Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 338
Filtrar
1.
Life Sci ; 352: 122857, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38914305

RESUMEN

AIM: AMPK can be considered as an important target molecule for cancer for its unique ability to directly recognize cellular energy status. The main aim of this study is to explore the role of different AMPK activators in managing cancer cell aggressiveness and to understand the mechanistic details behind the process. MAIN METHODS: First, we explored the AMPK expression pattern and its significance in different subtypes of lung cancer by accessing the TCGA data sets for LUNG, LUAD and LUSC patients and then established the correlation between AMPK expression pattern and overall survival of lung cancer patients using Kaplan-Meire plot. We further carried out several cell-based assays by employing different wet lab techniques including RT-PCR, Western Blot, proliferation, migration and invasion assays to fulfil the aim of the study. KEY FINDINGS: SIGNIFICANCE: This study identifies the importance of AMPK activators as a repurposing agent for combating lung and colon cancer cell aggressiveness. It also suggests SRT-1720 as a potent repurposing agent for cancer treatment especially in NSCLC patients where a point mutation is present in LKB1.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares , Monoaminooxidasa , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Monoaminooxidasa/metabolismo , Monoaminooxidasa/genética , Línea Celular Tumoral , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores de la Monoaminooxidasa/farmacología , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica
2.
Sci Adv ; 10(6): eadi4935, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38335292

RESUMEN

Prostate cancer (PC) develops in a microenvironment where the stromal cells modulate adjacent tumor growth and progression. Here, we demonstrated elevated levels of monoamine oxidase B (MAOB), a mitochondrial enzyme that degrades biogenic and dietary monoamines, in human PC stroma, which was associated with poor clinical outcomes of PC patients. Knockdown or overexpression of MAOB in human prostate stromal fibroblasts indicated that MAOB promotes cocultured PC cell proliferation, migration, and invasion and co-inoculated prostate tumor growth in mice. Mechanistically, MAOB induces a reactive stroma with activated marker expression, increased extracellular matrix remodeling, and acquisition of a protumorigenic phenotype through enhanced production of reactive oxygen species. Moreover, MAOB transcriptionally activates CXCL12 through Twist1 synergizing with TGFß1-dependent Smads in prostate stroma, which stimulates tumor-expressed CXCR4-Src/JNK signaling in a paracrine manner. Pharmacological inhibition of stromal MAOB restricted PC xenograft growth in mice. Collectively, these findings characterize the contribution of MAOB to PC and suggest MAOB as a potential stroma-based therapeutic target.


Asunto(s)
Monoaminooxidasa , Neoplasias de la Próstata , Animales , Humanos , Masculino , Ratones , Línea Celular Tumoral , Fibroblastos/metabolismo , Monoaminooxidasa/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transducción de Señal , Microambiente Tumoral
3.
J Biol Chem ; 300(3): 105760, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38367663

RESUMEN

In the cold, the absence of the mitochondrial uncoupling protein 1 (UCP1) results in hyper-recruitment of beige fat, but classical brown fat becomes atrophied. Here we examine possible mechanisms underlying this phenomenon. We confirm that in brown fat from UCP1-knockout (UCP1-KO) mice acclimated to the cold, the levels of mitochondrial respiratory chain proteins were diminished; however, in beige fat, the mitochondria seemed to be unaffected. The macrophages that accumulated massively not only in brown fat but also in beige fat of the UCP1-KO mice acclimated to cold did not express tyrosine hydroxylase, the norepinephrine transporter (NET) and monoamine oxidase-A (MAO-A). Consequently, they could not influence the tissues through the synthesis or degradation of norepinephrine. Unexpectedly, in the cold, both brown and beige adipocytes from UCP1-KO mice acquired an ability to express MAO-A. Adipose tissue norepinephrine was exclusively of sympathetic origin, and sympathetic innervation significantly increased in both tissues of UCP1-KO mice. Importantly, the magnitude of sympathetic innervation and the expression levels of genes induced by adrenergic stimulation were much higher in brown fat. Therefore, we conclude that no qualitative differences in innervation or macrophage character could explain the contrasting reactions of brown versus beige adipose tissues to UCP1-ablation. Instead, these contrasting responses may be explained by quantitative differences in sympathetic innervation: the beige adipose depot from the UCP1-KO mice responded to cold acclimation in a canonical manner and displayed enhanced recruitment, while the atrophy of brown fat lacking UCP1 may be seen as a consequence of supraphysiological adrenergic stimulation in this tissue.


