Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Expert Opin Pharmacother ; 25(8): 1051-1069, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935538

ABSTRACT

INTRODUCTION: The treatment landscape of non-small cell lung cancer (NSCLC) has seen significant advancements in recent years, marked by a shift toward target agents and immune checkpoint inhibitors (ICIs). However, chemotherapy remains a cornerstone of treatment, alone or in combination. Microtubule-targeting agents, such as taxanes and vinca alkaloids, play a crucial role in clinical practice in both early and advanced settings in NSCLC. AREA COVERED: This review outlines the mechanisms of action, present significance, and prospective advancements of microtubule-targeting agents (MTAs), with a special highlight on new combinations in phase 3 trials. The online databases PubMed, Web of Science, Cochrane Library, and ClinicalTrials.gov were searched using the terms 'Microtubule-targeting agents' and 'non-small cell lung cancer' or synonyms, with a special focus over the last 5 years of publications. EXPERT OPINION: Despite the emergence of immunotherapy, MTA remains crucial, often used alongside or after immunotherapy, especially in squamous cell lung cancer. Next-generation sequencing expands treatment options, but reliable biomarkers for immunotherapy are lacking. While antibody-drug conjugates (ADCs) show promise, managing toxicities remain vital. In the early stages, MTAs, possibly with ICIs, are standard, while ADCs may replace traditional chemotherapy in the advanced stages. Nevertheless, MTAs remain essential in subsequent lines or for patients with contraindications.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Tubulin Modulators , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Tubulin Modulators/therapeutic use , Antineoplastic Agents/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology
2.
Blood Adv ; 8(8): 1920-1933, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38359376

ABSTRACT

ABSTRACT: This works defines, to the best of our knowledge, for the first time a molecular circuit connecting nicotinamide mononucleoside phosphoribosyl transferase (NAMPT) activity to the B-cell receptor (BCR) pathway. Using 4 distinct xenograft models derived from patients with Richter syndrome (RS-PDX), we show that BCR cross-linking results in transcriptional activation of the nicotinamide adenine dinucleotide (NAD) biosynthetic enzyme NAMPT, with increased protein expression, in turn, positively affecting global cellular NAD levels and sirtuins activity. NAMPT blockade, by using the novel OT-82 inhibitor in combination with either BTK or PI3K inhibitors (BTKi or PI3Ki), induces rapid and potent apoptotic responses in all 4 models, independently of their mutational profile and the expression of the other NAD biosynthetic enzymes, including nicotinate phosphoribosyltransferase. The connecting link in the circuit is represented by AKT that is both tyrosine- and serine-phosphorylated by PI3K and deacetylated by sirtuin 1 and 2 to obtain full kinase activation. Acetylation (ie, inhibition) of AKT after OT-82 administration was shown by 2-dimensional gel electrophoresis and immunoprecipitation. Consistently, pharmacological inhibition or silencing of sirtuin 1 and 2 impairs AKT activation and induces apoptosis of RS cells in combination with PI3Ki or BTKi. Lastly, treatment of RS-PDX mice with the combination of PI3Ki and OT-82 results in significant inhibition of tumor growth, with evidence of in vivo activation of apoptosis. Collectively, these data highlight a novel application for NAMPT inhibitors in combination with BTKi or PI3Ki in aggressive lymphomas.


Subject(s)
Benzamides , Leukemia, Lymphocytic, Chronic, B-Cell , Lymphoma, Large B-Cell, Diffuse , Pyrazoles , Pyridines , Humans , Animals , Mice , NAD/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Nicotinamide Phosphoribosyltransferase
3.
Front Bioeng Biotechnol ; 11: 1297644, 2023.
Article in English | MEDLINE | ID: mdl-38162179

ABSTRACT

Introduction: The secretome of mesenchymal stromal cells (MSCs) serves as an innovative tool employed in the regenerative medicine approach. In this particular context, three-dimensional (3D) culture systems are widely utilized to better replicate in vivo conditions and facilitate prolonged cell maintenance during culture. The use of spheroids enables the preservation of the classical phenotypical characteristics of MSCs. However, the distinct microenvironment within the spheroid may impact the secretome, thereby enhancing the angiogenic properties of adult MSCs that typically possess a reduced angiogenic potential compared to MSCs derived from perinatal tissues due to the hypoxia created in the internal region of the spheroid. Methods: In this study, large spheroids (2,600 cells, ∼300 µm diameter) and small spheroids (1,000 cells, ∼200 µm diameter) were used to examine the role of spheroid diameter in the generation of nutrients and oxygen gradients, cellular senescence, and the angiogenic potential of secreted factors and extracellular vesicles (EVs). Results: In this study, we demonstrate that large spheroids showed increased senescence and a secretome enriched in pro-angiogenic factors, as well as pro-inflammatory and anti-angiogenic cytokines, while small spheroids exhibited decreased senescence and a secretome enriched in pro-angiogenic molecules. We also demonstrated that 3D culture led to a higher secretion of EVs with classical phenotypic characteristics. Soluble factors and EVs from small spheroids exhibited higher angiogenic potential in a human umbilical vein endothelial cell (HUVEC) angiogenic assay. Discussion: These findings highlighted the necessity of choosing the appropriate culture system for obtaining soluble factors and EVs for specific therapeutic applications.

4.
Acta Neuropathol ; 143(4): 453-469, 2022 04.
Article in English | MEDLINE | ID: mdl-35141810

ABSTRACT

The protein α-synuclein, a key player in Parkinson's disease (PD) and other synucleinopathies, exists in different physiological conformations: cytosolic unfolded aggregation-prone monomers and helical aggregation-resistant multimers. It has been shown that familial PD-associated missense mutations within the α-synuclein gene destabilize the conformer equilibrium of physiologic α-synuclein in favor of unfolded monomers. Here, we characterized the relative levels of unfolded and helical forms of cytosolic α-synuclein in post-mortem human brain tissue and showed that the equilibrium of α-synuclein conformations is destabilized in sporadic PD and DLB patients. This disturbed equilibrium is decreased in a brain region-specific manner in patient samples pointing toward a possible "prion-like" propagation of the underlying pathology and forms distinct disease-specific patterns in the two different synucleinopathies. We are also able to show that a destabilization of multimers mechanistically leads to increased levels of insoluble, pathological α-synuclein, while pharmacological stabilization of multimers leads to a "prion-like" aggregation resistance. Together, our findings suggest that these disease-specific patterns of α-synuclein multimer destabilization in sporadic PD and DLB are caused by both regional neuronal vulnerability and "prion-like" aggregation transmission enabled by the destabilization of local endogenous α-synuclein protein.


Subject(s)
Lewy Body Disease , Parkinson Disease , Prions , Synucleinopathies , Brain/pathology , Humans , Lewy Bodies/pathology , Lewy Body Disease/pathology , Parkinson Disease/pathology , Prions/metabolism , alpha-Synuclein/metabolism
5.
Cell Rep ; 36(1): 109333, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233191

ABSTRACT

While misfolding of alpha-synuclein (αSyn) is central to the pathogenesis of Parkinson's disease (PD), fundamental questions about its structure and function at the synapse remain unanswered. We examine synaptosomes from non-transgenic and transgenic mice expressing wild-type human αSyn, the E46K fPD-causing mutation, or an amplified form of E46K ("3K"). Synaptosomes from mice expressing the 3K mutant show reduced Ca2+-dependent vesicle exocytosis, altered synaptic vesicle ultrastructure, decreased SNARE complexes, and abnormal levels of certain synaptic proteins. With our intra-synaptosomal nuclear magnetic resonance (NMR) method, we reveal that WT αSyn participates in heterogeneous interactions with synaptic components dependent on endogenous αSyn and synaptosomal integrity. The 3K mutation markedly alters these interactions. The synaptic microenvironment is necessary for αSyn to reach its native conformations and establish a physiological interaction network. Its inability to populate diverse conformational ensembles likely represents an early step in αSyn dysfunction that contributes to the synaptotoxicity observed in synucleinopathies.


Subject(s)
Parkinson Disease/metabolism , Parkinson Disease/pathology , Synaptic Vesicles/pathology , Synaptosomes/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Exocytosis , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Biological , Protein Conformation , Protein Folding , Protein Multimerization , Recombinant Proteins/metabolism , SNARE Proteins/metabolism , Solubility , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomes/ultrastructure
6.
Brain Commun ; 2(1): fcaa010, 2020.
Article in English | MEDLINE | ID: mdl-32280944

ABSTRACT

Since researchers identified α-synuclein as the principal component of Lewy bodies and Lewy neurites, studies have suggested that it plays a causative role in the pathogenesis of dementia with Lewy bodies and other 'synucleinopathies'. While α-synuclein dyshomeostasis likely contributes to the neurodegeneration associated with the synucleinopathies, few direct biochemical analyses of α-synuclein from diseased human brain tissue currently exist. In this study, we analysed sequential protein extracts from a substantial number of patients with neuropathological diagnoses of dementia with Lewy bodies and corresponding controls, detecting a shift of cytosolic and membrane-bound physiological α-synuclein to highly aggregated forms. We then fractionated aqueous extracts (cytosol) from cerebral cortex using non-denaturing methods to search for soluble, disease-associated high molecular weight species potentially associated with toxicity. We applied these fractions and corresponding insoluble fractions containing Lewy-type aggregates to several reporter assays to determine their bioactivity and cytotoxicity. Ultimately, high molecular weight cytosolic fractions enhances phospholipid membrane permeability, while insoluble, Lewy-associated fractions induced morphological changes in the neurites of human stem cell-derived neurons. While the concentrations of soluble, high molecular weight α-synuclein were only slightly elevated in brains of dementia with Lewy bodies patients compared to healthy, age-matched controls, these observations suggest that a small subset of soluble α-synuclein aggregates in the brain may drive early pathogenic effects, while Lewy body-associated α-synuclein can drive neurotoxicity.

7.
Alzheimers Dement ; 15(9): 1183-1194, 2019 09.
Article in English | MEDLINE | ID: mdl-31416794

ABSTRACT

INTRODUCTION: There is keen interest in elucidating the biological mechanisms underlying recent failures of ß-site amyloid precursor protein-cleaving enzyme-1 (BACE1) inhibitors in Alzheimer's disease trials. METHODS: We developed a highly sensitive and specific immunoassay for BACE1 in cell lines and iPSC-derived human neurons to systematically analyze the effects of eight clinically relevant BACE1 inhibitors. RESULTS: Seven of 8 inhibitors elevated BACE1 protein levels. Among protease inhibitors tested, the elevation was specific to BACE1 inhibitors. The inhibitors did not increase BACE1 transcription but extended the protein's half-life. BACE1 became elevated at concentrations below the IC50 for amyloid ß (Aß). DISCUSSION: Elevation of BACE1 by 7 of 8 BACE1 inhibitors raises new concerns about advancing such ß-secretase inhibitors for AD. Chronic elevation could lead to intermittently uninhibited BACE1 when orally dosed inhibitors reach trough levels, abnormally increasing substrate processing. Compounds such as roburic acid that lower Aß by dissociating ß/γ secretase complexes are better candidates because they neither inhibit ß- and γ-secretase nor increase BACE1 levels.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Imidazoles , Neurons/metabolism , Spiro Compounds , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Culture Techniques , Enzyme-Linked Immunosorbent Assay , Half-Life , Humans , Mice
8.
J Biol Chem ; 294(25): 9799-9812, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31048377

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative disorders, and both genetic and histopathological evidence have implicated the ubiquitous presynaptic protein α-synuclein (αSyn) in its pathogenesis. Recent work has investigated how disrupting αSyn's interaction with membranes triggers trafficking defects, cellular stress, and apoptosis. Special interest has been devoted to a series of mutants exacerbating the effects of the E46K mutation (associated with autosomal dominant PD) through homologous Glu-to-Lys substitutions in αSyn's N-terminal region (i.e. E35K and E61K). Such E46K-like mutants have been shown to cause dopaminergic neuron loss and severe but L-DOPA-responsive motor defects in mouse overexpression models, presenting enormous translational potential for PD and other "synucleinopathies." In this work, using a variety of biophysical techniques, we characterize the molecular pathology of E46K-like αSyn mutants by studying their structure and membrane-binding and remodeling abilities. We find that, although a slight increase in the mutants' avidity for synaptic vesicle-like membranes can be detected, most of their deleterious effects are connected to their complete disruption of αSyn's curvature selectivity. Indiscriminate binding can shift αSyn's subcellular localization away from its physiological interactants at the synaptic bouton toward trafficking vesicles and organelles, as observed in E46K-like cellular and murine models, as well as in human pathology. In conclusion, our findings suggest that a loss of curvature selectivity, rather than increased membrane affinity, could be the critical dyshomeostasis in synucleinopathies.


Subject(s)
Cell Membrane/pathology , Glutamic Acid/chemistry , Lipids/analysis , Lysine/chemistry , Mutant Proteins/metabolism , Mutation , alpha-Synuclein/metabolism , Cell Membrane/metabolism , Glutamic Acid/genetics , Humans , Lipids/chemistry , Lysine/genetics , Mutant Proteins/genetics , alpha-Synuclein/genetics
9.
Methods Mol Biol ; 1948: 123-143, 2019.
Article in English | MEDLINE | ID: mdl-30771175

ABSTRACT

α-Synuclein's physiology and pathology have been linked by numerous reports to its ability to bind and remodel membranes, especially at synaptic terminals. It is therefore critical for researchers investigating the determinants of these interactions to rely on methods capable of providing an accurate and complete physicochemical snapshot of the binding events. Circular dichroism (CD) and isothermal titration calorimetry (ITC) are established techniques for the study of binding equilibria in biological systems and, especially when used in combination, allow a thorough characterization of the protein-lipid interplay.Here we provide general guidelines and describe some common pitfalls of these experiments. This protocol describes the preparation of small unilamellar vesicles (SUVs), mimicking the curved bilayers α-synuclein normally interacts with, the CD-monitored titration of α-synuclein with SUVs, the ITC (lipid-into-protein) experiment, and the subsequent data analysis using an n independent binding site model.


Subject(s)
Calorimetry , Cell Membrane/chemistry , Circular Dichroism , Temperature , alpha-Synuclein/chemistry , Cell Membrane/metabolism , Data Analysis , Lipid Bilayers/chemistry , Liposomes/chemistry , Protein Binding , alpha-Synuclein/metabolism
10.
J Cell Biol ; 218(2): 644-663, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30626721

ABSTRACT

Intramembrane proteolysis of transmembrane substrates by the presenilin-γ-secretase complex is preceded and regulated by shedding of the substrate's ectodomain by α- or ß-secretase. We asked whether ß- and γ-secretases interact to mediate efficient sequential processing of APP, generating the amyloid ß (Aß) peptides that initiate Alzheimer's disease. We describe a hitherto unrecognized multiprotease complex containing active ß- and γ-secretases. BACE1 coimmunoprecipitated and cofractionated with γ-secretase in cultured cells and in mouse and human brain. An endogenous high molecular weight (HMW) complex (∼5 MD) containing ß- and γ-secretases and holo-APP was catalytically active in vitro and generated a full array of Aß peptides, with physiological Aß42/40 ratios. The isolated complex responded properly to γ-secretase modulators. Alzheimer's-causing mutations in presenilin altered the Aß42/40 peptide ratio generated by the HMW ß/γ-secretase complex indistinguishably from that observed in whole cells. Thus, Aß is generated from holo-APP by a BACE1-γ-secretase complex that provides sequential, efficient RIP processing of full-length substrates to final products.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain/enzymology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Brain/drug effects , Enzyme Inhibitors/pharmacology , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/enzymology , Male , Mice, Inbred C57BL , Multienzyme Complexes , Mutation , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Binding , Proteolysis , Substrate Specificity
11.
PLoS One ; 13(7): e0198715, 2018.
Article in English | MEDLINE | ID: mdl-29995905

ABSTRACT

N-terminal acetylation is one of the most common co- and post-translational modifications of the eukaryotic proteome and regulates numerous aspects of cellular physiology, such as protein folding, localization and turnover. In particular α-synuclein, whose dyshomeostasis has been tied to the pathogenesis of several neurodegenerative disorders, is completely Nα-acetylated in nervous tissue. In this work, building on previous reports, we develop and characterize a bacterial N-terminal acetylation system based on the expression of the yeast N-terminal acetyltransferase B (NatB) complex under the control of the PBAD (L-arabinose-inducible) promoter. We show its functionality and the ability to completely Nα-acetylate our model substrate α-synuclein both upon induction of the construct with L-arabinose and also by only relying on the constitutive expression of the NatB genes.


Subject(s)
Acetyltransferases/genetics , Escherichia coli/genetics , Genetic Vectors/chemistry , Plasmids/chemistry , Protein Processing, Post-Translational , alpha-Synuclein/genetics , Acetylation/drug effects , Acetyltransferases/metabolism , Amino Acid Sequence , Arabinose/pharmacology , Cloning, Molecular , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/metabolism , Humans , Peptide Fragments/analysis , Plasmids/metabolism , Promoter Regions, Genetic/drug effects , Proteolysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , alpha-Synuclein/metabolism
12.
FEBS Lett ; 592(9): 1464-1472, 2018 05.
Article in English | MEDLINE | ID: mdl-29633780

ABSTRACT

α-Synuclein (αSyn) is a key player in the pathogenesis of Parkinson's disease and other synucleinopathies. Here, we report the existence of a novel soluble α-helical conformer of αSyn, obtained through transient interaction with lipid interfaces, and propose dynamic oligomerization as the mechanism underlying its stability. The conformational space of αSyn appears to be highly context-dependent, and lipid bilayers might thus play crucial roles as molecular chaperones in a cellular environment.


Subject(s)
Lipid Metabolism , Protein Refolding , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Solubility
13.
Front Mol Neurosci ; 10: 210, 2017.
Article in English | MEDLINE | ID: mdl-28713242

ABSTRACT

The microtubule (MT)-associated protein Tau is a natively unfolded protein, involved in a number of neurodegenerative disorders, collectively called tauopathies, aggregating in neurofibrillary tangles (NFT). It is an open question how the conversion from a MT bound molecule to an aggregation-prone Tau species occurs and, also, if and how tauopathy-related mutations affect its behavior in the cell. To address these points, we exploited a genetically encoded FRET sensor based on the full length Tau protein, to monitor in real time Tau conformational changes in different conditions in live cells. By studying the FRET signal we found that soluble Tau molecules, detached from MTs, display an unfolded structure. On the contrary, we observed an increased FRET signal generated by Tau monomers bound to MT, indicating that the association with MTs induced a folding of Tau protein, decreasing the distance between its N and C termini. We exploited the FRET sensor to investigate the impact of FTDP-17 mutations and of phosphorylation-site mutations on Tau folding and mobility in live cells. We demonstrated that the FTDP-17 Tau mutations weaken the interaction of Tau with cellular MTs, shifting the equilibrium towards the soluble pool while, conversely, phosphorylation site mutations shift the equilibrium of Tau towards the MT-bound state and a more closed conformation.

SELECTION OF CITATIONS
SEARCH DETAIL