Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 32
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
bioRxiv ; 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38854151

RÉSUMÉ

In response to central nervous system (CNS) injury, tissue resident immune cells such as microglia and circulating systemic neutrophils are often first responders. The degree to which these cells interact in response to CNS damage is poorly understood, and even less so, in the neural retina which poses a challenge for high resolution imaging in vivo. In this study, we deploy fluorescence adaptive optics scanning light ophthalmoscopy (AOSLO) to study fluorescent microglia and neutrophils in mice. We simultaneously track immune cell dynamics using label-free phase-contrast AOSLO at micron-level resolution. Retinal lesions were induced with 488 nm light focused onto photoreceptor (PR) outer segments. These lesions focally ablated PRs, with minimal collateral damage to cells above and below the plane of focus. We used in vivo (AOSLO, SLO and OCT) imaging to reveal the natural history of the microglial and neutrophil response from minutes-to-months after injury. While microglia showed dynamic and progressive immune response with cells migrating into the injury locus within 1-day after injury, neutrophils were not recruited despite close proximity to vessels carrying neutrophils only microns away. Post-mortem confocal microscopy confirmed in vivo findings. This work illustrates that microglial activation does not recruit neutrophils in response to acute, focal loss of photoreceptors, a condition encountered in many retinal diseases.

2.
J Neurosci ; 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830764

RÉSUMÉ

Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's Disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-ß (Aß) pathology (PS2APP) or combined Aß and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.Significance Statement Multiple TREM2 agonist antibodies are investigated in mouse models of Alzheimer's Disease and Multiple Sclerosis. Despite agonism in culture models and after acute dosing in mice, antibodies do not show benefit in overall AD pathology and worsen recovery after demyelination.

3.
J Med Chem ; 67(5): 3287-3306, 2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38431835

RÉSUMÉ

Transient receptor potential ankyrin 1 (TRPA1) is a nonselective calcium ion channel highly expressed in the primary sensory neurons, functioning as a polymodal sensor for exogenous and endogenous stimuli, and has been implicated in neuropathic pain and respiratory disease. Herein, we describe the optimization of potent, selective, and orally bioavailable TRPA1 small molecule antagonists with strong in vivo target engagement in rodent models. Several lead molecules in preclinical single- and short-term repeat-dose toxicity studies exhibited profound prolongation of coagulation parameters. Based on a thorough investigative toxicology and clinical pathology analysis, anticoagulation effects in vivo are hypothesized to be manifested by a metabolite─generated by aldehyde oxidase (AO)─possessing a similar pharmacophore to known anticoagulants (i.e., coumarins, indandiones). Further optimization to block AO-mediated metabolism yielded compounds that ameliorated coagulation effects in vivo, resulting in the discovery and advancement of clinical candidate GDC-6599, currently in Phase II clinical trials for respiratory indications.


Sujet(s)
Maladies de l'appareil respiratoire , Canaux cationiques TRP , Humains , Canaux cationiques TRP/métabolisme , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire , Aldehyde oxidase/métabolisme , Oxidoreductases/métabolisme , Protéines du cytosquelette/métabolisme
4.
EBioMedicine ; 102: 105045, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38471394

RÉSUMÉ

BACKGROUND: Schizophrenia, a debilitating psychiatric disorder, displays considerable interindividual variation in clinical presentations. The ongoing debate revolves around whether this heterogeneity signifies a continuum of severity linked to a singular causative factor or a collection of distinct subtypes with unique origins. Within the realm of schizophrenia, the functional impairment of GluN2A, a subtype of the NMDA receptor, has been associated with an elevated risk. Despite GluN2A's expression across various neuronal types throughout the brain, its specific contributions to schizophrenia and its involvement in particular cell types or brain regions remain unexplored. METHODS: We generated age-specific, cell type-specific or brain region-specific conditional knockout mice targeting GluN2A and conducted a comprehensive analysis using tests measuring phenotypes relevant to schizophrenia. FINDINGS: Through the induction of germline ablation of GluN2A, we observed the emergence of numerous schizophrenia-associated abnormalities in adult mice. Intriguingly, GluN2A knockout performed at different ages, in specific cell types and within distinct brain regions, we observed overlapping yet distinct schizophrenia-related phenotypes in mice. INTERPRETATION: Our interpretation suggests that the dysfunction of GluN2A is sufficient to evoke heterogeneous manifestations associated with schizophrenia, indicating that GluN2A stands as a prominent risk factor and a potential therapeutic target for schizophrenia. FUNDING: This project received support from the Shanghai Municipal Science and Technology Major Project (Grant No. 2019SHZDZX02) awarded to Y.C. and the Natural Science Foundation of Shanghai (Grant No. 19ZR1468600 and 201409003800) awarded to G.Y.


Sujet(s)
Récepteurs du N-méthyl-D-aspartate , Schizophrénie , Animaux , Souris , Encéphale/métabolisme , Neurones/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Récepteurs du N-méthyl-D-aspartate/métabolisme , Schizophrénie/génétique , Schizophrénie/métabolisme
5.
Cell Rep Methods ; 4(1): 100673, 2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38171361

RÉSUMÉ

While antisense oligonucleotides (ASOs) are used in the clinic, therapeutic development is hindered by the inability to assay ASO delivery and activity in vivo. Accordingly, we developed a dual-fluorescence, knockin mouse model that constitutively expresses mKate2 and an engineered EGFP that is alternatively spliced in the presence of ASO to induce expression. We first examined free ASO activity in the brain following intracerebroventricular injection revealing EGFP splice-switching is both ASO concentration and time dependent in major central nervous system cell types. We then assayed the impact of lipid nanoparticle delivery on ASO activity after intravenous administration. Robust EGFP fluorescence was observed in the liver and EGFP+ cells were successfully isolated using fluorescence-activated cell sorting. Together, these results show the utility of this animal model in quantifying both cell-type- and organ-specific ASO delivery, which can be used to advance ASO therapeutics for many disease indications.


Sujet(s)
Oligonucléotides antisens , Oligonucléotides , Souris , Animaux , Foie/métabolisme , Administration par voie intraveineuse , Agents colorants/métabolisme
6.
Front Cell Dev Biol ; 11: 1252547, 2023.
Article de Anglais | MEDLINE | ID: mdl-37691820

RÉSUMÉ

Rare DRAM2 coding variants cause retinal dystrophy with early macular involvement via unknown mechanisms. We found that DRAM2 is ubiquitously expressed in the human eye and expression changes were observed in eyes with more common maculopathy such as Age-related Macular Degeneration (AMD). To gain insights into pathogenicity of DRAM2-related retinopathy, we used a combination of in vitro and in vivo models. We found that DRAM2 loss in human pluripotent stem cell (hPSC)-derived retinal organoids caused the presence of additional mesenchymal cells. Interestingly, Dram2 loss in mice also caused increased proliferation of cells from the choroid in vitro and exacerbated choroidal neovascular lesions in vivo. Furthermore, we observed that DRAM2 loss in human retinal pigment epithelial (RPE) cells resulted in increased susceptibility to stress-induced cell death in vitro and that Dram2 loss in mice caused age-related photoreceptor degeneration. This highlights the complexity of DRAM2 function, as its loss in choroidal cells provided a proliferative advantage, whereas its loss in post-mitotic cells, such as photoreceptor and RPE cells, increased degeneration susceptibility. Different models such as human pluripotent stem cell-derived systems and mice can be leveraged to study and model human retinal dystrophies; however, cell type and species-specific expression must be taken into account when selecting relevant systems.

7.
Nat Commun ; 14(1): 4703, 2023 08 05.
Article de Anglais | MEDLINE | ID: mdl-37543621

RÉSUMÉ

TGFß signaling is associated with non-response to immune checkpoint blockade in patients with advanced cancers, particularly in the immune-excluded phenotype. While previous work demonstrates that converting tumors from excluded to inflamed phenotypes requires attenuation of PD-L1 and TGFß signaling, the underlying cellular mechanisms remain unclear. Here, we show that TGFß and PD-L1 restrain intratumoral stem cell-like CD8 T cell (TSCL) expansion and replacement of progenitor-exhausted and dysfunctional CD8 T cells with non-exhausted T effector cells in the EMT6 tumor model in female mice. Upon combined TGFß/PD-L1 blockade IFNγhi CD8 T effector cells show enhanced motility and accumulate in the tumor. Ensuing IFNγ signaling transforms myeloid, stromal, and tumor niches to yield an immune-supportive ecosystem. Blocking IFNγ abolishes the anti-PD-L1/anti-TGFß therapy efficacy. Our data suggest that TGFß works with PD-L1 to prevent TSCL expansion and replacement of exhausted CD8 T cells, thereby maintaining the T cell compartment in a dysfunctional state.


Sujet(s)
Antigène CD274 , Tumeurs du sein , Lymphocytes T CD8+ , Inhibiteurs de points de contrôle immunitaires , Facteur de croissance transformant bêta , Femelle , Animaux , Souris , Différenciation cellulaire , Lymphocytes T CD8+/immunologie , Cellules souches , Antigène CD274/antagonistes et inhibiteurs , Facteur de croissance transformant bêta/antagonistes et inhibiteurs , Interféron gamma/immunologie , Épuisement des cellules T , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Souris de lignée BALB C , Lignée cellulaire tumorale , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , RNA-Seq
8.
Nature ; 611(7934): 148-154, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36171287

RÉSUMÉ

Recent single-cell studies of cancer in both mice and humans have identified the emergence of a myofibroblast population specifically marked by the highly restricted leucine-rich-repeat-containing protein 15 (LRRC15)1-3. However, the molecular signals that underlie the development of LRRC15+ cancer-associated fibroblasts (CAFs) and their direct impact on anti-tumour immunity are uncharacterized. Here in mouse models of pancreatic cancer, we provide in vivo genetic evidence that TGFß receptor type 2 signalling in healthy dermatopontin+ universal fibroblasts is essential for the development of cancer-associated LRRC15+ myofibroblasts. This axis also predominantly drives fibroblast lineage diversity in human cancers. Using newly developed Lrrc15-diphtheria toxin receptor knock-in mice to selectively deplete LRRC15+ CAFs, we show that depletion of this population markedly reduces the total tumour fibroblast content. Moreover, the CAF composition is recalibrated towards universal fibroblasts. This relieves direct suppression of tumour-infiltrating CD8+ T cells to enhance their effector function and augments tumour regression in response to anti-PDL1 immune checkpoint blockade. Collectively, these findings demonstrate that TGFß-dependent LRRC15+ CAFs dictate the tumour-fibroblast setpoint to promote tumour growth. These cells also directly suppress CD8+ T cell function and limit responsiveness to checkpoint blockade. Development of treatments that restore the homeostatic fibroblast setpoint by reducing the population of pro-disease LRRC15+ myofibroblasts may improve patient survival and response to immunotherapy.


Sujet(s)
Fibroblastes associés au cancer , Protéines membranaires , Myofibroblastes , Tumeurs du pancréas , Cellules stromales , Animaux , Humains , Souris , Fibroblastes associés au cancer/métabolisme , Lymphocytes T CD8+/immunologie , Protéines membranaires/métabolisme , Myofibroblastes/métabolisme , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Récepteurs TGF-bêta , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral , Antigène CD274
9.
Nat Commun ; 12(1): 7150, 2021 12 09.
Article de Anglais | MEDLINE | ID: mdl-34887411

RÉSUMÉ

Tissue regeneration after injury requires coordinated regulation of stem cell activation, division, and daughter cell differentiation, processes that are increasingly well understood in many regenerating tissues. How accurate stem cell positioning and localized integration of new cells into the damaged epithelium are achieved, however, remains unclear. Here, we show that enteroendocrine cells coordinate stem cell migration towards a wound in the Drosophila intestinal epithelium. In response to injury, enteroendocrine cells release the N-terminal domain of the PTK7 orthologue, Otk, which activates non-canonical Wnt signaling in intestinal stem cells, promoting actin-based protrusion formation and stem cell migration towards a wound. We find that this migratory behavior is closely linked to proliferation, and that it is required for efficient tissue repair during injury. Our findings highlight the role of non-canonical Wnt signaling in regeneration of the intestinal epithelium, and identify enteroendocrine cell-released ligands as critical coordinators of intestinal stem cell migration.


Sujet(s)
Mouvement cellulaire , Drosophila/métabolisme , Cellules entéroendocrines/cytologie , Muqueuse intestinale/cytologie , Cellules souches/cytologie , Protéines de type Wingless/métabolisme , Plaies et blessures/physiopathologie , Animaux , Drosophila/cytologie , Drosophila/génétique , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Muqueuse intestinale/métabolisme , Intestins , Récepteurs à activité tyrosine kinase/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Cellules souches/métabolisme , Protéines de type Wingless/génétique , Voie de signalisation Wnt , Plaies et blessures/génétique , Plaies et blessures/métabolisme
10.
Cell Rep Med ; 2(8): 100381, 2021 08 17.
Article de Anglais | MEDLINE | ID: mdl-34467254

RÉSUMÉ

Anti-integrins are therapeutically effective for inflammatory bowel disease, yet the relative contribution of α4ß7 and αEß7 to gut lymphocyte trafficking is not fully elucidated. Here, we evaluate the effect of α4ß7 and αEß7 blockade using a combination of murine models of gut trafficking and longitudinal gene expression analysis in etrolizumab-treated patients with Crohn's disease (CD). Dual blockade of α4ß7 and αEß7 reduces CD8+ T cell accumulation in the gut to a greater extent than blockade of either integrin alone. Anti-αEß7 reduces epithelial:T cell interactions and promotes egress of activated T cells from the mucosa into lymphatics. Inflammatory gene expression is greater in human intestinal αEß7+ T cells. Etrolizumab-treated patients with CD display a treatment-specific reduction in inflammatory and cytotoxic intraepithelial lymphocytes (IEL) genes. Concurrent blockade of α4ß7 and αEß7 promotes reduction of cytotoxic IELs and inflammatory T cells in the gut mucosa through a stepwise inhibition of intestinal tissue entry and retention.


Sujet(s)
Maladies inflammatoires intestinales/immunologie , Intégrines/métabolisme , Lymphocytes/immunologie , Animaux , Anticorps monoclonaux humanisés/pharmacologie , Biopsie , Lymphocytes T CD8+ , Cadhérines/métabolisme , Communication cellulaire , Mouvement cellulaire , Côlon/anatomopathologie , Épitopes/immunologie , Femelle , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Inflammation/complications , Inflammation/anatomopathologie , Maladies inflammatoires intestinales/complications , Maladies inflammatoires intestinales/anatomopathologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/immunologie , Muqueuse intestinale/anatomopathologie , Noeuds lymphatiques/anatomopathologie , Souris de lignée C57BL , Souris transgéniques , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques
11.
Br J Clin Pharmacol ; 87(1): 129-139, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32415670

RÉSUMÉ

BACKGROUND AND PURPOSE: Develop a translational assay of Transient Receptor Potential Ankyrin 1 (TRPA1) activity for use as a preclinical and clinical biomarker. EXPERIMENTAL APPROACH: Allyl isothiocyanate (AITC), capsaicin or citric acid were applied to ears of wildtype and Trpa1-knock out (Trpa1 KO) rats, and changes in dermal blood flow (DBF) were measured by laser speckle contrast imaging. In humans, the DBF, pain and itch responses to 5-20% AITC applied to the forearm were measured and safety was evaluated. Reproducibility of the DBF, pain and itch responses to topically applied 10% and 15% AITC were assessed at two visits separated by 13-15 days. DBF changes were summarized at 5-minute intervals as areas under the curve (AUC) and maxima. Intraclass correlation coefficient (ICC) was calculated to assess arm-arm and period-period reproducibility. KEY RESULTS: AITC- and citric acid-induced DBF were significantly reduced in Trpa1 KO rats compared to wildtype (90 ± 2% and 65 ± 11% reduction, respectively), whereas capsaicin response did not differ. In humans, each AITC concentration significantly increased DBF compared to vehicle with the maximal increase occurring 5 minutes post application. Ten percent and 15% AITC were selected as safe and effective stimuli. AUC from 0 to 5 minutes was the most reproducible metric of AITC-induced DBF across arms (ICC = 0.92) and periods (ICC = 0.85). Subject-reported pain was more reproducible than itch across visits (ICC = 0.76 vs 0.17, respectively). CONCLUSION AND IMPLICATIONS: AITC-induced DBF is a suitable target engagement biomarker of TRPA1 activity for preclinical and clinical studies of TRPA1 antagonists.


Sujet(s)
Rodentia , Animaux , Marqueurs biologiques , Humains , Isothiocyanates , Rats , Reproductibilité des résultats , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire
12.
J Neurosci ; 40(9): 1956-1974, 2020 02 26.
Article de Anglais | MEDLINE | ID: mdl-31980586

RÉSUMÉ

TREM2 is an Alzheimer's disease (AD) risk gene expressed in microglia. To study the role of Trem2 in a mouse model of ß-amyloidosis, we compared PS2APP transgenic mice versus PS2APP mice lacking Trem2 (PS2APP;Trem2ko) at ages ranging from 4 to 22 months. Microgliosis was impaired in PS2APP;Trem2ko mice, with Trem2-deficient microglia showing compromised expression of proliferation/Wnt-related genes and marked accumulation of ApoE. Plaque abundance was elevated in PS2APP;Trem2ko females at 6-7 months; but by 12 or 19-22 months of age, it was notably diminished in female and male PS2APP;Trem2ko mice, respectively. Across all ages, plaque morphology was more diffuse in PS2APP;Trem2ko brains, and the Aß42:Aß40 ratio was elevated. The amount of soluble, fibrillar Aß oligomers also increased in PS2APP;Trem2ko hippocampi. Associated with these changes, axonal dystrophy was exacerbated from 6 to 7 months onward in PS2APP;Trem2ko mice, notwithstanding the reduced plaque load at later ages. PS2APP;Trem2ko mice also exhibited more dendritic spine loss around plaque and more neurofilament light chain in CSF. Thus, aggravated neuritic dystrophy is a more consistent outcome of Trem2 deficiency than amyloid plaque load, suggesting that the microglial packing of Aß into dense plaque is an important neuroprotective activity.SIGNIFICANCE STATEMENT Genetic studies indicate that TREM2 gene mutations confer increased Alzheimer's disease (AD) risk. We studied the effects of Trem2 deletion in the PS2APP mouse AD model, in which overproduction of Aß peptide leads to amyloid plaque formation and associated neuritic dystrophy. Interestingly, neuritic dystrophies were intensified in the brains of Trem2-deficient mice, despite these mice displaying reduced plaque accumulation at later ages (12-22 months). Microglial clustering around plaques was impaired, plaques were more diffuse, and the Aß42:Aß40 ratio and amount of soluble, fibrillar Aß oligomers were elevated in Trem2-deficient brains. These results suggest that the Trem2-dependent compaction of Aß into dense plaques is a protective microglial activity, limiting the exposure of neurons to toxic Aß species.


Sujet(s)
Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Axones/anatomopathologie , Épines dendritiques/anatomopathologie , Glycoprotéines membranaires/génétique , Fragments peptidiques/métabolisme , Plaque amyloïde/génétique , Récepteurs immunologiques/génétique , Facteur en trèfle-1/métabolisme , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Microglie/anatomopathologie , Neurites/anatomopathologie , Protéines neurofilamenteuses/liquide cérébrospinal , Plaque amyloïde/anatomopathologie
13.
Cell Rep ; 30(2): 381-396.e4, 2020 01 14.
Article de Anglais | MEDLINE | ID: mdl-31940483

RÉSUMÉ

NMDA receptors (NMDARs) play subunit-specific roles in synaptic function and are implicated in neuropsychiatric and neurodegenerative disorders. However, the in vivo consequences and therapeutic potential of pharmacologically enhancing NMDAR function via allosteric modulation are largely unknown. We examine the in vivo effects of GNE-0723, a positive allosteric modulator of GluN2A-subunit-containing NMDARs, on brain network and cognitive functions in mouse models of Dravet syndrome (DS) and Alzheimer's disease (AD). GNE-0723 use dependently potentiates synaptic NMDA receptor currents and reduces brain oscillation power with a predominant effect on low-frequency (12-20 Hz) oscillations. Interestingly, DS and AD mouse models display aberrant low-frequency oscillatory power that is tightly correlated with network hypersynchrony. GNE-0723 treatment reduces aberrant low-frequency oscillations and epileptiform discharges and improves cognitive functions in DS and AD mouse models. GluN2A-subunit-containing NMDAR enhancers may have therapeutic benefits in brain disorders with network hypersynchrony and cognitive impairments.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Encéphale/métabolisme , Cognition/effets des médicaments et des substances chimiques , Cyclopropanes/pharmacologie , Épilepsies myocloniques/traitement médicamenteux , Nitriles/pharmacologie , Récepteurs du N-méthyl-D-aspartate/métabolisme , Thiazoles/pharmacologie , Régulation allostérique/effets des médicaments et des substances chimiques , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Cellules CHO , Cricetulus , Modèles animaux de maladie humaine , Épilepsies myocloniques/génétique , Épilepsies myocloniques/métabolisme , Humains , Mâle , Souris , Souris de lignée C57BL , Pyrazoles/pharmacologie , Récepteurs du N-méthyl-D-aspartate/agonistes
14.
Cell Rep ; 28(8): 2111-2123.e6, 2019 08 20.
Article de Anglais | MEDLINE | ID: mdl-31433986

RÉSUMÉ

Complement pathway overactivation can lead to neuronal damage in various neurological diseases. Although Alzheimer's disease (AD) is characterized by ß-amyloid plaques and tau tangles, previous work examining complement has largely focused on amyloidosis models. We find that glial cells show increased expression of classical complement components and the central component C3 in mouse models of amyloidosis (PS2APP) and more extensively tauopathy (TauP301S). Blocking complement function by deleting C3 rescues plaque-associated synapse loss in PS2APP mice and ameliorates neuron loss and brain atrophy in TauP301S mice, improving neurophysiological and behavioral measurements. In addition, C3 protein is elevated in AD patient brains, including at synapses, and levels and processing of C3 are increased in AD patient CSF and correlate with tau. These results demonstrate that complement activation contributes to neurodegeneration caused by tau pathology and suggest that blocking C3 function might be protective in AD and other tauopathies.


Sujet(s)
Maladie d'Alzheimer/immunologie , Amyloïdose/immunologie , Complément C3/métabolisme , Dégénérescence nerveuse/immunologie , Tauopathies/immunologie , Maladie d'Alzheimer/génétique , Animaux , Atrophie , Comportement animal , Marqueurs biologiques/métabolisme , Encéphale/anatomopathologie , Complément C1q/métabolisme , Complément C3/liquide cérébrospinal , Complément C3/génétique , Modèles animaux de maladie humaine , Femelle , Délétion de gène , Régulation de l'expression des gènes , Humains , Mâle , Souris transgéniques , Dégénérescence nerveuse/génétique , Neurones/métabolisme , Neurones/anatomopathologie , Plaque amyloïde/métabolisme , Synapses/métabolisme
15.
Cancer Immunol Res ; 7(6): 963-976, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31064777

RÉSUMÉ

Exhausted T cells have been described in cancer patients and murine tumor models largely based on their expression of various inhibitory receptors. Understanding of the functional attributes of these cells is limited. Here, we report that among CD8+ T cells in commonly used syngeneic tumor models, the coexpression of inhibitory receptors PD-1, LAG3, and TIM3 defined a group of highly activated and functional effector cells. Coexpression of these receptors further enriched for antigen-specific cells with increased T-cell receptor clonality. Anti-PD-L1 treatment increased the number and activation of these triple-positive CD8+ T cells without affecting the density of PD-1- cells. The intratumoral density of CD8+ T cells coexpressing inhibitory receptors negatively correlated with tumor burden. The density ratio and pretreatment phenotype of CD8+ T cells coexpressing inhibitory receptors was positively correlated with response across a variety of tumor models. Our results demonstrate that coexpression of inhibitory receptors is not a signifier of exhausted T cells, but rather can define a group of activated and functional effector cells in syngeneic tumor models. In the cancer setting, these cells could represent a heterogeneous population of not only exhausted but also highly activated cells responsive to treatment.


Sujet(s)
Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Récepteurs costimulateurs et inhibiteurs des cellules T/génétique , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Tumeurs/étiologie , Tumeurs/métabolisme , Animaux , Antigène CD274/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux , Lignée cellulaire tumorale , Cytotoxicité immunologique , Modèles animaux de maladie humaine , Déterminants antigéniques des lymphocytes T/immunologie , Femelle , Facteur nucléaire hépatocytaire HNF-1 alpha/génétique , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Isogreffes , Souris , Tumeurs/anatomopathologie , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme
16.
Sci Rep ; 8(1): 13055, 2018 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-30143651

RÉSUMÉ

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

17.
Sci Rep ; 8(1): 7348, 2018 05 09.
Article de Anglais | MEDLINE | ID: mdl-29743491

RÉSUMÉ

Geographic atrophy (GA), the advanced form of dry age-related macular degeneration (AMD), is characterized by progressive loss of retinal pigment epithelium cells and photoreceptors in the setting of characteristic extracellular deposits and remains a serious unmet medical need. While genetic predisposition to AMD is dominated by polymorphisms in complement genes, it remains unclear how complement activation contributes to retinal atrophy. Here we demonstrate that complement is activated on photoreceptor outer segments (POS) in the retina peripheral to atrophic lesions associated with GA. When exposed to human serum following outer blood-retinal barrier breakdown, POS act as potent activators of the classical and alternative complement pathway. In mouse models of retinal degeneration, classical and alternative pathway complement activation on photoreceptors contributed to the loss of photoreceptor function. This was dependent on C5a-mediated recruitment of peripheral blood monocytes but independent of resident microglia. Genetic or pharmacologic inhibition of both classical and alternative complement C3 and C5 convertases was required to reduce progressive degeneration of photoreceptor rods and cones. Our study implicates systemic classical and alternative complement proteins and peripheral blood monocytes as critical effectors of localized retinal degeneration with potential relevance for the contribution of complement activation to GA.


Sujet(s)
Activation du complément/génétique , Atrophie géographique/physiopathologie , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Animaux , Atrophie/anatomopathologie , Activation du complément/physiologie , Complément C3/génétique , Complément C3/physiologie , Complément C4/génétique , Complément C4/physiologie , Atrophie géographique/génétique , Humains , Dégénérescence maculaire/physiopathologie , Souris , Souris de lignée C57BL , Souris knockout , Monocytes/métabolisme , Cellules photoréceptrices/métabolisme , Rétine/métabolisme , Dégénérescence de la rétine/anatomopathologie , Épithélium pigmentaire de la rétine/métabolisme
18.
J Exp Med ; 213(2): 189-207, 2016 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-26755704

RÉSUMÉ

Age-related macular degeneration (AMD), a leading cause of vision impairment in the ageing population, is characterized by irreversible loss of retinal pigment epithelial (RPE) cells and photoreceptors and can be associated with choroidal neovascularization. Mononuclear phagocytes are often present in AMD lesions, but the processes that direct myeloid cell recruitment remain unclear. Here, we identify IL-33 as a key regulator of inflammation and photoreceptor degeneration after retina stress or injury. IL-33(+) Müller cells were more abundant and IL-33 cytokine was elevated in advanced AMD cases compared with age-matched controls with no AMD. In rodents, retina stress resulted in release of bioactive IL-33 that in turn increased inflammatory chemokine and cytokine expression in activated Müller cells. Deletion of ST2, the IL-33 receptor α chain, or treatment with a soluble IL-33 decoy receptor significantly reduced release of inflammatory mediators from Müller cells, inhibited accumulation of mononuclear phagocytes in the outer retina, and protected photoreceptor rods and cones after a retina insult. This study demonstrates a central role for IL-33 in regulating mononuclear phagocyte recruitment to the photoreceptor layer and positions IL-33 signaling as a potential therapeutic target in macular degenerative diseases.


Sujet(s)
Immunité innée , Interleukine-33/métabolisme , Dégénérescence maculaire/immunologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Études cas-témoins , Noyau de la cellule/immunologie , Cytokines/métabolisme , Cellules épendymogliales/immunologie , Cellules épendymogliales/anatomopathologie , Femelle , Humains , Techniques in vitro , Protéine-1 analogue au récepteur de l'interleukin-1 , Interleukine-33/composition chimique , Interleukine-33/déficit , Interleukine-33/génétique , Macula/immunologie , Macula/anatomopathologie , Dégénérescence maculaire/génétique , Dégénérescence maculaire/anatomopathologie , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Maturation post-traductionnelle des protéines , Rats , Récepteurs de surface cellulaire/génétique , Récepteurs de surface cellulaire/métabolisme , Récepteurs aux interleukines/déficit , Récepteurs aux interleukines/génétique , Récepteurs aux interleukines/métabolisme , Épithélium pigmentaire de la rétine/immunologie , Épithélium pigmentaire de la rétine/anatomopathologie
19.
J Neurophysiol ; 112(2): 233-48, 2014 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-24740854

RÉSUMÉ

Whisker deflection evokes sparse, low-probability spiking among L2/3 pyramidal cells in rodent somatosensory cortex (S1), with spiking distributed nonuniformly between more and less responsive cells. The cellular and local circuit factors that determine whisker responsiveness across neurons are unclear. To identify these factors, we used two-photon calcium imaging and loose-seal recording to identify more and less responsive L2/3 neurons in S1 slices in vitro, during feedforward recruitment of the L2/3 network by L4 stimulation. We observed a broad gradient of spike recruitment thresholds within local L2/3 populations, with low- and high-threshold cells intermixed. This recruitment gradient was significantly correlated across different L4 stimulation sites, and between L4-evoked and whisker-evoked responses in vivo, indicating that a substantial component of responsiveness is independent of tuning to specific feedforward inputs. Low- and high-threshold L2/3 pyramidal cells differed in L4-evoked excitatory synaptic conductance and intrinsic excitability, including spike threshold and the likelihood of doublet spike bursts. A gradient of intrinsic excitability was observed across neurons. Cells that spiked most readily to L4 stimulation received the most synaptic excitation but had the lowest intrinsic excitability. Low- and high-threshold cells did not differ in dendritic morphology, passive membrane properties, or L4-evoked inhibitory conductance. Thus multiple gradients of physiological properties exist across L2/3 pyramidal cells, with excitatory synaptic input strength best predicting overall spiking responsiveness during network recruitment.


Sujet(s)
Potentiels évoqués somatosensoriels , Cellules pyramidales/physiologie , Cortex somatosensoriel/physiologie , Vibrisses/innervation , Animaux , Signalisation calcique , Potentiels post-synaptiques excitateurs , Potentiels post-synaptiques inhibiteurs , Souris , Souris de lignée C57BL , Cellules pyramidales/métabolisme , Rats , Rat Long-Evans , Seuils sensoriels , Cortex somatosensoriel/cytologie , Vibrisses/physiologie
20.
J Exp Med ; 211(2): 233-44, 2014 Feb 10.
Article de Anglais | MEDLINE | ID: mdl-24470444

RÉSUMÉ

Antibodies to transferrin receptor (TfR) have potential use for therapeutic entry into the brain. We have shown that bispecific antibodies against TfR and ß-secretase (BACE1 [ß-amyloid cleaving enzyme-1]) traverse the blood-brain barrier (BBB) and effectively reduce brain amyloid ß levels. We found that optimizing anti-TfR affinity improves brain exposure and BACE1 inhibition. Here we probe the cellular basis of this improvement and explore whether TfR antibody affinity alters the intracellular trafficking of TfR. Comparing high- and low-affinity TfR bispecific antibodies in vivo, we found that high-affinity binding to TfR caused a dose-dependent reduction of brain TfR levels. In vitro live imaging and colocalization experiments revealed that high-affinity TfR bispecific antibodies facilitated the trafficking of TfR to lysosomes and thus induced the degradation of TfR, an observation which was further confirmed in vivo. Importantly, high-affinity anti-TfR dosing induced reductions in brain TfR levels, which significantly decreased brain exposure to a second dose of low-affinity anti-TfR bispecific. Thus, high-affinity anti-TfR alters TfR trafficking, which dramatically impacts the capacity for TfR to mediate BBB transcytosis.


Sujet(s)
Anticorps bispécifiques/métabolisme , Encéphale/immunologie , Encéphale/métabolisme , Récepteurs à la transferrine/immunologie , Récepteurs à la transferrine/métabolisme , Amyloid precursor protein secretases/immunologie , Animaux , Affinité des anticorps , Aspartic acid endopeptidases/immunologie , Barrière hémato-encéphalique/immunologie , Barrière hémato-encéphalique/métabolisme , Femelle , Lysosomes/immunologie , Lysosomes/métabolisme , Souris , Souris de lignée C57BL , Transport des protéines , Transcytose/immunologie , Transcytose/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...