Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Glia ; 72(5): 857-871, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38234042

RESUMEN

Tumor-associated astrocytes (TAAs) in the glioblastoma microenvironment play an important role in tumor development and malignant progression initiated by glioma stem cells (GSCs). In the current study, normal human astrocytes (NHAs) were cultured and continuously treated with GSC-derived exosomes (GSC-EXOs) induction to explore the mechanism by which GSCs affect astrocyte remodeling. This study revealed that GSC-EXOs can induce the transformation of NHAs into TAAs, with relatively swollen cell bodies and multiple extended processes. In addition, high proliferation, elevated resistance to temozolomide (TMZ), and increased expression of TAA-related markers (TGF-ß, CD44, and tenascin-C) were observed in the TAAs. Furthermore, GSC-derived exosomal miR-3065-5p could be delivered to NHAs, and miR-3065-5p levels increased significantly in TAAs, as verified by miRNA expression profile sequencing and Reverse transcription polymerase chain reaction. Overexpression of miR-3065-5p also enhanced NHA proliferation, elevated resistance to TMZ, and increased the expression levels of TAA-related markers. In addition, both GSC-EXO-induced and miR-3065-5p-overexpressing NHAs promoted tumorigenesis of GSCs in vivo. Discs Large Homolog 2 (DLG2, downregulated in glioblastoma) is a direct downstream target of miR-3065-5p in TAAs, and DLG2 overexpression could partially reverse the transformation of NHAs into TAAs. Collectively, these data demonstrate that GSC-EXOs induce the transformation of NHAs into TAAs via the miR-3065-5p/DLG2 signaling axis and that TAAs can further promote the tumorigenesis of GSCs. Thus, precisely blocking the interactions between astrocytes and GSCs via exosomes may be a novel strategy to inhibit glioblastoma development, but more in-depth mechanistic studies are still needed.


Asunto(s)
Exosomas , Glioblastoma , Glioma , MicroARNs , Humanos , Glioblastoma/patología , Astrocitos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Exosomas/metabolismo , Glioma/patología , Temozolomida/farmacología , Temozolomida/metabolismo , Células Madre Neoplásicas/metabolismo , Carcinogénesis/genética , Proliferación Celular , Microambiente Tumoral , Proteínas Supresoras de Tumor/metabolismo , Guanilato-Quinasas/metabolismo
2.
Neurobiol Dis ; 191: 106396, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176570

RESUMEN

Studies from rodents to primates and humans indicate that individuals vary in how resilient they are to stress, and understanding the basis of these variations may help improve treatments for depression. Here we explored the potential contribution of the gut microbiome to such variation. Mice were exposed to chronic unpredictable mild stress (CUMS) for 4 weeks then allowed to recover for 3 weeks, after which they were subjected to behavioral tests and categorized as showing low or high stress resilience. The two types of mouse were compared in terms of hippocampal gene expression using RNA sequencing, fecal microbiomes using 16S RNA sequencing, and extent of neurogenesis in the hippocampus using immunostaining of brain sections. Fecal microbiota were transplanted from either type of mouse into previously stress-exposed and stress-naïve animals, and the effects of the transplantation on stress-induced behaviors and neurogenesis in the hippocampus were examined. Finally, we blocked neurogenesis using temozolomide to explore the role of neurogenesis promoted by fecal microbiota transplantation in enhancing resilience to stress. Results showed that highly stress-resilient mice, but not those with low resilience, improved significantly on measures of anhedonia, behavioral despair, and anxiety after 3-week recovery from CUMS. Their feces showed greater abundance of Lactobacillus, Bifidobacterium and Romboutsia than feces from mice with low stress resilience, as well as lower abundance of Staphylococcus, Psychrobacter and Corynebacterium. Similarly, highly stress-resilient mice showed greater neurogenesis in hippocampus than animals with low stress resilience. Transplanting fecal microbiota from mice with high stress resilience into previously CUMS-exposed recipients rescued neurogenesis in hippocampus, facilitating recovery from stress-induced depression and cognitive decline. Blockade of neurogenesis with temozolomide abolished recovery of recipients from CUMS-induced depression and cognitive decline in mice transplanted with fecal microbiota from mice with high stress resilience. In conclusion, our results suggested that remodeling of the gut microbiome after stress may reverse stress-induced impairment of hippocampal neurogenesis and thereby promote recovery from stress-induced depression.


Asunto(s)
Depresión , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Depresión/metabolismo , Microbioma Gastrointestinal/genética , Temozolomida/metabolismo , Temozolomida/farmacología , Hipocampo/metabolismo , Neurogénesis , Estrés Psicológico/psicología
3.
Mol Biol (Mosk) ; 57(6): 31-40, 2023.
Artículo en Ruso | MEDLINE | ID: mdl-38062972

RESUMEN

Temozolomide resistance is a major cause of recurrence and poor prognosis in neuroglioma. Recently, growing evidence has suggested that mitophagy is involved in drug resistance in various tumor types. However, the role and molecular mechanisms of mitophagy in temozolomide resistance in glioma remain unclear. In this study, mitophagy levels in temozolomide-resistant and -sensitive cell lines were evaluated. The mechanisms underlying the regulation of mitophagy were explored through RNA sequencing, and the roles of differentially expressed genes in mitophagy and temozolomide resistance were investigated. We found that mitophagy promotes temozolomide resistance in glioma. Specifically, small ubiquitin-like modifier specific protease 6 (SENP6) promoted temozolomide resistance in glioma by inducing mitophagy. Protein-protein interactions between SENP6 and the mitophagy executive protein PTEN-induced kinase 1 (PINK1) resulted in a reduction in small ubiquitin-like modifier 2 (SUMO2)ylation of PINK1, thereby enhancing mitophagy. Our study demonstrates that by inducing mitophagy, the interaction of SENP6 with PINK1 promotes temozolomide resistance in glioblastoma. Therefore, targeting SENP6 or directly regulating mitophagy could be a potential and novel therapeutic target for reversing temozolomide resistance in glioma.


Asunto(s)
Resistencia a Antineoplásicos , Glioma , Mitofagia , Humanos , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Mitocondrias/metabolismo , Mitofagia/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Temozolomida/farmacología , Temozolomida/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinas/metabolismo , Resistencia a Antineoplásicos/genética
4.
Behav Pharmacol ; 34(5): 263-274, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37401406

RESUMEN

Temozolomide is used commonly in the treatment of some types of cancers, but it may also result in cognitive impairments such as memory deficits. l -Dopa, a well known medicine for the central nervous system, has been shown to have positive effects on some cognitive disorders. Here we sought to investigate the effect of l -Dopa on temozolomide-induced cognitive impairments. BALB/c mice were subjected to 3-days temozolomide and 6-days concomitant l -Dopa/benserazide administration in six groups (control, l -Dopa 25 mg/kg, l -Dopa 75 mg/kg, temozolomide, temozolomide +  l -Dopa 25 mg/kg, and temozolomide +  l -Dopa 75 mg/kg). Open field test, object location recognition, novel object recognition test, and shuttle-box test were carried out to determine the locomotor, anxiety-like behavior, and memory function of subjects. TNF-α and brain-derived neurotrophic factor (BDNF) gene expression in the hippocampus was measured by real-time PCR. Mice treated with temozolomide showed recognition memory impairment, along with hippocampal TNF-α and BDNF mRNA expression level raise, and detection of histological insults in hematoxylin and eosin hippocampal slides. Mice that received temozolomide +  l -Dopa showed normal behavioral function and lower TNF-α and BDNF hippocampal mRNA expression levels, and histologically normal hippocampal CA1 region in comparison with mice in the temozolomide group. Our results provide evidence that l -Dopa prevents temozolomide-induced recognition memory deficit in mice at the acute phase probably via l -Dopa antineuroinflammatory effects.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Disfunción Cognitiva , Ratones , Masculino , Animales , Temozolomida/farmacología , Temozolomida/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Hipocampo , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , ARN Mensajero/metabolismo
5.
J Adv Res ; 43: 219-231, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36585110

RESUMEN

INTRODUCTION: Adult hippocampal neurogenesis (AHN) is acknowledged to play a critical role in depression. Emerging evidence suggests that the Wnt/ß-catenin pathway can modulate hippocampal neurogenesis. Crocin, a natural carotenoid, possesses antidepressant property. Yet, how it affects neurogenesis and exerts antidepressant response remains unknown. OBJECTIVE: To explore the role of AHN and Wnt/ß-catenin in the antidepressant action of crocin. METHODS: Depressive-related behaviors, including sucrose preference test (SPT), tail suspension test (TST), forced swimming test (FST), and sexual behaviors were performed following crocin treatment. Neurogenesis was characterized via immunohistochemistry, immunofluorescence, Golgi staining and electrophysiology approach. Wnt/ß-catenin signaling was examined with western blot analysis. The role of AHN Wnt/ß-catenin cascade in crocin's antidepressant response was assessed by conditional removal of glial fibrillary acidic protein (GFAP)-expressing newborn neural cells, temozolomide administration, microinfusion of Dkk1 or viral-mediated shRNA of Wnt3a. RESULTS: Crocin decreased the immobility duration in TST and FST without impairing the performance in sexual behaviors. Crocin boosted the proliferation and differentiation of progenitors, and promoted dendritic maturation and functional integration of hippocampal newborn neurons. Conditional removal of GFAP-expressing neural cells or temozolomide administration impaired the antidepressant response of crocin. Additionally, Wnt/ß-catenin signaling was promoted following crocin treatment. In chronic unpredictable mild stress (CUMS) murine model, crocin treatment displayed antidepressant response in SPT, FST and TST, and restored the neurogenesis levels and Wnt/ß-catenin signaling impaired by CUMS. Infusion of Dickkopf-1 (DKK1) or knockdown of Wnt3a in the hippocampus impaired the antidepressant response of crocin. CONCLUSION: Crocin exerted antidepressant response, which was dependent on enhancement of AHN and activation of the Wnt/ß-catenin pathway.


Asunto(s)
Carotenoides , Hipocampo , Neurogénesis , Animales , Ratones , Antidepresivos/farmacología , Antidepresivos/metabolismo , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo , beta Catenina/farmacología , Carotenoides/metabolismo , Carotenoides/farmacología , Carotenoides/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Temozolomida/metabolismo , Temozolomida/farmacología
6.
Probiotics Antimicrob Proteins ; 15(5): 1079-1091, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-35639268

RESUMEN

Chemotherapy-induced intestinal mucositis is one of the major toxic side effects in the treatment of cancer patients. The purpose of this study is to screen lactic acid bacteria which could alleviate intestinal inflammation and damage induced by chemotherapeutic agents and explore the possible underlying mechanisms. Lactobacillus salivarius CPU-01 was selected from traditional Chinese fermented foods due to its protective effects on the toxicity of temozolomide in Caenorhabditis elegans. Eighteen ICR mice were randomly divided into 3 groups including control group, temozolomide-induced intestinal mucositis group, and temozolomide + L. salivarius CPU-01 group, and were used to investigate the effect of L. salivarius CPU-01 on chemotherapy-induced intestinal mucositis. It has been demonstrated that the administration of L. salivarius CPU-01 can prevent colon shortening and alleviate colon tissue damage caused by temozolomide-induced intestinal mucositis in mice. L. salivarius CPU-01 relieved the intestinal microbiota disorders caused by temozolomide and contributed to the growth of beneficial bacteria, such as Lactobacillus, Clostridia UCG - 014_norank, and Akkermansia. In vivo experiments also indicated that L. salivarius CPU-01 can suppress the level of temozolomide-induced pro-inflammatory cytokines in serum and mRNA expression in the small intestine tissues. It was also found that L. salivarius CPU-01 significantly increased the expressions of intestinal tight junction (TJ) proteins, ZO-1, and Occludin proteins in mice treated with temozolomide. These findings suggest that L. salivarius CPU-01 can ameliorate temozolomide-induced intestinal mucositis by modulating gut microbiota, blocking pro-inflammatory cytokines, and repairing the intestinal barrier. These findings suggest probiotics may serve as a potential alternative therapeutic strategy for the prevention of chemotherapy-induced intestinal mucositis in the future.


Asunto(s)
Antineoplásicos , Microbioma Gastrointestinal , Ligilactobacillus salivarius , Mucositis , Ratones , Animales , Mucositis/inducido químicamente , Mucositis/metabolismo , Mucositis/microbiología , Citocinas/metabolismo , Temozolomida/efectos adversos , Temozolomida/metabolismo , Ratones Endogámicos ICR , Antineoplásicos/farmacología , Mucosa Intestinal/microbiología
7.
Int J Mol Sci ; 23(23)2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36499683

RESUMEN

The survival of patients with glioblastoma (GBM) is poor. The main cause is the presence of glioma stem cells (GSCs), exceptionally resistant to temozolomide (TMZ) treatment. This last may be related to the heterogeneous expression of ion channels, among them TRPML2. Its mRNA expression was evaluated in two different neural stem cell (NS/PC) lines and sixteen GBM stem-like cells by qRT-PCR. The response to TMZ was evaluated in undifferentiated or differentiated GSCs, and in TRPML2-induced or silenced GSCs. The relationship between TRPML2 expression and responsiveness to TMZ treatment was evaluated by MTT assay showing that increased TRPML2 mRNA levels are associated with resistance to TMZ. This research was deepened by qRT-PCR and western blot analysis. PI3K/AKT and JAK/STAT pathways as well as ABC and SLC drug transporters were involved. Finally, the relationship between TRPML2 expression and overall survival (OS) and progression-free survival (PFS) in patient-derived GSCs was evaluated by Kaplan-Meier analysis. The expression of TRPML2 mRNA correlates with worse OS and PFS in GBM patients. Thus, the expression of TRPML2 in GSCs influences the responsiveness to TMZ in vitro and affects OS and PFS in GBM patients.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Temozolomida/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/metabolismo , ARN Mensajero/metabolismo , Resistencia a Antineoplásicos , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico
8.
Cell Biol Int ; 46(9): 1519-1529, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35731168

RESUMEN

Glioma initiating cells (GICs), also known as glioma stem cells, display the capacity to recapitulate the functional diversity within the tumor. Despite the great progress achieved over the last decades, defining the key molecular regulators of GICs has represented a major obstacle in this field. In our study, data from The Cancer Genome Atlas database illustrated a relationship between C-X-C motif chemokine receptor 4 (CXCR4) expression and the survival of glioma patients. Mechanistically, we further indicated that CXCR4 mediated the upregulation of Kruppel like factor 5 (KLF5), a zinc-finger-containing transcription factor, to facilitate the proliferation of GICs. What's more, CXCR4 also enhanced the chemoresistance through KLF5/Bcl2-like 12 (BCl2L12) in glioma. The elevated expression of KLF5 and BCL2L12 induced by CXCR4 was dependent on phosphoinositide 3-kinases (PI3K)/serine/threonine kinase (AKT) signaling. Importantly, combined application of temozolomide and a CXCR4 inhibitor efficiently reversed CXCR4 mediated drugs resistance and improved anticancer effects in vivo. Collectively, our findings confirmed that CXCR4 promoted GICs proliferation via the KLF5/BCL2L12 dependent pathway, which may enrich the understanding of GICs and help drive the design of efficacious therapeutic strategies.


Asunto(s)
Neoplasias Encefálicas , Glioma , Receptores CXCR4 , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Proteínas Musculares/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Temozolomida/metabolismo , Temozolomida/farmacología
9.
Elife ; 102021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236314

RESUMEN

Temozolomide (TMZ), a DNA methylating agent, is the primary chemotherapeutic drug used in glioblastoma treatment. TMZ induces mostly N-alkylation adducts (N7-methylguanine and N3-methyladenine) and some O6-methylguanine (O6mG) adducts. Current models propose that during DNA replication, thymine is incorporated across from O6mG, promoting a futile cycle of mismatch repair (MMR) that leads to DNA double-strand breaks (DSBs). To revisit the mechanism of O6mG processing, we reacted plasmid DNA with N-methyl-N-nitrosourea (MNU), a temozolomide mimic, and incubated it in Xenopus egg-derived extracts. We have shown that in this system, MMR proteins are enriched on MNU-treated DNA and we observed robust, MMR-dependent, repair synthesis. Our evidence also suggests that MMR, initiated at O6mG:C sites, is strongly stimulated in cis by repair processing of other lesions, such as N-alkylation adducts. Importantly, MNU-treated plasmids display DSBs in extracts, the frequency of which increases linearly with the square of alkylation dose. We suggest that DSBs result from two independent repair processes, one involving MMR at O6mG:C sites and the other involving base excision repair acting at a nearby N-alkylation adduct. We propose a new, replication-independent mechanism of action of TMZ, which operates in addition to the well-studied cell cycle-dependent mode of action.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , ADN/metabolismo , Temozolomida/metabolismo , Animales , Reparación de la Incompatibilidad de ADN , Replicación del ADN , Expresión Génica , Humanos , Temozolomida/farmacología , Xenopus
10.
Int J Biol Macromol ; 187: 742-754, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34310997

RESUMEN

A surface modulated biodegradable transdermal strategy has been exploited for improving the biopharmaceutical properties of Temozolomide augmented in Poly Lactic-co-glycolic acid (PLGA) chitosan double walled nanogel (PCNGL). The PCNGL was synthesized by dual approach methodology showing consistent nanosize particle range of 210 nm and PDI 0.325 ± 0.43 with cationic zeta potential values +29.34 ± 0.79 mV. The PCNGL showed qualitative endothermic & exothermic temperature dependent degradation peaks by thermogravimetry analysis. Blood hemolysis and coagulation assay showed 3.37 ± 0.19 as hemolytic ratio, validating biologically safe margin for transdermal delivery. The in vitro drug release showed 85% transdermal release at slightly acidic pH mimicking skin microenvironment. The ex vivo studies displayed noteworthy penetration potential validated by concentration depth assay and confocal laser scanning microscopy, exhibiting 80% Temozolomide uptake in porcine epidermal tissue. The current research demonstrated the biodegradable controlled delivery of chemotherapeutic Temozolomide leading to biologically safe transdermal therapy.


Asunto(s)
Antineoplásicos Alquilantes/química , Portadores de Fármacos , Nanogeles , Poloxámero/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Temozolomida/química , Administración Cutánea , Animales , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/metabolismo , Preparaciones de Acción Retardada , Composición de Medicamentos , Liberación de Fármacos , Epidermis/metabolismo , Concentración de Iones de Hidrógeno , Nanotecnología , Absorción Cutánea , Propiedades de Superficie , Sus scrofa , Temozolomida/administración & dosificación , Temozolomida/metabolismo
11.
Pharm Res ; 38(6): 1067-1079, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34100216

RESUMEN

PURPOSE: Glioblastoma (GBM) is a malignant brain tumor with a poor long-term prognosis due to recurrence from highly resistant GBM cancer stem cells (CSCs), for which the current standard of treatment with temozolomide (TMZ) alone will unlikely produce a viable cure. In addition, CSCs regenerate rapidly and overexpress methyl transferase which overrides the DNA-alkylating mechanism of TMZ, leading to resistance. The objective of this research was to apply the concepts of nanotechnology to develop a multi-drug therapy, TMZ and idasanutlin (RG7388, a potent mouse double minute 2 (MDM2) antagonist), loaded in functionalized nanoparticles (NPs) that target the GBM CSC subpopulation, reduce the cell viability and provide possibility of in vivo preclinical imaging. METHODS: Polymer-micellar NPs composed of poly(styrene-b-ethylene oxide) (PS-b-PEO) and poly(lactic-co-glycolic) acid (PLGA) were developed by a double emulsion technique loading TMZ and/or RG7388. The NPs were covalently bound to a 15-nucleotide base-pair CD133 aptamer to target the CD133 antigen expressed on the surfaces of GBM CSCs. For diagnostic functionality, the NPs were labelled with radiotracer Zirconium-89 (89Zr). RESULTS: NPs maintained size range less than 100 nm, a low negative charge and exhibited the ability to target and kill the CSC subpopulation when TMZ and RG7388 were used in combination. The targeting function of CD133 aptamer promoted killing in GBM CSCs providing impetus for further development of targeted nanosystems for localized therapy in future in vivo models. CONCLUSIONS: This work has provided a potential clinical application for targeting GBM CSCs with simultaneous diagnostic imaging.


Asunto(s)
Antígeno AC133/metabolismo , Neoplasias Encefálicas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/metabolismo , Nanopartículas/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Desarrollo de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico , Humanos , Ratones , Micelas , Nanopartículas/administración & dosificación , Células Madre Neoplásicas/efectos de los fármacos , Polímeros/administración & dosificación , Polímeros/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirrolidinas/administración & dosificación , Pirrolidinas/metabolismo , Temozolomida/administración & dosificación , Temozolomida/metabolismo , para-Aminobenzoatos/administración & dosificación , para-Aminobenzoatos/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33846242

RESUMEN

Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.


Asunto(s)
Glioblastoma/metabolismo , Microglía/metabolismo , Temozolomida/farmacología , Adulto , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Resistencia a Antineoplásicos/fisiología , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Interleucina-11/inmunología , Interleucina-11/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Microglía/fisiología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Transducción de Señal/efectos de los fármacos , Temozolomida/metabolismo , Microambiente Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/fisiología
13.
Gene ; 776: 145445, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33484758

RESUMEN

Glioblastom Multiforme (GBM) is the most invasive and malignant member of the IV grade of the subclass Astrocytoma according to the last assessment of the 2016 WHO report. Due to the resistance to treatment and weak response, as well as the topographical structure of the blood brain barrier, the treatment is also difficult due to the severe clinical manifestation, and new treatment methods and new therapeutic agents are needed. Temozolomide (TMZ) is widely used in the treatment of glioblastoma and is considered as the primary treatment modality. TMZ, a member of the class of cognitive agents, is currently considered the most effective drug because it can easily pass through the blood brain barrier. Glucose metabolism is a complex energy producing machine that, a glucose molecule produces 38 molecules of ATP after full glycolytic catabolism. According to Otto Warburg's numerous studies cancer cells perform the first glycolytic step without entering the mitochondrial step. These cells produce lactic acid and make the micro-media more acidic even in aerobic conditions. This phenomenon is attributed to the Warburg hypothesis and either as aerobic glycolysis. Although glycolysis enzymes are the primary actors of this phenotypic expression, some genetic and epigenetic factors are no exception. We experimentally used KC7F2 active ingredient to target cancer metabolism. In our study, we evaluated cancer metabolism in combination with the effect of TMZ chemotherapeutic agent, examining the effect of two different agents separately and in combination to observe the effects of cancer cell proliferation, survival, apoptosis and expression of metabolism genes on expression. We observed that the combined effect of reduced the effective dose of the TMZ alkylating agent and that the effect was increased and the effect of the combined teraphy is assessed from a metabolic point of view and that it suppresses aerobic glycolysis.


Asunto(s)
Disulfuros/farmacología , Glioma/tratamiento farmacológico , Sulfonamidas/farmacología , Temozolomida/farmacología , Antineoplásicos/farmacología , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Disulfuros/metabolismo , Resistencia a Antineoplásicos/genética , Glioblastoma/patología , Glioma/patología , Glucosa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Sulfonamidas/metabolismo , Temozolomida/metabolismo
14.
Biotechnol Lett ; 43(2): 383-392, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33145669

RESUMEN

Glioma is one of the most aggressive and highly fatal diseases with an extremely poor prognosis. Considering the poor clinical response to therapy in glioma, it is urgent to establish an in vitro model to facilitate the screening and assessment of anti-brain-tumor drugs. The blood-brain barrier (BBB), as well as liver metabolism plays an important role in determining the pharmacological activity of many anti-brain-tumor drugs. In this work, we designed a multi-interface liver-brain chip integrating co-culture system to assess hepatic metabolism dependent cytotoxicity of anti-brain-tumor drug in vitro. This microdevice composed of three microchannels which were separated by porous membrane and collagen. HepG2 and U87 cells were cultured in separated channels as mimics of liver and glioblastoma. Brain microvascular endothelial cells (BMECS) and cerebral astrocytes were co-cultured on collagen to mimic the brain microvascular endothelial barrier. Three common anti-tumor drugs, paclitaxel (PTX), capecitabine (CAP) and temozolomide (TMZ), were evaluated on this chip. In integrated liver-brain chip, liver enhanced the cytotoxicity of CAP on U87 cells by 30%, but having no significant effect on TMZ. The BBB decreased the cytotoxicity of PTX by 20%, while no significant effects were observed on TMZ and CAP, indicating the importance of liver metabolism and blood-brain barrier on the evaluation of anti-brain-tumor drugs. This work provides a biomimetic liver-brain model to mimic the physiological and pharmacological processes in vitro and presents a simple platform for long-term cell co-culture, drug delivery and metabolism, and real-time analysis of drug effects on brain cancer.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Hígado/efectos de los fármacos , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Capecitabina/metabolismo , Capecitabina/farmacología , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Células Hep G2 , Humanos , Inactivación Metabólica/efectos de los fármacos , Dispositivos Laboratorio en un Chip , Hígado/metabolismo , Nanopartículas/química , Paclitaxel/metabolismo , Paclitaxel/farmacología , Temozolomida/metabolismo
15.
Int J Mol Sci ; 21(21)2020 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-33171763

RESUMEN

Extracellular vesicles (EVs) are widely investigated in glioblastoma multiforme (GBM) for their involvement in regulating GBM pathobiology as well as for their use as potential biomarkers. EVs, through cell-to-cell communication, can deliver proteins, nucleic acids, and lipids that are able to reprogram tumor-associated macrophages (TAMs). This research is aimed to concentrate, characterize, and identify molecular markers of EVs subtypes released by temozolomide (TMZ)-treated and non TMZ-treated four diverse GBM cells. Morphology, size distribution, and quantity of small (sEVs) and large (lEVs) vesicles were analyzed by cryo-TEM. Quality and quantity of EVs surface markers were evaluated, having been obtained by Western blotting. GBM cells shed a large amount of EVs, showing a cell line dependent molecular profile A comparative analysis distinguished sEVs and lEVs released by temozolomide (TMZ)-treated and non TMZ-treated GBM cells on the basis of quantity, size and markers expression. Finally, the GBM-derived sEVs and lEVs, irrespective of TMZ treatment, when challenged with macrophages, modulated cell activation toward a tendentially M2b-like phenotype.


Asunto(s)
Vesículas Extracelulares/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Temozolomida/farmacología , Línea Celular Tumoral , Microscopía por Crioelectrón/métodos , Resistencia a Antineoplásicos/genética , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , MicroARNs/genética , Temozolomida/metabolismo
16.
PLoS One ; 15(9): e0238238, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32881880

RESUMEN

The prognosis for patients with glioblastoma (GB) remains grim. Concurrent temozolomide (TMZ) radiation-the cornerstone of glioma control-extends the overall median survival of GB patients by only a few months over radiotherapy alone. While these survival gains could be partly attributed to radiosensitization, this benefit is greatly minimized in tumors expressing O6-methylguanine DNA methyltransferase (MGMT), which specifically reverses O6-methylguanine lesions. Theoretically, non-O6-methylguanine lesions (i.e., the N-methylpurine adducts), which represent up to 90% of TMZ-generated DNA adducts, could also contribute to radiosensitization. Unfortunately, at concentrations attainable in clinical practice, the alkylation capacity of TMZ cannot overwhelm the repair of N-methylpurine adducts to efficiently exploit these lesions. The current therapeutic application of TMZ therefore faces two main obstacles: (i) the stochastic presence of MGMT and (ii) a blunted radiosensitization potential at physiologic concentrations. To circumvent these limitations, we are developing a novel molecule called NEO212-a derivatization of TMZ generated by coupling TMZ to perillyl alcohol. Based on gas chromatography/mass spectrometry and high-performance liquid chromatography analyses, we determined that NEO212 had greater tumor cell uptake than TMZ. In mouse models, NEO212 was more efficient than TMZ at crossing the blood-brain barrier, preferentially accumulating in tumoral over normal brain tissue. Moreover, in vitro analyses with GB cell lines, including TMZ-resistant isogenic variants, revealed more potent cytotoxic and radiosensitizing activities for NEO212 at physiologic concentrations. Mechanistically, these advantages of NEO212 over TMZ could be attributed to its enhanced tumor uptake presumably leading to more extensive DNA alkylation at equivalent dosages which, ultimately, allows for N-methylpurine lesions to be better exploited for radiosensitization. This effect cannot be achieved with TMZ at clinically relevant concentrations and is independent of MGMT. Our findings establish NEO212 as a superior radiosensitizer and a potentially better alternative to TMZ for newly diagnosed GB patients, irrespective of their MGMT status.


Asunto(s)
Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Glioma/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Temozolomida/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Dacarbazina/análisis , Dacarbazina/metabolismo , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos/genética , Cromatografía de Gases y Espectrometría de Masas , Glioma/patología , Humanos , Ratones , Ratones Endogámicos C57BL , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Fármacos Sensibilizantes a Radiaciones/análisis , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Temozolomida/análisis , Temozolomida/metabolismo , Temozolomida/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
PLoS One ; 15(2): e0229534, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32101575

RESUMEN

Temozolomide (TMZ) is a drug of choice in glioblastoma treatment. Its therapeutic applications expand also beyond high grade gliomas. However, a significant number of recurrences and resistance to the drug is observed. The key factor in each chemotherapy is to achieve the therapeutic doses of a drug at the pathologic site. Nonetheless, the rate of temozolomide penetration from blood to cerebrospinal fluid is only 20-30%, and even smaller into brain intestinum. That makes a challenge for the therapeutic regimens to obtain effective drug concentrations with minimal toxicity and minor side effects. The aim of our research was to explore a novel epigenetic mechanism of temozolomide action in therapeutic conditions. We analyzed the epigenetic effects of TMZ influence on different glioblastoma cell lines in therapeutically achieved TMZ concentrations through total changes of the level of 5-methylcytosine in DNA, the main epigenetic marker. That was done with classical approach of radioactive nucleotide post-labelling and separation on thin-layer chromatography. In the range of therapeutically achieved temozolomide concentrations we observed total DNA hypomethylation. The significant hypermethylating effect was visible after reaching TMZ concentrations of 10-50 µM (depending on the cell line). Longer exposure time promoted DNA hypomethylation. The demethylated state of the glioblastoma cell lines was overcome by repeated TMZ applications, where dose-dependent increase in DNA 5-methylcytosine contents was observed. Those effects were not seen in non-cancerous cell line. The increase of DNA methylation resulting in global gene silencing and consecutive down regulation of gene expression after TMZ treatment may explain better glioblastoma patients' survival.


Asunto(s)
Glioblastoma/genética , Glioblastoma/metabolismo , Temozolomida/farmacología , 5-Metilcitosina , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/genética , Resistencia a Antineoplásicos/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Epigenómica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioma/patología , Humanos , Recurrencia Local de Neoplasia/genética , Temozolomida/metabolismo
18.
Cell Biol Toxicol ; 36(3): 273-278, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31758290

RESUMEN

Glioblastoma (GBM) is the most aggressive type of glioma. Temozolomide (TMZ) is currently the drug of choice used for post-operative chemotherapy of GBM. However, the presence of intrinsic and acquired resistance hinders the success of chemotherapy. To understand the TMZ resistant mechanisms in glioma, we investigated the alterations in cellular signaling pathways by performing transcriptome analysis of TMZ treated glioma cells. Gene Set Enrichment Analysis (GSEA) indicated a significant enrichment of Wnt/ß-catenin signaling besides many other pathways in TMZ treated cells. Further, we demonstrate that TMZ treatment increased the activity from TOPflash reporter, (a Wnt responsive reporter), enhanced the levels of pGSK-3ß (S9) and reduced the levels of p-ß-catenin (S33/37/T41) with a concomitant increase in transcript and protein levels of Wnt targets in a concentration and time-dependent manner. While TMZ treated cells did not show alteration in any of the Wnt ligands, PI3K inhibitor (LY294002) treatment repressed Akt activation and abolished the TMZ-mediated induction of Wnt/ß-catenin pathway. In addition, we show that Wnt/ß-catenin signaling activation by TMZ is independent of ATM/Chk2 pathway. Further, we also demonstrate the activation of mTOR pathway after TMZ treatment. Thus, our results demonstrate that activation of Wnt/ß-catenin pathway involves an ATM/Chk2- independent PI3K/Akt/GSK-3 cascade in TMZ treated cells and further provides mechanistic basis for the chemoresistance of glioma to TMZ.


Asunto(s)
Glioma/metabolismo , Temozolomida/farmacología , Vía de Señalización Wnt/fisiología , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/metabolismo , Glioma/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Temozolomida/metabolismo , Proteínas Wnt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
19.
Cells ; 8(11)2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31653091

RESUMEN

Glioblastoma (GBM) is the most common astrocytic-derived brain tumor in adults, characterized by a poor prognosis mainly due to the resistance to the available therapy. The study of mitochondria-derived oxidative stress, and of the biological events that orbit around it, might help in the comprehension of the molecular mechanisms at the base of GBM responsiveness to Temozolomide (TMZ). Sensitive and resistant GBM cells were used to test the role of mitochondrial ROS release in TMZ-resistance. Chaperone-Mediated Autophagy (CMA) activation in relation to reactive oxygen species (ROS) release has been measured by monitoring the expression of specific genes. Treatments with H2O2 were used to test their potential in reverting resistance. Fluctuations of cytoplasmic ROS levels were accountable for CMA induction and cytotoxic effects observed in TMZ sensitive cells after treatment. On the other hand, in resistant cells, TMZ failed in producing an increase in cytoplasmic ROS levels and CMA activation, preventing GBM cell toxicity. By increasing oxidative stress, CMA activation was recovered, as also cell cytotoxicity, especially in combination with TMZ treatment. Herein, for the first time, it is shown the relation between mitochondrial ROS release, CMA activation and TMZ-responsiveness in GBM.


Asunto(s)
Autofagia Mediada por Chaperones/fisiología , Glioblastoma/metabolismo , Estrés Oxidativo/fisiología , Apoptosis , Autofagia/efectos de los fármacos , Autofagia/fisiología , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Autofagia Mediada por Chaperones/efectos de los fármacos , Citoplasma/metabolismo , Resistencia a Antineoplásicos/genética , Glioblastoma/tratamiento farmacológico , Humanos , Peróxido de Hidrógeno , Mitocondrias/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Temozolomida/metabolismo , Temozolomida/farmacología
20.
Brain Res ; 1720: 146302, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31226325

RESUMEN

Despite the increased understanding of the oncological mechanisms underlying Glioblastoma multiforme (GBM) pathophysiology, and recent advances in therapeutic strategies such as maximal surgical resection and post-operative radiotherapy with concomitant and adjuvant temozolomide chemotherapy, the prognosis for patients with brain tumors remains limited. Evidences indicate that the assessment of DNA methylation status in cancer stem cells would allow identifying molecules expressed in these cells, to lead to targeted elimination of this critical population from brain tumors, making the glioblastoma treatment more effective. This study aimed to analyze the role of microRNA-181d associated with the methylation status of the O6-methylguanine methyl transferase (MGMT) gene in Glioblastoma multiforme cancer stem cells subjected to treatment with temozolomide and ionizing radiation. Such responses were analyzed in terms of cell survival, evaluation of the MGMT gene methylation status by MS-HRM (Methylation-Sensitive High Resolution Melting), and analysis of miRNA-181d and MGMT gene expression by relative quantification of mRNA levels in cancer stem cells subjected to treatment with temozolomide and ionizing radiation, isolated or combined. We showed that ionizing radiation and temozolomide reduced the viability of cancer stem cells from GBM patients, as well as modified MGMT gene and miRNA-181d expression in cancer stem cells, suggesting that miRNA-181d interferes in the glioblastoma cancer stem cell response to treatment with temozolomide and ionizing radiation.


Asunto(s)
Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioblastoma/genética , MicroARNs/genética , Proteínas Supresoras de Tumor/genética , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/metabolismo , Brasil , Metilación de ADN , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Femenino , Glioblastoma/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Pronóstico , Radiación Ionizante , Temozolomida/metabolismo , Temozolomida/uso terapéutico , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...