Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 120.743
Filtrer
1.
J Pineal Res ; 76(5): e12986, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38965880

RÉSUMÉ

This contribution reviews the role of inbred and transgenic mouse strains for deciphering the mammalian melatoninergic and circadian system. It focusses on the pineal organ as melatonin factory and two major targets of the melatoninergic system, the suprachiasmatic nuclei (SCN) and the hypophysial pars tuberalis (PT). Mammalian pinealocytes sharing molecular characteristics with true pineal and retinal photoreceptors synthesize and secrete melatonin into the blood and cerebrospinal fluid night by night. Notably, neuron-like connections exist between the deep pinealocytes and the habenular/pretectal region suggesting direct pineal-brain communication. Control of melatonin biosynthesis in rodents involves transcriptional regulation including phosphorylation of CREB and upregulation of mPer1. In the SCN, melatonin acts upon MT1 and MT2 receptors. Melatonin is not necessary to maintain the rhythm of the SCN molecular clockwork, but it has distinct effects on the synchronization of the circadian rhythm by light, facilitates re-entrainment of the circadian system to phase advances in the level of the SCN molecular clockwork by acting upon MT2 receptors and plays a stabilizing role in the circadian system as evidenced from locomotor activity recordings. While the effects in the SCN are subtle, melatonin is essential for PT functions. Via the MT1 receptor it drives the PT-intrinsic molecular clockwork and the retrograde and anterograde output pathways controlling seasonal rhythmicity. Although inbred and transgenic mice do not show seasonal reproduction, the pathways from the PT are fully intact if the animals are melatonin proficient. Thus, only melatonin-proficient strains are suited to investigate the circadian and melatoninergic systems.


Sujet(s)
Rythme circadien , Mélatonine , Animaux , Mélatonine/métabolisme , Rythme circadien/physiologie , Souris , Modèles animaux , Noyau suprachiasmatique/métabolisme , Souris transgéniques , Glande pinéale/métabolisme
2.
FASEB J ; 38(13): e23796, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967302

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is an orphan neurodegenerative disease. Immune system dysregulation plays an essential role in ALS onset and progression. Our preclinical studies have shown that the administration of exogenous allogeneic B cells improves outcomes in murine models of skin and brain injury through a process termed pligodraxis, in which B cells adopt an immunoregulatory and neuroprotective phenotype in an injured environment. Here, we investigated the effects of B-cell therapy in the SOD1G93A mouse preclinical model of ALS and in a person living with ALS. Purified splenic mature naïve B cells from haploidentical donor mice were administered intravenously in SOD1G93A mice for a total of 10 weekly doses. For the clinical study in a person with advanced ALS, IgA gammopathy of unclear significance, and B lymphopenia, CD19+ B cells were positively selected from a healthy haploidentical donor and infused intravenously twice, at a 60-day interval. Repeated intravenous B-cell administration was safe and significantly delayed disease onset, extended survival, reduced cellular apoptosis, and decreased astrogliosis in SOD1G93A mice. Repeated B-cell infusion in a person with ALS was safe and did not appear to generate a clinically evident inflammatory response. An improvement of 5 points on the ALSFRS-R scale was observed after the first infusion. Levels of inflammatory markers showed persistent reduction post-infusion. This represents a first demonstration of the efficacy of haploidentical B-cell infusion in the SOD1G93A mouse and the safety and feasibility of using purified haploidentical B lymphocytes as a cell-based therapeutic strategy for a person with ALS.


Sujet(s)
Sclérose latérale amyotrophique , Lymphocytes B , Sclérose latérale amyotrophique/thérapie , Sclérose latérale amyotrophique/immunologie , Animaux , Souris , Humains , Lymphocytes B/immunologie , Modèles animaux de maladie humaine , Souris transgéniques , Mâle , Femelle , Souris de lignée C57BL , Immunomodulation , Adulte d'âge moyen
3.
J Neuroinflammation ; 21(1): 168, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961498

RÉSUMÉ

BACKGROUND: The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive. RESULTS: In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes. CONCLUSION: Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.


Sujet(s)
Macrophages , Souris transgéniques , Corps vitré , Vésicule vitelline , Animaux , Souris , Corps vitré/cytologie , Vésicule vitelline/cytologie , Macrophages/métabolisme , Souris de lignée C57BL , Récepteur de facteur de croissance granulocyte-macrophage/métabolisme , Récepteur de facteur de croissance granulocyte-macrophage/génétique , Animaux nouveau-nés
4.
Front Immunol ; 15: 1389551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966642

RÉSUMÉ

Introduction: Pathogenesis of cutaneous leishmaniases involves parasite growth, persistent inflammation, and likely participation of lipoproteins (LP). The cholesteryl ester transfer protein (CETP), involved in LP remodeling, has been shown to participate in the inflammatory response and the evolution of infectious conditions. Methods: We evaluated the impact of the presence of CETP on infection by Leishmania (L.) amazonensis in an experimental model of cutaneous leishmaniasis using C57BL6/J mice transgenic for human CETP (CETP), having as control their littermates that do not express the protein, wild-type (WT) mice. The progression of the lesion after infection in the footpad was monitored for 12 weeks. Two groups of animals were formed to collect the plantar pad in the 4th and 12th week post-infection. Results: The lesion increased from the 3rd week onwards, in both groups, with a gradual decrease from the 10th week onwards in the CETP group compared to the WT group, showing a reduction in parasitism and an improvement in the healing process, a reduction in CD68+ cells, and an increase in CD163+ and CD206, characterizing a population of M2 macrophages. A reduction in ARG1+ cells and an increase in INOS+ cells were observed. During infection, the LP profile showed an increase in triglycerides in the VLDL fraction in the CETP group at 12 weeks. Gene expression revealed a decrease in the CD36 receptor in the CETP group at 12 weeks, correlating with healing and parasite reduction. In vitro, macrophages derived from bone marrow cells from CETP mice showed lower parasite load at 48 h and, a reduction in arginase activity at 4 h accompanied by increased NO production at 4 and 24 h compared to WT macrophages, corroborating the in vivo findings. Discussion: The data indicate that the presence of CETP plays an important role in resolving Leishmania (L.) amazonensis infection, reducing parasitism, and modulating the inflammatory response in controlling infection and tissue repair.


Sujet(s)
Protéines de transfert des esters de cholestérol , Leishmaniose cutanée , Macrophages , Souris de lignée C57BL , Souris transgéniques , Animaux , Protéines de transfert des esters de cholestérol/génétique , Protéines de transfert des esters de cholestérol/métabolisme , Leishmaniose cutanée/immunologie , Leishmaniose cutanée/parasitologie , Leishmaniose cutanée/métabolisme , Souris , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/parasitologie , Humains , Évolution de la maladie , Modèles animaux de maladie humaine
5.
Front Immunol ; 15: 1425938, 2024.
Article de Anglais | MEDLINE | ID: mdl-38953020

RÉSUMÉ

Introduction: P2X receptors are a family of homo- and heterotrimeric cation channels gated by extracellular ATP. The P2X4 and P2X7 subunits show overlapping expression patterns and have been involved in similar physiological processes, such as pain and inflammation as well as various immune cell functions. While formation of P2X2/P2X3 heterotrimers produces a distinct pharmacological phenotype and has been well established, functional identification of a P2X4/P2X7 heteromer has been difficult and evidence for and against a physical association has been found. Most of this evidence stems, however, from in vitro model systems. Methods: Here, we used a P2X7-EGFP BAC transgenic mouse model as well as P2X4 and P2X7 knock-out mice to re-investigate a P2X4-P2X7 interaction in mouse lung by biochemical and immunohistochemical experiments as well as quantitative expression analysis. Results: No detectable amounts of P2X4 could be co-purified from mouse lung via P2X7-EGFP. In agreement with these findings, immuno-histochemical analysis using a P2X7-specific nanobody revealed only limited overlap in the cellular and subcellular localizations of P2X4 and P2X7 in both the native lung tissue and primary cells. Comparison of P2X4 and P2X7 transcript and protein levels in the respective gene-deficient and wild type mice showed no mutual interrelation between their expression levels in whole lungs. However, a significantly reduced P2rx7 expression was found in alveolar macrophages of P2rx4 -/- mice. Discussion: In summary, our detailed analysis of the cellular and subcellular P2X4 and P2X7 localization and expression does not support a physiologically relevant direct association of P2X4 and P2X7 subunits or receptors in vivo.


