Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Curr Med Chem ; 31(11): 1361-1403, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37013427

RESUMEN

The purinergic P2X7 receptor (P2X7R), an ATP-gated non-selective cation channel, has emerged as a gatekeeper of inflammation that controls the release of proinflammatory cytokines. As a key player in initiating the inflammatory signaling cascade, the P2X7 receptor is currently under intense scrutiny as a target for the treatment of different pathologies, including chronic inflammatory disorders (rheumatoid arthritis and osteoarthritis), chronic neuropathic pain, mood disorders (depression and anxiety), neurodegenerative diseases, ischemia, cancer (leukemia), and many others. For these reasons, pharmaceutical companies have invested in discovering compounds able to modulate the P2X7R and filed many patent applications. This review article presents an account of P2X7R structure, function, and tissue distribution, emphasizing its role in inflammation. Next, we illustrate the different chemical classes of non-competitive P2X7R antagonists reported by highlighting their properties and qualities as clinical candidates for treating inflammatory disorders and neurodegenerative diseases. We also discuss the efforts to develop effective Positron Emission Tomography (PET) radioligands to progress the understanding of the pathomechanisms of neurodegenerative disorders, to provide evidence of drug-target engagement, and to assist clinical dose selection for novel drug therapies.


Asunto(s)
Neoplasias , Enfermedades Neurodegenerativas , Humanos , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad , Inflamación/tratamiento farmacológico , Inflamación/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Receptores Purinérgicos P2X7/uso terapéutico
2.
Acta Pharmacol Sin ; 44(12): 2504-2524, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37482570

RESUMEN

Sinomenine (SIN) is an isoquinoline alkaloid isolated from Sinomenii Caulis, a traditional Chinese medicine used to treat rheumatoid arthritis (RA). Clinical trials have shown that SIN has comparable efficacy to methotrexate in treating patients with RA but with fewer adverse effects. In this study, we explored the anti-inflammatory effects and therapeutic targets of SIN in LPS-induced RAW264.7 cells and in collagen-induced arthritis (CIA) mice. LPS-induced RAW264.7 cells were pretreated with SIN (160, 320, 640 µM); and CIA mice were administered SIN (25, 50 and 100 mg·kg-1·d-1, i.p.) for 30 days. We first conducted a solvent-induced protein precipitation (SIP) assay in LPS-stimulated RAW264.7 cells and found positive evidence for the direct binding of SIN to guanylate-binding protein 5 (GBP5), which was supported by molecular simulation docking, proteomics, and binding affinity assays (KD = 3.486 µM). More importantly, SIN treatment markedly decreased the expression levels of proteins involved in the GBP5/P2X7R-NLRP3 pathways in both LPS-induced RAW264.7 cells and the paw tissue of CIA mice. Moreover, the levels of IL-1ß, IL-18, IL-6, and TNF-α in both the supernatant of inflammatory cells and the serum of CIA mice were significantly reduced. This study illustrates a novel anti-inflammatory mechanism of SIN; SIN suppresses the activity of NLRP3-related pathways by competitively binding GBP5 and downregulating P2X7R protein expression, which ultimately contributes to the reduction of IL-1ß and IL-18 production. The binding specificity of SIN to GBP5 and its inhibitory effect on GBP5 activity suggest that SIN has great potential as a specific GBP5 antagonist.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Humanos , Ratones , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Interleucina-18/efectos adversos , Receptores Purinérgicos P2X7/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR , Lipopolisacáridos/farmacología , Transducción de Señal , Artritis Reumatoide/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Proteínas de Unión al GTP
3.
Cell Signal ; 106: 110641, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36858191

