Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 68(3): 288-301, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36252182

RESUMEN

Hypoxia contributes to the exaggerated yet ineffective airway inflammation that fails to oppose infections in cystic fibrosis (CF). However, the potential for impairment of essential immune functions by HIF-1α (hypoxia-inducible factor 1α) inhibition demands a better comprehension of downstream hypoxia-dependent pathways that are amenable for manipulation. We assessed here whether hypoxia may interfere with the activity of AhR (aryl hydrocarbon receptor), a versatile environmental sensor highly expressed in the lungs, where it plays a homeostatic role. We used murine models of Aspergillus fumigatus infection in vivo and human cells in vitro to define the functional role of AhR in CF, evaluate the impact of hypoxia on AhR expression and activity, and assess whether AhR agonism may antagonize hypoxia-driven inflammation. We demonstrated that there is an important interferential cross-talk between the AhR and HIF-1α signaling pathways in murine and human CF, in that HIF-1α induction squelched the normal AhR response through an impaired formation of the AhR:ARNT (aryl hydrocarbon receptor nuclear translocator)/HIF-1ß heterodimer. However, functional studies and analysis of the AhR genetic variability in patients with CF proved that AhR agonism could prevent hypoxia-driven inflammation, restore immune homeostasis, and improve lung function. This study emphasizes the contribution of environmental factors, such as infections, in CF disease progression and suggests the exploitation of hypoxia:xenobiotic receptor cross-talk for antiinflammatory therapy in CF.


Asunto(s)
Fibrosis Quística , Receptores de Hidrocarburo de Aril , Humanos , Ratones , Animales , Receptores de Hidrocarburo de Aril/metabolismo , Hipoxia/metabolismo , Transducción de Señal , Inflamación , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
2.
Int J Mol Sci ; 24(19)2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37834316

RESUMEN

The pathogenesis of coronavirus disease 2019 (COVID-19) is associated with a hyperinflammatory response. The mechanisms of SARS-CoV-2-induced inflammation are scantly known. Methylglyoxal (MG) is a glycolysis-derived byproduct endowed with a potent glycating action, leading to the formation of advanced glycation end products (AGEs), the main one being MG-H1. MG-H1 exerts strong pro-inflammatory effects, frequently mediated by the receptor for AGEs (RAGE). Here, we investigated the involvement of the MG-H1/RAGE axis as a potential novel mechanism in SARS-CoV-2-induced inflammation by resorting to human bronchial BEAS-2B and alveolar A549 epithelial cells, expressing different levels of the ACE2 receptor (R), exposed to SARS-CoV-2 spike protein 1 (S1). Interestingly, we found in BEAS-2B cells that do not express ACE2-R that S1 exerted a pro-inflammatory action through a novel MG-H1/RAGE-based pathway. MG-H1 levels, RAGE and IL-1ß expression levels in nasopharyngeal swabs from SARS-CoV-2-positive and -negative individuals, as well as glyoxalase 1 expression, the major scavenging enzyme of MG, seem to support the results obtained in vitro. Altogether, our findings reveal a novel mechanism involved in the inflammation triggered by S1, paving the way for the study of the MG-H1/RAGE inflammatory axis in SARS-CoV-2 infection as a potential therapeutic target to mitigate COVID-19-associated pathogenic inflammation.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Glicoproteína de la Espiga del Coronavirus , Piruvaldehído/farmacología , Piruvaldehído/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Enzima Convertidora de Angiotensina 2 , Inflamación/metabolismo
3.
Int J Mol Sci ; 22(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34638532

RESUMEN

Bone metastases from prostate cancer (PCa) result from a complex cross-talk between PCa cells and osteoblasts (OB). Thus, targeting this interplay has become an attractive strategy to interfere with PCa bone dissemination. The agents currently used in clinical trials have proved ineffective, boosting research to identify additional mechanisms that may be involved in this two-directional talk. Here, we investigated whether and how 5-hydro-5-methylimidazolone (MG-H1), a specific methylglyoxal (MG)-derived advanced glycation end product (AGE), was a novel player in the dialogue between PCa and OB to drive PCa bone metastases. Conditioned medium from osteotropic PC3 PCa cells, pre-treated or not with a specific MG scavenger, was administrated to human primary OB and cell morphology, mesenchymal trans-differentiation, pro-osteogenic determinants, PCa-specific molecules, and migration/invasion were studied by phase-contrast microscopy, real-time PCR, western blot and specific assays, respectively. We found that PC3 cells were able to release MG-H1 that, by binding to the receptor for AGEs (RAGE) on OB, reprogrammed them into a less-differentiate phenotype, endowed with some PCa-specific molecular features and malignant properties, in a mechanism involving reactive oxidative species (ROS) production and NF-kB pathway activation. These findings provide novel insights into the mechanisms of PCa osteoblastic metastases and foster in vivo research toward new therapeutic strategies interfering with PCa/OB cross-talk.


Asunto(s)
Neoplasias Óseas/secundario , Desdiferenciación Celular/fisiología , Imidazoles/metabolismo , Ornitina/análogos & derivados , Osteoblastos/citología , Neoplasias de la Próstata/patología , Antígenos de Neoplasias/metabolismo , Huesos/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Medios de Cultivo Condicionados/farmacología , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ornitina/metabolismo , Células PC-3 , Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
4.
Am J Pathol ; 188(11): 2553-2563, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30125541

RESUMEN

Because Sertoli cells (SCs) play a central role in germ cell survival, their death may result in marked germ cell loss and infertility. SCs are the only somatic cells within the seminiferous tubules and are essential for regulating spermatogenesis. Factors that enhance or diminish the viability of SCs may have profound effects on spermatogenesis. Yet the mechanisms underlying the maintenance of SC viability remain largely unknown. Glyoxalase 1 (Glo1) detoxifies methylglyoxal (MG), a highly reactive carbonyl species mainly formed during glycolysis, which is a potent precursor of cytotoxic advanced glycation end products (AGEs). Hydroimidazolone (MG-H1) and argpyrimidine (ArgPyr) are AGEs resulting from MG-mediated post-translational modification of arginine residues in various proteins. The role of Glo1 and MG-derived AGEs in regulating the fate of SCs has never been investigated. By using gene silencing and the specific MG scavenger, aminoguanidine, the authors demonstrate that Glo1, under testosterone and follicle-stimulating hormone control, sustains viability of porcine neonatal SCs through a mechanism involving the NF-κB pathway. Glo1 knockdown induces a mitochondrial apoptotic pathway driven by the intracellular accumulation of MG-H1 and ArgPyr that desensitizes NF-κB signaling by modifying the inhibitor of NF-κB kinase, IKKß. This is the first report describing a role for Glo1 and MG-derived AGEs in SC biology, providing valuable new insights into the potential involvement of this metabolic axis into spermatogenesis.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Lactoilglutatión Liasa/metabolismo , Ornitina/análogos & derivados , Pirimidinas/farmacología , Células de Sertoli/citología , Testosterona/metabolismo , Animales , Lactoilglutatión Liasa/genética , Masculino , Ornitina/farmacología , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Porcinos
5.
J Cell Mol Med ; 22(5): 2865-2883, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29504694

RESUMEN

Metastasis is the primary cause of death in prostate cancer (PCa) patients. Effective therapeutic intervention in metastatic PCa is undermined by our poor understanding of its molecular aetiology. Defining the mechanisms underlying PCa metastasis may lead to insights into how to decrease morbidity and mortality in this disease. Glyoxalase 1 (Glo1) is the detoxification enzyme of methylglyoxal (MG), a potent precursor of advanced glycation end products (AGEs). Hydroimidazolone (MG-H1) and argpyrimidine (AP) are AGEs originating from MG-mediated post-translational modification of proteins at arginine residues. AP is involved in the control of epithelial to mesenchymal transition (EMT), a crucial determinant of cancer metastasis and invasion, whose regulation mechanisms in malignant cells are still emerging. Here, we uncover a novel mechanism linking Glo1 to the maintenance of the metastatic phenotype of PCa cells by controlling EMT by engaging the tumour suppressor miR-101, MG-H1-AP and TGF-ß1/Smad signalling. Moreover, circulating levels of Glo1, miR-101, MG-H1-AP and TGF-ß1 in patients with metastatic compared with non-metastatic PCa support our in vitro results, demonstrating their clinical relevance. We suggest that Glo1, together with miR-101, might be potential therapeutic targets for metastatic PCa, possibly by metformin administration.


Asunto(s)
Transición Epitelial-Mesenquimal , Lactoilglutatión Liasa/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Regiones no Traducidas 3'/genética , Anciano , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Homoarginina/análogos & derivados , Homoarginina/sangre , Homoarginina/metabolismo , Humanos , Imidazoles/sangre , Imidazoles/metabolismo , Lactoilglutatión Liasa/sangre , Masculino , Metformina/farmacología , MicroARNs/sangre , MicroARNs/metabolismo , Persona de Mediana Edad , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Ornitina/análogos & derivados , Ornitina/sangre , Ornitina/metabolismo , Fenotipo , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/genética , Pirimidinas/sangre , Pirimidinas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Tioléster Hidrolasas/metabolismo , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/metabolismo
6.
Int J Mol Sci ; 19(2)2018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29385039

RESUMEN

Urological cancers include a spectrum of malignancies affecting organs of the reproductive and/or urinary systems, such as prostate, kidney, bladder, and testis. Despite improved primary prevention, detection and treatment, urological cancers are still characterized by an increasing incidence and mortality worldwide. While advances have been made towards understanding the molecular bases of these diseases, a complete understanding of the pathological mechanisms remains an unmet research goal that is essential for defining safer pharmacological therapies and prognostic factors, especially for the metastatic stage of these malignancies for which no effective therapies are currently being used. Glyoxalases, consisting of glyoxalase 1 (Glo1) and glyoxalase 2 (Glo2), are enzymes that catalyze the glutathione-dependent metabolism of cytotoxic methylglyoxal (MG), thus protecting against cellular damage and apoptosis. They are generally overexpressed in numerous cancers as a survival strategy by providing a safeguard through enhancement of MG detoxification. Increasing evidence suggests that glyoxalases, especially Glo1, play an important role in the initiation and progression of urological malignancies. In this review, we highlight the critical role of glyoxalases as regulators of tumorigenesis in the prostate through modulation of various critical signaling pathways, and provide an overview of the current knowledge on glyoxalases in bladder, kidney and testis cancers. We also discuss the promise and challenges for Glo1 inhibitors as future anti-prostate cancer (PCa) therapeutics and the potential of glyoxalases as biomarkers for PCa diagnosis.


Asunto(s)
Lactoilglutatión Liasa/metabolismo , Tioléster Hidrolasas/metabolismo , Neoplasias Urológicas/enzimología , Antineoplásicos/uso terapéutico , Carcinogénesis , Femenino , Humanos , Lactoilglutatión Liasa/antagonistas & inhibidores , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/metabolismo
7.
J Cell Mol Med ; 21(1): 165-176, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27696702

RESUMEN

The enzyme indoleamine 2,3-dioxygenase 1 (IDO1) catalyses the initial, rate-limiting step in tryptophan (Trp) degradation, resulting in tryptophan starvation and the production of immunoregulatory kynurenines. IDO1's catalytic function has long been considered as the one mechanism responsible for IDO1-dependent immune suppression by dendritic cells (DCs), which are master regulators of the balance between immunity and tolerance. However, IDO1 also harbours immunoreceptor tyrosine-based inhibitory motifs, (ITIM1 and ITIM2), that, once phosphorylated, bind protein tyrosine phosphatases, (SHP-1 and SHP-2), and thus trigger an immunoregulatory signalling in DCs. This mechanism leads to sustained IDO1 expression, in a feedforward loop, which is particularly important in restraining autoimmunity and chronic inflammation. Yet, under specific conditions requiring that early and protective inflammation be unrelieved, tyrosine-phosphorylated ITIMs will instead bind the suppressor of cytokine signalling 3 (SOCS3), which drives IDO1 proteasomal degradation and shortens the enzyme half-life. To dissect any differential roles of the two IDO1's ITIMs, we generated protein mutants by replacing one or both ITIM-associated tyrosines with phospho-mimicking glutamic acid residues. Although all mutants lost their enzymic activity, the ITIM1 - but not ITIM2 mutant - did bind SHPs and conferred immunosuppressive effects on DCs, making cells capable of restraining an antigen-specific response in vivo. Conversely, the ITIM2 mutant would preferentially bind SOCS3, and IDO1's degradation was accelerated. Thus, it is the selective phosphorylation of either ITIM that controls the duration of IDO1 expression and function, in that it dictates whether enhanced tolerogenic signalling or shutdown of IDO1-dependent events will occur in a local microenvironment.