Sujet(s)
Poumon , Souris knockout , Souris transgéniques , Récepteurs purinergiques P2X4 , Récepteurs purinergiques P2X7 , Animaux , Récepteurs purinergiques P2X4/métabolisme , Récepteurs purinergiques P2X4/génétique , Récepteurs purinergiques P2X7/génétique , Récepteurs purinergiques P2X7/métabolisme , Souris , Poumon/métabolisme , Poumon/immunologie , Souris de lignée C57BL , Liaison aux protéines
6.
Sci Rep ; 14(1): 15019, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951573

RÉSUMÉ

Circulating tumor cells (CTCs) represent a rare and heterogeneous population of cancer cells that are detached from the tumor site and entered blood or lymphatic circulation. Once disseminated in distant tissues, CTCs could remain dormant or create a tumor mass causing serious danger for patients. Many technologies exist to isolate CTCs from patients' blood samples, mostly based on microfluidic systems or by sorting them according to their surface antigens, notably EpCAM, and/or cytokeratins for carcinoma. ScreenCell has developed an easy-to-use, antigen-independent, rapid, cost-effective, and efficient technology that isolates CTCs according to their bigger size compared to the blood cells. This study provides the technical information necessary to isolate and characterize CTCs from mouse blood. By using blood samples from transgenic mice with breast cancer or from WT mice in which we spiked cancer cells, we showed that ScreenCell technology is compatible with standard EDTA blood collection tubes. Furthermore, the ScreenCell Cyto kit could treat up to 500 µl and the ScreenCell MB kit up to 200 µl of mouse blood. As the ScreenCell MB kit captures unaltered live CTCs, we have shown that their DNA could be efficiently extracted, and the isolated cells could be grown in culture. In conclusion, ScreenCell provides a rapid, easy, antigen-independent, cost-effective, and efficient technology to isolate and characterize CTCs from the blood samples of cancer patients and murine models. Thanks to this technology CTCs could be captured fixed or alive. Murine cancer models are extensively used in pre-clinical studies. Therefore, this study demonstrates the crucial technical points necessary while manipulating mouse blood samples using ScreenCell technology.


Sujet(s)
Séparation cellulaire , Souris transgéniques , Cellules tumorales circulantes , Cellules tumorales circulantes/anatomopathologie , Cellules tumorales circulantes/métabolisme , Animaux , Souris , Séparation cellulaire/méthodes , Femelle , Humains , Lignée cellulaire tumorale , Tumeurs du sein/anatomopathologie , Tumeurs du sein/sang
7.
Sci Transl Med ; 16(754): eadj5958, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38959324

RÉSUMÉ

Pathological tau aggregates cause cognitive decline in neurodegenerative tauopathies, including Alzheimer's disease (AD). These aggregates are prevalent within intracellular compartments. Current tau immunotherapies have shown limited efficacy in clearing intracellular tau aggregates and improving cognition in clinical trials. In this study, we developed toxic tau conformation-specific monoclonal antibody-2 (TTCM2), which selectively recognized pathological tau aggregates in brain tissues from patients with AD, dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). TTCM2 potently inhibited tau-seeding activity, an essential mechanism underlying tauopathy progression. To effectively target intracellular tau aggregates and ensure rapid delivery to the brain, TTCM2 was loaded in micelles (TTCM2-ms) and administered through the intranasal route. We found that intranasally administered TTCM2-ms efficiently entered the brain in hTau-tauopathy mice, targeting pathological tau in intracellular compartments. Moreover, a single intranasal dose of TTCM2-ms effectively cleared pathological tau, elevated synaptic proteins, and improved cognitive functions in aged tauopathy mice. Mechanistic studies revealed that TTCM2-ms cleared intracellular, synaptic, and seed-competent tau aggregates through tripartite motif-containing 21 (TRIM21), an intracellular antibody receptor and E3 ubiquitin ligase known to facilitate proteasomal degradation of cytosolic antibody-bound proteins. TRIM21 was found to be essential for TTCM2-ms-mediated clearance of tau pathology. Our study collectively provides evidence of the effectiveness of nasal tau immunotherapy in targeting and clearing intracellular tau pathology through TRIM21 and enhancing cognition in aged tauopathy mice. This study could be valuable in designing effective tau immunotherapies for AD and other tauopathies.


Sujet(s)
Administration par voie nasale , Cognition , Immunothérapie , Souris transgéniques , Tauopathies , Protéines tau , Animaux , Protéines tau/métabolisme , Tauopathies/thérapie , Tauopathies/anatomopathologie , Tauopathies/métabolisme , Immunothérapie/méthodes , Humains , Souris , Vieillissement/anatomopathologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Anticorps monoclonaux/pharmacologie , Modèles animaux de maladie humaine , Agrégats de protéines/effets des médicaments et des substances chimiques
8.
Sci Rep ; 14(1): 15260, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38956136

RÉSUMÉ

KCNQ4 is a voltage-gated K+ channel was reported to distribute over the basolateral surface of type 1 vestibular hair cell and/or inner surface of calyx and heminode of the vestibular nerve connected to the type 1 vestibular hair cells of the inner ear. However, the precise localization of KCNQ4 is still controversial and little is known about the vestibular phenotypes caused by KCNQ4 dysfunction or the specific role of KCNQ4 in the vestibular organs. To investigate the role of KCNQ4 in the vestibular organ, 6-g hypergravity stimulation for 24 h, which represents excessive mechanical stimulation of the sensory epithelium, was applied to p.W277S Kcnq4 transgenic mice. KCNQ4 was detected on the inner surface of calyx of the vestibular afferent in transmission electron microscope images with immunogold labelling. Vestibular function decrease was more severe in the Kcnq4p.W277S/p.W277S mice than in the Kcnq4+/+ and Kcnq4+/p.W277S mice after the stimulation. The vestibular function loss was resulted from the loss of type 1 vestibular hair cells, which was possibly caused by increased depolarization duration. Retigabine, a KCNQ activator, prevented hypergravity-induced vestibular dysfunction and hair cell loss. Patients with KCNQ4 mutations also showed abnormal clinical vestibular function tests. These findings suggest that KCNQ4 plays an essential role in calyx and afferent of type 1 vestibular hair cell preserving vestibular function against excessive mechanical stimulation.


Sujet(s)
Cellules ciliées vestibulaires , Canaux potassiques KNCQ , Souris transgéniques , Animaux , Canaux potassiques KNCQ/métabolisme , Canaux potassiques KNCQ/génétique , Cellules ciliées vestibulaires/métabolisme , Cellules ciliées vestibulaires/anatomopathologie , Souris , Phénylènediamines/pharmacologie , Carbamates/pharmacologie , Labyrinthe vestibulaire/métabolisme , Labyrinthe vestibulaire/anatomopathologie , Labyrinthe vestibulaire/physiopathologie
9.
Sci Rep ; 14(1): 15053, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38956389

RÉSUMÉ

Targeting intracellular inhibiting proteins has been revealed to be a promising strategy to improve CD8+ T cell anti-tumor efficacy. Here, we are focusing on intracellular inhibiting proteins specific to TCR signaling: DOK1 and DOK2 expressed in T cells. We hypothesized that depletion of intracellular inhibition checkpoint DOK1 and DOK2 could improve CD8+ T-cell based cancer therapies. To evaluate the role of DOK1 and DOK2 depletion in physiology and effector function of CD8+ T lymphocytes and in cancer progression, we established a transgenic T cell receptor mouse model specific to melanoma antigen hgp100 (pmel-1 TCR Tg) in WT and Dok1/Dok2 DKO (double KO) mice. We showed that both DOK1 and DOK2 depletion in CD8+ T cells after an in vitro pre-stimulation induced a higher percentage of effector memory T cells as well as an up regulation of TCR signaling cascade- induced by CD3 mAbs, including the increased levels of pAKT and pERK, two major phosphoproteins involved in T cell functions. Interestingly, this improved TCR signaling was not observed in naïve CD8+ T cells. Despite this enhanced TCR signaling essentially shown upon stimulation via CD3 mAbs, pre-stimulated Dok1/Dok2 DKO CD8+ T cells did not show any increase in their activation or cytotoxic capacities against melanoma cell line expressing hgp100 in vitro. Altogether we demonstrate here a novel aspect of the negative regulation by DOK1 and DOK2 proteins in CD8+ T cells. Indeed, our results allow us to conclude that DOK1 and DOK2 have an inhibitory role following long term T cell stimulations.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Lymphocytes T CD8+ , Protéines de liaison à l'ADN , Mémoire immunologique , Souris knockout , Phosphoprotéines , Protéines de liaison à l'ARN , Récepteurs aux antigènes des cellules T , Transduction du signal , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Lignée cellulaire tumorale , Souris transgéniques
10.
J Neuroinflammation ; 21(1): 167, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956605

RÉSUMÉ

BACKGROUND: Deposition of amyloid ß, which is produced by amyloidogenic cleavage of APP by ß- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology. RESULTS: In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aß were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aß phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets. CONCLUSIONS: Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.


Sujet(s)
Maladie d'Alzheimer , Chimiotaxie , Souris de lignée C57BL , microARN , Microglie , microARN/métabolisme , microARN/génétique , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Souris , Humains , Microglie/métabolisme , Microglie/anatomopathologie , Mâle , Chimiotaxie/physiologie , Femelle , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Souris transgéniques , Sujet âgé , Régulation de l'expression des gènes
11.
Ren Fail ; 46(2): 2373276, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38967134

RÉSUMÉ

BACKGROUND: Podocytes, as intrinsic renal cells, can also express MHC-II and costimulatory molecules under inflammatory conditions, suggesting that they may act as antigen-presenting cells (APCs) to activate immune cell responses and then lead to immune-mediated renal injury. They are already recognized as main targets in the pathogenic mechanism of hepatitis B virus (HBV)-associated glomerulonephritis (HBV-GN). Previous studies also have indicated that inflammatory cells infiltration and immune-mediated tissue injury are evident in the kidney samples of patients with HBV-GN. However, the role of podocytes immune disorder in the pathogenic mechanism of HBV-GN remains unclear. METHODS: Renal function and inflammatory cells infiltration were measured in HBV transgenic (HBV-Tg) mice. In vitro, podocytes/CD4+ T cells or macrophages co-culture system was established. Then, the expression of HBx, CD4, and CD68 was determined by immunohistochemistry, while the expression of MHC-II, CD40, and CD40L was determined by immunofluorescence. Co-stimulatory molecules expression was examined by flow cytometry. The levels of inflammatory factors were detected by ELISA. RESULTS: In vivo, renal function was obviously impaired in HBV-Tg mice. HBx was significantly upregulated and immune cells infiltrated in the glomerulus of HBV-Tg mice. Expression of MHC-II and costimulatory molecule CD40 increased in the podocytes of HBV-Tg mice; CD4+ T cells exhibited increased CD40L expression in glomerulus. In vitro, CD40 expression was markedly elevated in HBx-podocytes. In co-culture systems, HBx-podocytes stimulated CD4+ T cells activation and caused the imbalance between IFN-γ and IL-4. HBx-podocytes also enhanced the adhesion ability of macrophages and induced the release of proinflammatory mediators. CONCLUSION: Taken together, these podocyte-related immune disorder may be involved in the pathogenic mechanism of HBV-GN.


Sujet(s)
Glomérulonéphrite , Virus de l'hépatite B , Souris transgéniques , Podocytes , Transactivateurs , Protéines virales régulatrices ou accessoires , Animaux , Podocytes/immunologie , Podocytes/anatomopathologie , Podocytes/métabolisme , Souris , Transactivateurs/métabolisme , Transactivateurs/génétique , Glomérulonéphrite/immunologie , Glomérulonéphrite/anatomopathologie , Glomérulonéphrite/virologie , Virus de l'hépatite B/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Hépatite B/immunologie , Hépatite B/complications , Humains , Techniques de coculture , Mâle , Modèles animaux de maladie humaine , Souris de lignée C57BL
12.
Sci Rep ; 14(1): 15873, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38982272

RÉSUMÉ

Apolipoprotein E (APOE) is a major cholesterol carrier responsible for lipid transport and injury repair in the brain. The human APOE gene (h-APOE) has 3 naturally occurring alleles: ε3, the common allele; ε4, which increases Alzheimer's disease (AD) risk up to 15-fold; and ε2, the rare allele which protects against AD. Although APOE4 has negative effects on neurocognition in old age, its persistence in the population suggests a survival advantage. We investigated the relationship between APOE genotypes and fertility in EFAD mice, a transgenic mouse model expressing h-APOE. We show that APOE4 transgenic mice had the highest level of reproductive performance, followed by APOE3 and APOE2. Intriguingly, APOE3 pregnancies had more fetal resorptions and reduced fetal weights relative to APOE4 pregnancies. In conclusion, APOE genotypes impact fertility and pregnancy outcomes in female mice, in concordance with findings in human populations. These mouse models may help elucidate how h-APOE4 promotes reproductive fitness at the cost of AD in later life.


Sujet(s)
Maladie d'Alzheimer , Apolipoprotéines E , Modèles animaux de maladie humaine , Fécondité , Souris transgéniques , Animaux , Maladie d'Alzheimer/génétique , Femelle , Souris , Fécondité/génétique , Humains , Apolipoprotéines E/génétique , Apolipoprotéine E4/génétique , Polymorphisme génétique , Grossesse , Génotype , Apolipoprotéine E3/génétique , Allèles
13.
Proc Natl Acad Sci U S A ; 121(29): e2401420121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38995966

RÉSUMÉ

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/pTau, however, appears to vary depending on the animal model. Our prior work suggested that antigen-specific memory CD8 T ("hiT") cells act upstream of Aß/pTau after brain injury. Here, we examine whether hiT cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hiT mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. We identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD.


Sujet(s)
Maladie d'Alzheimer , Lymphocytes T CD8+ , Modèles animaux de maladie humaine , Maladie d'Alzheimer/immunologie , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Animaux , Lymphocytes T CD8+/immunologie , Souris , Humains , Plaque amyloïde/anatomopathologie , Plaque amyloïde/immunologie , Peptides bêta-amyloïdes/métabolisme , Souris transgéniques , Encéphale/anatomopathologie , Encéphale/immunologie , Mâle , Interféron gamma/métabolisme , Interféron gamma/immunologie , Vieillissement/immunologie , Mémoire immunologique , Cellules T mémoire/immunologie , Perforine/métabolisme , Perforine/génétique , Femelle
14.
Sci Immunol ; 9(97): eado5295, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996008

RÉSUMÉ

αß T cell receptor (TCR) V(D)J genes code for billions of TCR combinations. However, only some appear on peripheral T cells in any individual because, to mature, thymocytes must react with low affinity but not high affinity with thymus expressed major histocompatibility (MHC)/peptides. MHC proteins are very polymorphic. Different alleles bind different peptides. Therefore, any individual might express many different MHC alleles to ensure that some peptides from an invader are bound to MHC and activate T cells. However, most individuals express limited numbers of MHC alleles. To explore this, we compared the TCR repertoires of naïve CD4 T cells in mice expressing one or two MHC alleles. Unexpectedly, the TCRs in heterozygotes were less diverse that those in the sum of their MHC homozygous relatives. Our results suggest that thymus negative selection cancels out the advantages of increased thymic positive selection in the MHC heterozygotes.


Sujet(s)
Lymphocytes T CD4+ , Hétérozygote , Animaux , Souris , Lymphocytes T CD4+/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Complexe majeur d'histocompatibilité/immunologie , Complexe majeur d'histocompatibilité/génétique , Souris de lignée C57BL , Thymus (glande)/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/immunologie , Souris transgéniques
15.
Nat Commun ; 15(1): 5563, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982047

RÉSUMÉ

The spatial organization of a neuronal circuit is critically important for its function since the location of neurons is often associated with function. In the cerebellum, the major output of the cerebellar cortex are synapses made from Purkinje cells onto neurons in the cerebellar nuclei, yet little has been known about the spatial organization of these synapses. We explored this question using whole-cell electrophysiology and optogenetics in acute sagittal cerebellar slices to produce spatial connectivity maps of cerebellar cortical output in mice. We observed non-random connectivity where Purkinje cell inputs clustered in cerebellar transverse zones: while many nuclear neurons received inputs from a single zone, several multi-zonal connectivity motifs were also observed. Single neurons receiving input from all four zones were overrepresented in our data. These findings reveal that the output of the cerebellar cortex is spatially structured and represents a locus for multimodal integration in the cerebellum.


Sujet(s)
Cortex cérébelleux , Optogénétique , Cellules de Purkinje , Synapses , Animaux , Cortex cérébelleux/physiologie , Cellules de Purkinje/physiologie , Souris , Synapses/physiologie , Mâle , Noyaux du cervelet/physiologie , Techniques de patch-clamp , Souris de lignée C57BL , Voies nerveuses/physiologie , Femelle , Neurones/physiologie , Cervelet/physiologie , Souris transgéniques
16.
Proc Natl Acad Sci U S A ; 121(28): e2403635121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38950371

RÉSUMÉ

While the intracellular-extracellular distribution of lactate has been suggested to play a critical role in the healthy and diseased brain, tools are lacking to noninvasively probe lactate in intracellular and extracellular spaces. Here, we show that, by measuring the diffusion of lactate with diffusion-weighted magnetic resonance (MR) spectroscopy in vivo and comparing it to the diffusion of purely intracellular metabolites, noninvasive quantification of extracellular and intracellular lactate fractions becomes possible. More specifically, we detect alterations of lactate diffusion in the APP/PS1 mouse model of Alzheimer's disease. Data modeling allows quantifying decreased extracellular lactate fraction in APP/PS1 mice as compared to controls, which is quantitatively confirmed with implanted enzyme-microelectrodes. The capability of diffusion-weighted MR spectroscopy to quantify extracellular-intracellular lactate fractions opens a window into brain metabolism, including in Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Encéphale , Acide lactique , Animaux , Acide lactique/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Souris , Souris transgéniques , Imagerie par résonance magnétique de diffusion/méthodes , Espace extracellulaire/métabolisme , Modèles animaux de maladie humaine , Spectroscopie par résonance magnétique/méthodes , Mâle , Précurseur de la protéine bêta-amyloïde/métabolisme
17.
Theranostics ; 14(9): 3653-3673, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948066

RÉSUMÉ

Rationale: Recent evidence highlights the pivotal role of mitochondrial dysfunction in mood disorders, but the mechanism involved remains unclear. We studied whether the Hippo/YAP/14-3-3η signaling pathway mediates mitochondrial abnormalities that result in the onset of major depressive disorder (MDD) in a mouse model. Methods: The ROC algorithm was used to identify a subpopulation of mice that were exposed to chronic unpredictable mild stress (CUMS) and exhibited the most prominent depressive phenotype (Dep). Electron microscopy, biochemical assays, quantitative PCR, and immunoblotting were used to evaluate synaptic and mitochondrial changes in the basolateral amygdala (BLA). RNA sequencing was used to explore changes in the Hippo pathway and downstream target genes. In vitro pharmacological inhibition and immunoprecipitation was used to confirm YAP/14-3-3η interaction and its role in neuronal mitochondrial dysfunction. We used virus-mediated gene overexpression and knockout in YAP transgenic mice to verify the regulatory effect of the Hippo/YAP/14-3-3η pathway on depressive-like behavior. Results: Transcriptomic data identified a large number of genes and signaling pathways that were specifically altered from the BLA of Dep mice. Dep mice showed notable synaptic impairment in BLA neurons, as well as mitochondrial damage characterized by abnormal mitochondrial morphology, compromised function, impaired biogenesis, and alterations in mitochondrial marker proteins. The Hippo signaling pathway was activated in Dep mice during CUMS, and the transcriptional regulatory activity of YAP was suppressed by phosphorylation of its Ser127 site. 14-3-3η was identified as an important co-regulatory factor of the Hippo/YAP pathway, as it can respond to chronic stress and regulate cytoplasmic retention of YAP. Importantly, the integrated Hippo/YAP/14-3-3η pathway mediated neuronal mitochondrial dysfunction and depressive behavior in Dep mice. Conclusion: The integrated Hippo/YAP/14-3-3η pathway in the BLA neuron is critical in mediating depressive-like behaviors in mice, suggesting a causal role for this pathway in susceptibility to chronic stress-induced depression. This pathway therefore may present a therapeutic target against mitochondrial dysfunction and synaptic impairment in MDD.


Sujet(s)
Groupe nucléaire basolatéral , Modèles animaux de maladie humaine , Voie de signalisation Hippo , Mitochondries , Protein-Serine-Threonine Kinases , Transduction du signal , Protéines de signalisation YAP , Animaux , Souris , Mitochondries/métabolisme , Protéines de signalisation YAP/métabolisme , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Mâle , Stress psychologique/complications , Stress psychologique/métabolisme , Protéines 14-3-3/métabolisme , Protéines 14-3-3/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Trouble dépressif majeur/métabolisme , Trouble dépressif majeur/anatomopathologie , Dépression/métabolisme , Souris de lignée C57BL , Neurones/métabolisme , Neurones/anatomopathologie , Souris transgéniques
18.
Eur J Histochem ; 68(3)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38963135

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder involving motor neuron (MN) loss in the motor cortex, brainstem and spinal cord leading to progressive paralysis and death. Due to the pathogenetic complexity, there are no effective therapies available. In this context the use of mesenchymal stem cells and their vesicular counterpart is an emerging therapeutic strategy to counteract neurodegeneration. The extracellular vesicles derived from adipose stem cells (ASC-EVs) recapitulate and ameliorate the neuroprotective effect of stem cells and, thanks to their small dimensions, makes their use suitable to develop novel therapeutic approaches for neurodegenerative diseases as ALS. Here we investigate a therapeutic regimen of ASC-EVs injection in SOD1(G93A) mice, the most widely used murine model of ALS. Repeated intranasal administrations of high doses of ASC-EVs were able to ameliorate motor performance of injected SOD1(G93A) mice at the early stage of the disease and produce a significant improvement at the end-stage in the lumbar MNs rescue. Moreover, ASC-EVs preserve the structure of neuromuscular junction without counteracting the muscle atrophy. The results indicate that the intranasal ASC-EVs administration acts in central nervous system sites rather than at peripheral level in SOD1(G93A) mice. These considerations allow us to identify future applications of ASC-EVs that involve different targets simultaneously to maximize the clinical and neuropathological outcomes in ALS in vivo models.


Sujet(s)
Sclérose latérale amyotrophique , Vésicules extracellulaires , Cellules souches mésenchymateuses , Superoxide dismutase-1 , Animaux , Vésicules extracellulaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , Souris , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/thérapie , Sclérose latérale amyotrophique/anatomopathologie , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme , Souris transgéniques , Modèles animaux de maladie humaine , Tissu adipeux/métabolisme , Motoneurones/métabolisme , Jonction neuromusculaire/métabolisme
19.
FASEB J ; 38(13): e23797, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963344

RÉSUMÉ

The role of N-glycosylation in the myogenic process remains poorly understood. Here, we evaluated the impact of N-glycosylation inhibition by Tunicamycin (TUN) or by phosphomannomutase 2 (PMM2) gene knockdown, which encodes an enzyme essential for catalyzing an early step of the N-glycosylation pathway, on C2C12 myoblast differentiation. The effect of chronic treatment with TUN on tibialis anterior (TA) and extensor digitorum longus (EDL) muscles of WT and MLC/mIgf-1 transgenic mice, which overexpress muscle Igf-1Ea mRNA isoform, was also investigated. TUN-treated and PMM2 knockdown C2C12 cells showed reduced ConA, PHA-L, and AAL lectin binding and increased ER-stress-related gene expression (Chop and Hspa5 mRNAs and s/uXbp1 ratio) compared to controls. Myogenic markers (MyoD, myogenin, and Mrf4 mRNAs and MF20 protein) and myotube formation were reduced in both TUN-treated and PMM2 knockdown C2C12 cells. Body and TA weight of WT and MLC/mIgf-1 mice were not modified by TUN treatment, while lectin binding slightly decreased in the TA muscle of WT (ConA and AAL) and MLC/mIgf-1 (ConA) mice. The ER-stress-related gene expression did not change in the TA muscle of WT and MLC/mIgf-1 mice after TUN treatment. TUN treatment decreased myogenin mRNA and increased atrogen-1 mRNA, particularly in the TA muscle of WT mice. Finally, the IGF-1 production and IGF1R signaling pathways activation were reduced due to N-glycosylation inhibition in TA and EDL muscles. Decreased IGF1R expression was found in TUN-treated C2C12 myoblasts which was associated with lower IGF-1-induced IGF1R, AKT, and ERK1/2 phosphorylation compared to CTR cells. Chronic TUN-challenge models can help to elucidate the molecular mechanisms through which diseases associated with aberrant N-glycosylation, such as Congenital Disorders of Glycosylation (CDG), affect muscle and other tissue functions.


Sujet(s)
Différenciation cellulaire , Chaperonne BiP du réticulum endoplasmique , Muscles squelettiques , Myoblastes , Récepteur IGF de type 1 , Transduction du signal , Tunicamycine , Animaux , Souris , Glycosylation , Myoblastes/métabolisme , Chaperonne BiP du réticulum endoplasmique/métabolisme , Tunicamycine/pharmacologie , Récepteur IGF de type 1/métabolisme , Récepteur IGF de type 1/génétique , Muscles squelettiques/métabolisme , Développement musculaire/physiologie , Lignée cellulaire , Souris transgéniques , Stress du réticulum endoplasmique , Facteur de croissance IGF-I/métabolisme , Facteur de croissance IGF-I/génétique
20.
Proc Natl Acad Sci U S A ; 121(29): e2310421121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976733

RÉSUMÉ

We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Tests de neutralisation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Humains , Anticorps neutralisants/immunologie , Souris , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Anticorps antiviraux/immunologie , Tests de neutralisation/méthodes , Souris transgéniques , Coronavirus humain OC43/immunologie , Coronavirus humain OC43/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/immunologie , Chlorocebus aethiops , Cellules Vero , Macaca mulatta
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...