Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 42(8): 2673-2695, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34415465

ABSTRACT

The chiral molecule, apomorphine, is currently used for the treatment of Parkinson's disease (PD). As a potent dopamine receptor agonist, this lipophilic compound is especially effective for treating motor fluctuations in advanced PD patients. In addition to its receptor-mediated actions, apomorphine has also antioxidant and free radical scavenger activities. Neuroinflammation, oxidative stress, and microglia reactivity have emerged as central players in PD. Thus, modulating microglia activation in PD may be a valid therapeutic strategy. We previously reported that murine microglia are strongly activated upon exposure to A53T mutant α-synuclein. The present study was designed to investigate whether apomorphine enantiomers could modulate this A53T-induced microglial activation. Taken together, the results provided evidence that apomorphine enantiomers decrease A53T-induced microgliosis, through the activation of the NRF2 signalling pathway, leading to a lower pro-inflammatory state and restoring the phagocytic activity. Suppressing NRF2 recruitment (trigonelline exposure) or silencing specifically Nfe2l2 gene (siRNA treatment) abolished or strongly decreased the anti-inflammatory activity of apomorphine. In conclusion, apomorphine, which is already used in PD patients to mimic dopamine activity, may also be suitable to decrease α-synuclein-induced microglial reactivity.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Antioxidants/pharmacology , Apomorphine/metabolism , Apomorphine/pharmacology , Dopamine/metabolism , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Free Radical Scavengers/pharmacology , Humans , Mice , Microglia/metabolism , NF-E2-Related Factor 2/metabolism , Parkinson Disease/metabolism , RNA, Small Interfering/metabolism , alpha-Synuclein/metabolism
2.
Hemodial Int ; 2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33709483

ABSTRACT

INTRODUCTION: Fluid management is an important goal of dialysis treatment. The accurate assessment of fluid status is still a challenge for clinical nephrologists. Bioimpedance analysis (BIA) has been proposed as an objective tool to assess hydration. METHODS: This was a prospective randomized controlled study to compare hydration status measured by clinical assessment compared to BIA using a body composition monitor (BCM). The primary outcome was defined as the decline of cardiac biomarker N-terminal pro brain natriuretic peptide (NT-proBNP) from baseline to the end of the study. FINDINGS: About 281 chronic hemodialysis patients were assessed for eligibility, and 132 patients provided written informed consent to participate (65 BIA group, 67 clinical group). Predialytic NT-proBNP, and decline of NT-proBNP were similar in both groups. The amount of overhydration (2.18 ± 2.11 L vs. 1.29 ± 1.97 L; p 0.016) and the number of patients with severe overhydration (46.0% vs. 30.6%, p = 0.04) were significantly higher in the BIA group at the end of the study. Fluid accumulation in the interdialytic period was significantly lower in the clinical group (p = 0.013). Adverse events occurred more often in the BIA group (p = 0.032). The cumulative number of hypovolemic events was significantly higher in the BIA group (p = 0.002). DISCUSSION: Fluid management by BIA does not lead to a better cardiac outcome (appraised by surrogate markers) than fluid management by careful clinical assessment. Adapting the dry weight according to BIA results increases the risk of adverse events, especially hypovolemic episodes. Careful clinical fluid assessment is important for optimal care of chronic hemodialysis patients.

3.
Cancer Lett ; 450: 32-41, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30790680

ABSTRACT

Cancer stem cells, also known as tumor-initiating cells (TICs), are a population of aggressive and self-renewing cells that are responsible for the initiation and progression of many cancers, including colorectal carcinoma. Intratumoral hypoxia, i.e. reduced oxygen supply following uncontrolled proliferation of cancer cells, is thought to support TIC activity by inducing specific hypoxia-responsive mechanisms that are not yet entirely understood. Using previously established and fully characterized patient-derived TIC cultures, we could observe increased sphere and colony formation under hypoxic conditions. Mechanistically, microRNA (miRNA)-profiling experiments allowed us to identify miR-215 as one of the main hypoxia-induced miRNAs in primary colon TICs. Through stable overexpression of miR-215, followed by a set of functional in vitro and in vivo investigations, miR-215 was pinpointed as a negative feedback regulator, working against the TIC-promoting effects of hypoxia. Furthermore, we could single out LGR5, a bona fide marker of non-neoplastic intestinal stem cells, as a downstream target of hypoxia/miR-215 signaling. The strong tumor- and TIC-suppressor potential of miR-215 and the regulatory role of the hypoxia/miR-215/LGR5 axis may thus represent interesting points of attack for the development of innovative anti-CSC therapy approaches.


Subject(s)
Cell Hypoxia/physiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Colonic Neoplasms/genetics , Genes, Tumor Suppressor , Heterografts , Humans , Mice , Mice, Inbred NOD , MicroRNAs/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Spheroids, Cellular , Tumor Cells, Cultured , Up-Regulation
4.
J Exp Clin Cancer Res ; 38(1): 56, 2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30728057

ABSTRACT

BACKGROUND: Melanoma is the most aggressive and deadly form of skin cancer with increasing case numbers worldwide. The development of inhibitors targeting mutated BRAF (found in around 60% of melanoma patients) has markedly improved overall survival of patients with late-stage tumors, even more so when combined with MEK inhibitors targeting the same signaling pathway. However, invariably patients become resistant to this targeted therapy resulting in rapid progression with treatment-refractory disease. The purpose of this study was the identification of new kinase inhibitors that do not lead to the development of resistance in combination with BRAF inhibitors (BRAFi), or that could be of clinical benefit as a 2nd line treatment for late-stage melanoma patients that have already developed resistance. METHODS: We have screened a 274-compound kinase inhibitor library in 3 BRAF mutant melanoma cell lines (each one sensitive or made resistant to 2 distinct BRAFi). The screening results were validated by dose-response studies and confirmed the killing efficacies of many kinase inhibitors. Two different tools were applied to investigate and quantify potential synergistic effects of drug combinations: the Chou-Talalay method and the Synergyfinder application. In order to exclude that resistance to the new treatments might occur at later time points, synergistic combinations were administered to fluorescently labelled parental and resistant cells over a period of > 10 weeks. RESULTS: Eight inhibitors targeting Wee1, Checkpoint kinase 1/2, Aurora kinase, MEK, Polo-like kinase, PI3K and Focal adhesion kinase killed melanoma cells synergistically when combined with a BRAFi. Additionally, combination of a Wee1 and Chk inhibitor showed synergistic killing effects not only on sensitive cell lines, but also on intrinsically BRAFi- and treatment induced-resistant melanoma cells. First in vivo studies confirmed these observations. Interestingly, continuous treatment with several of these drugs, alone or in combination, did not lead to emergence of resistance. CONCLUSIONS: Here, we have identified new, previously unexplored (in the framework of BRAFi resistance) inhibitors that have an effect not only on sensitive but also on BRAFi-resistant cells. These promising combinations together with the new immunotherapies could be an important step towards improved 1st and 2nd line treatments for late-stage melanoma patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Melanoma/physiopathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Small Molecule Libraries
5.
Mol Cancer ; 17(1): 145, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30290811

ABSTRACT

BACKGROUND: Drug resistance remains an unsolved clinical issue in oncology. Despite promising initial responses obtained with BRAF and MEK kinase inhibitors, resistance to treatment develops within months in virtually all melanoma patients. METHODS: Microarray analyses were performed in BRAF inhibitor-sensitive and resistant cell lines to identify changes in the transcriptome that might play a role in resistance. siRNA approaches and kinase inhibitors were used to assess the involvement of the identified Anaplastic Lymphoma Kinase (ALK) in drug resistance. The capability of extracellular vesicles (EVs) to transfer drug resistant properties was investigated in co-culture assays. RESULTS: Here, we report a new mechanism of acquired drug resistance involving the activation of a novel truncated form of ALK. Knock down or inhibition of ALK re-sensitised resistant cells to BRAF inhibition and induced apoptosis. Interestingly, truncated ALK was also secreted into EVs and we show that EVs were the vehicle for transferring drug resistance. CONCLUSIONS: To our knowledge, this is the first report demonstrating the functional involvement of EVs in melanoma drug resistance by transporting a truncated but functional form of ALK, able to activate the MAPK signalling pathway in target cells. Combined inhibition of ALK and BRAF dramatically reduced tumour growth in vivo. These findings make ALK a promising clinical target in melanoma patients.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , Melanoma/metabolism , Anaplastic Lymphoma Kinase/genetics , Animals , Biological Transport , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isoenzymes , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
6.
Cancer Res ; 78(14): 3793-3808, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29748374

ABSTRACT

The vast majority of colorectal cancer-related deaths can be attributed to metastatic spreading of the disease. Therefore, deciphering molecular mechanisms of metastatic dissemination is a key prerequisite to improve future treatment options. With this aim, we took advantage of different colorectal cancer cell lines and recently established primary cultures enriched in colon cancer stem cells, also known as tumor-initiating cells (TIC), to identify genes and miRNAs with regulatory functions in colorectal cancer progression. We show here that metastasis-derived TICs display increased capacity for self-renewal, TGFß signaling activity, and reduced expression of the miR-371∼373 cluster compared with nonmetastatic cultures. TGFß receptor 2 (TGFBR2) and aldehyde dehydrogenase A1 (ALDH1A1) were identified as important target genes of the miR-371∼373 cluster. In addition, TGFBR2 repression, either by direct knockdown or indirectly via overexpression of the entire miR-371∼373 cluster, decreased tumor-initiating potential of TICs. We observed significantly reduced in vitro self-renewal activity as well as lowered tumor initiation and metastatic outgrowth capacity in vivo following stable overexpression of the miR-371∼373 cluster in different colon TIC cultures. Inhibitor of DNA binding 1 (ID1) was affected by both TGFBR2 and miR-371∼373 cluster alterations. Functional sphere and tumor formation as well as metastatic dissemination assays validated the link between miR-371∼373 and ID1. Altogether, our results establish the miR-371∼373/TGFBR2/ID1 signaling axis as a novel regulatory mechanism of TIC self-renewal and metastatic colonization.Significance: These findings establish the miR-371∼373/TGFBR2/ID1 signaling axis as a novel mechanism regulating self-renewal of tumor-initiating cell and metastatic colonization, potentially opening new concepts for therapeutic targeting of cancer metastasis.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3793/F1.large.jpg Cancer Res; 78(14); 3793-808. ©2018 AACR.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Inhibitor of Differentiation Protein 1/genetics , MicroRNAs/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction/genetics , Animals , Cell Line, Tumor , Cell Self Renewal/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology
7.
PLoS One ; 11(9): e0162717, 2016.
Article in English | MEDLINE | ID: mdl-27622765

ABSTRACT

Parkinson's disease (PD) is histologically described by the deposition of α-synuclein, whose accumulation in Lewy bodies causes dopaminergic neuronal death. Although most of PD cases are sporadic, point mutations of the gene encoding the α-synuclein protein cause inherited forms of PD. There are currently six known point mutations that result in familial PD. Oxidative stress and neuroinflammation have also been described as early events associated with dopaminergic neuronal degeneration in PD. Though it is known that microglia are activated by wild-type α-synuclein, little is known about its mutated forms and the signaling cascades responsible for this microglial activation. The present study was designed to investigate consequences of wild-type and mutant α-synuclein (A53T, A30P and E46K) exposure on microglial reactivity. Interestingly, we described that α-synuclein-induced microglial reactivity appeared to be peptide-dependent. Indeed, the A53T protein activated more strongly microglia than the wild-type α-synuclein and other mutants. This A53T-induced microglial reactivity mechanism was found to depend on phosphorylation mechanisms mediated by MAPKs and on successive NFkB/AP-1/Nrf2 pathways activation. These results suggest that the microgliosis intensity during PD might depend on the type of α-synuclein protein implicated. Indeed, mutated forms are more potent microglial stimulators than wild-type α-synuclein. Based on these data, anti-inflammatory and antioxidant therapeutic strategies may be valid in order to reduce microgliosis but also to subsequently slow down PD progression, especially in familial cases.


Subject(s)
Microglia/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Gene Expression , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Microglia/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Point Mutation , Reactive Oxygen Species/metabolism , Signal Transduction
8.
PLoS One ; 10(6): e0130624, 2015.
Article in English | MEDLINE | ID: mdl-26091541

ABSTRACT

Neuroinflammation is the local reaction of the brain to infection, trauma, toxic molecules or protein aggregates. The brain resident macrophages, microglia, are able to trigger an appropriate response involving secretion of cytokines and chemokines, resulting in the activation of astrocytes and recruitment of peripheral immune cells. IL-1ß plays an important role in this response; yet its production and mode of action in the brain are not fully understood and its precise implication in neurodegenerative diseases needs further characterization. Our results indicate that the capacity to form a functional NLRP3 inflammasome and secretion of IL-1ß is limited to the microglial compartment in the mouse brain. We were not able to observe IL-1ß secretion from astrocytes, nor do they express all NLRP3 inflammasome components. Microglia were able to produce IL-1ß in response to different classical inflammasome activators, such as ATP, Nigericin or Alum. Similarly, microglia secreted IL-18 and IL-1α, two other inflammasome-linked pro-inflammatory factors. Cell stimulation with α-synuclein, a neurodegenerative disease-related peptide, did not result in the release of active IL-1ß by microglia, despite a weak pro-inflammatory effect. Amyloid-ß peptides were able to activate the NLRP3 inflammasome in microglia and IL-1ß secretion occurred in a P2X7 receptor-independent manner. Thus microglia-dependent inflammasome activation can play an important role in the brain and especially in neuroinflammatory conditions.


Subject(s)
Brain/cytology , Carrier Proteins/metabolism , Inflammasomes/metabolism , Microglia/metabolism , Amyloid beta-Peptides/toxicity , Animals , Astrocytes/metabolism , Carrier Proteins/genetics , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Interleukin-18/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/analysis , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Peptide Fragments/toxicity , Receptors, Purinergic P2X7/metabolism , alpha-Synuclein/pharmacology
9.
J Neurochem ; 114(2): 576-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20456016

ABSTRACT

Alzheimer's disease (AD) is characterized by the presence of extracellular deposits referred to beta-amyloid (Abeta) complexes or senile plaques. Abeta peptide is firstly produced as monomers, readily aggregating to form multimeric complexes, of which the smallest aggregates are known to be the most neurotoxic. In AD patients, abundant reactive microglia migrate to and surround the Abeta plaques. Though it is well known that microglia are activated by Abeta, little is known about the peptide conformation and the signaling cascades responsible for this activation. In this study, we have stimulated murine microglia with different Abeta(1-42) forms, inducing an inflammatory state, which was peptide conformation-dependent. The lightest oligomeric forms induced a more violent inflammatory response, whereas the heaviest oligomers and the fibrillar conformation were less potent inducers. BocMLF, a formylpeptide chemotactic receptor 2 antagonist, decreased the oligomeric Abeta-induced inflammatory response. The Abeta-induced signal transduction was found to depend on phosphorylation mechanisms mediated by MAPKs and on activator protein 1/nuclear factor kappa-light-chain-enhancer of activated B cells pathways activation. These results suggest that the reactive microgliosis intensity during AD might depend on the disease progression and consequently on the Abeta conformation production. The recognition of Abeta by the formylpeptide chemotactic receptor 2 seems to be a starting point of the signaling cascade inducing an inflammatory state.


Subject(s)
Amyloid beta-Peptides/chemistry , Microglia/physiology , Peptide Fragments/chemistry , Receptors, Formyl Peptide/physiology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/physiology , Animals , Biopolymers , Cell Line , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Microglia/immunology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Peptide Fragments/immunology , Peptide Fragments/physiology , Phosphorylation , Protein Conformation , Signal Transduction , Transcription Factor AP-1/physiology
10.
Glia ; 57(16): 1741-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19455581

ABSTRACT

The Notch pathway is implicated in many aspects of the central nervous system (CNS) development and functions. Recently, we and others identified the Notch pathway to be involved in inflammatory events of the CNS. To understand the implication of this pathway on astrocytes, we have studied the Jagged-Notch-Hes pathway under inflammatory conditions. LPS exposure induced an upregulation of Jagged1 expression on cultured astrocytes. To address the role of Jagged1 in the modulation of inflammation, we used a siRNA mediated silencing of Jagged1 (siRNA J1). Jagged1 inhibition induced important variations on the Notch pathway components like Hes1, Hes5, Notch3, and RBP-Jkappa. siRNA J1 repressed the mRNA expression of genes known as hallmarks of the gliosis like GFAP and endothelin(B) receptor. On activated astrocytes, the inhibition of Jagged1 had antiinflammatory effects and resulted in a decrease of LPS-induced proinflammatory cytokines (IL1beta, IL1alpha, and TNFalpha) as well as the iNOS expression. The inhibition of Jagged1 induced a modulation of the JAK/STAT/SOCS signaling pathway. Most interestingly, the siRNA J1 decreased the LPS-induced translocation of NFkappaB p65 and this could be correlated to the phosphorylation of IkappaBalpha. These results suggest that during inflammatory and gliotic events of the CNS, Jagged1/Notch signaling sustains the inflammation mainly through NFkappaB and in part through JAK/STAT/SOCS signaling pathways.


Subject(s)
Astrocytes/metabolism , Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Janus Kinases/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Astrocytes/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Silencing , Gliosis/metabolism , Homeodomain Proteins/metabolism , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , Membrane Proteins/genetics , Nitric Oxide/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor HES-1 , Transfection , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...