Asunto(s)
Inmunosupresores/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Tirosina/inmunología , Animales , Citocinas/inmunología , Células Dendríticas/inmunología , Femenino , Semivida , Tolerancia Inmunológica/inmunología , Quinurenina/inmunología , Ratones , Ratones Endogámicos C57BL , Fosforilación/inmunología , Dominios Proteicos/inmunología , Transducción de Señal/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Triptófano/inmunología
8.
Prostate ; 77(2): 196-210, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27696457

RESUMEN

BACKGROUND: Glyoxalase 2 (Glo2), together with glyoxalase 1 (Glo1), forms the main scavenging system of methylglyoxal, a potent pro-apoptotic agent mainly generated by glycolysis. An increased rate of glycolysis is a well known signature of cancer cells. As a survival strategy, Glo1 is overexpressed in many human malignant cells, including prostate cancer (PCa), where it plays a crucial role in progression. No information is available on the role of Glo2 in the same ambit. PCa is the most common malignancy affecting men in the western world. Progression to a lethal hormone-refractory PCa represents the major concern in this pathology. Therefore, a deeper understanding of the molecular mechanisms underlying PCa invasiveness and metastasis is urgently needed in order to develop novel therapeutic targets for this incurable state of the malignancy. METHODS: Glo2 and Glo1 expression was examined in clinical samples of PCa by immunohistochemistry and in different PCa cell models by western blotting and quantitative real-time polymerase chain reaction. Gene silencing/overexpression and scavenging/inhibitory agents were used for functional analyses. RESULTS: We demonstrated that Glo2, together with Glo1, represents a novel mechanism in PCa progression as part of a pathway driven by PTEN/PI3K/AKT/mTOR signaling with involvement of PKM2 and ERα. Importantly, Glo1/Glo2 silencing did not alter the behavior of benign cells. CONCLUSIONS: Targeting glyoxalases metabolic pathway may represent a strategy to selectively inhibit advanced PCa. Prostate 77:196-210, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptor alfa de Estrógeno/metabolismo , Lactoilglutatión Liasa/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Tioléster Hidrolasas/metabolismo , Hormonas Tiroideas/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Transformada , Progresión de la Enfermedad , Humanos , Masculino , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión a Hormona Tiroide
9.
Mol Carcinog ; 56(9): 2112-2126, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28470764

RESUMEN

Glyoxalase 2 (Glo2), a metabolic enzyme, is overexpressed in some human cancers which suggests this enzyme may play a role in human tumorigenesis. In prostate cancer (PCa), the role of Glo2 has been scarcely investigated and there are no studies addressing a causative involvement of this protein in this neoplasia. Here, we examined the immunohistochemical profile of Glo2 in human PCa and benign adjacent tissues and investigated Glo2 involvement in PCa development in human prostate cell lines. PCa and matched adjacent normal tissues were obtained from paraffin sections of primary PCa from 20 patients who had undergone radical prostatectomy. Histopathological diagnosis was confirmed for each sample. Glo2 expression analysis was performed by immunohistochemistry in prostate tissues, and by qRT-PCR and immunoblotting in prostate cell lines. The causative and mechanistic role of Glo2 in prostate tumorigenesis was demonstrated by Glo2 ectopic expression/silencing and employing specific activators/inhibitors. Our results showed that Glo2 was selectively expressed in PCa but not in the luminal compartment of the adjacent benign epithelium consistently in all the examined 20 cases. Glo2 expression in PCa was dependent on androgen receptor (AR) and was aimed at stimulating cell proliferation and eluding apoptosis through a mechanism involving the p53-p21 axis. Glo2 was intensely expressed in the basal cells of benign glands but was not involved in PCa genesis. Our results demonstrate for the first time that Glo2 drives prostate tumorigenesis and suggest that it may represent a novel adjuvant marker in the pathological diagnosis of early PCa.


Asunto(s)
Carcinogénesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Tioléster Hidrolasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Técnicas de Silenciamiento del Gen , Humanos , Lactoilglutatión Liasa/metabolismo , Masculino , Próstata/metabolismo , Transducción de Señal , Tioléster Hidrolasas/genética
10.
BMC Cancer ; 17(1): 502, 2017 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-28747156

RESUMEN

BACKGROUND: We investigated whether GSTT1 ("null" allele), GSTM1 ("null"allele), GSTP1 (A313G), RFC1 (G80A), MTHFR (C677T), TS (2R/3R) polymorphisms were associated with toxicity and survival in patients with early breast cancer (EBC) treated with adjuvant chemotherapy (CT). METHODS: This prospective trial included patients with stage I-III BC subjected to CT with CMF or FEC regimens. PCR-RFLP was performed for MTHFR, RFC1 and GSTP1, while PCR for TS, GSTT1 and GSTM1 genes. RESULTS: Among the 244 patients consecutively enrolled, 48.7% were treated with FEC and 51.3% with CMF. Patients with TS2R/3R genotype showed less frequently severe neutropenia (G3/G4) than those with TS2R/2R and 3R/3R genotype (p = 0.038). Patients with MTHFRCT genotype had a higher probability of developing severe neutropenia than those with MTHFR CC genotype (p = 0.043). Patients with RFC1GG or GSTT1-null genotype or their combination (GSTT1-null/RFC1GG) were significantly associated with a shorter disease free survival (DFS) (p = 0.009, p = 0.053, p = 0.003, respectively) and overall survival (OS) (p = 0.036, p = 0.015, p = 0.005, respectively). Multivariate analysis confirmed the association of RFC1GG genotype with a shorter DFS (p = 0.018) and of GSTT1-null genotype of a worse OS (p = 0.003), as well as for the combined genotypes GSTT1-null/RFC1GG, (DFS: p = 0.004 and OS: p = 0.003). CONCLUSIONS: Our data suggest that TS2R/2R and 3R/3R or MTHFR CT genotypes have a potential role in identifying patients with greater risk of toxicity to CMF/FEC and that RFC1 GG and GSTT1-null genotypes alone or in combination could be important markers in predicting clinical outcome in EBC patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/tratamiento farmacológico , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/mortalidad , Quimioterapia Adyuvante , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Supervivencia sin Enfermedad , Epirrubicina/efectos adversos , Epirrubicina/uso terapéutico , Femenino , Fluorouracilo/efectos adversos , Fluorouracilo/uso terapéutico , Frecuencia de los Genes , Estudios de Asociación Genética , Genotipo , Gutatión-S-Transferasa pi/genética , Glutatión Transferasa/genética , Humanos , Estimación de Kaplan-Meier , Metotrexato/efectos adversos , Metotrexato/uso terapéutico , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Persona de Mediana Edad , Análisis Multivariante , Modelos de Riesgos Proporcionales , Estudios Prospectivos
11.
Mediators Inflamm ; 2017: 5858315, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28331244

RESUMEN

Interleukin-1ß (IL-1ß) is a pleiotropic cytokine and a crucial mediator of inflammatory and immune responses. IL-1ß processing and release are tightly controlled by complex pathways such as NF-kB/ERK1/2, to produce pro-IL-1ß, and NALP3/ASC/Caspase-1 inflammasome, to produce the active secreted protein. Dysregulation of both IL-1ß and its related pathways is involved in inflammatory/autoimmune disorders and in a wide range of other diseases. Identifying molecules modulating their expression is a crucial need to develop new therapeutic agents. IL-1ß is a strong regulator of Brain Natriuretic Peptide (BNP), a hormone involved in cardiovascular homeostasis by guanylyl cyclase Natriuretic Peptide Receptor (NPR-1). An emerging role of BNP in inflammation and immunity, although proposed, remains largely unexplored. Here, we newly demonstrated that, in human THP-1 monocytes, LPS/ATP-induced IL-1ß secretion is strongly inhibited by BNP/NPR-1/cGMP axis at all the molecular mechanisms that tightly control its production and release, NF-kB, ERK 1/2, and all the elements of NALP3/ASC/Caspase-1 inflammasome cascade, and that NALP3 inflammasome inhibition is directly related to BNP deregulatory effect on NF-kB/ERK 1/2 activation. Our findings reveal a novel potent anti-inflammatory and immunomodulatory role for BNP and open new alleys of investigation for a possible employment of this endogenous agent in the treatment of inflammatory/immune-related and IL-1ß/NF-kB/ERK1/2/NALP3/ASC/Caspase-1-associated diseases.


Asunto(s)
Interleucina-1beta/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Péptido Natriurético Encefálico/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Supervivencia Celular , Regulación hacia Abajo , Activación Enzimática , Homeostasis , Humanos , Inflamasomas/metabolismo , Inflamación , Interleucina-1beta/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , Monocitos/citología
12.
Apoptosis ; 19(1): 102-16, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24002659

