Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nanomaterials (Basel) ; 14(7)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38607114

RESUMEN

Cadmium sulfide (CdS)-based photocatalysts are prepared following a hydrothermal procedure (with CdCl2 and thiourea as precursors). The HydroThermal material annealed (CdS-HTa) is crystalline with a band gap of 2.31 eV. Photoelectrochemical investigation indicates a very reducing photo-potential of -0.9 V, which is very similar to that of commercial CdS. CdS-HTa, albeit having similar reducing properties, is more active than commercial CdS in the reductive dehalogenation of 2,2-dichloropropionic acid (dalapon) to propionic acid. Spectroscopic, electro-, and photoelectrochemical investigation show that photocatalytic properties of CdS are correlated to its electronic structure. The reductive dehalogenation of dalapon has a double significance: on one hand, it represents a demanding reductive process for a photocatalyst, and on the other hand, it has a peculiar interest in water treatment because dalapon can be considered a representative molecule of persistent organic pollutants and is one of the most important disinfection by products, whose removal from the water is the final obstacle to its complete reuse. HPLC-MS investigation points out that complete disappearance of dalapon passes through 2-monochloropropionic acid and leads to propionic acid as the final product. CdS-HTa requires very mild working conditions (room temperature, atmospheric pressure, natural pH), and it is stable and recyclable without significant loss of activity.

2.
J Pharm Biomed Anal ; 234: 115503, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37295189

RESUMEN

Metformin hydrochloride (MH) has recently been repurposed as an anticancer agent, showing antiproliferative activity in vitro and in vivo. In particular, experimental evidence has suggested its potential clinical efficacy in glioblastoma (GBM), a very aggressive tumor frequently characterized by gloomy prognosis. Unfortunately, the published literature concerning experimental applications of MH in glioblastoma animal models report no data on metformin levels reached in the brain, which, considering the high hydrophilicity of the drug, are likely very low. Therefore, new sensitive analytical methods to be applied on biological tissues are necessary to improve our knowledge of MH in vivo biodistribution and biological effects on tumors. In this research work, a GC-MS method for MH quantification in brain tissues is proposed. MH has been derivatized using N-methyl-bis(trifluoroacetamide), as already described in the literature, but the derivatization conditions have been optimized; moreover, deuterated MH has been selected as the best internal standard, after a comparative evaluation including other internal standards employed in published methods. After ascertaining method linearity, its accuracy, precision, specificity, repeatability, LOD and LOQ (0.373 µM and 1.242 µM, respectively, corresponding to 0.887 and 2.958 pmol/mg of wet tissue) have been evaluated on mouse brain tissue samples, obtained through a straightforward preparation procedure involving methanolic extraction from lyophilized brain homogenates and solid phase purification. The method has been validated on brain samples obtained from mice, either healthy or xenografted with GBM cells, receiving metformin dissolved in the drinking water. This analytical method can be usefully applied in preclinical studies aiming at clarifying MH mechanism of action in brain tumors.


Asunto(s)
Glioblastoma , Metformina , Animales , Ratones , Cromatografía de Gases y Espectrometría de Masas/métodos , Metformina/análisis , Glioblastoma/tratamiento farmacológico , Distribución Tisular , Encéfalo
4.
Nanomaterials (Basel) ; 13(3)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36770375

RESUMEN

A good photocatalyst maximizes the absorption of excitation light while reducing the recombination of photogenerated carriers. Among visible light responsive materials, CdS has good carrier transport capacity; however, its photostability is poor and limits its use. Here, the synthesis of a new hydrothermal CdS is reported, and post-synthesis annealing determines crystal properties and spectroscopic characteristics. The introduction of sulfur vacancies as intra band gap states is the key factor for the enhancement of photocatalytic activity. In fact, by spectroscopic and photo-electrochemical experiments, we demonstrate that sulfur vacancies act as an electron sink, favoring the charge transfer process to methyl orange. In addition, the studied hydrothermal CdS is characterized by very high stability, thus enabling a visible-light active photocatalyst that is overall recyclable, stable and more efficient than the commercial benchmark.

5.
Mol Psychiatry ; 27(11): 4790-4799, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36138130

RESUMEN

As a prime mover in Alzheimer's disease (AD), microglial activation requires membrane translocation, integration, and activation of the metamorphic protein chloride intracellular channel 1 (CLIC1), which is primarily cytoplasmic under physiological conditions. However, the formation and activation mechanisms of functional CLIC1 are unknown. Here, we found that the human antimicrobial peptide (AMP) LL-37 promoted CLIC1 membrane translocation and integration. It also activates CLIC1 to cause microglial hyperactivation, neuroinflammation, and excitotoxicity. In mouse and monkey models, LL-37 caused significant pathological phenotypes linked to AD, including elevated amyloid-ß, increased neurofibrillary tangles, enhanced neuronal death and brain atrophy, enlargement of lateral ventricles, and impairment of synaptic plasticity and cognition, while Clic1 knockout and blockade of LL-37-CLIC1 interactions inhibited these phenotypes. Given AD's association with infection and that overloading AMP may exacerbate AD, this study suggests that LL-37, which is up-regulated upon infection, may be a driving force behind AD by acting as an endogenous agonist of CLIC1.


Asunto(s)
Enfermedad de Alzheimer , Catelicidinas , Canales de Cloruro , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Catelicidinas/metabolismo , Catelicidinas/farmacología , Canales de Cloruro/metabolismo , Microglía/metabolismo
6.
Prog Neurobiol ; 216: 102313, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35760142

RESUMEN

We have uncovered a novel role for astrocytes-derived extracellular vesicles (EVs) in controlling intraneuronal Ca2+ concentration ([Ca2+]i) and identified transglutaminase-2 (TG2) as a surface-cargo of astrocytes-derived EVs. Incubation of hippocampal neurons with primed astrocyte-derived EVs have led to an increase in [Ca2+]i, unlike EVs from TG2-knockout astrocytes. Exposure of neurons or brain slices to extracellular TG2 promoted a [Ca2+]i rise, which was reversible upon TG2 removal and was dependent on Ca2+ influx through the plasma membrane. Patch-clamp and calcium imaging recordings revealed TG2-dependent neuronal membrane depolarization and activation of inward currents, due to the Na+/Ca2+-exchanger (NCX) operating in the reverse mode and indirect activation of L-type VOCCs, as indicated by VOCCs/NCX pharmacological inhibitors. A subunit of Na+/K+-ATPase was selected by comparative proteomics and identified as being functionally inhibited by extracellular TG2, implicating Na+/K+-ATPase inhibition in NCX reverse mode-switching leading to Ca2+ influx and higher basal [Ca2+]i. These data suggest that reactive astrocytes control intraneuronal [Ca2+]i through release of EVs with TG2 as responsible cargo, which could have a significant impact on synaptic activity in brain inflammation.


Asunto(s)
Astrocitos , Vesículas Extracelulares , Adenosina Trifosfatasas , Astrocitos/metabolismo , Calcio/metabolismo , Vesículas Extracelulares/metabolismo , Homeostasis , Humanos , Neuronas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Intercambiador de Sodio-Calcio/metabolismo
7.
Int J Mol Sci ; 23(10)2022 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-35628602

RESUMEN

Soluble amyloid ß (Aß) oligomers have been shown to be highly toxic to neurons and are considered to be a major cause of the neurodegeneration underlying Alzheimer's disease (AD). That makes soluble Aß oligomers a promising drug target. In addition to eliminating these toxic species from the patients' brain with antibody-based drugs, a new class of drugs is emerging, namely Aß aggregation inhibitors or modulators, which aim to stop the formation of toxic Aß oligomers at the source. Here, pharmacological data of the novel Aß aggregation modulator GAL-201 are presented. This small molecule (288.34 g/mol) exhibits high binding affinity to misfolded Aß1-42 monomers (KD = 2.5 ± 0.6 nM). Pharmacokinetic studies in rats using brain microdialysis are supportive of its oral bioavailability. The Aß oligomer detoxifying potential of GAL-201 has been demonstrated by means of single cell recordings in isolated hippocampal neurons (perforated patch experiments) as well as in vitro and in vivo extracellular monitoring of long-term potentiation (LTP, in rat transverse hippocampal slices), a cellular correlate for synaptic plasticity. Upon preincubation, GAL-201 efficiently prevented the detrimental effect on LTP mediated by Aß1-42 oligomers. Furthermore, the potential to completely reverse an already established neurotoxic process could also be demonstrated. Of particular note in this context is the self-propagating detoxification potential of GAL-201, leading to a neutralization of Aß oligomer toxicity even if GAL-201 has been stepwise removed from the medium (serial dilution), likely due to prion-like conformational changes in Aß1-42 monomer aggregates (trigger effect). The authors conclude that the data presented strongly support the further development of GAL-201 as a novel, orally available AD treatment with potentially superior clinical profile.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo , Plasticidad Neuronal , Ratas
8.
Molecules ; 27(9)2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35566275

RESUMEN

New composite photocatalysts have been obtained by chemical bath deposition of CdS on top of either nanostructured crystalline ZrO2 or TiO2 films previously deposited on conductive glass FTO. Their morphological, photoelectrochemical and photochemical properties have been investigated and compared. Time resolved spectroscopic, techniques show that in FTO/TiO2/CdS films the radiative recombination of charges, separated by visible illumination of CdS, is faster than in FTO/ZrO2/CdS, evidencing that carrier dynamics in the two systems is different. Photoelectrochemical investigation evidence a suppression of electron collection in ZrO2/CdS network, whereas electron injection from CdS to TiO2 is very efficient since trap states of TiO2 act as a reservoir for long lived electrons storage. This ability of FTO/TiO2/CdS films is used in the reductive cleavage of N=N bonds of some azo-dyes by visible light irradiation, with formation and accumulation of reduced aminic intermediates, identified by ESI-MS analysis. Needed protons are provided by sodium formate, a good hole scavenger that leaves no residue upon oxidation. FTO/TiO2/CdS has an approximately 100 meV driving force larger than FTO/ZrO2/CdS under illumination for azo-dye reduction and it is always about 10% more active than the seconds. The films showed very high stability and recyclability, ease of handling and recovering.


Asunto(s)
Compuestos Azo , Titanio , Catálisis , Colorantes , Luz , Titanio/química
9.
J Exp Clin Cancer Res ; 41(1): 53, 2022 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-35135603

RESUMEN

BACKGROUND: Chloride intracellular channel-1 (CLIC1) activity controls glioblastoma proliferation. Metformin exerts antitumor effects in glioblastoma stem cells (GSCs) inhibiting CLIC1 activity, but its low potency hampers its translation in clinical settings. METHODS: We synthesized a small library of novel biguanide-based compounds that were tested as antiproliferative agents for GSCs derived from human glioblastomas, in vitro using 2D and 3D cultures and in vivo in the zebrafish model. Compounds were compared to metformin for both potency and efficacy in the inhibition of GSC proliferation in vitro (MTT, Trypan blue exclusion assays, and EdU labeling) and in vivo (zebrafish model), migration (Boyden chamber assay), invasiveness (Matrigel invasion assay), self-renewal (spherogenesis assay), and CLIC1 activity (electrophysiology recordings), as well as for the absence of off-target toxicity (effects on normal stem cells and toxicity for zebrafish and chick embryos). RESULTS: We identified Q48 and Q54 as two novel CLIC1 blockers, characterized by higher antiproliferative potency than metformin in vitro, in both GSC 2D cultures and 3D spheroids. Q48 and Q54 also impaired GSC self-renewal, migration and invasion, and displayed low systemic in vivo toxicity. Q54 reduced in vivo proliferation of GSCs xenotransplanted in zebrafish hindbrain. Target specificity was confirmed by recombinant CLIC1 binding experiments using microscale thermophoresis approach. Finally, we characterized GSCs from GBMs spontaneously expressing low CLIC1 protein, demonstrating their ability to grow in vivo and to retain stem-like phenotype and functional features in vitro. In these GSCs, Q48 and Q54 displayed reduced potency and efficacy as antiproliferative agents as compared to high CLIC1-expressing tumors. However, in 3D cultures, metformin and Q48 (but not Q54) inhibited proliferation, which was dependent on the inhibition dihydrofolate reductase activity. CONCLUSIONS: These data highlight that, while CLIC1 is dispensable for the development of a subset of glioblastomas, it acts as a booster of proliferation in the majority of these tumors and its functional expression is required for biguanide antitumor class-effects. In particular, the biguanide-based derivatives Q48 and Q54, represent the leads to develop novel compounds endowed with better pharmacological profiles than metformin, to act as CLIC1-blockers for the treatment of CLIC1-expressing glioblastomas, in a precision medicine approach.


Asunto(s)
Biguanidas/uso terapéutico , Canales de Cloruro/metabolismo , Glioblastoma/genética , Glioma/genética , Células Madre Neoplásicas/metabolismo , Biguanidas/farmacología , Línea Celular Tumoral , Glioblastoma/patología , Glioma/patología , Humanos
10.
Anticancer Res ; 40(12): 6879-6884, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33288580

RESUMEN

BACKGROUND/AIM: Invasive bladder cancer mortality remains high despite progresses made in early diagnosis and surgical procedures. Thus, there is a need to define new markers for bladder cancer. CLIC1 has not been previously studied in bladder cancer and thus, we aimed to assess its immunohistochemical expression in relation to different stages of bladder cancer development. MATERIALS AND METHODS: Immunohistochemistry for CLIC1 was applied in 50 cases of muscle invasive bladder cancer. RESULTS: CLIC1 was not expressed in the normal urothelium, but a strong reaction was observed in dysplastic urothelium, carcinoma in situ and in 94% of the cases with invasive urothelial carcinoma; however, it was not expressed in squamous cell carcinoma cases. No correlation was found between the immunohistochemical expression of CLIC1 and the stage and grade of the tumour. CONCLUSION: CLIC1 was overexpressed in urinary bladder dysplastic epithelium, carcinoma in situ and invasive carcinoma. CLIC1 constitutes a new potential marker of invasive bladder cancer.


Asunto(s)
Canales de Cloruro/genética , Expresión Génica , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Anciano , Biomarcadores de Tumor , Canales de Cloruro/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad
11.
Nanomaterials (Basel) ; 10(11)2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33126556

RESUMEN

UV-photoexcitation of TiO2 in contact with aqueous solutions of azo dyes does not imply only its photocatalytic degradation, but the reaction fate of the dye depends on the experimental conditions. In fact, we demonstrate that the presence of sodium formate is the switch from a degradative pathway of the dye to its transformation into useful products. Laser flash photolysis experiments show that charge separation is extremely long lived in nanostructured TiO2 thin films, making them suitable to drive both oxidation and reduction reactions. ESR spin trapping and photoluminescence experiments demonstrate that formate anions are very efficient in intercepting holes, thereby inhibiting OH radicals formation. Under these conditions, electrons promoted in the conduction band of TiO2 and protons deriving from the oxidation of formate on photogenerated holes lead to the reductive cleavage of N=N bonds with formation and accumulation of reduced intermediates. Negative ion ESI-MS findings provide clear support to point out this new mechanism. This study provides a facile solution for realizing together wastewater purification and photocatalytic conversion of a waste (discharged dye) into useful products (such as sulfanilic acid used again for synthesis of new azo dyes). Moreover, the use of TiO2 deposited on an FTO (Fluorine Tin Oxide) glass circumvents all the difficulties related to the use of slurries. The obtained photocatalyst is easy to handle and to recover and shows an excellent stability allowing complete recyclability.

12.
Mol Neurobiol ; 57(12): 5336-5351, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32880860

RESUMEN

PCDH19 encodes for protocadherin-19 (PCDH19), a cell-adhesion molecule of the cadherin superfamily preferentially expressed in the brain. PCDH19 mutations cause a neurodevelopmental syndrome named epileptic encephalopathy, early infantile, 9 (EIEE9) characterized by seizures associated with cognitive and behavioral deficits. We recently reported that PCDH19 binds the alpha subunits of GABAA receptors (GABAARs), modulating their surface availability and miniature inhibitory postsynaptic currents (mIPSCs). Here, we investigated whether PCDH19 regulatory function on GABAARs extends to the extrasynaptic receptor pool that mediates tonic current. In fact, the latter shapes neuronal excitability and network properties at the base of information processing. By combining patch-clamp recordings in whole-cell and cell-attached configurations, we provided a functional characterization of primary hippocampal neurons from embryonic rats of either sex expressing a specific PCDH19 short hairpin (sh)RNA. We first demonstrated that PCDH19 downregulation reduces GABAAR-mediated tonic current, evaluated by current shift and baseline noise analysis. Next, by single-channel recordings, we showed that PCDH19 regulates GABAARs kinetics without altering their conductance. In particular, GABAARs of shRNA-expressing neurons preferentially exhibit brief openings at the expense of long ones, thus displaying a flickering behavior. Finally, we showed that PCDH19 downregulation reduces the rheobase and increases the frequency of action potential firing, thus indicating neuronal hyperexcitability. These findings establish PCDH19 as a critical determinant of GABAAR-mediated tonic transmission and GABAARs gating, and provide the first mechanistic insights into PCDH19-related hyperexcitability and comorbidities.


Asunto(s)
Potenciales de Acción , Cadherinas/metabolismo , Epilepsia/metabolismo , Epilepsia/fisiopatología , Hipocampo/patología , Inhibición Neural/fisiología , Neuronas/patología , Receptores de GABA-A/metabolismo , Animales , Regulación hacia Abajo , Cinética , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley
14.
Int J Mol Sci ; 21(4)2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-32098256

RESUMEN

Pathologies that lead to neurodegeneration in the central nervous system (CNS) represent a major contemporary medical challenge. Neurodegenerative processes, like those that occur in Alzheimer's disease (AD) are progressive, and at the moment, they are unstoppable. Not only is an adequate therapy missing but diagnosis is also extremely complicated. The most reliable method is the measurement of beta amyloid and tau peptides concentration in the cerebrospinal fluid (CSF). However, collecting liquid samples from the CNS is an invasive procedure, thus it is not suitable for a large-scale prevention program. Ideally, blood testing is the most manageable and appropriate diagnostic procedure for a massive population screening. Recently, a few candidates, including proteins or microRNAs present in plasma/serum have been identified. The aim of the present work is to propose the chloride intracellular channel 1 (CLIC1) protein as a potential marker of neurodegenerative processes. CLIC1 protein accumulates in peripheral blood mononuclear cells (PBMCs), and increases drastically when the CNS is in a chronic inflammatory state. In AD patients, both immunolocalization and mRNA quantification are able to show the behavior of CLIC1 during a persistent inflammatory state of the CNS. In particular, confocal microscopy analysis and electrophysiological measurements highlight the significant presence of transmembrane CLIC1 (tmCLIC1) in PBMCs from AD patients. Recent investigations suggest that tmCLIC1 has a very specific role. This provides an opportunity to use blood tests and conventional technologies to discriminate between healthy individuals and patients with ongoing neurodegenerative processes.


Asunto(s)
Enfermedad de Alzheimer/sangre , Membrana Celular/metabolismo , Canales de Cloruro/sangre , Monocitos/metabolismo , Anciano , Enfermedad de Alzheimer/patología , Biomarcadores/sangre , Membrana Celular/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/patología
15.
Cells ; 8(11)2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31752162

RESUMEN

Cellular prion protein (PrPC) is a membrane-anchored glycoprotein representing the physiological counterpart of PrP scrapie (PrPSc), which plays a pathogenetic role in prion diseases. Relatively little information is however available about physiological role of PrPC. Although PrPC ablation in mice does not induce lethal phenotypes, impairment of neuronal and bone marrow plasticity was reported in embryos and adult animals. In neurons, PrPC stimulates neurite growth, prevents oxidative stress-dependent cell death, and favors antiapoptotic signaling. However, PrPC activity is not restricted to post-mitotic neurons, but promotes cell proliferation and migration during embryogenesis and tissue regeneration in adult. PrPC acts as scaffold to stabilize the binding between different membrane receptors, growth factors, and basement proteins, contributing to tumorigenesis. Indeed, ablation of PrPC expression reduces cancer cell proliferation and migration and restores cell sensitivity to chemotherapy. Conversely, PrPC overexpression in cancer stem cells (CSCs) from different tumors, including gliomas-the most malignant brain tumors-is predictive for poor prognosis, and correlates with relapses. The mechanisms of the PrPC role in tumorigenesis and its molecular partners in this activity are the topic of the present review, with a particular focus on PrPC contribution to glioma CSCs multipotency, invasiveness, and tumorigenicity.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas PrPC/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/genética , Humanos , Invasividad Neoplásica , Regeneración Nerviosa , Proteínas PrPC/genética
16.
Cancer Genomics Proteomics ; 16(4): 299-307, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31243111

RESUMEN

BACKGROUND/AIM: Chloride intracellular channel 1 (CLIC1) represents a promising target for personalized therapy. Our aim was to assess CLIC1 expression in clear cell renal cell carcinoma (cc RCC) and identify its possible prognostic role. MATERIALS AND METHODS: Fifty cases of cc RCC were evaluated and selected for immunohistochemistry. CLIC1 expression was correlated with tumor grade, invasion and heterogeneity. RESULTS: A total of 87.5% of the cases were CLIC1 positive, with either a homogeneous (31.42%) or a heterogeneous (68.57%) pattern. Low, mild and strong CLIC1 expressing tumors were defined based on nuclear (N), cytoplasmic (C), membrane (M) or combinations of them (NC, NM, CM, NCM) in terms of CLIC1 distribution. A significant correlation was found between tumor grade and percent of positive tumor cells (p=0.017). For G3 tumors, CLIC1 cytoplasmic expression was strongly correlated with high expression status (p=0.025) and tumor heterogeneity (p=0.004). CLIC1 expression was also correlated with metastasis (p=0.046). CONCLUSION: We defined four cc RCC groups depending on G, CLIC1 expression and pattern: i) G3/NM/low CLIC1+, ii) G2/CM/mild CLIC1+ iii) G1 or G2/NM or CM /high CLIC1+, and iv) G2/M /high CLIC1.


Asunto(s)
Carcinoma de Células Renales/genética , Canales de Cloruro/metabolismo , Carcinoma de Células Renales/patología , Femenino , Humanos , Masculino
17.
Front Oncol ; 9: 135, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30918838

RESUMEN

The lack of in-depth knowledge about the molecular determinants of glioblastoma (GBM) occurrence and progression, combined with few effective and BBB crossing-targeted compounds represents a major challenge for the discovery of novel and efficacious drugs for GBM. Among relevant molecular factors controlling the aggressive behavior of GBM, chloride intracellular channel 1 (CLIC1) represents an emerging prognostic and predictive biomarker, as well as a promising therapeutic target. CLIC1 is a metamorphic protein, co-existing as both soluble cytoplasmic and membrane-associated conformers, with the latter acting as chloride selective ion channel. CLIC1 is involved in several physiological cell functions and its abnormal expression triggers tumor development, favoring tumor cell proliferation, invasion, and metastasis. CLIC1 overexpression is associated with aggressive features of various human solid tumors, including GBM, in which its expression level is correlated with poor prognosis. Moreover, increasing evidence shows that modification of microglia ion channel activity, and CLIC1 in particular, contributes to the development of different neuropathological states and brain tumors. Intriguingly, CLIC1 is constitutively active within cancer stem cells (CSCs), while it seems less relevant for the survival of non-CSC GBM subpopulations and for normal cells. CSCs represent GBM development and progression driving force, being endowed with stem cell-like properties (self-renewal and differentiation), ability to survive therapies, to expand and differentiate, causing tumor recurrence. Downregulation of CLIC1 results in drastic inhibition of GBM CSC proliferation in vitro and in vivo, making the control of the activity this of channel a possible innovative pharmacological target. Recently, drugs belonging to the biguanide class (including metformin) were reported to selectively inhibit CLIC1 activity in CSCs, impairing their viability and invasiveness, but sparing normal stem cells, thus representing potential novel antitumor drugs with a safe toxicological profile. On these premises, we review the most recent insights into the biological role of CLIC1 as a potential selective pharmacological target in GBM. Moreover, we examine old and new drugs able to functionally target CLIC1 activity, discussing the challenges and potential development of CLIC1-targeted therapies.

18.
Respir Res ; 19(1): 198, 2018 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-30290809

RESUMEN

BACKGROUND: Although pharmacological treatment has increased the average life expectancy of patients with cystic fibrosis, the median survival of females is shorter than that of males. In vitro and in vivo studies have shown that estrogens play a relevant role in the disease progression. The aim of this study was to investigate the effects of 17ß-estradiol and tamoxifen citrate (TMX) on calcium-activated chloride channel (CaCC) currents in human bronchial epithelial cells carrying the ΔPhe508-CFTR mutation both in homozygosis and in heterozygosis. METHODS: Perforated patch clamp experiments were performed on single cells of the immortalized cell lines CFBE and IB3-1. Gramicidin (10 or 20 µM) was added to the electrode solution to reach the whole cell configuration. The electrical stimulation protocol consisted of square voltages ranging from - 80 to + 80 mV, in steps of 20 mV and with a duration of 800 msec. RESULTS: The presence of 17ß-estradiol significantly reduced the CaCC currents, both in basal conditions and in the presence of ATP (100 µM). The addition of TMX (10 µM) completely restored the currents abolished by 17ß-estradiol, in basal conditions and after stimulation with ATP in both CFBE and IB3-1 cells. TMX had a strong, direct action on membrane current density, which significantly increased more than 4-fold in both cases. The membrane current stimulation produced by TMX was further enhanced by the addition of ATP. CFBE cells incubated for 24 h with 3 µM VX-809 (a CFTR corrector) and then acutely stimulated with VX-770 (a CFTR potentiator) in the presence of forskolin, showed an increase of chloride currents which were abolished by Inh-172. The chloride current density induced by TMX + ATP was, on average, greater than that obtained with VX-809 + VX-770 + forskolin. The currents elicited by TMX + ATP were abolished by the addition of NPPB, a CaCC inhibitor. The combined administration of TMX/ATP and VXs/FSK had an additional effect on chloride currents. CONCLUSIONS: Our results show that TMX restores CaCC currents inhibited by 17ß-estradiol and directly activates the transmembrane chloride currents potentiated by ATP, an effect which is mutation independent. The combined effect of TMX with current used treatments for cystic fibrosis could be of benefit to patients.


Asunto(s)
Canales de Cloruro/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Moduladores de los Receptores de Estrógeno/farmacología , Mutación Puntual/genética , Mucosa Respiratoria/efectos de los fármacos , Tamoxifeno/farmacología , Línea Celular Transformada , Canales de Cloruro/fisiología , Estradiol/farmacología , Humanos , Mucosa Respiratoria/fisiología
19.
Front Pharmacol ; 9: 899, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30186163

RESUMEN

The antidiabetic biguanide metformin exerts antiproliferative effects in different solid tumors. However, during preclinical studies, metformin concentrations required to induce cell growth arrest were invariably within the mM range, thus difficult to translate in a clinical setting. Consequently, the search for more potent metformin derivatives is a current goal for new drug development. Although several cell-specific intracellular mechanisms contribute to the anti-tumor activity of metformin, the inhibition of the chloride intracellular channel 1 activity (CLIC1) at G1/S transition is a key events in metformin antiproliferative effect in glioblastoma stem cells (GSCs). Here we tested several known biguanide-related drugs for the ability to affect glioblastoma (but not normal) stem cell viability, and in particular: phenformin, a withdrawn antidiabetic drug; moroxydine, a former antiviral agent; and proguanil, an antimalarial compound, all of them possessing a linear biguanide structure as metformin; moreover, we evaluated cycloguanil, the active form of proguanil, characterized by a cyclized biguanide moiety. All these drugs caused a significant impairment of GSC proliferation, invasiveness, and self-renewal reaching IC50 values significantly lower than metformin, (range 0.054-0.53 mM vs. 9.4 mM of metformin). All biguanides inhibited CLIC1-mediated ion current, showing the same potency observed in the antiproliferative effects, with the exception of proguanil which was ineffective. These effects were specific for GSCs, since no (or little) cytotoxicity was observed in normal umbilical cord mesenchymal stem cells, whose viability was not affected by metformin and moroxydine, while cycloguanil and phenformin induced toxicity only at much higher concentrations than required to reduce GSC proliferation or invasiveness. Conversely, proguanil was highly cytotoxic also for normal mesenchymal stem cells. In conclusion, the inhibition of CLIC1 activity represents a biguanide class-effect to impair GSC viability, invasiveness, and self-renewal, although dissimilarities among different drugs were observed as far as potency, efficacy and selectivity as CLIC1 inhibitors. Being CLIC1 constitutively active in GSCs, this feature is relevant to grant the molecules with high specificity toward GSCs while sparing normal cells. These results could represent the basis for the development of novel biguanide-structured molecules, characterized by high antitumor efficacy and safe toxicological profile.

20.
Mol Cancer Ther ; 17(11): 2451-2461, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30135216

RESUMEN

Glioblastoma (GB) is the most lethal, aggressive, and diffuse brain tumor. The main challenge for successful treatment is targeting the cancer stem cell (CSC) subpopulation responsible for tumor origin, progression, and recurrence. Chloride Intracellular Channel 1 (CLIC1), highly expressed in CSCs, is constitutively present in the plasma membrane where it is associated with chloride ion permeability. In vitro, CLIC1 inhibition leads to a significant arrest of GB CSCs in G1 phase of the cell cycle. Furthermore, CLIC1 knockdown impairs tumor growth in vivo Here, we demonstrate that CLIC1 membrane localization and function is specific for GB CSCs. Mesenchymal stem cells (MSC) do not show CLIC1-associated chloride permeability, and inhibition of CLIC1 protein function has no influence on MSC cell-cycle progression. Investigation of the basic functions of GB CSCs reveals a constitutive state of oxidative stress and cytoplasmic alkalinization compared with MSCs. Both intracellular oxidation and cytoplasmic pH changes have been reported to affect CLIC1 membrane functional expression. We now report that in CSCs these three elements are temporally linked during CSC G1-S transition. Impeding CLIC1-mediated chloride current prevents both intracellular ROS accumulation and pH changes. CLIC1 membrane functional impairment results in GB CSCs resetting from an allostatic tumorigenic condition to a homeostatic steady state. In contrast, inhibiting NADPH oxidase and NHE1 proton pump results in cell death of both GB CSCs and MSCs. Our results show that CLIC1 membrane protein is crucial and specific for GB CSC proliferation, and is a promising pharmacologic target for successful brain tumor therapies. Mol Cancer Ther; 17(11); 2451-61. ©2018 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , Canales de Cloruro/metabolismo , Fase G1 , Glioblastoma/patología , Células Madre Neoplásicas/patología , Especies Reactivas de Oxígeno/metabolismo , Fase S , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Ciclina D1/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Persona de Mediana Edad , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Células Madre Neoplásicas/metabolismo , Intercambiador 1 de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...