RESUMEN

The purinergic signaling pathway is the oldest evolutionary transmitter system that regulates a wide array of physiological and pathophysiological processes in central nervous system. However, the question of how the purinergic compounds interact with administrated drugs is rarely addressed. We aimed to clarify the interplay between purinergic signaling and chemotherapeutic drug temozolomide (TMZ) in human glioma cell line. We applied an initial retinoic acid-induced differentiation of A172 glioma cells and tested the P2X7 receptor expression in undifferentiated and differentiated gliomas. We compared the P2X7 receptor agonists/antagonists influence and their co-action with TMZ in both cell types through assessment of cell proliferation, viability and migrative properties. Molecular docking allowed to indicate the potential binding site for TMZ in the structure of hP2X7 receptor. Differentiated cells turned out to be more susceptible to ATP and TMZ alone but also to the concerted action of TMZ and ATP. Enhanced effects triggered by ATP and TMZ treatment include the decreased by 70% viability, and reduced migration ability of differentiated A172 glioma cells. Noteworthy, these results can be achieved already at low non-toxic ATP concentration and at reduced to 125 µM effective concentration of TMZ. Therefore, ATP molecules must be present and maintained at appropriate concentration in glioma cells microenvironment to achieve their co-action with TMZ and enhanced anti-cancer activity. All that, in turn, could shorten the therapy, increase its efficacy and limit the side effects for the patient. Our purinergic approach creates a promising perspective for developing novel combined oncological therapies.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Receptores Purinérgicos P2X7/uso terapéutico , Simulación del Acoplamiento Molecular , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Adenosina Trifosfato , Línea Celular Tumoral , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis , Microambiente Tumoral
4.
J Transl Med ; 21(1): 132, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36803784

RESUMEN

BACKGROUND: Osteosarcoma is the most common malignant tumor in bone and its prognosis has reached a plateau in the past few decades. Recently, metabolic reprogramming has attracted increasing attention in the field of cancer research. In our previous study, P2RX7 has been identified as an oncogene in osteosarcoma. However, whether and how P2RX7 promotes osteosarcoma growth and metastasis through metabolic reprogramming remains unexplored. METHODS: We used CRISPR/Cas9 genome editing technology to establish P2RX7 knockout cell lines. Transcriptomics and metabolomics were performed to explore metabolic reprogramming in osteosarcoma. RT-PCR, western blot and immunofluorescence analyses were used to determine gene expression related to glucose metabolism. Cell cycle and apoptosis were examined by flowcytometry. The capacity of glycolysis and oxidative phosphorylation were assessed by seahorse experiments. PET/CT was carried out to assess glucose uptake in vivo. RESULTS: We demonstrated that P2RX7 significantly promotes glucose metabolism in osteosarcoma via upregulating the expression of genes related to glucose metabolism. Inhibition of glucose metabolism largely abolishes the ability of P2RX7 to promote osteosarcoma progression. Mechanistically, P2RX7 enhances c-Myc stabilization by facilitating nuclear retention and reducing ubiquitination-dependent degradation. Furthermore, P2RX7 promotes osteosarcoma growth and metastasis through metabolic reprogramming in a predominantly c-Myc-dependent manner. CONCLUSIONS: P2RX7 plays a key role in metabolic reprogramming and osteosarcoma progression via increasing c-Myc stability. These findings provide new evidence that P2RX7 might be a potential diagnostic and/or therapeutic target for osteosarcoma. Novel therapeutic strategies targeting metabolic reprogramming appear to hold promise for a breakthrough in the treatment of osteosarcoma.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Osteosarcoma/genética , Osteosarcoma/patología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Glucosa , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/uso terapéutico
5.
Epilepsia ; 64(2): 511-523, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36507708