RESUMEN

Reactive oxygen species (ROS) are implicated in the regulation of apoptosis through a number of distinct mechanisms depending on cell type and stimulation conditions. Glyoxalase I (GI) metabolizes methylglyoxal (MG) and MG-derived advanced glycation end products (AGEs) known to cause apoptosis. This study examined the possible role of GI among the mechanisms of ROS-driven apoptosis in human bronchial epithelial BEAS-2B cells exposed to wood dust and signaling pathways by which these reactive species regulate GI expression. Our results showed that wood dust generated distinct ROS (superoxide anion, and hydrogen peroxide) by selectively inhibiting the enzymatic activity of superoxide dismutase or glutathione peroxidase and catalase enzymes. These ROS caused a dramatic inhibition of the antiglycation GI enzyme, leading to the intracellular accumulation of the pro-apoptotic AGE, argpyrimidine (AP) and programmed cell death via a mitochondrial pathway. Pre-treatment with N-acetyl-L-cysteine (NAC), a ROS scavenger, prevented these events. Hence, ROS-induced apoptosis in BEAS-2B cells occurred via a novel mechanism relying on GI inhibition and AP accumulation. We interestingly found that superoxide anion and hydrogen peroxide induced a diverse apoptosis level by differently inhibiting GI via NF-κB pathway. Since maintenance of an intact epithelium is a critically important determinant of normal respiratory function, the knowledge of the mechanisms underlying its disruption may provide insight into the genesis of a number of pathological conditions commonly occurring in wood dust occupational exposure. Our findings suggest that the antioxidant NAC may merit investigation as a potential preventive agent in wood dust exposure-induced respiratory diseases.


Asunto(s)
Apoptosis , Células Epiteliales/enzimología , Peróxido de Hidrógeno/metabolismo , Lactoilglutatión Liasa/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo , Bronquios/citología , Bronquios/enzimología , Bronquios/metabolismo , Línea Celular , Regulación hacia Abajo , Polvo/análisis , Células Epiteliales/citología , Células Epiteliales/metabolismo , Glicosilación , Humanos , Exposición Profesional/efectos adversos , Pinus/efectos adversos , Quercus/efectos adversos , Especies Reactivas de Oxígeno/toxicidad
13.
Pharmaceuticals (Basel) ; 17(3)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38543153

RESUMEN

Patients with cystic fibrosis (PwCF) have recently experienced an unprecedented breakthrough with the adoption of modulator therapy in clinical practice. This remarkable achievement has led to the reconsideration of disease management as the increased life expectancy has gradually shifted the attention over a spectrum of extra-pulmonary manifestations that become prevalent in the aging population. It comes to be that complementary approaches that target patient co-morbidities are needed for the optimal clinical management of PwCF. A strategy would be to adjuvate the cystic fibrosis transmembrane conductance regulator (CFTR) in performing its functions in the different organs in which it is expressed. Solute carrier family 26 (SLC26) members appear ideal in this context. Indeed, they not only cooperate with CFTR in the organ-dependent regulation of ion fluxes but physically interact with it to reciprocally modulate their function. In this opinion, we summarize available evidence pointing to a physical and functional interaction between CFTR and SLC26 members, with a particular focus on SLC26A6 for its wider expression and broader anion selectivity, and then discuss how restoring the physical interaction between CFTR and SLC26A6 might be beneficial in the treatment of PwCF in the era of modulator therapy.

14.
Int J Pharm ; 661: 124393, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38942183

RESUMEN

This paper focuses on the preparation and characterization of antibacterial alginate microparticles containing silver@hydroxyapatite functionalized calcium carbonate composites for tissue engineering. Microparticles were prepared by cross-linking a silver@composite sodium alginate dispersion with CaCl2. This method showed a very good silver efficiency loading and the presence of silver chloride nanoparticles was detected. Silver free microparticles, containing hydroxyapatite functionalized calcium carbonates and neat alginate microparticles were prepared as well. All microparticles were characterized for water absorption and for in vitro bioactivity by immersion in simulated body fluid (SBF). Finally, antimicrobial and antibiofilm activities as well as cytotoxicity were evaluated. Microparticles containing silver@composites exhibited good antimicrobial and antibiofilm activities against Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa and Candida albicans, but exerted a certain cytotoxicity against the tested cell models (fibroblasts and osteoblasts). Microparticles containing hydroxyapatite functionalized calcium carbonates were found to be always less cytotoxic, also in comparison to neat alginate microparticles, proving that the presence of the inorganic matrices exerts a protective effect on microparticle cytotoxicity.