Asunto(s)
Tejido Adiposo Beige , Tejido Adiposo Pardo , Sistema Nervioso Simpático , Termogénesis , Proteína Desacopladora 1 , Animales , Ratones , Tejido Adiposo Beige/inervación , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adrenérgicos/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Norepinefrina/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Ratones Noqueados , Aclimatación/genética , Sistema Nervioso Simpático/fisiología , Macrófagos/metabolismo
4.
Aquat Toxicol ; 267: 106800, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38183773

RESUMEN

Bisphenol S (BPS) is a common endocrine-disrupting chemical globally used in several consumer and industrial products. Although previous studies suggested that BPS induces multiple effects in exposed organisms, very little is known about its intergenerational effect on offspring behavior and/or the potential underlying mechanisms. To this end, adult female zebrafish Danio rerio were exposed to BPS (0, 10, 30 µg/L) and 1 µg/L of 17-ß-estradiol (E2) as a positive control for 60 days. Afterwards, female fish were bred with untreated males, and their offspring were raised to 6 months old in control water. Maternal exposure to BPS decreased male offspring anxiety and antipredator behaviors while boldness remained unaffected. Specifically, maternal exposure to 10 and 30 µg/L BPS and 1 µg/L E2 were found to impact male offspring anxiety levels as they decreased the total time that individuals spent in the dark zone in the light/dark box test and increased the total track length in the center of the open field test. In addition, maternal exposure to all concentrations of BPS and E2 disrupted antipredator responses of male offspring by decreasing shoal cohesion in the presence of chemical alarm cues derived from conspecifics, which communicated high risk. To elucidate the possible molecular mechanism underlying these neuro-behavioral effects of BPS, we assessed the serotonergic system via changes in mRNA expression of serotonin receptors, including the 5-HT1A, 5-HT1B, and 5-HT1D subtypes, the serotonin transporter and monoamine oxidase (MAO). The impaired anxiety and antipredator responses were associated with reduced levels of 5-HT1A subtype and MAO mRNA expression within the brain of adult male offspring. Collectively, the results of this study demonstrate that maternal exposure to environmental concentrations of BPS can interfere with the serotonergic signaling pathway in the developing brain, subsequently leading to the onset of a suite of behavioral deficits in adult offspring.


Asunto(s)
Fenoles , Sulfonas , Contaminantes Químicos del Agua , Pez Cebra , Humanos , Animales , Masculino , Femenino , Pez Cebra/metabolismo , Exposición Materna , Serotonina/metabolismo , Contaminantes Químicos del Agua/toxicidad , Ansiedad/inducido químicamente , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , ARN Mensajero/metabolismo
5.
FEBS J ; 291(5): 849-864, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37814408

RESUMEN

Monoamine oxidases (MAOs) are pivotal regulators of neurotransmitters in mammals, while microbial MAOs have been shown to be valuable biocatalysts for enantioselective synthesis of pharmaceutical compounds or precursors thereof. To extend the knowledge of how MAOs function at the molecular level and in order to provide more biocatalytic tools, we set out to identify and study a robust bacterial variant: a MAO from the thermophile Thermoanaerobacterales bacterium (MAOTb ). MAOTb is highly thermostable with melting temperatures above 73 °C and is well expressed in Escherichia coli. Substrate screening revealed that the oxidase is most efficient with n-alkylamines with n-heptylamine being the best substrate. Presteady-state kinetic analysis shows that reduced MAOTb rapidly reacts with molecular oxygen, confirming that it is a bona fide oxidase. The crystal structure of MAOTb was resolved at 1.5 Å and showed an exceptionally high similarity with the two human MAOs, MAO A and MAO B. The active site of MAOTb resembles mostly the architecture of human MAO A, including the cysteinyl protein-FAD linkage. Yet, the bacterial MAO lacks a C-terminal extension found in human MAOs, which explains why it is expressed and purified as a soluble protein, while the mammalian counterparts are anchored to the membrane through an α-helix. MAOTb also displays a slightly different active site access tunnel, which may explain the specificity toward long aliphatic amines. Being an easy-to-express, thermostable enzyme, for which a high-resolution structure was elucidated, this bacterial MAO may develop into a valuable biocatalyst for synthetic chemistry or biosensing.


Asunto(s)
Bacterias , Monoaminooxidasa , Humanos , Animales , Cinética , Monoaminooxidasa/genética , Biocatálisis , Aminas , Escherichia coli/genética , Mamíferos
6.
Biosci Biotechnol Biochem ; 88(3): 322-332, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38066695

RESUMEN

This study was to investigate the inhibitory activity of small hairtail-related peptides (VFEVFW, LPNSLYQQ, LPNSLYQK, and FADAME) on intracellular monoamine oxidase-A (MAO-A) and their protective effects in a cell model. Specifically, the inhibition activity in SH-SY5Y cells indicated that VFEVFW and LPNSLYQK reduced ∼50% of MAO-A activity in cells, at 0.5 m m. The survival experiment demonstrated that the toxic effect of dexamethasone (DEX) on cells can be significantly alleviated in the presence of peptides, and these peptides can restore (>20%) the mitochondrial membrane potential of SH-SY5Y cells reduced by DEX. Circular dichroism displayed that peptides affected the secondary structure of MAO-A in a concentration-dependent manner. Finally, the real-time quantitative polymerase chain reaction assay revealed that the MAO-A inhibitory activity of the peptides was associated with the upregulation of brain derived neurotrophic factor/cAMP (Cyclic adenosine monophosphate) response element binding protein)/B-cell lymphoma-2 mRNA levels.


Asunto(s)
Monoaminooxidasa , Neuroblastoma , Humanos , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Inhibidores de la Monoaminooxidasa/metabolismo , Línea Celular Tumoral , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuronas , Péptidos/farmacología
7.
Biomed Res Int ; 2023: 2592691, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37841082

RESUMEN

The catalytic activity of monoamine oxidase A (MAO-A) has been linked to tumorigenesis due to the production of reactive oxygen species (ROS) and the resulting oxidative stress. MAO-A inhibition revealed a beneficial role in prostate and lung cancer treatment. This study is aimed at evaluating the effect of different monoamine oxidase A inhibitors (MAO-AIs) on the proliferation and progression of breast cancer cell lines. The cell viability assay was used to evaluate the antiproliferative and combined effects of MAO-AIs. Cell migration was evaluated using wound healing, invasion, and colony formation assays. The underlying mechanism of cell death was studied using flow cytometry. The real-time polymerase chain reaction was used to determine the relative gene expression. Finally, MAO-A activity in breast cancer cells was evaluated using an MAO-A activity assay. According to the results, the examined MAO-AIs significantly inhibited the proliferation of breast cancer cells in a dose-dependent manner. In breast cancer cells, the combination of anticancer drugs (doxorubicin or raloxifene) with MAO-AIs resulted in a synergistic effect. MAO-AIs significantly reduced wound closure and invasion ability in breast cancer cells. Also, MAO-AIs reduced the colony count and size of breast cancer cells. MAO-AIs resulted in significant proapoptotic activity in breast cancer cells. Finally, the MAO-AIs suppressed MAO-A, Bcl-2, and VEGF gene expressions in breast cancer cells relative to untreated cells. This study provides solid evidence supporting the anticancer effect of MAO-A inhibitors in breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Inhibidores de la Monoaminooxidasa , Masculino , Humanos , Inhibidores de la Monoaminooxidasa/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
8.
Oncology ; 101(7): 415-424, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37231904

RESUMEN

INTRODUCTION: Gastric cancer is one of the common causes of cancer-related death in the world. Neurotransmitters have recently been related to the proliferation of cancer cells, but the role of neurotransmitters in the progression of gastric cancer is still unexplored. The cross-talk between the nervous system and immune cells through serotonin and its receptors in the tumor microenvironment can impact tumor progress. Our purpose is to expose probable changes in serotonin receptors, acetylcholinesterase, and monoamine oxidase A gene expression in gastric cancer. METHODS: Transcript of serotonin receptors (5-HTR2A, 5-HTR2B, 5-HTR3A, 5-HTR7) and monoamine oxidase A genes in the peripheral blood mononuclear cells (40 patients and 40 control) and tissue (21 tumors and 21 normal adjacent tissues) were assessed. The gene expression was analyzed by quantitative real-time PCR using suitable primers. Statistical analysis was performed using appropriate software (REST, Prism). RESULTS: Significantly higher amounts of 5-HTR2A, 5-HTR2B, 5-HTR3A, 5-HTR7, and acetylcholinesterase gene transcripts were found in the peripheral blood of gastric cancer patients compared with healthy individuals. The expression of 5-HTR2B and 5-HTR3A genes was significantly higher (p = 0.0250, p = 0.0005, respectively) and the acetylcholinesterase gene was lower in the tissue of patients (p = 0.0119) compared with adjacent normal tissue. CONCLUSION: This study highlights the role of serotonin receptors in gastric cancer that might have suggestions for the development of novel therapeutics and defensive approaches that target factors associated with the link between the nervous system, cancer cells, and the tumor microenvironment.


Asunto(s)
Acetilcolinesterasa , Neoplasias Gástricas , Humanos , Acetilcolinesterasa/genética , Neoplasias Gástricas/genética , Microambiente Tumoral/genética , Leucocitos Mononucleares , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Expresión Génica , Monoaminooxidasa/genética
9.
Basic Res Cardiol ; 118(1): 4, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36670288

RESUMEN

During embryonic development, cardiomyocytes undergo differentiation and maturation, processes that are tightly regulated by tissue-specific signaling cascades. Although redox signaling pathways involved in cardiomyogenesis are established, the exact sources responsible for reactive oxygen species (ROS) formation remain elusive. The present study investigates whether ROS produced by the mitochondrial flavoenzyme monoamine oxidase A (MAO-A) play a role in cardiomyocyte differentiation from human induced pluripotent stem cells (hiPSCs). Wild type (WT) and MAO-A knock out (KO) hiPSCs were generated by CRISPR/Cas9 genome editing and subjected to cardiomyocyte differentiation. Mitochondrial ROS levels were lower in MAO-A KO compared to the WT cells throughout the differentiation process. MAO-A KO hiPSC-derived cardiomyocytes (hiPSC-CMs) displayed sarcomere disarray, reduced α- to ß-myosin heavy chain ratio, GATA4 upregulation and lower macroautophagy levels. Functionally, genetic ablation of MAO-A negatively affected intracellular Ca2+ homeostasis in hiPSC-CMs. Mechanistically, MAO-A generated ROS contributed to the activation of AKT signaling that was considerably attenuated in KO cells. In addition, MAO-A ablation caused a reduction in WNT pathway gene expression consistent with its reported stimulation by ROS. As a result of WNT downregulation, expression of MESP1 and NKX2.5 was significantly decreased in MAO-A KO cells. Finally, MAO-A re-expression during differentiation rescued expression levels of cardiac transcription factors, contractile structure, and intracellular Ca2+ homeostasis. Taken together, these results suggest that MAO-A mediated ROS generation is necessary for the activation of AKT and WNT signaling pathways during cardiac lineage commitment and for the differentiation of fully functional human cardiomyocytes.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Diferenciación Celular/fisiología , Vía de Señalización Wnt
10.
Free Radic Biol Med ; 195: 309-328, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36592660

RESUMEN

This study depicted the effect of IL-13 and 13(S)HpODE (the endogenous product during IL-13 activation) in the process of cancer cell apoptosis. We examined the role of both IL-13 and 13(S)HpODE in mediating apoptotic pathway in three different in vitro cellular models namely A549 lung cancer, HCT116 colorectal cancer and CCF52 GBM cells. Our data showed that IL-13 promotes apoptosis of A549 lung carcinoma cells through the involvement of 15-LO, PPARγ and MAO-A. Our observations demonstrated that IL-13/13(S)HpODE stimulate MAO-A-mediated intracellular ROS production and p53 as well as p21 induction which play a crucial role in IL-13-stimulated A549 cell apoptosis. We further showed that 13(S)HpODE promotes apoptosis of HCT116 and CCF52 cells through the up-regulation of p53 and p21 expression. Our data delineated that IL-13 stimulates p53 and p21 induction which is mediated through 15-LO and MAO-A in A549 cells. In addition, we observed that PPARγ plays a vital role in apoptosis as well as in p53 and p21 expression in A549 cells in the presence of IL-13. We validated our observations in case of an in vivo colon cancer tumorigenic study using syngeneic mice model and demonstrated that 13(S)HpODE significantly reduces solid tumor growth through the activation of apoptosis. These data thus confirmed that IL-13 > 15-LO>13(S)HpODE > PPARγ>MAO-A > ROS > p53>p21 axis has a major contribution in regulating cancer cell apoptosis and further identified 13(S)HpODE as a potential chemo-preventive agent which can improve the efficacy of cancer treatment as a combination compound.


Asunto(s)
Apoptosis , Interleucina-13 , Neoplasias Pulmonares , Proteína p53 Supresora de Tumor , Animales , Ratones , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Interleucina-13/farmacología , Neoplasias Pulmonares/patología , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Humanos , Células A549
11.
Int J Mol Sci ; 23(19)2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-36233054

RESUMEN

Bladder cancer is the 10th most common cancer in the world and has a high risk of recurrence and metastasis. In order to sustain high energetic needs, cancer cells undergo complex metabolic adaptations, such as a switch toward aerobic glycolysis, that can be exploited therapeutically. Reactive oxygen species (ROS) act as key regulators of cancer metabolic reprogramming and tumorigenesis, but the sources of ROS remain unidentified. Monoamine oxidases (MAOs) are mitochondrial enzymes that generate H2O2 during the breakdown of catecholamines and serotonin. These enzymes are particularly important in neurological disorders, but recently, a new link between MAOs and cancer has been uncovered, involving their production of ROS. At present, the putative role of MAOs in bladder cancer has never been evaluated. We observed that human urothelial tumor explants and the bladder cancer cell line AY27 expressed both MAO-A and MAO-B isoforms. Selective inhibition of MAO-A or MAO-B limited mitochondrial ROS accumulation, cell cycle progression and proliferation of bladder cancer cells, while only MAO-A inhibition prevented cell motility. To test whether ROS contributed to MAO-induced tumorigenesis, we used a mutated form of MAO-A which was unable to produce H2O2. Adenoviral transduction of the WT MAO-A stimulated the proliferation and migration of AY27 cells while the Lys305Met MAO-A mutant was inactive. This was consistent with the fact that the antioxidant Trolox strongly impaired proliferation and cell cycle progression. Most interestingly, AY27 cells were highly dependent on glucose metabolism to sustain their growth, and MAO inhibitors potently reduced glycolysis and oxidative phosphorylation, due to pyruvate depletion. Accordingly, MAO inhibitors decreased the expression of proteins involved in glucose transport (GLUT1) and transformation (HK2). In conclusion, urothelial cancer cells are characterized by a metabolic shift toward glucose-dependent metabolism, which is important for cell growth and is under the regulation of MAO-dependent oxidative stress.


Asunto(s)
Carcinoma , Neoplasias de la Vejiga Urinaria , Antioxidantes/metabolismo , Carcinogénesis/metabolismo , Carcinoma/metabolismo , Catecolaminas/metabolismo , Proliferación Celular , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Estrés Oxidativo , Piruvatos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serotonina/metabolismo , Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo
12.
Biochem Biophys Res Commun ; 634: 100-107, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-36242915

RESUMEN

We have previously shown that monoamine oxidase A (MAO A) mediates prostate cancer growth and metastasis. Further, MAO A/Pten double knockout (DKO) mice were generated and demonstrated that the deletion of MAO A delayed prostate tumor development in the Pten knockout mouse model of prostate adenocarcinoma. Here, we investigated its effect on immune cells in the tumor microenvironment in MAO A/Pten DKO mouse model. Our results shows that Paraffin embedded prostate tissues from MAO A/Pten DKO mice had elevated markers of immune stimulation (CD8+ cytotoxic T cells, granzyme B, and IFNγ) and decreased expression of markers of immune suppression (FoxP3, CD11b, HIF-1-alpha, and arginase 1) compared to parental Pten knockouts (MAO A wildtype). CD11b+ myeloid derived suppressor cells (MDSC) were the primary immunosuppressive cell types in these tumors. The data suggest that deletion of MAO A reduces immune suppression in prostate tumors to enhance antitumor immunity in prostate cancer. Thus, MAO A inhibitor may alleviate immune suppression, increase the antitumor immune response and be used for cancer immunotherapy.


Asunto(s)
Próstata , Neoplasias de la Próstata , Humanos , Masculino , Ratones , Animales , Próstata/patología , Monoaminooxidasa/genética , Neoplasias de la Próstata/patología , Terapia de Inmunosupresión , Microambiente Tumoral , Línea Celular Tumoral
13.
Cells ; 11(17)2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36078109

RESUMEN

Diabetes leads to cardiomyopathy and heart failure, the leading cause of death for diabetic patients. Monoamine oxidase (MAO) inhibition in diabetic cardiomyopathy prevents oxidative stress, mitochondrial and endoplasmic reticulum stress and the development of diastolic dysfunction. However, it is unclear whether, in addition to the direct effects exerted on the mitochondria, MAO activity is able to post-transcriptionally regulate cardiomyocyte function and survival in diabetes. To this aim, we performed gene and miRNA expression profiling in cardiac tissue from streptozotocin-treated mice (model of type 1 diabetes (T1D)), administered with either vehicle or MAOs inhibitor pargyline for 12 weeks. We found that inhibition of MAO activity in T1D hearts leads to profound transcriptomic changes, affecting autophagy and pro-survival pathways activation. MAO activity in T1D hearts increased miR-133a-3p, -193a-3p and -27a-3p expression. These miRNAs target insulin-like growth factor receptor 1 (Igf1r), growth factor receptor bound protein 10 and inositol polyphosphate 4 phosphatase type 1A, respectively, all components of the IGF1R/PI3K/AKT signaling pathway. Indeed, AKT activation was significantly downregulated in T1D hearts, whereas MAO inhibition restored the activation of this pro-survival pathway. The present study provides an important link between MAO activity, transcriptomic changes and activation of pro-survival signaling and autophagy in diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus Tipo 1 , Cardiomiopatías Diabéticas , MicroARNs , Animales , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/metabolismo , Ratones , MicroARNs/genética , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
14.
Hypertens Res ; 45(10): 1582-1598, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35941358

RESUMEN

Renalase is a ~38 kDa flavin-adenine dinucleotide (FAD) domain-containing protein that can function as a cytokine and an anomerase. It is emerging as a novel regulator of cardiometabolic diseases. Expressed mainly in the kidneys, renalase has been reported to have a hypotensive effect and may control blood pressure through regulation of sympathetic tone. Furthermore, genetic variations in the renalase gene, such as a functional missense polymorphism (Glu37Asp), have implications in the cardiovascular and renal systems and can potentially increase the risk of cardiometabolic disorders. Research on the physiological functions and biochemical actions of renalase over the years has indicated a role for renalase as one of the key proteins involved in various disease states, such as diabetes, impaired lipid metabolism, and cancer. Recent studies have identified three transcription factors (viz., Sp1, STAT3, and ZBP89) as key positive regulators in modulating the expression of the human renalase gene. Moreover, renalase is under the post-transcriptional regulation of two microRNAs (viz., miR-29b, and miR-146a), which downregulate renalase expression. While renalase supplementation may be useful for treating hypertension, inhibition of renalase signaling may be beneficial to patients with cancerous tumors. However, more incisive investigations are required to unravel the potential therapeutic applications of renalase. Based on the literature pertaining to the function and physiology of renalase, this review attempts to consolidate and comprehend the role of renalase in regulating cardiometabolic and renal disorders.


Asunto(s)
Hipertensión , Enfermedades Renales , MicroARNs , Humanos , Hipertensión/genética , Enfermedades Renales/genética , MicroARNs/genética , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo
15.
Chem Biol Interact ; 365: 110082, 2022 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-35940281

RESUMEN

Worldwide usage of caffeine results in its constant release into the aquatic environment and growing concerns related to associated risks. We assessed (neuro)toxicity of environmentally relevant concentrations of caffeine, using novel biomarkers of neural function in SH-SY5Y cells and markers of general toxicity also in HepG2 cells. The RQ-PCR analyses showed that caffeine disturbs the expression of genes encoding several key elements of neurotransmitter pathways, with the most prominent responses observed for serotonin receptor 3A, dopamine receptor D2, monoamine oxidase B and GABA-transaminase. Expression of genes encoding synaptotagmin 10 involved in exocytosis of neurotransmitters and ATPase Na+/K+ transporting subunit alpha 3 was also disturbed. Caffeine stimulated the activity of monoamine oxidase, while cytotoxicity and effects on mitochondrial membrane potential were not observed. Our study points out the new possible molecular targets of caffeine and suggests that the raising concerns related to its growing environmental presence are justified.


Asunto(s)
Neuroblastoma , Síndromes de Neurotoxicidad , Biomarcadores/metabolismo , Cafeína/toxicidad , Línea Celular Tumoral , Humanos , Monoaminooxidasa/genética , Neurotransmisores
16.
Biol Reprod ; 107(5): 1279-1295, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-35871545

RESUMEN

The one-carbon metabolism (OCM) pathway provides purines and thymidine for synthesis of nucleic acids required for cell division, and S-adenosyl methionine for polyamine and creatine syntheses and the epigenetic regulation of gene expression. This study aimed to determine if serine hydroxymethyltransferase 2 (SHMT2), a key enzyme in the OCM pathway, is critical for ovine trophectoderm (oTr) cell function and conceptus development by inhibiting translation of SHMT2 mRNA using a morpholino antisense oligonucleotide (MAO). In vitro treatment of oTr cells with MAO-SHMT2 decreased expression of SHMT2 protein, which was accompanied by reduced proliferation (P = 0.053) and migration (P < 0.05) of those cells. Intrauterine injection of MAO-SHMT2 in ewes on Day 11 post-breeding tended to decrease the overall pregnancy rate (on Days 16 and 18) compared with MAO-control (3/10 vs. 7/10, P = 0.07). The three viable conceptuses (n = 2 on Day 16 and n = 1 on Day 18) recovered from MAO-SHMT2 ewes had only partial inhibition of SHMT2 mRNA translation. Conceptuses from the three pregnant MAO-SHMT2 ewes had similar levels of expression of mRNAs and proteins involved in OCM as compared with conceptuses from MAO-control ewes. These results indicate that knockdown of SHMT2 protein reduces proliferation and migration of oTr cells (in vitro) to decrease elongation of blastocysts from spherical to elongated forms. These in vitro effects suggest that increased embryonic deaths in ewes treated with MAO-SHMT2 are the result of decreased SHMT2-mediated trophectoderm cell proliferation and migration supporting a role for the OCM pathway in survival and development of ovine conceptuses.


Asunto(s)
Implantación del Embrión , Epigénesis Genética , Embarazo , Ovinos , Animales , Femenino , Implantación del Embrión/fisiología , Biosíntesis de Proteínas , Embrión de Mamíferos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología
17.
Bioconjug Chem ; 33(6): 1166-1178, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35587267

RESUMEN

Inhibitors for monoamine oxidase-B (MAO-B) were screened from an FV library with a randomized complementarity-determining region 3 (CDR3) region using a monoclonal antibody against dopamine. As the first step, the FV library was expressed on the outer membrane of E. coli by site-directed mutagenesis of the randomized CDR3 region. Among the FV library, variants with a binding affinity to monoclonal antibodies against dopamine were screened and cloned. From the comparison of the binding activity of the screened clones to a control clone with a modified FV antibody (only with CDR1 and CDR2), the CDR3 regions of screened clones were determined to directly interact with the monoclonal antibody against dopamine. These CDR3 sequences were then synthesized as mimotopes (mimicking peptides) of dopamine. The inhibitory activity of two mimotopes against MAO-B was analyzed using HeLa cells overexpressing MAO-B, as well as using activated human astrocytes; their inhibitory activity was compared to that of a commercial inhibitor of MAO-B, selegiline. The inhibition efficiency of the two mimotopes (in comparison with selegiline) was estimated to be 67.2% and 69.4% in the HeLa cells and 64.4% and 58.0% in the human astrocytes. The gene expression pattern in astrocytes after treatment with the two mimotopes was also analyzed and compared with that in the human astrocytes treated with selegiline. Finally, the interaction between two mimotopes and MAO-B was analyzed using docking simulation, and the candidate regions of MAO-B for the interaction with each mimotope were explored through the docking simulation.


Asunto(s)
Monoaminooxidasa , Selegilina , Anticuerpos Monoclonales , Dopamina/metabolismo , Escherichia coli/metabolismo , Células HeLa , Humanos , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Péptidos , Selegilina/farmacología
18.
Int Immunopharmacol ; 109: 108772, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35461155

RESUMEN

OBJECTIVES: Osteoarthritis (OA) is a prevalent degenerative joint disorder characterized by cartilage destruction and extracellular matrix (ECM) degeneration. Here, we studied the potential function of monoamine oxidase A (MAOA) in OA pathogenesis. METHODS: Cartilage tissue samples were collected from 33 patients with knee OA and nine normal healthy controls. Sprague-Dawley rats with anterior cruciate ligament transection (ACLT) and primary chondrocytes treated with interleukin (IL)-1ß were used as OA animal and cell models, respectively. The effects of adenovirus-mediated MAOA overexpression in OA models were studied using Safranin-O staining, immunohistochemistry, CCK-8 assay, EdU assay, flow cytometry, qRT-PCR, western blotting, and immunofluorescence. RESULTS: MAOA was identified as an overlapping downregulating gene in the GSE82107, GSE1919, GSE169077, and GSE29746 datasets. MAOA expression was negatively correlated with OA severity. MAOA downregulation was confirmed in ACLT rats and IL-1ß-treated chondrocytes. Notably, MAOA overexpression significantly inhibited ACLT-induced OA pathogenesis in rats, as was evidenced by the reduced Osteoarthritis Research Society International (OARSI) score and serum crosslinked C-telopeptides of type II collagen (CTX-II) and cartilage oligomeric matrix protein (COMP) levels. These findings show that MAOA overexpression inhibits extracellular matrix (ECM) degradation and promotes ACLT-induced autophagy. The effects of MAOA on ECM degradation and autophagy were also confirmed in IL-1ß-treated primary chondrocytes. Additionally, MAOA protects chondrocytes against IL-1ß-induced apoptosis. Furthermore, treating chondrocytes with 3-MA significantly attenuated the protective effects of MAOA. CONCLUSION: MAOA was identified as a downregulated gene in OA. Restoring MAOA expression protects against chondrocyte loss and ECM degradation through autophagy regulation.


Asunto(s)
Cartílago Articular , Condrocitos , Matriz Extracelular , Monoaminooxidasa , Osteoartritis de la Rodilla , Animales , Autofagia/genética , Cartílago Articular/patología , Células Cultivadas , Condrocitos/patología , Matriz Extracelular/patología , Humanos , Interleucina-1beta/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Ratas , Ratas Sprague-Dawley
19.
Biochem Biophys Res Commun ; 606: 135-141, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35349822

RESUMEN

Neuroendocrine transdifferentiation (NED) of prostate cancer (PCa) is the main cause of failure of androgen receptor inhibitor treatment. However, the molecular mechanisms underlying the development of NEPC, especially treatment-induced NEPC, remain unclear. Emerging evidence indicates that elevated monoamine oxidase A (MAOA) contribute to the proliferation, cell stemness, and bone metastasis in PCa. Here, we generated an enzalutamide-induced NED cell model to assess the role of MAOA during NED. Overall, MAOA expression was significantly increased upon Enz long-term exposure and was required for neuroendocrine marker expression. In particular, Enz was found to induce NED via the MAOA/mTOR/HIF-1α signaling axis. Further analyses revealed that the MAOA inhibitor clorgyline(CLG) may bring multiple benefits to CRPC patients, including better therapeutic effect and delays NED. These findings suggest that MAOA may be an important target for the development of anti-NED therapies, thereby providing a novel strategy for the combined application of CLG and AR inhibitors in the clinic.


Asunto(s)
Transdiferenciación Celular , Monoaminooxidasa , Neoplasias de la Próstata , Línea Celular Tumoral , Humanos , Masculino , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal
20.
Bioengineered ; 13(1): 1049-1061, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35112970

RESUMEN

As a leading complication of sepsis, sepsis-induced cardiac dysfunction (SICD) contributed to the high mortality of patients with sepsis. Long non-coding RNA (LncRNA) LINC00472 has been reported to be in sepsis-induced disease. Nonetheless, its biological function and underlying molecular in SICD remain largely unknown. In this study, in vivo and in vitro SICD models were established via LPS treatment. H&E staining was employed for the evaluation of myocardial injury. ELISA assay was performed to detect cardiac Troponin I (cTnI), creatine kinase-MB (CK-MB), interleukin (IL)-1ß, and tumor necrosis factor-α (TNF-α) levels. Cardiomyocyte viability and apoptosis were assessed via CCK-8 and flow cytometry assays. The transcriptional regulation of YY1 on LINC00472 was demonstrated via ChIP assay. Besides, the interaction between YY1 and LINC00472, as well as the association between miR-335-3p and LINC00472 or MAOA were verified via luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Herein, highly expressed LINC00472 was observed in both in vivo and in vitro SICD models. LINC00472 knockdown substantially attenuated LPS-induced inhibition on cardiomyocyte viability and reversed cardiomyocyte apoptosis and inflammatory response mediated by LPS treatment. YY1 induced LINC00472 upregulation, thereby promoting cardiomyocyte dysfunction induced by LPS. In addition, MAOA upregulation or miR-335-3p inhibition could partly reverse the suppressive effect on LPS-induced cardiomyocyte dysfunction mediated by LINC00472 knockdown. Based on our results, it seemed that YY1-activated LINC00472 might contribute to SICD progression via the miR-335-3p/MAOA pathway.


Asunto(s)
Cardiopatías , MicroARNs , ARN Largo no Codificante , Sepsis , Factor de Transcripción YY1 , Animales , Modelos Animales de Enfermedad , Cardiopatías/etiología , Cardiopatías/genética , Cardiopatías/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Sepsis/complicaciones , Sepsis/genética , Sepsis/metabolismo , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA