Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
ACS Nano ; 18(26): 16505-16515, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38875527

ABSTRACT

Cyclic oligoadenylates (cOAs) are small second messenger molecules produced by the type III CRISPR-Cas system as part of the prokaryotic immune response. The role of cOAs is to allosterically activate downstream effector proteins that induce dormancy or cell death, and thus abort viral spread through the population. Interestingly, different type III systems have been reported to utilize different cOA stoichiometries (with 3 to 6 adenylate monophosphates). However, so far, their characterization has only been possible in bulk and with sophisticated equipment, while a portable assay with single-molecule resolution has been lacking. Here, we demonstrate the label-free detection of single cOA molecules using a simple protein nanopore assay. It sensitively identifies the stoichiometry of individual cOA molecules and their mixtures from synthetic and enzymatic origin. To achieve this, we trained a convolutional neural network (CNN) and validated it with a series of experiments on mono- and polydisperse cOA samples. Ultimately, we determined the stoichiometric composition of cOAs produced enzymatically by the CRISPR type III-A and III-B variants of Thermus thermophilus and confirmed the results by liquid chromatography-mass spectroscopy (LC-MS). Interestingly, both variants produce cOAs of nearly identical composition (within experimental uncertainties), and we discuss the biological implications of this finding. The presented nanopore-CNN workflow with single cOA resolution can be adapted to many other signaling molecules (including eukaryotic ones), and it may be integrated into portable handheld devices with potential point-of-care applications.


Subject(s)
CRISPR-Cas Systems , Nanopores , CRISPR-Cas Systems/genetics
2.
Cell Death Dis ; 15(4): 296, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670941

ABSTRACT

Genes of the Sprouty family (Spry1-4) restrain signaling by certain receptor tyrosine kinases. Consequently, these genes participate in several developmental processes and function as tumor suppressors in adult life. Despite these important roles, the biology of this family of genes still remains obscure. Here we show that Sprouty proteins are general mediators of cellular senescence. Induction of cellular senescence by several triggers in vitro correlates with upregulation of Sprouty protein levels. More importantly, overexpression of Sprouty genes is sufficient to cause premature cellular senescence, via a conserved N-terminal tyrosine (Tyrosine 53 of Sprouty1). Accordingly, fibroblasts from knockin animals lacking that tyrosine escape replicative senescence. In vivo, heterozygous knockin mice display delayed induction of cellular senescence during cutaneous wound healing and upon chemotherapy-induced cellular senescence. Unlike other functions of this family of genes, induction of cellular senescence appears to be independent of activation of the ERK1/2 pathway. Instead, we show that Sprouty proteins induce cellular senescence upstream of the p38 pathway in these in vitro and in vivo paradigms.


Subject(s)
Cellular Senescence , Fibroblasts , Membrane Proteins , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Fibroblasts/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Wound Healing
4.
Mol Oncol ; 17(1): 98-118, 2023 01.
Article in English | MEDLINE | ID: mdl-36409196

ABSTRACT

Uterine serous carcinoma (USC) is an aggressive form of endometrial cancer (EC), characterized by its high propensity for metastases. In fact, while endometrioid endometrial carcinoma (EEC), which accounts for 85% of EC, presents a good prognosis, USC is the most frequently fatal. Herein, we used for the first time a peptide-based tyrosine-kinase-activity profiling approach to quantify the changes in tyrosine kinase activation between USC and EEC. Among the tyrosine kinases highly activated in USC, we identified focal adhesion kinase (FAK). We conducted mechanistic studies using cellular models. In a USC cell line, targeting FAK either by inhibitors PF-573228 and defactinib (VS-6063) or by gene silencing limits 3D cell growth and reduces cell migration. Moreover, results from our studies suggest that oxidative stress is increased in USC tumors compared to EEC ones. Reactive oxygen species (ROS) induce tyrosine phosphorylation of FAK and a concomitant tyrosine phosphorylation of paxillin, a mediator of FAK signal transduction. Mechanistically, by tracking hundreds of individual cells per condition, we show that ROS increased cell distance and migration velocity, highlighting the role of ROS-FAK-PAX signaling in cell migration. Both defactinib and ROS scavenger N-acetylcysteine (NAC) revert this effect, pointing toward ROS as potential culprits for the increase in USC cell motility. A proof of concept of the role of FAK in controlling cell growth was obtained in in vivo experiments using cancer-tissue-originated spheroids (CTOS) and a patient-derived orthotopic xenograft model (orthoxenograft/PDOX). Defactinib reduces cell proliferation and protein oxidation, supporting a pro-tumoral antioxidant role of FAK, whereas antioxidant NAC reverts FAK inhibitor effects. Overall, our data points to ROS-mediated FAK activation in USC as being responsible for the poor prognosis of this tumor type and emphasize the potential of FAK inhibition for USC treatment.


Subject(s)
Antioxidants , Cystadenocarcinoma, Serous , Focal Adhesion Kinase 1 , Humans , Antioxidants/metabolism , Cell Line, Tumor , Cell Movement , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , Focal Adhesion Kinase 1/metabolism , Oxidative Stress , Phosphorylation , Reactive Oxygen Species , Tyrosine/metabolism , Animals
5.
Adv Sci (Weinh) ; : e2204211, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36373729

ABSTRACT

Clinical management of endometrial cancer (EC) is handicapped by the limited availability of second line treatments and bona fide molecular biomarkers to predict recurrence. These limitations have hampered the treatment of these patients, whose survival rates have not improved over the last four decades. The advent of coordinated studies such as The Cancer Genome Atlas Uterine Corpus Endometrial Carcinoma (TCGA_UCEC) has partially solved this issue, but the lack of proper experimental systems still represents a bottleneck that precludes translational studies from successful clinical testing in EC patients. Within this context, the first study reporting the generation of a collection of endometrioid-EC-patient-derived orthoxenograft (PDOX) mouse models is presented that is believed to overcome these experimental constraints and pave the way toward state-of-the-art precision medicine in EC. The collection of primary tumors and derived PDOXs is characterized through an integrative approach based on transcriptomics, mutational profiles, and morphological analysis; and it is demonstrated that EC tumors engrafted in the mouse uterus retain the main molecular and morphological features from analogous tumor donors. Finally, the molecular properties of these tumors are harnessed to assess the therapeutic potential of trastuzumab, a human epidermal growth factor receptor 2 (HER2) inhibitor with growing interest in EC, using patient-derived organotypic multicellular tumor spheroids and in vivo experiments.

6.
J Exp Clin Cancer Res ; 41(1): 285, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36163066

ABSTRACT

BACKGROUND: Gasdermin B (GSDMB) over-expression promotes poor prognosis and aggressive behavior in HER2 breast cancer by increasing resistance to therapy. Decoding the molecular mechanism of GSDMB-mediated drug resistance is crucial to identify novel effective targeted treatments for HER2/GSDMB aggressive tumors. METHODS: Different in vitro approaches (immunoblot, qRT-PCR, flow cytometry, proteomic analysis, immunoprecipitation, and confocal/electron microscopy) were performed in HER2 breast and gastroesophageal carcinoma cell models. Results were then validated using in vivo preclinical animal models and analyzing human breast and gastric cancer samples. RESULTS: GSDMB up-regulation renders HER2 cancer cells more resistant to anti-HER2 agents by promoting protective autophagy. Accordingly, the combination of lapatinib with the autophagy inhibitor chloroquine increases the therapeutic response of GSDMB-positive cancers in vitro and in zebrafish and mice tumor xenograft in vivo models. Mechanistically, GSDMB N-terminal domain interacts with the key components of the autophagy machinery LC3B and Rab7, facilitating the Rab7 activation during pro-survival autophagy in response to anti-HER2 therapies. Finally, we validated these results in clinical samples where GSDMB/Rab7/LC3B co-expression associates significantly with relapse in HER2 breast and gastric cancers. CONCLUSION: Our findings uncover for the first time a functional link between GSDMB over-expression and protective autophagy in response to HER2-targeted therapies. GSDMB behaves like an autophagy adaptor and plays a pivotal role in modulating autophagosome maturation through Rab7 activation. Finally, our results provide a new and accessible therapeutic approach for HER2/GSDMB + cancers with adverse clinical outcome.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Animals , Autophagy , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Chloroquine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Lapatinib/pharmacology , Mice , Neoplasm Recurrence, Local , Proteomics , Receptor, ErbB-2/genetics , Zebrafish
7.
Mol Cancer ; 21(1): 175, 2022 09 03.
Article in English | MEDLINE | ID: mdl-36057593

ABSTRACT

BACKGROUND: Epigenetic programming during development is essential for determining cell lineages, and alterations in this programming contribute to the initiation of embryonal tumour development. In neuroblastoma, neural crest progenitors block their course of natural differentiation into sympathoadrenergic cells, leading to the development of aggressive and metastatic paediatric cancer. Research of the epigenetic regulators responsible for oncogenic epigenomic networks is crucial for developing new epigenetic-based therapies against these tumours. Mammalian switch/sucrose non-fermenting (mSWI/SNF) ATP-dependent chromatin remodelling complexes act genome-wide translating epigenetic signals into open chromatin states. The present study aimed to understand the contribution of mSWI/SNF to the oncogenic epigenomes of neuroblastoma and its potential as a therapeutic target. METHODS: Functional characterisation of the mSWI/SNF complexes was performed in neuroblastoma cells using proteomic approaches, loss-of-function experiments, transcriptome and chromatin accessibility analyses, and in vitro and in vivo assays. RESULTS: Neuroblastoma cells contain three main mSWI/SNF subtypes, but only BRG1-associated factor (BAF) complex disruption through silencing of its key structural subunits, ARID1A and ARID1B, impairs cell proliferation by promoting cell cycle blockade. Genome-wide chromatin remodelling and transcriptomic analyses revealed that BAF disruption results in the epigenetic repression of an extensive invasiveness-related expression program involving integrins, cadherins, and key mesenchymal regulators, thereby reducing adhesion to the extracellular matrix and the subsequent invasion in vitro and drastically inhibiting the initiation and growth of neuroblastoma metastasis in vivo. CONCLUSIONS: We report a novel ATPase-independent role for the BAF complex in maintaining an epigenomic program that allows neuroblastoma invasiveness and metastasis, urging for the development of new BAF pharmacological structural disruptors for therapeutic exploitation in metastatic neuroblastoma.


Subject(s)
Chromatin , Neuroblastoma , Animals , Child , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Epigenomics , Humans , Mammals/metabolism , Neuroblastoma/genetics , Proteomics
8.
JCI Insight ; 7(14)2022 07 22.
Article in English | MEDLINE | ID: mdl-35737463

ABSTRACT

The ribosomal protein S6 kinase 1 (S6K1) is a relevant effector downstream of the mammalian target of rapamycin complex 1 (mTORC1), best known for its role in the control of lipid homeostasis. Consistent with this, mice lacking the S6k1 gene have a defect in their ability to induce the commitment of fat precursor cells to the adipogenic lineage, which contributes to a significant reduction of fat mass. Here, we assess the therapeutic blockage of S6K1 in diet-induced obese mice challenged with LY2584702 tosylate, a specific oral S6K1 inhibitor initially developed for the treatment of solid tumors. We show that diminished S6K1 activity hampers fat mass expansion and ameliorates dyslipidemia and hepatic steatosis, while modifying transcriptome-wide gene expression programs relevant for adipose and liver function. Accordingly, decreased mTORC1 signaling in fat (but increased in the liver) segregated with defective epithelial-mesenchymal transition and the impaired expression of Cd36 (coding for a fatty acid translocase) and Lgals1 (Galectin 1) in both tissues. All these factors combined align with reduced adipocyte size and improved lipidomic signatures in the liver, while hepatic steatosis and hypertriglyceridemia were improved in treatments lasting either 3 months or 6 weeks.


Subject(s)
Fatty Liver , TOR Serine-Threonine Kinases , Animals , Diet , Fatty Liver/drug therapy , Fatty Liver/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
Cancers (Basel) ; 14(12)2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35740505

ABSTRACT

Metabolomic profiling analysis has the potential to highlight new molecules and cellular pathways that may serve as potential therapeutic targets for disease treatment. In this study, we used an LC-MS/MS platform to define, for the first time, the specific metabolomic signature of uterine serous carcinoma (SC), a relatively rare and aggressive variant of endometrial cancer (EC) responsible for 40% of all endometrial cancer-related deaths. A metabolomic analysis of 31 ECs (20 endometrial endometrioid carcinomas (EECs) and 11 SCs) was performed. Following multivariate statistical analysis, we identified 232 statistically different metabolites among the SC and EEC patient samples. Notably, most of the metabolites identified (89.2%) were lipid species and showed lower levels in SCs when compared to EECs. In addition to lipids, we also documented metabolites belonging to amino acids and purine nucleotides (such as 2-Oxo-4-methylthiobutanoic acid, synthesised by acireductone dioxygenase 1 (ADI1) enzyme), which showed higher levels in SCs. To further investigate the role of ADI1 in SC, we analysed the expression protein levels of ADI1 in 96 ECs (67 EECs and 29 SCs), proving that the levels of ADI1 were higher in SCs compared to EECs. We also found that ADI1 mRNA levels were higher in p53 abnormal ECs compared to p53 wild type tumours. Furthermore, elevated ADI1 mRNA levels showed a statistically significant negative correlation with overall survival and progression-free survival among EEC patients. Finally, we tested the ability of ADI1 to induce migration and invasion capabilities in EC cell lines. Altogether, these results suggest that ADI1 could be a potential therapeutic target in poor-prognosis SCs and other Ecs with abnormal p53 expression.

10.
Mol Oncol ; 16(11): 2235-2259, 2022 06.
Article in English | MEDLINE | ID: mdl-35167193

ABSTRACT

AT-rich interactive domain-containing protein 1A (ARID1A) loss-of-function mutation accompanied by a loss of ARID1A protein expression is frequently observed in endometrial carcinomas. However, the molecular mechanisms linking these genetic changes to the altered pathways regulating tumour initiation, maintenance and/or progression remain poorly understood. Thus, the main aim of this study was to analyse the role of ARID1A loss of function in endometrial tumorigenesis. Here, using different endometrial in vitro and in vivo models, such as tumoral cell lines, 3D primary cultures and metastatic or genetically modified mouse models, we show that altered expression of ARID1A is not enough to initiate endometrial tumorigenesis. However, in an established endometrial cancer context, ARID1A loss of function accelerates tumoral progression and metastasis through the disruption of the G2/M cell cycle checkpoint and ATM/ATR-mediated DNA damage checkpoints, increases epithelial cell proliferation rates and induces epithelial mesenchymal transition through the activation of histone deacetylase 6 (HDAC6). Next, we demonstrated that the inhibition of HDAC6 function, using the HDAC6-specific inhibitor ACY1215 or by transfection with HDAC6 short hairpin RNA (shRNA), can reverse the migratory and invasive phenotype of ARID1A-knockdown cells. Further, we also show that inhibition of HDAC6 activity causes an apoptotic vulnerability to etoposide treatments in ARID1A-deficient cells. In summary, the findings exposed in this work indicate that the inhibition of HDAC6 activity is a potential therapeutic strategy for patients suffering from ARID1A-mutant endometrial cancer diagnosed in advanced stages.


Subject(s)
Endometrial Neoplasms , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition , Female , Histone Deacetylase 6/genetics , Humans , Mice , Transcription Factors/genetics
11.
Adv Sci (Weinh) ; 9(4): e2104759, 2022 02.
Article in English | MEDLINE | ID: mdl-34898027

ABSTRACT

The H19X-encoded miR-424(322)/503 cluster regulates multiple cellular functions. Here, it is reported for the first time that it is also a critical linchpin of fat mass expansion. Deletion of this miRNA cluster in mice results in obesity, while increasing the pool of early adipocyte progenitors and hypertrophied adipocytes. Complementary loss and gain of function experiments and RNA sequencing demonstrate that miR-424(322)/503 regulates a conserved genetic program involved in the differentiation and commitment of white adipocytes. Mechanistically, it is demonstrated that miR-424(322)/503 targets γ-Synuclein (SNCG), a factor that mediates this program rearrangement by controlling metabolic functions in fat cells, allowing adipocyte differentiation and adipose tissue enlargement. Accordingly, diminished miR-424(322) in mice and obese humans co-segregate with increased SNCG in fat and peripheral blood as mutually exclusive features of obesity, being normalized upon weight loss. The data unveil a previously unknown regulatory mechanism of fat mass expansion tightly controlled by the miR-424(322)/503 through SNCG.


Subject(s)
Adipose Tissue/metabolism , Cell Differentiation , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , gamma-Synuclein/metabolism , Adipogenesis , Animals , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Proteins/genetics , gamma-Synuclein/genetics
12.
Nanomaterials (Basel) ; 11(12)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34947779

ABSTRACT

Nanopatterning to fabricate advanced nanostructured materials is a widely employed technology in a broad spectrum of applications going from spintronics and nanoelectronics to nanophotonics. This work reports on an easy route for nanopatterning making use of ordered porous templates with geometries ranging from straight lines to square, triangular or rhombohedral lattices, to be employed for the designed growth of sputtered materials with engineered properties. The procedure is based on large-scale nanoimprinting using patterned low-cost commercial disks, as 1-D grating stamps, followed by a single electrochemical process that allows one to obtain 1-D ordered porous anodic templates. Multiple imprinting steps at different angles enable more complex 2-D patterned templates. Subsequently, sputtering facilitates the growth of ferromagnetic antidot thin films (e.g., from 20 to 100 nm Co thick layers) with designed symmetries. This technique constitutes a non-expensive method for massive mold production and pattern generation avoiding standard lithographical techniques. In addition, it overcomes current challenges of the two-stage electrochemical porous anodic alumina templates: (i) allowing the patterning of large areas with high ordering and/or complex antidot geometries, and (ii) being less-time consuming.

13.
EMBO Rep ; 22(12): e53201, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34633138

ABSTRACT

During the female lifetime, the expansion of the epithelium dictated by the ovarian cycles is supported by a transient increase in the mammary epithelial stem cell population (MaSCs). Notably, activation of Wnt/ß-catenin signaling is an important trigger for MaSC expansion. Here, we report that the miR-424/503 cluster is a modulator of canonical Wnt signaling in the mammary epithelium. We show that mammary tumors of miR-424(322)/503-depleted mice exhibit activated Wnt/ß-catenin signaling. Importantly, we show a strong association between miR-424/503 deletion and breast cancers with high levels of Wnt/ß-catenin signaling. Moreover, miR-424/503 cluster is required for Wnt-mediated MaSC expansion induced by the ovarian cycles. Lastly, we show that miR-424/503 exerts its function by targeting two binding sites at the 3'UTR of the LRP6 co-receptor and reducing its expression. These results unveil an unknown link between the miR-424/503, regulation of Wnt signaling, MaSC fate, and tumorigenesis.


Subject(s)
Epithelium , Low Density Lipoprotein Receptor-Related Protein-6 , Mammary Glands, Animal/cytology , MicroRNAs , Wnt Signaling Pathway , Animals , Breast Neoplasms , Carcinogenesis , Cell Line, Tumor , Epithelial Cells/cytology , Epithelium/metabolism , Female , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Menstrual Cycle , Mice , MicroRNAs/genetics , Stem Cells/cytology
14.
Cancers (Basel) ; 13(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638328

ABSTRACT

Neuroblastoma is a pediatric tumor of the peripheral nervous system that accounts for up to ~15% of all cancer-related deaths in children. Recently, it has become evident that epigenetic deregulation is a relevant event in pediatric tumors such as high-risk neuroblastomas, and a determinant for processes, such as cell differentiation blockade and sustained proliferation, which promote tumor progression and resistance to current therapies. Thus, a better understanding of epigenetic factors implicated in the aggressive behavior of neuroblastoma cells is crucial for the development of better treatments. In this study, we characterized the role of ZRF1, an epigenetic activator recruited to genes involved in the maintenance of the identity of neural progenitors. We combined analysis of patient sample expression datasets with loss- and gain-of-function studies on neuroblastoma cell lines. Functional analyses revealed that ZRF1 is functionally dispensable for those cellular functions related to cell differentiation, proliferation, migration, and invasion, and does not affect the cellular response to chemotherapeutic agents. However, we found that high levels of ZRF1 mRNA expression are associated to shorter overall survival of neuroblastoma patients, even when those patients with the most common molecular alterations used as prognostic factors are removed from the analyses, thereby suggesting that ZRF1 expression could be used as an independent prognostic factor in neuroblastoma.

15.
Nanomaterials (Basel) ; 11(10)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34685176

ABSTRACT

Multi-segmented bilayered Fe/Cu nanowires have been fabricated through the electrodeposition in porous anodic alumina membranes. We have assessed, with the support of micromagnetic simulations, the dependence of fabricated nanostructures' magnetic properties either on the number of Fe/Cu bilayers or on the length of the magnetic layers, by fixing both the nonmagnetic segment length and the wire diameter. The magnetic reversal, in the segmented Fe nanowires (NWs) with a 300 nm length, occurs through the nucleation and propagation of a vortex domain wall (V-DW) from the extremities of each segment. By increasing the number of bilayers, the coercive field progressively increases due to the small magnetostatic coupling between Fe segments, but the coercivity found in an Fe continuous nanowire is not reached, since the interactions between layers is limited by the Cu separation. On the other hand, Fe segments 30 nm in length have exhibited a vortex configuration, with around 60% of the magnetization pointing parallel to the wires' long axis, which is equivalent to an isolated Fe nanodisc. By increasing the Fe segment length, a magnetic reversal occurred through the nucleation and propagation of a V-DW from the extremities of each segment, similar to what happens in a long cylindrical Fe nanowire. The particular case of the Fe/Cu bilayered nanowires with Fe segments 20 nm in length revealed a magnetization oriented in opposite directions, forming a synthetic antiferromagnetic system with coercivity and remanence values close to zero.

17.
Sci Rep ; 11(1): 43, 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33420134

ABSTRACT

CoFeB-based ultrathin films with perpendicular magnetic anisotropy are promising for different emerging technological applications such as nonvolatile memories with low power consumption and high-speed performance. In this work, the dynamical properties of [CoFeB (tCoFeB)/Pd (10 Å)]5 multilayered ultrathin films (1 Å ≤ tCoFeB ≤ 5 Å) are studied by using two complementary methods: time-resolved magneto-optical Kerr effect and broadband ferromagnetic resonance. The perpendicular magnetization is confirmed for multilayers with tCoFeB ≤ 4 Å. The effective perpendicular magnetic anisotropy reaches a clear maximum at tCoFeB = 3 Å. Further increase of CoFeB layer thickness reduces the perpendicular magnetic anisotropy and the magnetization became in-plane oriented for tCoFeB ≥ 5 Å. This behaviour is explained by considering competing contributions from surface and magnetoelastic anisotropies. It was also found that the effective damping parameter αeff decreases with CoFeB layer thickness and for tCoFeB = 4 Å reaches a value of ~ 0.019 that is suitable for microwave applications.

18.
Autophagy ; 17(5): 1077-1095, 2021 05.
Article in English | MEDLINE | ID: mdl-32401642

ABSTRACT

Autophagy is a highly conserved catabolic process and a major cellular pathway for the degradation of long-lived proteins and cytoplasmic organelles. An increasing body of evidence has unveiled autophagy as an indispensable biological function that helps to maintain normal tissue homeostasis and metabolic fitness that can also lead to severe consequences for the normal cellular functioning when altered. Recent accumulating data point to autophagy as a key player in a wide variety of physiological and pathophysiological conditions in the human endometrium, one of the most proficient self-regenerating tissues in the human body and an instrumental player in placental species reproductive function. The current review highlights the most recent findings regarding the process of autophagy in the normal and cancerous endometrial tissue. Current research efforts aiming to therapeutically exploit autophagy and the methodological approaches used are discussed.Abbreviations: 3-MA: 3-methyladenine; ACACA (acetyl-CoA carboxylase alpha); AICAR: 5-aminoimidazole-4-carboximide riboside; AKT: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; ATG3: autophagy related 3; ATG4C: autophagy related 4C cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG9: autophagy related 9; Baf A1: bafilomycin A1; BAX: BCL2 associated X, apoptosis regulator; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; CACNA1D: calcium voltage-gated channel subunit alpha1 D; CASP3: caspase 3; CASP7: caspase 7; CASP8: caspase 8; CASP9: caspase 9; CD44: CD44 molecule (Indian blood group); CDH1: cadherin 1; CDKN1A: cyclin dependent kinase inhibitor 1A; CDKN2A: cyclin dependent kinase inhibitor 2A; CMA: chaperone-mediated autophagy; CQ: chloroquine; CTNNB1: catenin beta 1; DDIT3: DNA damage inducible transcript 3; EC: endometrial cancer; EGFR: epidermal growth factor receptor; EH: endometrial hyperplasia; EIF4E: eukaryotic translation initiation factor 4E; EPHB2/ERK: EPH receptor B2; ER: endoplasmic reticulum; ERBB2: er-b2 receptor tyrosine kinase 2; ERVW-1: endogenous retrovirus group W member 1, envelope; ESR1: estrogen receptor 1; FSH: follicle-stimulating hormone; GCG/GLP1: glucagon; GFP: green fluorescent protein; GIP: gastric inhibitory polypeptide; GLP1R: glucagon-like peptide-1 receptor; GLS: glutaminase; H2AX: H2A.X variant histone; HIF1A: hypoxia inducible factor 1 alpha; HMGB1: high mobility group box 1; HOTAIR: HOX transcript antisense RNA; HSPA5: heat shock protein family A (HSP70) member 5; HSPA8: heat shock protein family A (HSP70) member 8; IGF1: insulin like growth factor 1; IL27: interleukin 27; INS: insulin; ISL: isoliquiritigenin; KRAS: KRAS proto-oncogene, GTPase; LAMP2: lysosomal-associated membrane protein 2; lncRNA: long-non-coding RNA; MAP1LC3A/LC3A: microtubule associated protein 1 light chain 3 alpha; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK8: mitogen-activated protein kinase 8; MAPK9: mitogen-activated protein kinase 9; MPA: medroxyprogesterone acetate; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; MYCBP: MYC-binding protein; NFE2L2: nuclear factor, erythroid 2 like 2; NFKB: nuclear factor kappa B; NFKBIA: NFKB inhibitor alpha; NK: natural killer; NR5A1: nuclear receptor subfamily 5 group A member 1; PARP1: poly(ADP-ribose) polymerase 1; PAX2: paired box 2; PDK1: pyruvate dehydrogenase kinase 1; PDX: patient-derived xenograft; PIK3C3/Vps34: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3CA: phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PIK3R1: phosphoinositide-3-kinase regulatory subunit 1; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PPD: protopanaxadiol; PRKCD: protein kinase C delta; PROM1/CD133: prominin 1; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PTEN: phosphatase and tensin homolog; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RFP: red fluorescent protein; RPS6KB1/S6K1: ribosomal protein S6 kinase B1; RSV: resveratrol; SGK1: serum/glucocorticoid regulated kinase 1; SGK3: serum/glucocorticoid regulated kinase family member 3; SIRT: sirtuin; SLS: stone-like structures; SMAD2: SMAD family member 2; SMAD3: SMAD family member 3; SQSTM1: sequestosome 1; TALEN: transcription activator-like effector nuclease; TGFBR2: transforming growth factor beta receptor 2; TP53: tumor protein p53; TRIB3: tribbles pseudokinase 3; ULK1: unc-51 like autophagy activating kinase 1; ULK4: unc-51 like kinase 4; VEGFA: vascular endothelial growth factor A; WIPI2: WD repeat domain, phosphoinositide interacting 2; XBP1: X-box binding protein 1; ZFYVE1: zinc finger FYVE domain containing 1.


Subject(s)
Autophagy/physiology , Endometrium/metabolism , Neoplasms/metabolism , Placenta/metabolism , Apoptosis Regulatory Proteins/metabolism , Female , Humans , Hyperplasia/metabolism , Pregnancy
19.
Nanoscale ; 12(41): 21207-21217, 2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33057527

ABSTRACT

The increasing demand for nanoscale magnetic devices requires development of 3D magnetic nanostructures. In this regard, focused electron beam induced deposition (FEBID) is a technique of choice for direct-writing of complex nano-architectures with applications in nanomagnetism, magnon spintronics, and superconducting electronics. However, intrinsic properties of nanomagnets are often poorly known and can hardly be assessed by local optical probe techniques. Here, an original spatially resolved approach is demonstrated for spin-wave spectroscopy of individual circular magnetic elements with sample volumes down to about 10-3 µm3. The key component of the setup is a coplanar waveguide whose microsized central part is placed over a movable substrate with well-separated CoFe-FEBID nanodisks which exhibit standing spin-wave resonances. The circular symmetry of the disks allows for the deduction of the saturation magnetization and the exchange stiffness of the material using an analytical theory. A good correspondence between the results of analytical calculations and micromagnetic simulations is revealed, indicating a validity of the used analytical model going beyond the initial thin-disk approximation used in the theoretical derivation. The presented approach is especially valuable for the characterization of direct-write magnetic elements opening new horizons for 3D nanomagnetism and magnonics.

20.
ACS Appl Mater Interfaces ; 11(19): 17654-17662, 2019 May 15.
Article in English | MEDLINE | ID: mdl-31007012

ABSTRACT

Local modification of magnetic properties of nanoelements is a key to design future-generation magnonic devices in which information is carried and processed via spin waves. One of the biggest challenges here is to fabricate simple and miniature phase-controlling elements with broad tunability. Here, we successfully realize such spin-wave phase shifters upon a single nanogroove milled by a focused ion beam in a Co-Fe microsized magnonic waveguide. By varying the groove depth and the in-plane bias magnetic field, we continuously tune the spin-wave phase and experimentally evidence a complete phase inversion. The microscopic mechanism of the phase shift is based on the combined action of the nanogroove as a geometrical defect and the lower spin-wave group velocity in the waveguide under the groove where the magnetization is reduced due to the incorporation of Ga ions during the ion-beam milling. The proposed phase shifter can easily be on-chip integrated with spin-wave logic gates and other magnonic devices. Our findings are crucial for designing nanomagnonic circuits and for the development of spin-wave nano-optics.

SELECTION OF CITATIONS
SEARCH DETAIL
...