Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Virol ; 98(5): e0041124, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38567952

ABSTRACT

Influenza A virus infection activates the NLRP3 inflammasome, a multiprotein signaling complex responsible for the proteolytic activation and release of the proinflammatory cytokine IL-1ß from monocytes and macrophages. Some influenza A virus (IAV) strains encode a short 90-amino acid peptide (PB1-F2) on an alternative open reading frame of segment 2, with immunomodulatory activity. We recently demonstrated that contemporary IAV PB1-F2 inhibits the activation of NLRP3, potentially by NEK7-dependent activation. PB1-F2 binds to NLRP3 with its C-terminal 50 amino acids, but the exact binding motif was unknown. On the NLRP3 side, the interface is formed through the leucine-rich-repeat (LRR) domain, potentially in conjunction with the pyrin domain. Here, we took advantage of PB1-F2 sequences from IAV strains with either weak or strong NLRP3 interaction. Sequence comparison and structure prediction using Alphafold2 identified a short four amino acid sequence motif (TQGS) in PB1-F2 that defines NLRP3-LRR binding. Conversion of this motif to that of the non-binding PB1-F2 suffices to lose inhibition of NLRP3 dependent IL-1ß release. The TQGS motif further alters the subcellular localization of PB1-F2 and its colocalization with NLRP3 LRR and pyrin domain. Structural predictions suggest the establishment of additional hydrogen bonds between the C-terminus of PB1-F2 and the LRR domain of NLRP3, with two hydrogen bonds connecting to threonine and glutamine of the TQGS motif. Phylogenetic data show that the identified NLRP3 interaction motif in PB1-F2 is widely conserved among recent IAV-infecting humans. Our data explain at a molecular level the specificity of NLRP3 inhibition by influenza A virus. IMPORTANCE: Influenza A virus infection is accompanied by a strong inflammatory response and high fever. The human immune system facilitates the swift clearance of the virus with this response. An essential signal protein in the proinflammatory host response is IL-1b. It is released from inflammatory macrophages, and its production and secretion depend on the function of NLRP3. We had previously shown that influenza A virus blocks NLRP3 activation by the expression of a viral inhibitor, PB1-F2. Here, we demonstrate how this short peptide binds to NLRP3 and provide evidence that a four amino acid stretch in PB1-F2 is necessary and sufficient to mediate this binding. Our data identify a new virus-host interface required to block one signaling path of the innate host response against influenza A virus.


Subject(s)
Influenza A virus , NLR Family, Pyrin Domain-Containing 3 Protein , Viral Proteins , Humans , Amino Acid Motifs , Amino Acid Sequence , HEK293 Cells , Inflammasomes/metabolism , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza, Human/virology , Influenza, Human/immunology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Binding , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry
2.
Int J Mol Sci ; 24(16)2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37629041

ABSTRACT

One of the most prevalent causes of olfactory loss includes traumatic brain injury with subsequent shearing of olfactory axons at the level of the cribriform plate (anterior skull base). Scar tissue at this level may prevent axonal regrowth toward the olfactory bulb. Currently, there is no cure for this debilitating and often permanent condition. One promising therapeutic concept is to implant a synthetic scaffold with growth factors through the cribriform plate/scar tissue to induce neuroregeneration. The first step toward this goal is to investigate the optimum conditions (growth factors, extracellular matrix proteins) to boost this regeneration. However, the lack of a specifically tailored in vitro model and an automated procedure for quantifying axonal length limits our ability to address this issue. The aim of this study is to create an automated quantification tool to measure axonal length and to determine the ideal growth factors and extracellular proteins to enhance axonal regrowth of olfactory sensory neurons in a mouse organotypic 2D model. We harvested olfactory epithelium (OE) of C57BL/6 mice and cultured them during 15 days on coverslips coated with various extracellular matrix proteins (Fibronectin, Collagen IV, Laminin, none) and different growth factors: fibroblast growth factor 2 (FGF2), brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), retinoic acid (RA), transforming growth factor ß (TGFß), and none. We measured the attachment rate on coverslips, the presence of cellular and axonal outgrowth, and finally, the total axonal length with a newly developed automated high-throughput quantification tool. Whereas the coatings did not influence attachment and neuronal outgrowth rates, the total axonal length was enhanced on fibronectin and collagen IV (p = 0.001). The optimum growth factor supplementation media to culture OE compared to the control condition were as follows: FGF2 alone and FGF2 from day 0 to 7 followed by FGF2 in combination with NGF from day 7 to 15 (p < 0.0001). The automated quantification tool to measure axonal length outperformed the standard Neuron J application by reducing the average analysis time from 22 to 3 min per specimen. In conclusion, robust regeneration of murine olfactory neurons in vitro can be induced, controlled, and efficiently measured using an automated quantification tool. These results will help advance the therapeutic concept closer toward preclinical studies.


Subject(s)
Olfactory Receptor Neurons , Animals , Mice , Mice, Inbred C57BL , Fibronectins , Cicatrix , Fibroblast Growth Factor 2/pharmacology , Nerve Growth Factor , Axons , Extracellular Matrix Proteins , Collagen Type IV , Culture Media
3.
Int J Cancer ; 153(9): 1568-1578, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37306359

ABSTRACT

The spatial distribution of tumor-infiltrating lymphocytes (TIL) predicts breast cancer outcome and response to systemic therapy, highlighting the importance of an intact tissue structure for characterizing tumors. Here, we present ST-FFPE, a spatial transcriptomics method for the analysis of formalin-fixed paraffin-embedded samples, which opens the possibility of interrogating archival tissue. The method involves extraction, exome capture and sequencing of RNA from different tumor compartments microdissected by laser-capture, and can be used to study the cellular composition of tumor microenvironment. Focusing on triple-negative breast cancer (TNBC), we characterized T cells, B cells, dendritic cells, fibroblasts and endothelial cells in both stromal and intra-epithelial compartments. We found a highly variable spatial distribution of immune cell subsets among tumors. This analysis revealed that the immune repertoires of intra-epithelial T and B cells were consistently less diverse and more clonal than those of stromal T and B cells. T-cell receptor (TCR) sequencing confirmed a reduced diversity and higher clonality of intra-epithelial T cells relative to the corresponding stromal T cells. Analysis of the top 10 dominant clonotypes in the two compartments showed a majority of shared but also some unique clonotypes both in stromal and intra-epithelial T cells. Hyperexpanded clonotypes were more abundant among intra-epithelial than stromal T cells. These findings validate the ST-FFPE method and suggest an accumulation of antigen-specific T cells within tumor core. Because ST-FFPE is applicable for analysis of previously collected tissue samples, it could be useful for rapid assessment of intratumoral cellular heterogeneity in multiple disease and treatment settings.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Endothelial Cells , Transcriptome , Receptors, Antigen, T-Cell , Gene Expression Profiling , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment/genetics
4.
Biomater Investig Dent ; 10(1): 2282523, 2023.
Article in English | MEDLINE | ID: mdl-38979096

ABSTRACT

Objectives: To compare infiltration, sealing and microleakage in root dentin with a self-conditioning adhesive system combined with dual curing resin (resin-based cement) to a conventional epoxy-resin-based sealer using confocal microscopy imaging. Methods: 26 roots were enlarged and disinfected. Dentin tubules of 24 teeth were labelled with a red fluorophore (Rhodamine B) (two samples served as controls). Root canal samples were sealed in group AH (n = 11) with a conventional sealer (AH Plus Root Canal Sealer, Dentsply DeTrey) and in group RC (n = 11) with a resin-based cement (Parabond combined with Paracore, Coltène). Roots were then sectioned horizontally and immersed in H2O2 to remove the Rhodamine B not fixed by the sealers. The empty dentin spaces were labeled with a green fluorophore (Fluorescein) enabling the evaluation of infiltration as well as microleakage by confocal microscopy. Two additional samples were fractured in vertical direction for observation under SEM. Results: Group RC presented significantly more infiltration in the middle third than in the middle and apical thirds of group AH. Microleakage was significantly higher in group AH than in group RC. SEM images revealed more dentin plugs and a homogenous resin layer in group RC in contrast to group AH. Conclusion: The resin-based cement revealed promising outcomes compared to a traditional epoxy resin based sealer.


Infiltration and microleakage in infiltrated and sealed root dentin samples are higher in middle than apical root thirds. Root dentin infiltration and sealing with a self-conditioning adhesive system and a dual-curing resin cement revealed less microleakage than with an epoxy-resin-based sealer.

5.
Materials (Basel) ; 15(13)2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35806828

ABSTRACT

Angiogenesis and bone regeneration are closely interconnected processes. Whereas type-H blood vessels are abundantly found in the osteogenic zones during endochondral long bone development, their presence in flat bones' development involving intramembranous mechanisms remains unclear. Here, we hypothesized that type-H-like capillaries that highly express CD31 and Endomucin (EMCN), may be present at sites of intramembranous bone development and participate in the control of osteogenesis. A rabbit model of calvarial bone augmentation was used in which bone growth was controlled over time (2-4 weeks) using a particulate bone scaffold. The model allowed the visualization of the entire spectrum of stages throughout bone growth in the same sample, i.e., active ossification, osteogenic activity, and controlled inflammation. Using systematic mRNA hybridization, the formation of capillaries subpopulations (CD31-EMCN staining) over time was studied and correlated with the presence of osteogenic precursors (Osterix staining). Type-H-like capillaries strongly expressing CD31 and EMCN were identified and described. Their presence increased gradually from the regenerative zone up to the osteogenic zone, at 2 and 4 weeks. Type-H-like capillaries may thus represent the initial vascular support encountered in flat bones' development and which organize osteogenic niches.

6.
Sci Adv ; 8(23): eabl5162, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35675399

ABSTRACT

Tumor-associated lymphatic vessels promote metastasis and regulate antitumor immune responses. Here, we assessed the impact of cytotoxic T cells on the local lymphatic vasculature and concomitant tumor dissemination during an antitumor response. Interferon-γ (IFN-γ) released by effector T cells enhanced the expression of immunosuppressive markers by tumor-associated lymphatic endothelial cells (LECs). However, at higher effector T cell densities within the tumor, T cell-based immunotherapies induced LEC apoptosis and decreased tumor lymphatic vessel density. As a consequence, lymphatic flow was impaired, and lymph node metastasis was reduced. Mechanistically, T cell-mediated tumor cell death induced the release of tumor antigens and cross-presentation by tumor LECs, resulting in antigen-specific LEC killing by T cells. When LECs lacked the IFN-γ receptor expression, LEC killing was abrogated, indicating that IFN-γ is indispensable for reducing tumor-associated lymphatic vessel density and drainage. This study provides insight into how cytotoxic T cells modulate tumor lymphatic vessels and may help to improve immunotherapeutic protocols.


Subject(s)
Endothelial Cells , Interferon-gamma , Antigens, Neoplasm , Cross-Priming , Endothelial Cells/metabolism , Humans , Interferon-gamma/metabolism , Lymphatic Metastasis
7.
Cells ; 11(10)2022 05 19.
Article in English | MEDLINE | ID: mdl-35626726

ABSTRACT

Muscle regeneration is essential for proper muscle homeostasis and relies primarily on muscle stem cells (MuSC). MuSC are maintained quiescent in their niche and can be activated following muscle injury. Using an in vitro model of primary human quiescent MuSC (called reserve cells, RC), we analyzed their Ca2+ response following their activation by fetal calf serum and assessed the role of Ca2+ in the processes of RC activation and migration. The results showed that RC displayed a high response heterogeneity in a cell-dependent manner following serum stimulation. Most of these responses relied on inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ release associated with Ca2+ influx, partly due to store-operated calcium entry. Our study further found that blocking the IP3 production, Ca2+ influx, or both did not prevent the activation of RC. Intra- or extracellular Ca2+ chelation did not impede RC activation. However, their migration potential depended on Ca2+ responses displayed upon stimulation, and Ca2+ blockers inhibited their movement. We conclude that the two major steps of muscle regeneration, namely the activation and migration of MuSC, differently rely on Ca2+ signals.


Subject(s)
Calcium Channels , Calcium , Calcium/metabolism , Calcium Channels/metabolism , Humans , Ion Transport , Muscle Fibers, Skeletal/metabolism , Stem Cells/metabolism
8.
Sci Transl Med ; 14(640): eabl6058, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417190

ABSTRACT

In chronic inflammatory diseases of the central nervous system (CNS), immune cells persisting behind the blood-brain barrier are supposed to promulgate local tissue destruction. The drivers of such compartmentalized inflammation remain unclear, but tissue-resident memory T cells (TRM) represent a potentially important cellular player in this process. Here, we investigated whether resting CD8+ TRM persisting after cleared infection with attenuated lymphocytic choriomeningitis virus (LCMV) can initiate immune responses directed against cognate self-antigen in the CNS. We demonstrated that time-delayed conditional expression of the LCMV glycoprotein as neo-self-antigen by glia cells reactivated CD8+ TRM. Subsequently, CD8+ TRM expanded and initiated CNS inflammation and immunopathology in an organ-autonomous manner independently of circulating CD8+ T cells. However, in the absence of CD4+ T cells, TCF-1+ CD8+ TRM failed to expand and differentiate into terminal effectors. Similarly, in human demyelinating CNS autoimmune lesions, we found CD8+ T cells expressing TCF-1 that predominantly exhibited a TRM-like phenotype. Together, our study provides evidence for CD8+ TRM-driven CNS immunopathology and sheds light on why inflammatory processes may evade current immunomodulatory treatments in chronic autoimmune CNS conditions.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Autoantigens , CD4-Positive T-Lymphocytes , Central Nervous System , Humans , Inflammation , Lymphocytic choriomeningitis virus
9.
Neoplasia ; 23(8): 775-782, 2021 08.
Article in English | MEDLINE | ID: mdl-34237504

ABSTRACT

The mechanisms of adaptive resistance to genetic-based targeted therapies of solid malignancies have been the subject of intense research. These studies hold great promise for finding co-targetable hub/pathways which in turn would control the downstream non-genetic mechanisms of adaptive resistance. Many such mechanisms have been described in the paradigmatic BRAF-mutated melanoma model of adaptive response to BRAF inhibition. Currently, a major challenge for these mechanistic studies is to confirm in vivo, at the single-cell proteomic level, the existence of dependencies between the co-targeted hub/pathways and their downstream effectors. Moreover, the drug-induced in vivo modulation of these dependencies needs to be demonstrated. Here, we implement such single-cell-based in vivo expression dependency quantification using immunohistochemistry (IHC)-based analyses of sequential biopsies in two xenograft models. These mimic phase 2 and 3 trials in our own therapeutic strategy to prevent the adaptive response to BRAF inhibition. In this mechanistic model, the dependencies between the targeted Li2CO3-inducible hub HuR and the resistance effectors are more likely time-shifted and transient since the minority of HuRLow cells, which act as a reservoir of adaptive plasticity, switch to a HuRHigh state as they paradoxically proliferate under BRAF inhibition. Nevertheless, we show that a copula/kernel density estimator (KDE)-based quantification of mutual information (MI) efficiently captures, at the individual level, the dependencies between HuR and two relevant resistance markers pERK and EGFR, and outperforms classic expression correlation coefficients. Ultimately, the validation of MI as a predictive IHC-based metric of response to our therapeutic strategy will be carried in clinical trials.


Subject(s)
Melanoma/drug therapy , Models, Theoretical , Molecular Targeted Therapy , Algorithms , Animals , Biomarkers, Tumor , Disease Management , Disease Models, Animal , Disease Susceptibility , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Melanoma/diagnosis , Melanoma/etiology , Melanoma/metabolism , Mice , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Single-Cell Analysis/methods , Treatment Outcome , Xenograft Model Antitumor Assays
10.
J Cell Biol ; 220(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34137788

ABSTRACT

Mutations in the WDR62 gene cause primary microcephaly, a pathological condition often associated with defective cell division that results in severe brain developmental defects. The precise function and localization of WDR62 within the mitotic spindle is, however, still under debate, as it has been proposed to act either at centrosomes or on the mitotic spindle. Here we explored the cellular functions of WDR62 in human epithelial cell lines using both short-term siRNA protein depletions and long-term CRISPR/Cas9 gene knockouts. We demonstrate that WDR62 localizes at spindle poles, promoting the recruitment of the microtubule-severing enzyme katanin. Depletion or loss of WDR62 stabilizes spindle microtubules due to insufficient microtubule minus-end depolymerization but does not affect plus-end microtubule dynamics. During chromosome segregation, WDR62 and katanin promote efficient poleward microtubule flux and favor the synchronicity of poleward movements in anaphase to prevent lagging chromosomes. We speculate that these lagging chromosomes might be linked to developmental defects in primary microcephaly.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation , Microtubules/enzymology , Nerve Tissue Proteins/metabolism , Spindle Poles/enzymology , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , HeLa Cells , Humans , Microcephaly/genetics , Microcephaly/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/genetics , Nerve Tissue Proteins/genetics , Protein Binding , Protein Transport , Signal Transduction , Spindle Poles/genetics , Time Factors
11.
Commun Biol ; 4(1): 380, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33782527

ABSTRACT

The LIM domain-dependent localization of the adapter protein paxillin to ß3 integrin-positive focal adhesions (FAs) is not mechanistically understood. Here, by combining molecular biology, photoactivation and FA-isolation experiments, we demonstrate specific contributions of each LIM domain of paxillin and reveal multiple paxillin interactions in adhesion-complexes. Mutation of ß3 integrin at a putative paxillin binding site (ß3VE/YA) leads to rapidly inward-sliding FAs, correlating with actin retrograde flow and enhanced paxillin dissociation kinetics. Induced mechanical coupling of paxillin to ß3VE/YA integrin arrests the FA-sliding, thereby disclosing an essential structural function of paxillin for the maturation of ß3 integrin/talin clusters. Moreover, bimolecular fluorescence complementation unveils the spatial orientation of the paxillin LIM-array, juxtaposing the positive LIM4 to the plasma membrane and the ß3 integrin-tail, while in vitro binding assays point to LIM1 and/or LIM2 interaction with talin-head domain. These data provide structural insights into the molecular organization of ß3 integrin-FAs.


Subject(s)
Fibroblasts/metabolism , Focal Adhesions/metabolism , Integrin alphaVbeta3/metabolism , Paxillin/metabolism , Animals , Binding Sites , Fluorescence Recovery After Photobleaching , Focal Adhesions/genetics , Integrin alphaVbeta3/genetics , Kinetics , Mice , Microscopy, Confocal , Microscopy, Fluorescence , NIH 3T3 Cells , Paxillin/genetics , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Structure-Activity Relationship
12.
FASEB J ; 34(2): 2625-2640, 2020 02.
Article in English | MEDLINE | ID: mdl-31908048

ABSTRACT

Primary cilia are nonmotile sensory organelles found on the surface of almost all kidney tubule epithelial cells. Being exposed to the tubular lumen, primary cilia are thought to be chemo- and mechanosensors of luminal composition and flux, respectively. We hypothesized that, Na+ transport and primary cilia exist in a sensory functional connection in mature renal tubule epithelial cells. Our results demonstrate that primary cilium length is reduced in mineralocorticoid receptor (MR) knockout (KO) mice in a cell autonomous manner along the aldosterone-sensitive distal nephron (ADSN) compared with wild type (as µm ± SEM; 3.1 ± 0.2 vs 4.0 ± 0.1). In mouse cortical collecting duct (mCCD)cl1 cells, which are a model of collecting duct (CD) principal cells, changes in Na+ transport intensity were found to mediate primary cilium length in response to aldosterone (as µm ± SEM: control: 2.7 ± 0.9 vs aldosterone treated: 3.8 ± 0.8). Cilium length was positively correlated with the availability of IFT88, a major intraflagellar anterograde transport complex B component, which is stabilized in response to exposure to aldosterone treatment. This suggests that the abundance of IFT88 is a regulated, rate limiting factor in the elongation of primary cilia. As previously observed in vivo, aldosterone treatment increased cell volume of cultured CD principal cells. Knockdown of IFT88 prevents ciliogenesis and inhibits the adaptive increase in cell size that was observed in response to aldosterone treatment. In conclusion, our results reveal a functional connection between Na+ transport, primary cilia, and cell size, which may play a key role in the morphological and functional adaptation of the CD to sustained changes in active Na+ reabsorption due to variations in aldosterone secretion.


Subject(s)
Aldosterone/pharmacology , Biological Transport/drug effects , Cilia/drug effects , Epithelial Cells/drug effects , Kidney Tubules, Collecting/drug effects , Aldosterone/metabolism , Animals , Cilia/metabolism , Epithelial Cells/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney Tubules, Collecting/cytology , Mice , Nephrons/drug effects , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/metabolism , Sodium/metabolism
13.
Curr Biol ; 29(21): 3563-3578.e6, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31668617

ABSTRACT

During mitosis, centrosomes affect the length of kinetochore fibers (k-fibers) and the stability of kinetochore-microtubule attachments, implying that they regulate k-fiber dynamics. However, the exact cellular and molecular mechanisms of this regulation remain unknown. Here, we created human cells with only one centrosome to investigate these mechanisms. Such cells formed asymmetric bipolar spindles that resulted in asymmetric cell divisions. K-fibers in the acentrosomal half-spindles were shorter, more stable, and had a reduced poleward microtubule flux at minus ends and more frequent pausing events at their plus ends. This indicates that centrosomes regulate k-fiber dynamics both locally at minus ends and far away at plus ends. At the molecular level, we find that the microtubule-stabilizing protein HURP is enriched on the k-fiber plus ends in the acentrosomal half-spindles of cells with only one centrosome. HURP depletion rebalances k-fiber stability and plus-end dynamics in such cells and improves spindle and cell division symmetry. Our data from 3 different cell lines indicate that HURP accumulates on k-fibers inversely proportionally to half-spindle length. We therefore propose that centrosomes regulate k-fiber plus ends indirectly via length-dependent accumulation of HURP.


Subject(s)
Centrosome/metabolism , Kinetochores/metabolism , Neoplasm Proteins/metabolism , Spindle Apparatus/metabolism , Cell Line , Humans
14.
Biochem Biophys Res Commun ; 517(2): 181-187, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31279529

ABSTRACT

Strategies that aim to limit the adaptive response to pathway inhibition in BRAF-mutated melanoma face the inherent limit of signaling redundancy and multiplicity of possible bypass mechanisms. Drug-induced expression of selected RNA-binding proteins, like the ubiquitously expressed HuR, has the potential to differentially stabilize the expression of many genes involved in the compensatory mechanisms of adaptive response. Here, we detect in BRAF-mutated melanoma cell lines having a higher propensity for adaptive response and in non-responding melanoma tumors, a larger proportion of HuRLow cells in the expression distribution of HuR. Using knockdown experiments, we demonstrate, through expression profiling and phenotypic assays, that increasing the proportion of HuRLow cells favors the adaptive response to BRAF inhibition, provided that the HuRLow state stays reversible. The MAPK dependency of melanoma cells appears to be diminished as the proportion of HuRLow cells increases. In single-cell assays, we demonstrate that the HuRLow cells display plasticity in their growth expression profile. Importantly, the adaptive over-proliferating cells emerge in the subpopulation containing the HuRLow cells. Therapeutic concentrations of lithium salts, although they moderately increase the global expression of HuR, are sufficient to suppress the HuRLow cells, induce an overall less resistant expression profile and attenuate in a HuR-dependent manner the adaptive response of melanoma cells in ex vivo assays. The therapeutic effectiveness of this approach is also demonstrated in vivo in mice xenografts. This study has immediate clinical relevance for melanoma therapy and opens a new avenue of strategies to prevent the adaptive response to targeted cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , ELAV-Like Protein 1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Lithium/pharmacology , Melanoma/genetics , Mice , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Up-Regulation/drug effects
15.
Nat Commun ; 9(1): 2042, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795284

ABSTRACT

Kinetochores are multi-protein complexes that power chromosome movements by tracking microtubules plus-ends in the mitotic spindle. Human kinetochores bind up to 20 microtubules, even though single microtubules can generate sufficient force to move chromosomes. Here, we show that high microtubule occupancy at kinetochores ensures robust chromosome segregation by providing a strong mechanical force that favours segregation of merotelic attachments during anaphase. Using low doses of the microtubules-targeting agent BAL27862 we reduce microtubule occupancy and observe that spindle morphology is unaffected and bi-oriented kinetochores can still oscillate with normal intra-kinetochore distances. Inter-kinetochore stretching is, however, dramatically reduced. The reduction in microtubule occupancy and inter-kinetochore stretching does not delay satisfaction of the spindle assembly checkpoint or induce microtubule detachment via Aurora-B kinase, which was so far thought to release microtubules from kinetochores under low stretching. Rather, partial microtubule occupancy slows down anaphase A and increases incidences of lagging chromosomes due to merotelically attached kinetochores.


Subject(s)
Aurora Kinase B/metabolism , Chromosome Segregation/physiology , Kinetochores/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism , Anaphase/drug effects , Anaphase/physiology , Benzimidazoles/pharmacology , Cell Line , Chromosome Segregation/drug effects , Humans , Intravital Microscopy , Kinetochores/ultrastructure , Microscopy, Electron , Microtubules/ultrastructure , Oxadiazoles/pharmacology , Spindle Apparatus/drug effects
16.
J Cutan Pathol ; 45(5): 333-336, 2018 May.
Article in English | MEDLINE | ID: mdl-29393978

ABSTRACT

BACKGROUND: Prognostic markers for melanoma, particularly for stage II disease, are needed for the risk-benefit evaluation of future adjuvant therapies. The mainly nuclear RNA-binding protein human antigen R (HuR) regulates the protein expression of thousands of mRNAs, its own heterogeneous expression could therefore reflect tumor heterogeneity and plasticity. Here, we evaluate its quantification in primary melanoma as a marker of metastatic outcome. METHODS: We conducted an immunohistochemistry-based automated quantification of HuR nuclear expression heterogeneity in primary melanomas, most with Breslow thickness ≥ 1 mm and calculated the dimensionless fourth moment, that is, the kurtosis of HuR (HuR K) expression distribution. Twelve tumors from patients with no metastatic disease were compared to a similar number of tumors from patients who had metastatic disease at 2 years follow up. RESULTS: HuR K value appeared significantly higher in the non-metastatic group comparatively to the metastatic group (P = 2.84 × 10-3 , 1-tailed Wilcoxon rank-sum test). Moreover, compared to the Breslow thickness, HuR K value appeared as a more robust marker of metastatic outcome (respective areas under receiver operating characteristic curves 0.84 and 0.87). CONCLUSION: Our data need confirmation on a large cohort, however strongly suggest that HuR expression heterogeneity quantification using kurtosis, could be used as a prognostic marker in melanoma.


Subject(s)
Biomarkers, Tumor/analysis , ELAV-Like Protein 1/biosynthesis , Melanoma/pathology , Skin Neoplasms/pathology , ELAV-Like Protein 1/analysis , Humans , Prognosis
17.
Science ; 356(6339)2017 05 19.
Article in English | MEDLINE | ID: mdl-28522470

ABSTRACT

Astrocyte communication is typically studied by two-dimensional calcium ion (Ca2+) imaging, but this method has not yielded conclusive data on the role of astrocytes in synaptic and vascular function. We developed a three-dimensional two-photon imaging approach and studied Ca2+ dynamics in entire astrocyte volumes, including during axon-astrocyte interactions. In both awake mice and brain slices, we found that Ca2+ activity in an individual astrocyte is scattered throughout the cell, largely compartmented between regions, preponderantly local within regions, and heterogeneously distributed regionally and locally. Processes and endfeet displayed frequent fast activity, whereas the soma was infrequently active. In awake mice, activity was higher than in brain slices, particularly in endfeet and processes, and displayed occasional multifocal cellwide events. Astrocytes responded locally to minimal axonal firing with time-correlated Ca2+ spots.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Calcium Signaling , Imaging, Three-Dimensional , Animals , Axons/metabolism , Hippocampus/cytology , Mice , Neuroanatomical Tract-Tracing Techniques , Wakefulness
18.
Cell ; 163(7): 1730-41, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26686654

ABSTRACT

The occurrence of cognitive disturbances upon CNS inflammation or infection has been correlated with increased levels of the cytokine tumor necrosis factor-α (TNFα). To date, however, no specific mechanism via which this cytokine could alter cognitive circuits has been demonstrated. Here, we show that local increase of TNFα in the hippocampal dentate gyrus activates astrocyte TNF receptor type 1 (TNFR1), which in turn triggers an astrocyte-neuron signaling cascade that results in persistent functional modification of hippocampal excitatory synapses. Astrocytic TNFR1 signaling is necessary for the hippocampal synaptic alteration and contextual learning-memory impairment observed in experimental autoimmune encephalitis (EAE), an animal model of multiple sclerosis (MS). This process may contribute to the pathogenesis of cognitive disturbances in MS, as well as in other CNS conditions accompanied by inflammatory states or infections.


Subject(s)
Astrocytes/metabolism , Dentate Gyrus/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Memory , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Learning , Mice , Multiple Sclerosis/physiopathology , Piperidines , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
19.
Nat Rev Neurosci ; 15(5): 327-35, 2014 05.
Article in English | MEDLINE | ID: mdl-24739787

ABSTRACT

Astrocyte Ca(2+) signalling has been proposed to link neuronal information in different spatial-temporal dimensions to achieve a higher level of brain integration. However, some discrepancies in the results of recent studies challenge this view and highlight key insufficiencies in our current understanding. In parallel, new experimental approaches that enable the study of astrocyte physiology at higher spatial-temporal resolution in intact brain preparations are beginning to reveal an unexpected level of compartmentalization and sophistication in astrocytic Ca(2+) dynamics. This newly revealed complexity needs to be attentively considered in order to understand how astrocytes may contribute to brain information processing.


Subject(s)
Astrocytes/physiology , Calcium Signaling/physiology , Calcium/metabolism , Animals , Astrocytes/ultrastructure , Brain/cytology , Humans , Synapses/physiology
20.
Nat Neurosci ; 14(10): 1276-84, 2011 Sep 11.
Article in English | MEDLINE | ID: mdl-21909085

ABSTRACT

Astrocytes communicate with synapses by means of intracellular calcium ([Ca(2+)](i)) elevations, but local calcium dynamics in astrocytic processes have never been thoroughly investigated. By taking advantage of high-resolution two-photon microscopy, we identify the characteristics of local astrocyte calcium activity in the adult mouse hippocampus. Astrocytic processes showed intense activity, triggered by physiological transmission at neighboring synapses. They encoded synchronous synaptic events generated by sparse action potentials into robust regional (∼12 µm) [Ca(2+)](i) elevations. Unexpectedly, they also sensed spontaneous synaptic events, producing highly confined (∼4 µm), fast (millisecond-scale) miniature Ca(2+) responses. This Ca(2+) activity in astrocytic processes is generated through GTP- and inositol-1,4,5-trisphosphate-dependent signaling and is relevant for basal synaptic function. Thus, buffering astrocyte [Ca(2+)](i) or blocking a receptor mediating local astrocyte Ca(2+) signals decreased synaptic transmission reliability in minimal stimulation experiments. These data provide direct evidence that astrocytes are integrated in local synaptic functioning in adult brain.


Subject(s)
Astrocytes/physiology , Calcium Signaling/physiology , Calcium/metabolism , Synapses/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/ultrastructure , Biophysics , Calcium Signaling/drug effects , Cell Communication/drug effects , Cell Communication/physiology , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heparin/pharmacology , Hippocampus/cytology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Patch-Clamp Techniques , Purinergic P2Y Receptor Antagonists/pharmacology , Sodium Channel Blockers/pharmacology , Sucrose/pharmacology , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...