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16478723

ABSTRACT

Endolymph, a high K(+)/low Na(+) fluid, participates in mechanoelectrical transduction in inner ear. Molecular mechanisms controlling endolymph ion homeostasis remain elusive, hampered by the lack of appropriate cellular models. We established an inner ear cell line by targeted oncogenesis. The expression of SV40 T antigen was driven by the proximal promoter of the human mineralocorticoid receptor (MR) gene, a receptor expressed in the inner ear. The EC5v cell line, microdissected from the semicircular canal, grew as a monolayer of immortalized epithelial cells forming domes. EC5v cells exhibited on filters of high transepithelial resistance and promoted K(+) secretion and Na(+) absorption. Functional MR and the 11beta-hydroxysteroid dehydrogenase type 2, a key enzyme responsible for MR selectivity were identified. Expression of the epithelial sodium channel and serum glucocorticoid-regulated kinase 1 was shown to be up-regulated by aldosterone, indicating that EC5v represents a novel corticosteroid-sensitive cell line. Ionic measurements and (86)Rb transport assays revealed an apical secretion of K(+) at least in part through the I(sK)/KvLQT1 potassium channel under standard culture conditions. However, when cells were exposed to high apically K(+)/low Na(+) fluid, mimicking endolymph exposure, I(sK)/KvLQT1 actually functioned as a strict apical to basolateral K(+) channel inhibited by clofilium. Quantitative reverse transcriptase-PCR further demonstrated that expression of KvLQT1 but not of I(sK) was down-regulated by high K(+) concentration. This first vestibular cellular model thus constitutes a valuable system to further investigate the molecular mechanisms controlling ionic transports in the inner ear and the pathophysiological consequences of their dysfunctions in vertigo and hearing loss.


Subject(s)
Adrenal Cortex Hormones/metabolism , Cell Culture Techniques/methods , Cell Line , Ear, Inner/cytology , KCNQ1 Potassium Channel/physiology , Potassium Channels, Voltage-Gated/physiology , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Animals , Antigens, Polyomavirus Transforming/metabolism , Biological Transport , Blotting, Western , Catalysis , Cells, Cultured , DNA Primers/chemistry , Ear, Inner/metabolism , Endolymph/metabolism , Immunohistochemistry , Ions , KCNQ1 Potassium Channel/metabolism , Kinetics , Mice , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Potassium/chemistry , Potassium/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Binding , Quaternary Ammonium Compounds/chemistry , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium/chemistry , Time Factors , Transgenes
14.
Acta Otolaryngol ; 124(4): 449-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15224873

ABSTRACT

OBJECTIVE: To detect and localize aquaporin-2 (AQP-2), a water channel regulated by the antidiuretic hormone, in human endolymphatic sac. MATERIAL AND METHODS: Human endolymphatic sacs were sampled during removal of vestibular schwannomas via a translabyrinthine approach. Samples were immediately fixed in 10% formalin (24 h) and embedded in paraffin; in situ hybridization and immunohistochemistry were performed with an AQP-2-specific probe and a polyclonal antibody. RESULTS: Both AQP-2 mRNA and protein were detected in the epithelium of the endolymphatic sac. AQP-2 immunostaining was mainly cytoplasmic, suggesting that most AQP-2 was located in intracellular pools. CONCLUSIONS: In the endolymphatic sac, AQP-2 probably participates in the homeostasis of endolymph; the possibility of reducing the volume of endolymph by inhibiting its expression and membranous insertion using an antidiuretic hormone inhibitor represents a new therapeutic approach for the treatment of Ménière's disease.


Subject(s)
Aquaporins/analysis , Endolymphatic Sac/chemistry , Aquaporin 2 , Cytoplasm/chemistry , Epithelium/chemistry , Humans , Immunohistochemistry , In Situ Hybridization , Vestibule, Labyrinth/chemistry
15.
Med Sci (Paris) ; 20(3): 304-10, 2004 Mar.
Article in French | MEDLINE | ID: mdl-15067575

ABSTRACT

The endolymphatic sac is a non-sensory organ of the inner ear. It is connected to the endolymphatic compartment that is filled with endolymph, a potassium-rich fluid that bathes the apical side of inner ear sensory cells. The main functions ascribed to the endolymphatic sac are the regulation of the volume and pressure of endolymph, the immune response of the inner ear, and the elimination of endolymphatic waste products by phagocytosis. Functional alteration of these functions, leading to deficient endolymph homeostasis and/or altered inner ear immune response, may participate to the pathophysiology of Ménière's disease, an inner ear pathology that causes episodes of vertigo, sensorineural hearing loss and tinnitus, and is characterized by an increase in volume of the cochleo-vestibular endolymph (endolymphatic hydrops).


Subject(s)
Endolymphatic Sac/physiology , Endolymph/chemistry , Endolymphatic Sac/immunology , Hearing Loss, Sensorineural/physiopathology , Homeostasis , Humans , Meniere Disease/physiopathology , Pressure , Tinnitus/physiopathology
16.
Acta Otolaryngol ; 123(2): 199-202, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12701740

ABSTRACT

OBJECTIVE: To improve the efficacy of intratympanic therapy using perilymphatic entry through the round window membrane. MATERIAL AND METHODS: The perilymphatic entry characteristics of 2 neutral molecules, mannitol (182.2 Da) and inulin (7,000 Da), were studied. A polyethylene catheter was placed in contact with the guinea pig round window membrane and sealed with cyanoacrylate glue. This catheter was linked to an osmotic minipump that delivered 100 microl portions of 3H-mannitol or 3H-inulin solutions over a 7-day period at a constant rate (0.5 microl/h). Perilymph in the scala vestibuli and scala tympani, cerebrospinal fluid (CSF) and plasma were sampled after 4-7 days of delivery. RESULTS: Despite the constant rate of infusion, perilymphatic radioactivity varied widely from one animal to the other, probably as a function of the position of the microcatheter within the round window niche and/or the permeability of the round window. Even a large molecule such as inulin entered the perilymphatic space. Seven days after the beginning of 3H-mannitol administration, the radioactivity was higher in the perilymph of the scala tympani than in that of the scala vestibuli. The perilymphatic radioactivity on Day 7 was approximately 50% lower than that measured on Day 4 (p = NS). Finally, round window membrane delivery did not preclude distant spread of the molecules into the blood and CSF. CONCLUSIONS: Using round window membrane delivery, the perilymphatic entry of mannitol and inulin depended on their molecular weight. Intratympanic delivery induced a high inter-individual heterogeneity of the drug concentration within the inner ear, with subsequent variability of the therapeutic effects.


Subject(s)
Inulin/pharmacology , Mannitol/pharmacology , Perilymph/drug effects , Round Window, Ear/drug effects , Administration, Topical , Animals , Biological Availability , Disease Models, Animal , Drug Administration Schedule , Guinea Pigs , Inulin/pharmacokinetics , Male , Mannitol/pharmacokinetics , Perilymph/metabolism , Probability , Radioisotopes , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...