15.
Front Biosci (Landmark Ed) ; 29(2): 59, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38420819

RESUMEN

BACKGROUND: The recent Coronavirus Disease 2019 (COVID-19) pandemic has dramatically exposed our gap in understanding the pathogenesis of airborne infections. Within such a context, it is increasingly clear that the nasal cavity represents a critical checkpoint not only in the initial colonization phase but also in shaping any infectious sequelae. This is particularly relevant to COVID-19 in that the nasal cavity is characterized by high-level expression of the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) receptor, Angiotensin-Converting Enzyme 2 (ACE2), all along the respiratory tract. As part of the nasal mucosa, commensal microbes harbored by the nasal cavity likely are far more than just innocent bystanders in the interaction between SARS-CoV-2 and the local microenvironment. Yet the role of the qualitative composition of the nasal microbiome is unclear, as is its function, whether protective or not. METHODS: In this study, individuals undergoing SARS-CoV-2 molecular testing at the Hospital of Perugia (Italy) were recruited, with their residual material from the nasopharyngeal swabs being collected for microbiome composition analysis and short-chain fatty acid (SCFA) measurements (by 16S rRNA sequencing and gas chromatography-mass spectrometry), respectively. RESULTS: After stratification by age, gender, and viral load, the composition of the nasopharyngeal microbiome appeared to be influenced by age and gender, and SARS-CoV-2 infection further determined compositional changes. Notwithstanding this variability, a restricted analysis of female subjects-once SARS-CoV-2-infected-unraveled a shared expansion of Lachnospirales-Lachnospiraceae, irrespective of the viral load and age. This was associated with a reduction in the branched SCFA isobutanoic acid, as well as in the SCFAs with longer chains. CONCLUSIONS: Our results indicate that the nasopharyngeal microbiome is influenced by age, gender, and viral load, with consistent patterns of microbiome changes being present across specific groups. This may help in designing a personalized medicine approach in COVID-19 patients with specific patterns of nasal microbial communities.


Asunto(s)
COVID-19 , Microbiota , Humanos , Femenino , SARS-CoV-2 , ARN Ribosómico 16S/genética , Nasofaringe
16.
J Extracell Vesicles ; 13(6): e12446, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38844736

RESUMEN

Dendritic cells (DCs) are essential orchestrators of immune responses and represent potential targets for immunomodulation in autoimmune diseases. Human amniotic fluid secretome is abundant in immunoregulatory factors, with extracellular vesicles (EVs) being a significant component. However, the impact of these EVs on dendritic cells subsets remain unexplored. In this study, we investigated the interaction between highly purified dendritic cell subsets and EVs derived from amniotic fluid stem cell lines (HAFSC-EVs). Our results suggest that HAFSC-EVs are preferentially taken up by conventional dendritic cell type 2 (cDC2) through CD29 receptor-mediated internalization, resulting in a tolerogenic DC phenotype characterized by reduced expression and production of pro-inflammatory mediators. Furthermore, treatment of cDC2 cells with HAFSC-EVs in coculture systems resulted in a higher proportion of T cells expressing the regulatory T cell marker Foxp3 compared to vehicle-treated control cells. Moreover, transfer of HAFSC-EV-treated cDC2s into an EAE mouse model resulted in the suppression of autoimmune responses and clinical improvement. These results suggest that HAFSC-EVs may serve as a promising tool for reprogramming inflammatory cDC2s towards a tolerogenic phenotype and for controlling autoimmune responses in the central nervous system, representing a potential platform for the study of the effects of EVs in DC subsets.


Asunto(s)
Líquido Amniótico , Células Dendríticas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental , Vesículas Extracelulares , Esclerosis Múltiple , Animales , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Líquido Amniótico/citología , Líquido Amniótico/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Femenino , Células Madre/metabolismo , Células Madre/citología , Ratones Endogámicos C57BL
17.
Prostate ; 73(2): 121-32, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22653787

RESUMEN

BACKGROUND: Glyoxalase I (GLOI) detoxifies reactive dicarbonyls, as methylglyoxal (MG) that, directly or through the formation of MG-derived adducts, is a growth inhibitor and apoptosis inducer. GLOI has been considered a general marker of cell proliferation, but a direct link between the two has yet to be demonstrated. The aim of the present work was to clarify whether GLOI was involved in the proliferation control of LNCaP and PC3 human prostate cancer cells or might play a different role in the growth regulation of these cells. METHODS: RNA interference was used to study the role of GLOI in cell proliferation or apoptosis. Cell proliferation was evaluated by [3H]thymidine incorporation assay and flow cytometry, that was also used to analyze apoptosis. Real-time TaqMan polymerase chain reaction and spectrophotometric analyses were used to study transcript levels or specific activity, respectively. Proteins levels were analyzed by Western blot. MG was measured by high-performance liquid chromatography. RESULTS: We found that GLOI is not implicated in the proliferation control of LNCaP and PC3 cells but plays a role in the apoptosis of invasive prostate cancer PC3 cells, through a mechanism involving a specific MG-adduct and NF-kB signaling pathway. CONCLUSIONS: Our data represent the first systematic demonstration that GLOI cannot be considered a general marker of cell proliferation and that acts as a pro-survival factor in invasive PC3 cells by elusing apoptosis. GLOI may be involved in prostate cancer progression, via the control of key molecules in the mitochondrial apoptotic mechanism, through NF-kB signaling pathway.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/fisiología , Proliferación Celular , Lactoilglutatión Liasa/fisiología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Ciclo Celular/fisiología , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Masculino , Proteínas Mitocondriales/fisiología , FN-kappa B/fisiología , Neoplasias de la Próstata/etiología , Transducción de Señal/fisiología
18.
Mediators Inflamm ; 2013: 159349, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24369440

RESUMEN

Postoperative-fluid retention is a severe complication frequently reported in patients undergoing major surgical procedures. The complex network of molecules involved in such a severe surgery-induced condition remains poorly understood. Inflammation has been proposed among the various causes of fluid retention. Since TNF-α is one of the main proinflammatory cytokine initially released after major surgery, it is reasonable to assume its involvement in fluid overload. Here, we showed that TNF-α selectively regulates key molecules involved in fluids balance, such as natriuretic peptides (NPs) and aquaporins, in human bronchial epithelial cells BEAS-2B. In particular, we found that TNF-α induced a decrease of arial natriuretic peptide, natriuretic peptide receptor-1, aquaporin-1 and aquaporin-5 and an increase of brain natriuretic peptide with a different involvement of nuclear factor-κB and mitogen-activated protein kinases signaling pathway activation. Moreover, the observed changes in NPs expression, demonstrate inflammation as an additional cause of brain natriuretic peptide elevation, adding an important piece of information in the novel area of study regarding NPs and inflammation. Finally, we suggest that inflammation is one of the mechanisms of Aquaporin-1 and aquaporin-5 expression regulation. Therefore, in this exploratory study, we speculate that TNF-α might be involved in postoperative-fluid retention related to major surgery.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Células Epiteliales/citología , Péptido Natriurético Encefálico/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Acuaporina 1/metabolismo , Acuaporina 5/metabolismo , Bronquios/citología , Línea Celular , Supervivencia Celular , Medios de Cultivo/química , Dexametasona/química , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hemodinámica , Homeostasis , Humanos , Inflamación/metabolismo , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal
19.
Polymers (Basel) ; 15(10)2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37242821

RESUMEN

Biodegradable metal alloys may be successfully used to support bone repair, avoiding second surgery commonly needed when inert metal alloys are used. Combining a biodegradable metal alloy with a suitable pain relief agent could improve patient quality of life. AZ31 alloy was coated using a poly(lactic-co-glycolic) acid (PLGA) polymer loaded with ketorolac tromethamine using the solvent casting method. The ketorolac release profile from the polymeric film and the coated AZ31 samples, the PLGA mass loss of polymeric film, and the cytotoxicity of the optimized coated alloy were assessed. The coated sample showed a ketorolac release that was prolonged for two weeks, which was slower than that of just the polymeric film, in simulated body fluid. PLGA mass loss was complete after a 45-day immersion in simulated body fluid. The PLGA coating was able to lower AZ31 and ketorolac tromethamine cytotoxicity observed in human osteoblasts. PLGA coating also prevents AZ31 cytotoxicity, which was identified in human fibroblasts. Therefore, PLGA was able to control ketorolac release and protect AZ31 from premature corrosion. These characteristics allow us to hypothesize that the use of ketorolac tromethamine-loaded PLGA coating on AZ31 in the management of bone fractures can favor osteosynthesis and relief pain.

20.
Antioxidants (Basel) ; 12(11)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38001829

RESUMEN

Hemoglobin is one of the proteins that are more susceptible to S-glutathionylation and the levels of its modified form, glutathionyl hemoglobin (HbSSG), increase in several human pathological conditions. The scope of the present review is to provide knowledge about how hemoglobin is subjected to S-glutathionylation and how this modification affects its functionality. The different diseases that showed increased levels of HbSSG and the methods used for its quantification in clinical investigations will be also outlined. Since there is a growing need for precise and reliable methods for markers of oxidative stress in human blood, this review highlights how HbSSG is emerging more and more as a good indicator of severe oxidative stress but also as a key pathogenic factor in several diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA