Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 22(1): 1308, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36513991

RESUMO

BACKGROUND: The discovery of the importance of the immune system and its role in oncogenesis led to the development of immunotherapy, a treatment that represents a major advance in oncology management. Due to the recent nature of immunotherapy, little is known about its side effects and their impact on quality of life. To date, there is no published study that accurately assesses the impact of immunotherapy on cognition, mood and/or fatigue in patients treated for cancer, despite potential neurological toxicities. The purpose of this study is to prospectively assess the incidence of cognitive impairment and cognitive complaints among cancer patients naïve for immunotherapy without concomitant anti-cancer treatment. METHODS: The Cog-Immuno trial is a multicentre longitudinal study addressing patients with cancer candidate to receive immunotherapy alone (n = 100). Immunotherapy treatment will include either anti-PD1/PDL1 or anti-CTLA4 monotherapy or combination therapy. Cognitive and quality of life assessment, electrocardiogram (ECG) and biological tests will be performed at baseline, thereafter 3, and 6 months after immunotherapy initiation. The primary endpoint is the proportion of patients treated by immunotherapy who will experience a decline in cognitive performances or in Montreal Cognitive Assessment (MoCA) score within 3 months after inclusion. Secondary endpoints concern: anxiety, depression, fatigue, clinical characteristics, biological data and neurophysiological measures (heart rate variability and hemispheric lateralization). A pre-clinical study will be conducted in cancer bearing mice receiving checkpoint inhibitors (ICI) with the evaluation of cognitive functions and emotional reactivity, collection of blood samples and investigation of neurobiological mechanisms from brain slices. DISCUSSION: Assessing and understanding the incidence and the severity of cognitive impairment and its impact on quality of life in cancer patients treated by immunotherapy is a major issue. The results of this study will provide information on the impact of these treatments on cognitive functions in order to help the physicians in the choice of the treatment. TRIAL REGISTRATION: NCT03599830, registered July 26, 2018. PROTOCOL VERSION: Version 5.1 dated from 2020/10/02.


Assuntos
Neoplasias , Qualidade de Vida , Animais , Camundongos , Estudos Prospectivos , Estudos Longitudinais , Imunoterapia/efeitos adversos , Cognição , Neoplasias/terapia , Fadiga/etiologia
2.
Cancers (Basel) ; 14(18)2022 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-36139563

RESUMO

Chemotherapy-related cognitive impairment (CRCI) and fatigue constitute common complaints among cancer patient survivors. Panax quinquefolius has been shown to be effective against fatigue in treated cancer patients. We developed a behavioral C57Bl/6j mouse model to study the role of a Panax quinquefolius-based solution containing vitamin C (Qiseng®) or vitamin C alone in activity/fatigue, emotional reactivity and cognitive functions impacted by 5-Fluorouracil (5-FU) chemotherapy. 5-FU significantly reduces the locomotor/exploration activity potentially associated with fatigue, evokes spatial cognitive impairments and leads to a decreased neurogenesis within the hippocampus (Hp). Qiseng® fully prevents the impact of chemotherapy on activity/fatigue and on neurogenesis, specifically in the ventral Hp. We observed that the chemotherapy treatment induces intestinal damage and inflammation associated with increased levels of Lactobacilli in mouse gut microbiota and increased expression of plasma pro-inflammatory cytokines, notably IL-6 and MCP-1. We demonstrated that Qiseng® prevents the 5-FU-induced increase in Lactobacilli levels and further compensates the 5-FU-induced cytokine release. Concomitantly, in the brains of 5-FU-treated mice, Qiseng® partially attenuates the IL-6 receptor gp130 expression associated with a decreased proliferation of neural stem cells in the Hp. In conclusion, Qiseng® prevents the symptoms of fatigue, reduced chemotherapy-induced neuroinflammation and altered neurogenesis, while regulating the mouse gut microbiota composition, thus protecting against intestinal and systemic inflammation.

3.
Autophagy ; 18(12): 3037-3039, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35468023

RESUMO

Accumulating data indicate that several components of the macroautophagy/autophagy machinery mediate additional functions, which do not depend on autophagosome biogenesis or lysosomal cargo degradation. In this context, we found that the core autophagy protein ATG9A participates in the chemotactic movement of several cell lines, including highly invasive glioblastoma cells. Accordingly, ATG9A-depleted cells are unable to form large and persistent leading-edge protrusions. By the design of an ATG9A-pHluorin construct and TIRF imaging, we established that ATG9A-positive vesicles are targeted toward the migration front, where their exocytosis is synchronized with protrusive activity. We finally demonstrated that ATG9A, through its interaction with clathrin adaptor complexes, controls the delivery of ITGB1 (integrin subunit beta 1) to the migration front and normal adhesion dynamics. Together, our work indicates that ATG9A protein has a wider role than anticipated and constitutes a critical component of vesicular trafficking allowing the expansion of cell protrusions and their anchorage to the extracellular matrix.


Assuntos
Autofagia , Proteínas de Transporte Vesicular , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Membrana/metabolismo , Movimento Celular
4.
J Cell Biol ; 221(3)2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35180289

RESUMO

Chemotactic migration is a fundamental cellular behavior relying on the coordinated flux of lipids and cargo proteins toward the leading edge. We found here that the core autophagy protein ATG9A plays a critical role in the chemotactic migration of several human cell lines, including highly invasive glioma cells. Depletion of ATG9A protein altered the formation of large and persistent filamentous actin (F-actin)-rich lamellipodia that normally drive directional migration. Using live-cell TIRF microscopy, we demonstrated that ATG9A-positive vesicles are targeted toward the migration front of polarized cells, where their exocytosis correlates with protrusive activity. Finally, we found that ATG9A was critical for efficient delivery of ß1 integrin to the leading edge and normal adhesion dynamics. Collectively, our data uncover a new function for ATG9A protein and indicate that ATG9A-positive vesicles are mobilized during chemotactic stimulation to facilitate expansion of the lamellipodium and its anchorage to the extracellular matrix.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Autofagia , Movimento Celular , Extensões da Superfície Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Actinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Quimiotaxia , Exocitose , Proteínas de Fluorescência Verde , Humanos , Integrina beta1/metabolismo , Glicoproteínas de Membrana/metabolismo , Pseudópodes/metabolismo , Reprodutibilidade dos Testes
5.
Soft Matter ; 17(48): 10846-10861, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34806746

RESUMO

Over the last few years, significant interest has emerged in the development of localised therapeutic strategies for the treatment of glioblastoma (GBM). The concept of attracting and trapping residual tumour cells within a confined area to facilitate their eradication has developed progressively. Herein, we propose a new design of hyaluronic acid-based hydrogel which can be utilized as a matrix containing a soluble chemoattractant to attract residual glioma cells and chemotherapeutic agents to eradicate them in a less invasive and more efficient way compared to the currently available methods. Hydrogels were prepared at different crosslinking densities, e.g. low and high density, by crosslinking hyaluronic acid with various concentrations of adipic acid dihydrazide and U87MG GBM cell morphology, survival and CD44 expression were evaluated. As a proof-of-concept, hydrogels were loaded with a small peptide chemokine, human urotensin II (hUII), and the migration and survival of U87MG GBM cells were studied. Chemoattractant-containing hydrogels were also loaded with chemotherapeutic drugs to promote cell death in culture. The results showed that U87MG cells were able to invade the hydrogel network and to migrate in response to the chemoattractant hUII. In addition, in static condition, hydrogels loaded with doxorubicin demonstrated significant cytotoxicity leading to less than 80% U87MG cell viability after 48 hours when compared to the control sample. In addition, in in vitro invasive assays, it was originally shown that the chemoattractant effect of hUII can be effective before the cytotoxic action of doxorubicin on the U87MG cells trapped in the hydrogel. Our results provide new insights into a promising approach which can be readily translated in vivo for the treatment of one of the most devastating brain tumours.


Assuntos
Antineoplásicos , Glioma , Antineoplásicos/farmacologia , Fatores Quimiotáticos , Glioma/tratamento farmacológico , Humanos , Ácido Hialurônico , Hidrogéis
6.
Front Cell Dev Biol ; 9: 652544, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33937253

RESUMO

Glioblastomas (GBMs) are the most common primary brain tumors characterized by strong invasiveness and angiogenesis. GBM cells and microenvironment secrete angiogenic factors and also express chemoattractant G protein-coupled receptors (GPCRs) to their advantage. We investigated the role of the vasoactive peptide urotensin II (UII) and its receptor UT on GBM angiogenesis and tested potential ligand/therapeutic options based on this system. On glioma patient samples, the expression of UII and UT increased with the grade with marked expression in the vascular and peri-necrotic mesenchymal hypoxic areas being correlated with vascular density. In vitro human UII stimulated human endothelial HUV-EC-C and hCMEC/D3 cell motility and tubulogenesis. In mouse-transplanted Matrigel sponges, mouse (mUII) and human UII markedly stimulated invasion by macrophages, endothelial, and smooth muscle cells. In U87 GBM xenografts expressing UII and UT in the glial and vascular compartments, UII accelerated tumor development, favored hypoxia and necrosis associated with increased proliferation (Ki67), and induced metalloproteinase (MMP)-2 and -9 expression in Nude mice. UII also promoted a "tortuous" vascular collagen-IV expressing network and integrin expression mainly in the vascular compartment. GBM angiogenesis and integrin αvß3 were confirmed by in vivo 99mTc-RGD tracer imaging and tumoral capture in the non-necrotic area of U87 xenografts in Nude mice. Peptide analogs of UII and UT antagonist were also tested as potential tumor repressor. Urotensin II-related peptide URP inhibited angiogenesis in vitro and failed to attract vascular and inflammatory components in Matrigel in vivo. Interestingly, the UT antagonist/biased ligand urantide and the non-peptide UT antagonist palosuran prevented UII-induced tubulogenesis in vitro and significantly delayed tumor growth in vivo. Urantide drastically prevented endogenous and UII-induced GBM angiogenesis, MMP, and integrin activations, associated with GBM tumoral growth. These findings show that UII induces GBM aggressiveness with necrosis and angiogenesis through integrin activation, a mesenchymal behavior that can be targeted by UT biased ligands/antagonists.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...