يعرض 1 - 9 نتائج من 9 نتيجة بحث عن '"Alza, Lía"', وقت الاستعلام: 0.82s تنقيح النتائج
  1. 1
    دورية أكاديمية

    مصطلحات موضوعية: Cancer microenvironment, Cancer models, Microfluidics

    الوصف: Glioblastoma (GBM) is a highly malignant brain tumour characterised by limited treatment options and poor prognosis. The tumour microenvironment, particularly the central hypoxic region of the tumour, is known to play a pivotal role in GBM progression. Cells within this region adapt to hypoxia by stabilising transcription factor HIF1-α, which promotes cell proliferation, dedifferentiation and chemoresistance. In this study we sought to examine the effects of NNC-55-0396, a tetralol compound which overactivates the unfolded protein response inducing apoptosis, using the organ-on-chip technology. We identified an increased sensitivity of the hypoxic core of the chip to NNC, which correlates with decreasing levels of HIF1-α in vitro. Moreover, NNC blocks the macroautophagic process that is unleashed by hypoxia as revealed by increased levels of autophagosomal constituent LC3-II and autophagy chaperone p62/SQSTM1. The specific effects of NNC in the hypoxic microenvironment unveil additional anti-cancer abilities of this compound and further support investigations on its use in combined therapies against GBM. ; This work was supported by the European Union’s Horizon 2020 research and innovation programme under grant agreement No 829010 (PRIME H2020-FETOPEN-2018-2019-2020-01). We also acknowledge the financial support from the Spanish Ministry of Economy and Competitiveness (MINECO fellowship, DIN 2020-011544); Ministry of Science and Innovation, the European Regional Development Fund (Proyect PID2021-126051OB-C41 funded by MCIN/AEI/10.13039/501100011033/FEDER, UE); MINECO Retos Program RTI2018-094739-B-I00 to CC and JH) and Fundació La Marató de TV3 (201909-30 to CC). CB would like to thank the Government of Aragon (DGA) for the predoctoral funding. AV is funded by Asociación Española Contra el Cáncer and LA is a recipient of an FI-AGAUR fellowship. We are grateful to Maria Casals and David Argilés for technical assistance. Cell culture experiments were performed in the Cell Culture Technical Scientific Service, ...

    وصف الملف: application/pdf

    العلاقة: info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2017-2020/RTI2018-094739-B-I00/ES/NUEVOS MECANISMOS DE DESREGULACION DE LA SEÑALIZACION POR CALCIO CONTRA EL GLIOBLASTOMA/; Reproducció del document publicat a: https://doi.org/10.1038/s41419-024-06492-1Test; Cell Death & Disease, 2024, vol. 15, num. 127; info:eu-repo/grantAgreement/EC/H2020/829010/EU/FETOPEN; https://doi.org/10.1038/s41419-024-06492-1Test; https://repositori.udl.cat/handle/10459.1/465078Test

  2. 2
    دورية أكاديمية

    مصطلحات موضوعية: Calcium, Glioblastoma, ER stress, Cell death

    الوصف: Mibefradil and NNC-55-0396, tetralol derivatives with a proven ability to block T-type calcium channels in excitable cells, reduce cancer cell viability in vitro, causing cell death. Furthermore, they reduce tumor growth in preclinical models of Glioblastoma multiforme (GBM), a brain tumor of poor prognosis. Here we found that GBM cells treated with cytotoxic concentrations of NNC-55-0396 paradoxically increased cytosolic calcium levels through the activation of inositol triphosphate receptors (IP3R) and ER stress. We used pharmacological inhibitors and gene silencing to dissect the cell death pathway stimulated by NNC-55-0396 in GBM cell lines and biopsy-derived cultures. Calcium chelation or IP3R inhibition prevented NNC-55-0396-mediated cytotoxicity, indicating that ER calcium efflux is the cause of cell death. Upstream of calcium mobilization, NNC-55-0396 activated the IRE1α arm of the Unfolded Protein Response (UPR) resulting in the nuclear translocation of pro-apoptotic CHOP. Consistent with these findings, silencing IRE1α or JNK1 rescued the cell death elicited by NNC-55-0396. Therefore, we demonstrate that activation of IRE1α and calcium signaling accounts for the cytotoxicity of NNC-55-0396 in GBM cells. The delineation of the signaling pathway that mediates the abrupt cell death triggered by this compound can help the development of new therapies for GBM. ; We are grateful to Dr. S. Shaikh for help with initial calcium experiments and to Dr. E. Vilaprinyó for statistical analysis. We acknowledge the technical support of D. Argilés and assistance from the personnel of the Cell Culture and the Flow Cytometry Services of IRBLleida/UdL. This work was funded by grants from the Spanish Ministry of Science and Innovation/FEDER “Una manera de hacer Europa” (Retos Program, number RTI2018-094739-B-I00 to JH & CC) and Fundació La Marató de TV3 (number 235/C/2019 to CC). AV was funded by UdL, IRBLleida-Diputació de Lleida and La Marató de TV3. LA is a recipient of an FI-AGAUR predoctoral fellowship. Work ...

    وصف الملف: application/pdf

    العلاقة: info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2017-2020/RTI2018-094739-B-I00/ES/NUEVOS MECANISMOS DE DESREGULACION DE LA SEÑALIZACION POR CALCIO CONTRA EL GLIOBLASTOMA/; Reproducció del document publicat a: https://doi.org/10.1016/j.biopha.2022.112881Test; Biomedicine & Pharmacotherapy, 2022, vol. 149, p. 112881; http://hdl.handle.net/10459.1/73515Test; https://doi.org/10.1016/j.biopha.2022.112881Test

  3. 3
    دورية أكاديمية

    الوصف: In the strongly polarized membranes of excitable cells, activation of T-type Ca2+ channels (TTCCs) by weak depolarizing stimuli allows the influx of Ca2+ which further amplifies membrane depolarization, thus 'recruiting' higher threshold voltage-gated channels to promote action potential firing. Nonetheless, TTCCs perform other functions in the plasma membrane of both excitable and non-excitable cells, in which they regulate a number of biochemical pathways relevant for cell cycle and cell fate. Furthermore, data obtained in the last 20 years have shown the involvement of TTCCs in tumor biology, designating them as promising chemotherapeutic targets. However, their activity in the steadily-depolarized membranes of cancer cells, in which most voltage-gated channels are in the inactivated (nonconducting) state, is counter-intuitive. Here we discuss that in cancer cells weak hyperpolarizing stimuli increase the fraction of open TTCCs which, in association with Ca2+-dependent K+ channels, may critically boost membrane hyperpolarization and driving force for Ca2+ entry through different voltage-independent Ca2+ channels. Available evidence also shows that TTCCs participate in positive feedback circuits with signaling effectors, which may warrant a switch-like activation of pro-proliferative and pro-survival pathways in spite of their low availability. Unravelling TTCC modus operandi in the context of non-excitable membranes may facilitate the development of novel anticancer approaches. ; This work was funded by grants from the Spanish Ministry of Science and Innovation/FEDER “Una manera de hacer Europa” (Retos Program, No. RTI2018-094739-B-I00 to JH & CC) and Fundacio La Marató de TV3 (No. 235/C/2019 to CC). LA is a recipient of an FI-AGAUR predoctoral fellowship. AV was funded by La Marató de TV3.

    وصف الملف: application/pdf

    العلاقة: info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2017-2020/RTI2018-094739-B-I00/ES/NUEVOS MECANISMOS DE DESREGULACION DE LA SEÑALIZACION POR CALCIO CONTRA EL GLIOBLASTOMA/; Reproducció del document publicat a: https://doi.org/10.1016/j.ceca.2022.102610Test; Cell Calcium, 2022, vol. 105, p. 102610; http://hdl.handle.net/10459.1/83504Test; https://doi.org/10.1016/j.ceca.2022.102610Test

  4. 4
    دورية أكاديمية

    المساهمون: Gobierno de España Ministerio de Ciencia e Innovación, Fundació la Marató de TV3, Ministerio de Ciencia e Innovación, European Regional Development Fund

    المصدر: Drug Discovery Today ; volume 27, issue 3, page 743-758 ; ISSN 1359-6446

    مصطلحات موضوعية: Drug Discovery, Pharmacology

  5. 5
    دورية أكاديمية

    الوصف: Over the last 20 years several studies have demonstrated a pivotal role of T‐type calcium channels (TTCCs) in tumor progression. Cytotoxic effects of TTCC pharmacological blockers have been reported in vitro and in preclinical models. However, the roles of TTCCs in cancer physiology are only beginning to be understood. We discuss current evidence for the signaling pathways and cellular processes stemming from TTCC activity, mainly inferred by inverse reasoning from pharmacological block and, only a few, by gene silencing or channel activation. A thorough analysis indicates that drug‐induced cytotoxicity is partially an off‐target effect. Dissection of on/off‐target activity is paramount in order to elucidate TTCC physiological roles, and to deliver efficacious therapies suited to different cancer types and stages. ; This work was funded by the Spanish Ministry of Science and Innovation/FEDER “Una manera de hacer Europa” (“Retos” Program, grant RTI2018‐094739‐BI00 to JH and CC) and Fundació La Marató de TV3 (grant 201909 to C.C.). LA held a predoctoral FI AGAUR fellowship and AV was funded from Fundació La Marató de TV3.

    وصف الملف: application/pdf

    العلاقة: info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2017-2020/RTI2018-094739-B-I00/ES/NUEVOS MECANISMOS DE DESREGULACION DE LA SEÑALIZACION POR CALCIO CONTRA EL GLIOBLASTOMA/; Reproducció del document publicat a: https://doi.org/10.1016/j.drudis.2021.11.021Test; Drug Discovery Today, 2022, vol. 27, núm. 3, p. 743-758; https://doi.org/10.1016/j.drudis.2021.11.021Test; http://hdl.handle.net/10459.1/72454Test

  6. 6
    دورية أكاديمية

    مصطلحات موضوعية: FAK, Glioblastoma, Proliferation, p62/SQSTM-1, p27/CDKN1B

    الوصف: Focal adhesion kinase (FAK) is a central component of focal adhesions that regulate cancer cell proliferation and migration. Here, we studied the effects of FAK inhibition in glioblastoma (GBM), a fast growing brain tumor that has a poor prognosis. Treating GBM cells with the FAK inhibitor PF-573228 induced a proliferative arrest and increased cell size. PF-573228 also reduced the growth of GBM neurospheres. These effects were associated with increased p27/CDKN1B levels and β-galactosidase activity, compatible with acquisition of senescence. Interestingly, FAK inhibition repressed the expression of the autophagy cargo receptor p62/SQSTM-1. Moreover, depleting p62 in GBM cells also induced a senescent-like phenotype through transcriptional upregulation of p27. Our results indicate that FAK inhibition arrests GBM cell proliferation, resulting in cell senescence, and pinpoint p62 as being key to this process. These findings highlight the possible therapeutic value of targeting FAK in GBM. ; This research was funded by Spanish Ministry of Science & Innovation (“Retos” Program), grant number RTI2018-094739-B-I00 to JH and CC. The APC was funded by Spanish Ministry of Science & Innovation

    وصف الملف: application/pdf

    العلاقة: info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2017-2020/RTI2018-094739-B-I00/ES/NUEVOS MECANISMOS DE DESREGULACION DE LA SEÑALIZACION POR CALCIO CONTRA EL GLIOBLASTOMA/; Reproducció del document publicat a https://doi.org/10.3390/cancers12051086Test; Cancers, 2020, vol. 12, núm. 5, p. E1086; http://hdl.handle.net/10459.1/68900Test; https://doi.org/10.3390/cancers12051086Test

  7. 7
    دورية أكاديمية

    الوصف: Hyperactivation of the Mitogen Activated Protein Kinase (MAPK) pathway is prevalent in melanoma, principally due to mutations in the B-Raf and NRas genes. MAPK inhibitors are effective only shortterm, and recurrence occurs due to functional redundancies or intertwined pathways. The remodeling of Ca2+ signaling is also common in melanoma cells, partly through the increased expression of T-type channels (TTCCs). Here we summarize current knowledge about the prognostic value and molecular targeting of TTCCs. Furthermore, we discuss recent evidence pointing to TTCCs as molecular switches for melanoma chemoresistance, which set the grounds for novel combined therapies against the advanced disease. ; This work was funded by the Spanish Ministry of Science, Innovation & Universities (“Retos” Program, grant RTI2018-094739-B-I00 to JH and CC). LA held a predoctoral fellowship from University of Lleida (UdL) and AV from UdL and IRBLleida (Diputació de Lleida).

    وصف الملف: application/pdf

    العلاقة: Versió postprint del document publicat a: https://doi.org/10.1016/j.bbcan.2020.188364Test; Biochimica et Biophysica Acta-Reviews on Cancer, 2020, vol. 1873, núm. 2, p. 188364; http://hdl.handle.net/10459.1/69470Test; https://doi.org/10.1016/j.bbcan.2020.188364Test

  8. 8
    دورية أكاديمية

    الوصف: T-type Ca2+ channels (TTCC) have been identified as key regulators of cancer cell cycle and survival. In vivo studies in glioblastoma (GBM) murine xenografts have shown that drugs able to block TTCC in vitro (such as tetralol derivatives mibefradil/NNC-55-096, or different 3,4-dihydroquinazolines) slow tumor progression. However, currently available TTCC pharmacological blockers have limited selectivity for TTCC, and are unable to distinguish between TTCC isoforms. Here we analyzed the expression of TTCC transcripts in human GBM cells and show a prevalence of Cav3.1 mRNAs. Infection of GBM cells with lentiviral particles carrying shRNA against Cav3.1 resulted in GBM cell death by apoptosis. We generated a murine GBM xenograft via subcutaneous injection of U87-MG GBM cells and found that tumor size was reduced when Cav3.1 expression was silenced. Furthermore, we developed an in vitro model of temozolomide-resistant GBM that showed increased expression of Cav3.1 accompanied by activation of macroautophagy. We confirmed a positive correlation between Cav3.1 and autophagic markers in both GBM cultures and biopsies. Of note, Cav3.1 knockdown resulted in transcriptional downregulation of p62/SQSTM1 and deficient autophagy. Together, these data identify Cav3.1 channels as potential targets for slowing GBM progression and recurrence based on their role in regulating autophagy. ; Paired biopsies of primary and recurrent GBMs were obtained through Biobank Networkfrom Carlos III Health Institute (NavarraBiomed, Toledo and Basque Biobanks). This work was funded by Instituto de Salud Carlos III/FEDER (“Una manera de hacer Europa”; PI13/01980 to JH). Work supported by IRBLleida Biobank (B.0000682) and PLATAFORMA BIOBANCOS PT17/0015/0027/. MCS and AV held predoctoral fellowships from University of Lleida (UdL).

    وصف الملف: application/pdf

    العلاقة: Versió postprint del document publicat a: https://doi.org/10.1158/0008-5472.CAN-18-1924Test; Cancer Research, 2019, vol. 79, núm. 8, p. 1857-1868; http://hdl.handle.net/10459.1/65789Test; https://doi.org/10.1158/0008-5472.CAN-18-1924Test

  9. 9
    دورية أكاديمية

    الوصف: T-Type calcium channels (TTCCs) are key regulators of membrane excitability, which is the reason why TTCC pharmacology is subject to intensive research in the neurological and cardiovascular fields. TTCCs also play a role in cancer physiology, and pharmacological blockers such as tetralols and dihydroquinazolines (DHQs) reduce the viability of cancer cells in vitro and slow tumor growth in murine xenografts. However, the available compounds are better suited to blocking TTCCs in excitable membranes rather than TTCCs contributing window currents at steady potentials. Consistently, tetralols and dihydroquinazolines exhibit cytostatic/cytotoxic activities at higher concentrations than those required for TTCC blockade, which may involve off-target effects. Gene silencing experiments highlight the targetability of TTCCs, but further pharmacological research is required for TTCC blockade to become a chemotherapeutic option. ; A.V. was the recipient of a predoctoral fellowship from the University of Lleida. S.S. was funded by the Marie Curie COFUND Programme. This work was supported by Instituto de Salud Carlos III/FEDER (PI13/01980 to J.H.).

    وصف الملف: application/pdf

    العلاقة: Versió postprint del document publicat a: https://doi.org/10.1016/j.molmed.2019.03.001Test; Trends in Molecular Medicine, 2019, vol. 25, núm. 7, p. 571-584; http://hdl.handle.net/10459.1/66361Test; https://doi.org/10.1016/j.molmed.2019.03.001Test