Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 227
Filtrar
1.
J Phys Chem B ; 128(15): 3621-3630, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38578255

RESUMEN

8-Thiomethyladenine (ASCH3), a potentially radiosensitizing modified nucleobase, has been synthesized in a reaction between 8-thioadenine and methyl iodide. Despite favorable dissociative electron attachment (DEA) characteristics, the radiolysis of an aqueous solution of ASCH3 with a dose of X-ray amounting to as much as 300 Gy leads to no effects. Nevertheless, crossed electron-molecule beam experiments in the gas phase on ASCH3 confirm the theoretical findings regarding the stability of its radical anion, namely, the most abundant reaction channel is related to the dissociation of the S-CH3 bond in the respective anion. Furthermore, electron-induced degradation of ASCH3 has been observed in aprotic acetonitrile, which is strong evidence for the involvement of proton transfer (PT) in stabilizing the radical anion in an aqueous solution. These findings demonstrate that PT in water can be the main player in deciding the radiosensitizing properties of modified nucleobases/nucleosides.

2.
Nat Commun ; 15(1): 2865, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570528

RESUMEN

Targeting neovascularization in glioblastoma (GBM) is hampered by poor understanding of the underlying mechanisms and unclear linkages to tumour molecular landscapes. Here we report that different molecular subtypes of human glioma stem cells (GSC) trigger distinct endothelial responses involving either angiogenic or circumferential vascular growth (vasectasia). The latter process is selectively triggered by mesenchymal (but not proneural) GSCs and is mediated by a subset of extracellular vesicles (EVs) able to transfer EGFR/EGFRvIII transcript to endothelial cells. Inhibition of the expression and phosphorylation of EGFR in endothelial cells, either pharmacologically (Dacomitinib) or genetically (gene editing), abolishes their EV responses in vitro and disrupts vasectasia in vivo. Therapeutic inhibition of EGFR markedly extends anticancer effects of VEGF blockade in mice, coupled with abrogation of vasectasia and prolonged survival. Thus, vasectasia driven by intercellular transfer of oncogenic EGFR may represent a new therapeutic target in a subset of GBMs.


Asunto(s)
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Glioma , Humanos , Animales , Ratones , Células Endoteliales/metabolismo , Glioma/metabolismo , Glioblastoma/metabolismo , Receptores ErbB/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Encefálicas/metabolismo
4.
J Enzyme Inhib Med Chem ; 39(1): 2290910, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38093611

RESUMEN

In the present work, we report a new series of potent SARS-CoV-2 Main Protease (Mpro) inhibitors based on maleimide derivatives. The inhibitory activities were tested in an enzymatic assay using recombinant Mpro (3CL Protease from coronavirus SARS-CoV-2). Within the set of new Mpro inhibitors, 6e demonstrated the highest activity in the enzymatic assay with an IC50 value of 8.52 ± 0.44 µM. The IC50 value for Nirmatrelvir (PF-07321332, used as a reference) was 0.84 ± 0.37 µM. The cytotoxic properties were determined in the MTT assay using MRC-5 and HEK-293 cell lines. In the course of the investigation, we found that the newly obtained maleimide derivatives are not substantially cytotoxic (IC50 values for most compounds were above 200 µM).


Asunto(s)
COVID-19 , Humanos , Células HEK293 , SARS-CoV-2 , Maleimidas/farmacología , Lactamas , Leucina , Nitrilos , Inhibidores de Proteasas/farmacología , Simulación del Acoplamiento Molecular , Antivirales/farmacología
5.
Adv Sci (Weinh) ; 10(34): e2304389, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37867228

RESUMEN

Efficient and targeted delivery of therapeutic agents remains a bottleneck in modern medicine. Here, biochemical engineering approaches to advance the repurposing of extracellular vesicles (EVs) as drug delivery vehicles are explored. Targeting ligands such as the sugar GalNAc are displayed on the surface of EVs using a HaloTag-fused to a protein anchor that is enriched on engineered EVs. These EVs are successfully targeted to human primary hepatocytes. In addition, the authors are able to decorate EVs with an antibody that recognizes a GLP1 cell surface receptor by using an Fc and Fab region binding moiety fused to an anchor protein, and they show that this improves EV targeting to cells that overexpress the receptor. The authors also use two different protein-engineering approaches to improve the loading of Cre recombinase into the EV lumen and demonstrate that functional Cre protein is delivered into cells in the presence of chloroquine, an endosomal escape enhancer. Lastly, engineered EVs are well tolerated upon intravenous injection into mice without detectable signs of liver toxicity. Collectively, the data show that EVs can be engineered to improve cargo loading and specific cell targeting, which will aid their transformation into tailored drug delivery vehicles.


Asunto(s)
Vesículas Extracelulares , Ratones , Animales , Humanos , Ligandos , Vesículas Extracelulares/metabolismo , Proteínas/metabolismo , Sistemas de Liberación de Medicamentos , Comunicación Celular
6.
J Phys Chem Lett ; 14(40): 8948-8955, 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37769041

RESUMEN

In the search for effective radiosensitizers for tumor cells, halogenated uracils have attracted more attention due to their large cross section for dissociation upon the attachment of low-energy electrons. In this study, we investigated dissociative electron attachment (DEA) to 5-iodo-4-thio-2'-deoxyuridine, a potential radiosensitizer using a crossed electron-molecule beam experiment coupled with quadrupole mass spectrometry. The experimental results were supported by calculations on the threshold energies of formed anions and transition state calculations. We show that low-energy electrons with kinetic energies near 0 eV may effectively decompose the molecule upon DEA. The by far most abundant anion observed corresponds to the iodine anion (I-). Due to the associated bond cleavage, a radical site is formed at the C5 position, which may initiate strand break formation if the molecule is incorporated into a DNA strand. Our results reflect the conclusion from previous radiolysis studies with the title compound, suggesting its potential as a radiosensitizer.


Asunto(s)
Electrones , Fármacos Sensibilizantes a Radiaciones , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Tiouridina , Aniones
7.
ACS Nano ; 17(13): 12052-12071, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37366177

RESUMEN

Extracellular vesicles (EVs) are continually released from cancer cells into biofluids, carrying actionable molecular fingerprints of the underlying disease with considerable diagnostic and therapeutic potential. The scarcity, heterogeneity and intrinsic complexity of tumor EVs present a major technological challenge in real-time monitoring of complex cancers such as glioblastoma (GBM). Surface-enhanced Raman spectroscopy (SERS) outputs a label-free spectroscopic fingerprint for EV molecular profiling. However, it has not been exploited to detect known biomarkers at the single EV level. We developed a multiplex fluidic device with embedded arrayed nanocavity microchips (MoSERS microchip) that achieves 97% confinement of single EVs in a minute amount of fluid (<10 µL) and enables molecular profiling of single EVs with SERS. The nanocavity arrays combine two featuring characteristics: (1) An embedded MoS2 monolayer that enables label-free isolation and nanoconfinement of single EVs due to physical interaction (Coulomb and van der Waals) between the MoS2 edge sites and the lipid bilayer; and (2) A layered plasmonic cavity that enables sufficient electromagnetic field enhancement inside the cavities to obtain a single EV level signal resolution for stratifying the molecular alterations. We used the GBM paradigm to demonstrate the diagnostic potential of the SERS single EV molecular profiling approach. The MoSERS multiplexing fluidic achieves parallel signal acquisition of glioma molecular variants (EGFRvIII oncogenic mutation and MGMT expression) in GBM cells. The detection limit of 1.23% was found for stratifying these key molecular variants in the wild-type population. When interfaced with a convolutional neural network (CNN), MoSERS improved diagnostic accuracy (87%) with which GBM mutations were detected in 12 patient blood samples, on par with clinical pathology tests. Thus, MoSERS demonstrates the potential for molecular stratification of cancer patients using circulating EVs.


Asunto(s)
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Glioma , Humanos , Glioblastoma/diagnóstico , Glioblastoma/genética , Glioblastoma/metabolismo , Molibdeno/metabolismo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Glioma/patología , Vesículas Extracelulares/química , Espectrometría Raman
8.
Int J Mol Sci ; 24(10)2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37240053

RESUMEN

When modified uridine derivatives are incorporated into DNA, radical species may form that cause DNA damage. This category of molecules has been proposed as radiosensitizers and is currently being researched. Here, we study electron attachment to 5-bromo-4-thiouracil (BrSU), a uracil derivative, and 5-bromo-4-thio-2'-deoxyuridine (BrSdU), with an attached deoxyribose moiety via the N-glycosidic (N1-C) bond. Quadrupole mass spectrometry was used to detect the anionic products of dissociative electron attachment (DEA), and the experimental results were supported by quantum chemical calculations performed at the M062X/aug-cc-pVTZ level of theory. Experimentally, we found that BrSU predominantly captures low-energy electrons with kinetic energies near 0 eV, though the abundance of bromine anions was rather low compared to a similar experiment with bromouracil. We suggest that, for this reaction channel, proton-transfer reactions in the transient negative ions limit the release of bromine anions.


Asunto(s)
Desoxirribosa , Electrones , Desoxirribosa/química , Bromo , Aniones , Bromodesoxiuridina
9.
J Am Chem Soc ; 145(16): 9059-9071, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37040588

RESUMEN

Single-strand breaks (SSBs) induced via electron attachment were previously observed in dry DNA under ultrahigh vacuum (UHV), while hydrated electrons were found not able to induce this DNA damage in an aqueous solution. To explain these findings, crossed electron-molecular beam (CEMB) and anion photoelectron spectroscopy (aPES) experiments coupled to density functional theory (DFT) modeling were used to demonstrate the fundamental importance of proton transfer (PT) in radical anions formed via electron attachment. Three molecular systems were investigated: 5'-monophosphate of 2'-deoxycytidine (dCMPH), where PT in the electron adduct is feasible, and two ethylated derivatives, 5'-diethylphosphate and 3',5'-tetraethyldiphosphate of 2'-deoxycytidine, where PT is blocked due to substitution of labile protons with the ethyl residues. CEMB and aPES experiments confirmed the cleavage of the C3'/C5'-O bond as the main dissociation channel related to electron attachment in the ethylated derivatives. In the case of dCMPH, however, electron attachment (in the aPES experiments) yielded its parent (intact) radical anion, dCMPH-, suggesting that its dissociation was inhibited. The aPES-measured vertical detachment energy of the dCMPH- was found to be 3.27 eV, which agreed with its B3LYP/6-31++G(d,p)-calculated value and implied that electron-induced proton transfer (EIPT) had occurred during electron attachment to the dCMPH model nucleotide. In other words, EIPT, subduing dissociation, appeared to be somewhat protective against SSB. While EIPT is facilitated in solution compared to the dry environment, the above findings are consistent with the stability of DNA against hydrated electron-induced SSB in solution versus free electron-induced SSB formation in dry DNA.


Asunto(s)
Hominidae , Protones , Animales , Modelos Moleculares , Electrones , ADN/química , Aniones/química , Daño del ADN
10.
Neuro Oncol ; 25(8): 1381-1394, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37100086

RESUMEN

Patients with diffuse glioma are at high risk of developing venous thromboembolism (VTE) over the course of the disease, with up to 30% incidence in patients with glioblastoma (GBM) and a lower but nonnegligible risk in lower-grade gliomas. Recent and ongoing efforts to identify clinical and laboratory biomarkers of patients at increased risk offer promise, but to date, there is no proven role for prophylaxis outside of the perioperative period. Emerging data suggest a higher risk of VTE in patients with isocitrate dehydrogenase (IDH) wild-type glioma and the potential mechanistic role of IDH mutation in the suppression of production of the procoagulants tissue factor and podoplanin. According to published guidelines, therapeutic anticoagulation with low molecular weight heparin (LMWH) or alternatively, direct oral anticoagulants (DOACs) in patients without increased risk of gastrointestinal or genitourinary bleeding is recommended for VTE treatment. Due to the elevated risk of intracranial hemorrhage (ICH) in GBM, anticoagulation treatment remains challenging and at times fraught. There are conflicting data on the risk of ICH with LMWH in patients with glioma; small retrospective studies suggest DOACs may convey lower ICH risk than LMWH. Investigational anticoagulants that prevent thrombosis without impairing hemostasis, such as factor XI inhibitors, may carry a better therapeutic index and are expected to enter clinical trials for cancer-associated thrombosis.


Asunto(s)
Glioblastoma , Glioma , Neoplasias , Tromboembolia Venosa , Humanos , Heparina de Bajo-Peso-Molecular/uso terapéutico , Tromboembolia Venosa/epidemiología , Tromboembolia Venosa/etiología , Tromboembolia Venosa/prevención & control , Estudios Retrospectivos , Anticoagulantes/uso terapéutico , Neoplasias/tratamiento farmacológico , Glioma/complicaciones , Glioma/epidemiología , Glioma/terapia , Glioblastoma/tratamiento farmacológico , Biología
11.
J Phys Chem B ; 127(11): 2565-2574, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36893332

RESUMEN

Previous density functional theory (DFT) studies on 6-brominated pyrimidine nucleosides suggest that 6-iodo-2'-deoxyuridine (6IdU) should act as a better radiosensitizer than its 5-iodosubstituted 2'-deoxyuridine analogue. In this work, we show that 6IdU is unstable in an aqueous solution. Indeed, a complete disappearance of the 6IdU signal was observed during its isolation by reversed-phase high-performance liquid chromatography (RP-HPLC). As indicated by the thermodynamic characteristics for the SN1-type hydrolysis of 6IdU obtained at the CAM-B3LYP/DGDZVP++ level and the polarizable continuum model (PCM) of water, 6-iodouracil (6IU) was already released quantitatively at ambient temperatures. The simulation of the hydrolysis kinetics demonstrated that a thermodynamic equilibrium was reached within seconds for the title compound. To assess the reliability of the calculations carried out, we synthesized 6-iodouridine (6IUrd), which was, unlike 6IdU, sufficiently stable in an aqueous solution at room temperature. The activation barrier for the N-glycosidic bond dissociation in 6IUrd was estimated experimentally using an Arrhenius plot. The stabilities in water calculated for 6IdU, 6IUrd, and 5-iodo-2'-deoxyuridine (5IdU) could be explained by the electronic and steric effects of the 2'-hydroxy group present in the ribose moiety. Our studies highlight the issue of the hydrolytic stability of potentially radiosensitizing nucleotides which, besides having favorable dissociative electron attachment (DEA) characteristics, must be stable in water to have any practical application.


Asunto(s)
Daño del ADN , Fármacos Sensibilizantes a Radiaciones , Reproducibilidad de los Resultados , Fármacos Sensibilizantes a Radiaciones/farmacología , Desoxiuridina/química , Agua/química
13.
Arch Immunol Ther Exp (Warsz) ; 71(1): 5, 2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36729313

RESUMEN

Extracellular vesicles (EVs) and particles (EPs) serve as unique carriers of complex molecular information with increasingly recognized roles in health and disease. Individual EVs/EPs collectively contribute to the molecular fingerprint of their producing cell, reflecting its identity, state, function and phenotype. This property is of particular interest in cancer where enormous heterogeneity of cancer cells is compounded by the presence of altered stromal, vascular and immune cell populations, which is further complicated by systemic responses elicited by the disease in individual patients. These diverse and interacting cellular compartments are dynamically represented by myriads of EVs/EPs released into the circulating biofluids (blood) during cancer progression and treatment. Current approaches of liquid biopsy seek to follow specific elements of the EV/EP cargo that may have diagnostic utility (as biomarkers), such as cancer cell-derived mutant oncoproteins or nucleic acids. However, with emerging technologies enabling high-throughput EV/EP analysis at a single particle level, a more holistic approach may be on the horizon. Indeed, each EV/EP carries multidimensional information (molecular "voxel") that could be integrated across thousands of particles into a larger and unbiased landscape (EV/EP "hologram") reflecting the true cellular complexity of the disease, along with cellular interactions, systemic responses and effects of treatment. Thus, the longitudinal molecular mapping of EV/EP populations may add a new dimension to crucial aspects of cancer biology, personalized diagnostics, and therapy.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Neoplasias/genética , Biomarcadores , Fenotipo
14.
Front Med (Lausanne) ; 10: 1252417, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38188342

RESUMEN

Single cell analysis of cancer cell transcriptome may shed a completely new light on cancer-associated thrombosis (CAT). CAT causes morbid, and sometimes lethal complications in certain human cancers known to be associated with high risk of venous thromboembolism (VTE), pulmonary embolism (PE) or arterial thromboembolism (ATE), all of which worsen patients' prognosis. How active cancers drive these processes has long evaded scrutiny. While "unspecific" microenvironmental effects and consequences of patient care (e.g., chemotherapy) have been implicated in pathogenesis of CAT, it has also been suggested that oncogenic pathways driven by either genetic (mutations), or epigenetic (methylation) events may influence the coagulant phenotype of cancer cells and stroma, and thereby modulate the VTE/PE risk. Consequently, the spectrum of driver events and their downstream effector mechanisms may, to some extent, explain the heterogeneity of CAT manifestations between cancer types, molecular subtypes, and individual cases, with thrombosis-promoting, or -protective mutations. Understanding this molecular causation is important if rationally designed countermeasures were to be deployed to mitigate the clinical impact of CAT in individual cancer patients. In this regard, multi-omic analysis of human cancers, especially at a single cell level, has brought a new meaning to concepts of cellular heterogeneity, plasticity, and multicellular complexity of the tumour microenvironment, with profound and still relatively unexplored implications for the pathogenesis of CAT. Indeed, cancers may contain molecularly distinct cellular subpopulations, or dynamic epigenetic states associated with different profiles of coagulant activity. In this article we discuss some of the relevant lessons from the single cell "omics" and how they could unlock new potential mechanisms through which cancer driving oncogenic lesions may modulate CAT, with possible consequences for patient stratification, care, and outcomes.

15.
Semin Cancer Biol ; 87: 196-213, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371024

RESUMEN

Cancer progression impacts and exploits the vascular system in several highly consequential ways. Among different types of vascular cells, blood cells and mediators that are engaged in these processes, endothelial cells are at the centre of the underlying circuitry, as crucial constituents of angiogenesis, angiocrine stimulation, non-angiogenic vascular growth, interactions with the coagulation system and other responses. Tumour-vascular interactions involve soluble factors, extracellular matrix molecules, cell-cell contacts, as well as extracellular vesicles (EVs) carrying assemblies of molecular effectors. Oncogenic mutations and transforming changes in the cancer cell genome, epigenome and signalling circuitry exert important and often cancer-specific influences upon pathways of tumour-vascular interactions, including the biogenesis, content, and biological activity of EVs and responses of cancer cells to them. Notably, EVs may carry and transfer bioactive, oncogenic macromolecules (oncoproteins, RNA, DNA) between tumour and vascular cells and thereby elicit unique functional changes and forms of vascular growth and remodeling. Cancer EVs influence the state of the vasculature both locally and systemically, as exemplified by cancer-associated thrombosis. EV-mediated communication pathways represent attractive targets for therapies aiming at modulation of the tumour-vascular interface (beyond angiogenesis) and could also be exploited for diagnostic purposes in cancer.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Células Endoteliales , Vesículas Extracelulares/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes , Neovascularización Patológica/metabolismo
16.
Thromb Res ; 213 Suppl 1: S29-S34, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-36210557

RESUMEN

Cancer has long been known to incur an increased risk of venous thromboembolism (VTE). Multiple risk factors for cancer-associated thrombosis (CAT) have been identified, and several pathophysiological mechanisms elucidated. However, until recently there was scant data available about the influence of cancer-specific somatic genetic alterations on the risk of venous thromboembolism. In the last few years, several gene loci were found to modulate the risk of CAT, usually causing an increase in risk but sometimes found to have a protective effect. Notably, cancer-specific somatic genetic alterations in KRAS, IDH1, ALK and ROS1 have been found to alter the risk of CAT by independent groups. Work in this field is limited by the high-dimensionality and often sparse nature of genomic datasets. Also, early data suggest that for certain genes the effect on VTE risk can be tumor type-specific, which suggests that predictive models must factor such interactions in order to optimally leverage genetic information. Notably, individual gene effects appear to be often small and no one gene explains most of the variability of CAT risk. Ultimately, improved knowledge of the genetic determinants of CAT will help ameliorate risk stratification for this complication and hopefully provide mechanistic insights. Better risk stratification could lead to enhanced pharmacological VTE prophylaxis, while advancements in the understanding of the biology of CAT could conceivably lead to non-anticoagulant targeted therapies for thrombosis.


Asunto(s)
Neoplasias , Trombosis , Tromboembolia Venosa , Humanos , Neoplasias/complicaciones , Neoplasias/genética , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas , Proteínas Proto-Oncogénicas p21(ras) , Proteínas Tirosina Quinasas Receptoras , Medición de Riesgo , Factores de Riesgo , Trombosis/complicaciones , Tromboembolia Venosa/etiología
17.
Nat Commun ; 13(1): 5494, 2022 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123372

RESUMEN

Glioblastoma (GBM) is an incurable form of primary astrocytic brain tumor driven by glioma stem cell (GSC) compartment closely associated with the vascular niche. GSC phenotypes are heterogeneous and range from proneural to mesenchymal-like, the latter characterised by greater invasiveness. Here we document the secretory (angiocrine) role of endothelial cells and their derived extracellular vesicles (EVs) as drivers of proneural-to-mesenchymal reprogramming of GSCs. These changes involve activation of matrix metalloproteinases (MMPs) and NFκB, and inactivation of NOTCH, while altering responsiveness to chemotherapy and driving infiltrative growth in the brain. Our findings suggest that EV-mediated angiocrine interactions impact the nature of cellular stemness in GBM with implications for disease biology and therapy.


Asunto(s)
Vesículas Extracelulares , Glioblastoma , Glioma , Células Endoteliales/patología , Vesículas Extracelulares/patología , Glioblastoma/patología , Glioma/patología , Humanos , Células Madre Neoplásicas/patología
18.
Best Pract Res Clin Haematol ; 35(1): 101349, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-36030072

RESUMEN

Cancer associated thrombosis (CAT) including venous and arterial thromboembolism (VTE and ATE respectively), as well as subclinical hypercoagulable states pose a risk of serious morbidity and mortality and poor outcomes in cancer patients. It is increasingly clear that rather than being unspecific aftermaths of tumour growth, CAT is causally linked to the molecular phenotype of cancer cells and its genetic and epigenetic oncogenic drivers. Emerging data suggest that mutational events and factors modifying chromatin architecture in cancer cells influence the repertoire of genes (coagulome) the products of which may interact with the hemostatic system either directly or through modification of inflammatory system or release of cancer-related prothrombotic extracellular vesicles (EVs). Single cell transcriptomic analysis of brain tumours reveals the coexistence of multiple coagulant mechanisms associated with different cancer cell subpopulations and sites. These observations may suggest that a multipronged, biologically based approach may be needed to effectively predict and manage CAT.


Asunto(s)
Coagulación Sanguínea , Neoplasias , Oncogenes , Trombosis , Epigenómica , Humanos , Neoplasias/complicaciones , Trombosis/etiología
19.
Int J Mol Sci ; 23(15)2022 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-35955461

RESUMEN

We investigate dissociative electron attachment to 5-fluorouracil (5-FU) employing a crossed electron-molecular beam experiment and quantum chemical calculations. Upon the formation of the 5-FU- anion, 12 different fragmentation products are observed, the most probable dissociation channel being H loss. The parent anion, 5-FU-, is not stable on the experimental timescale (~140 µs), most probably due to the low electron affinity of FU; simple HF loss and F- formation are seen only with a rather weak abundance. The initial dynamics upon electron attachment seems to be governed by hydrogen atom pre-dissociation followed by either its full dissociation or roaming in the vicinity of the molecule, recombining eventually into the HF molecule. When the HF molecule is formed, the released energy might be used for various ring cleavage reactions. Our results show that higher yields of the fluorine anion are most probably prevented through both faster dissociation of an H atom and recombination of F- with a proton to form HF. Resonance calculations indicate that F- is formed upon shape as well as core-excited resonances.


Asunto(s)
Electrones , Ácido Fluorhídrico , Aniones , Fluorouracilo/química , Hidrógeno/química
20.
Neurooncol Adv ; 4(1): vdac076, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795471

RESUMEN

Background: Despite aggressive upfront treatment in glioblastoma (GBM), recurrence remains inevitable for most patients. Accumulating evidence has identified hypermutation induced by temozolomide (TMZ) as an emerging subtype of recurrent GBM. However, its biological and therapeutic significance has yet to be described. Methods: We combined GBM patient and derive GBM stem cells (GSCs) from tumors following TMZ to explore response of hypermutant and non-hypermutant emergent phenotypes and explore the immune relevance of hypermutant and non-hypermutant states in vivo. Results: Hypermutation emerges as one of two possible mutational subtypes following TMZ treatment in vivo and demonstrates distinct phenotypic features compared to non-hypermutant recurrent GBM. Hypermutant tumors elicited robust immune rejection in subcutaneous contexts which was accompanied by increased immune cell infiltration. In contrast, immune rejection of hypermutant tumors were stunted in orthotopic settings where we observe limited immune infiltration. Use of anti-PD-1 immunotherapy showed that immunosuppression in orthotopic contexts was independent from the PD-1/PD-L1 axis. Finally, we demonstrate that mutational burden can be estimated from DNA contained in extracellular vesicles (EVs). Conclusion: Hypermutation post-TMZ are phenotypically distinct from non-hypermutant GBM and requires personalization for appropriate treatment. The brain microenvironment may be immunosuppressive and exploration of the mechanisms behind this may be key to improving immunotherapy response in this subtype of recurrent GBM.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...