RESUMEN

OBJECTIVE: The P2X7 receptor (P2X7R) is an important contributor to neuroinflammation, responding to extracellularly released adenosine triphosphate. Expression of the P2X7R is increased in the brain in experimental and human epilepsy, and genetic or pharmacologic targeting of the receptor can reduce seizure frequency and severity in preclinical models. Experimentally induced seizures also increase levels of the P2X7R in blood. Here, we tested 18 F-JNJ-64413739, a positron emission tomography (PET) P2X7R antagonist, as a potential noninvasive biomarker of seizure-damage and epileptogenesis. METHODS: Status epilepticus was induced via an intra-amygdala microinjection of kainic acid. Static PET studies (30 min duration, initiated 30 min after tracer administration) were conducted 48 h after status epilepticus via an intravenous injection of 18 F-JNJ-64413739. PET images were coregistered with a brain magnetic resonance imaging atlas, tracer uptake was determined in the different brain regions and peripheral organs, and values were correlated to seizure severity during status epilepticus. 18 F-JNJ-64413739 was also applied to ex vivo human brain slices obtained following surgical resection for intractable temporal lobe epilepsy. RESULTS: P2X7R radiotracer uptake correlated strongly with seizure severity during status epilepticus in brain structures including the cerebellum and ipsi- and contralateral cortex, hippocampus, striatum, and thalamus. In addition, a correlation between radiotracer uptake and seizure severity was also evident in peripheral organs such as the heart and the liver. Finally, P2X7R radiotracer uptake was found elevated in brain sections from patients with temporal lobe epilepsy when compared to control. SIGNIFICANCE: Taken together, our data suggest that P2X7R-based PET imaging may help to identify seizure-induced neuropathology and temporal lobe epilepsy patients with increased P2X7R levels possibly benefitting from P2X7R-based treatments.


Asunto(s)
Epilepsia del Lóbulo Temporal , Estado Epiléptico , Ratones , Humanos , Masculino , Animales , Epilepsia del Lóbulo Temporal/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Estado Epiléptico/inducido químicamente , Estado Epiléptico/diagnóstico por imagen , Estado Epiléptico/metabolismo , Convulsiones/tratamiento farmacológico
6.
Phytother Res ; 37(5): 1771-1786, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36444395

RESUMEN

Triple-negative breast cancer (TNBC) accounts for 10-20% of all human ductal adenocarcinomas and has a poor prognosis relative to other subtypes because of its high propensity to develop metastases. Here, the anticancer effects of asiaticoside (AC) against TNBC and the possible underlying mechanism were examined. We found that AC inhibited the TGF-ß1 expression and the SMAD2/3 phosphorylation in TNBC cells, thereby impairing the TGF-ß/SMAD signaling. AC inhibited the migration, invasion, and epithelial-mesenchymal transition (EMT) of TNBC cells by suppressing the TGF-ß/SMAD signaling. Meanwhile, AC inhibited the lung metastasis of TNBC cells in vivo and the expression of p-SMAD2/3 and vimentin, and increased the expression of E-cadherin and ZO-1 in the lung. Peroxisome proliferator activated receptor gamma (PPARG) was identified as a potential target of AC. AC increased PPARG expression, while PPARG knockdown attenuated the therapeutic effect of AC. AC-mediated PPARG overexpression suppressed the transcription of P2X purinoceptor 7 (P2RX7). The restoration of P2RX7 reversed the therapeutic effect of AC. These results suggested that AC blocked P2RX7-mediated TGF-ß/SMAD signaling by increasing PPARG expression, thereby suppressing EMT in TNBC.


Asunto(s)
PPAR gamma , Neoplasias de la Mama Triple Negativas , Humanos , PPAR gamma/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Transición Epitelial-Mesenquimal , Línea Celular Tumoral , Receptores Purinérgicos P2X7/uso terapéutico
7.
Pharmacol Ther ; 237: 108228, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35716953

RESUMEN

The purinoceptor 7 receptor (P2X7R) plays an important role in promoting inflammation in response to accumulating damage-associated molecular patterns (DAMPs) released from stressed or apoptotic cells and has been connected to various pathological conditions. The initial investment by large pharmaceutical companies such as AstraZeneca and Pfizer led to the development of several classes of P2X7R antagonists for the treatment of rheumatoid arthritis and Crohn's disease. While these compounds showed early promise as therapeutic agents and were found to potently inhibit adenosine triphosphate (ATP)-induced release of interleukin 1 beta (IL-1ß) in patient-derived monocytes primed with lipopolysaccharide (LPS), they failed to elicit a therapeutic benefit in phase II clinical trials. Within the last 10 years, a wealth of strong preclinical and clinical evidence has implicated IL-1ß as an aggressor in the development and progression of cardiovascular diseases, a cytokine modulated by the P2X7R. On account of the immune-mediated events that regulate atherosclerosis, antagonism of the P2X7R has been proposed as a therapeutic strategy due to the unique functionality of the receptor as an instigator of sterile inflammation. Here, we review the success and failures in P2X7R drug development to evaluate the major barriers to successful clinical translation of P2X7R antagonists. These avenues should be addressed by researchers and pharmaceutical companies to ensure future clinical success in the treatment of CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria , Antagonistas del Receptor Purinérgico P2X , Adenosina Trifosfato , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Humanos , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Preparaciones Farmacéuticas , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Receptores Purinérgicos P2X7/uso terapéutico
8.
J Vet Med Sci ; 84(4): 610-617, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35249909

RESUMEN

Postoperative ileus (POI) is a postsurgical gastrointestinal motility dysfunction caused by mechanical stress to the intestine during abdominal surgery. POI leads to nausea and vomiting reduced patient quality of life, as well as high medical costs and extended hospitalization. Intestinal inflammation caused by macrophages and neutrophils is thought to be important in the mechanism of POI. Surgery-associated tissue injury and inflammation induce the release of adenosine triphosphate (ATP) from injured cells. Released ATP binds the purinergic P2X7 receptor (P2X7R) expressed on inflammatory cells, inducing the secretion of inflammatory mediators. P2X7R antagonists are thought to be important mediators of the first step in the inflammation process, and studies in chemically induced colitis models confirmed that P2X7R antagonists exhibit anti-inflammatory effects. Therefore, we hypothesized that P2X7R plays an important role in POI. POI models were generated from C57BL/6J mice. Mice were treated with P2X7R antagonist A438079 (34 mg/kg) 30 min before and 2 hr after intestinal manipulation (IM). Inflammatory cell infiltration and gastrointestinal transit were measured. A438079 ameliorated macrophage and neutrophil infiltration in the POI model. Impaired intestinal transit improved following A438079 treatment. P2X7R was expressed on both infiltrating and resident macrophages in the inflamed ileal muscle layer. The P2X7R antagonist A438079 exhibits anti-inflammatory effects via P2X7R expressed on macrophages and therefore could be a target in the treatment of POI.


Asunto(s)
Ileus , Enfermedades de los Roedores , Adenosina Trifosfato , Animales , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Ileus/tratamiento farmacológico , Ileus/etiología , Ileus/metabolismo , Ileus/veterinaria , Inflamación/tratamiento farmacológico , Inflamación/veterinaria , Ratones , Ratones Endogámicos C57BL , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/veterinaria , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Calidad de Vida , Receptores Purinérgicos P2X7/uso terapéutico
9.
Prog Neurobiol ; 208: 102173, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34516970

RESUMEN

Tauopathies are neurodegenerative diseases characterized by the presence of aberrant intraneuronal aggregates of hyperphosphorylated Tau protein. Recent studies suggest that associated chronic neuroinflammation may contribute to the pathological Tau dissemination. However, the underlying molecular mechanisms remain unknown. Since purinergic P2X7 receptors (P2X7) can sense the rise of extracellular ATP levels associated with neuroinflammation, its involvement in neurodegeneration-associated inflammation was suggested. We found a P2X7 upregulation in patients diagnosed with different tauopathies and in a tauopathy mouse model, P301S mice. In vivo pharmacological or genetic blockade of P2X7 reverted microglial activation in P301S mice leading to a reduction in microglial migratory, secretory, and proliferative capacities, and promoting phagocytic function. Furthermore, it reduced the intraneuronal phosphorylated Tau levels in a GSK3-dependent way and increased extracellular phosphorylated Tau levels by reducing the expression of ectoenzyme TNAP. Accordingly, pharmacological or genetic blockade of P2X7 improved the cellular survival, motor and memory deficits and anxiolytic profile in P301S mice. Contrary, P2X7 overexpression caused a significant worsening of Tau-induced toxicity and aggravated the deteriorated motor and memory deficits in P301S mice. Our results indicate that P2X7 plays a deleterious role in tauopathies and suggest that its blockade may be a promising approach to treat Tauopathies.


Asunto(s)
Receptores Purinérgicos P2X7 , Tauopatías , Animales , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3/uso terapéutico , Humanos , Ratones , Ratones Transgénicos , Receptores Purinérgicos P2X7/uso terapéutico , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Proteínas tau/metabolismo
10.
Phytother Res ; 33(9): 2319-2328, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31264271

RESUMEN

P2X7 is a purinergic receptor involved in important physiological functions and pathological processes, such as inflammation, neurodegeneration, and pain. Despite its relevance, there is no selective antagonist useful in the treatment of diseases related to this receptor. In this context, research for a selective, safe, and potent antagonist compound that can be used in clinical therapy has been growing. In this work, we evaluated the potential antagonistic activity of three fungal extracts, namely, Vishniacozyma victoriae, Metschnikowia australis, and Ascomycota sp., which were discovered in a high-throughput screening campaign to search for new antagonists for P2X7R from natural products. First, the IC50 values of these fungal extracts were determined in J774.G8 (murine macrophage cell line) and U937 (human monocyte cell line) cells through dye uptake assays. The IC50 values of V. victoriae were 2.6 and 0.92 µg/mL, M. australis has IC50 values of 3.8 and 1.5 µg/mL, and Ascomycota sp. showed values of 2.1 and 0.67 µg/mL in J774.G8 and U937 cells, respectively. These extracts also significantly inhibited propidium iodide and Lucifer yellow uptake via P2X7R pore, P2X7R currents in electrophysiology, IL-1ß release, and the production of oxide nitric and reactive oxygen species. The extracts did not cause cytotoxicity within a period of 24 h. The results showed the promising antagonistic activity of these extracts toward P2X7R, thereby indicating that they can be future candidates for phytomedicines with potential clinical applicability.


Asunto(s)
Hongos/química , Receptores Purinérgicos P2X7/uso terapéutico , Animales , Técnicas de Cultivo de Célula , Descubrimiento de Drogas , Humanos , Ratones
11.
J Bioenerg Biomembr ; 48(4): 397-411, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27422545

RESUMEN

The second-generation photosensitizer methylene blue (MB) exhibits photochemical and photophysical properties suitable for photodynamic therapy (PDT)-based cancer treatment. However, the clinical application of MB is limited because of its high hydrophilicity, which hinders its penetration into tumor tissues. Therefore, new methods to improve the entry of MB into the cytoplasm of target cells are necessary. Because MB has a mass of 319 Da, transient pores on the plasma membrane, such as the pore induced by the P2X7 receptor (P2X7R) that allows the passage of molecules up to 900 Da, could be used. Using MTT viability assays, flow cytometry experiments, and fluorescence microscopy, we evaluated the toxicity and phototoxicity of MB and potentiation effects of ATP and MB on cell death processes in the J774 cell line (via a P2X7-associated pore). We observed that treatment with 5 µM MB for 15 min promoted the rate of entry of MB into the cytoplasm to 4.7 %. However, treatment with 5 µM MB and 1 mM ATP for the same amount of time increased this rate to 90.2 %. However, this effect was inhibited by pretreatment with a P2X7 antagonist. We used peritoneal macrophages and a cell line that does not express P2X7R as controls. These cells were more resistant to PDT with MB under the same experimental conditions. Taken together, these results suggest the use of the pore associated with P2X7R as a drug delivery system to increase the passage of hydrophilic drugs into cells that express this receptor, thus facilitating PDT.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fotoquimioterapia/métodos , Receptores Purinérgicos P2X7/uso terapéutico , Adenosina Trifosfato/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Macrófagos/citología , Azul de Metileno/farmacocinética , Azul de Metileno/toxicidad , Ratones , Porosidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA