Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 332
Filtrar
1.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008991

RESUMEN

Aging is a degenerative process involving cell function deterioration, leading to altered metabolic pathways, increased metabolite diversity, and dysregulated metabolism. Previously, we reported that human placenta-derived mesenchymal stem cells (hPD-MSCs) have therapeutic effects on ovarian aging. This study aimed to identify hPD-MSC therapy-induced responses at the metabolite and protein levels and serum biomarker(s) of aging and/or rejuvenation. We observed weight loss after hPD-MSC therapy. Importantly, insulin-like growth factor-I (IGF-I), known prolongs healthy life spans, were markedly elevated in serum. Capillary electrophoresis-time-of-flight mass spectrometry (CE-TOF/MS) analysis identified 176 metabolites, among which the levels of 3-hydroxybutyric acid, glycocholic acid, and taurine, which are associated with health and longevity, were enhanced after hPD-MSC stimulation. Furthermore, after hPD-MSC therapy, the levels of vitamin B6 and its metabolite pyridoxal 5'-phosphate were markedly increased in the serum and liver, respectively. Interestingly, hPD-MSC therapy promoted serotonin production due to increased vitamin B6 metabolism rates. Increased liver serotonin levels after multiple-injection therapy altered the expression of mRNAs and proteins associated with hepatocyte proliferation and mitochondrial biogenesis. Changes in metabolites in circulation after hPD-MSC therapy can be used to identify biomarker(s) of aging and/or rejuvenation. In addition, serotonin is a valuable therapeutic target for reversing aging-associated liver degeneration.


Asunto(s)
Reprogramación Celular , Metabolismo Energético , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Placenta/citología , Rejuvenecimiento , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Biomarcadores , Proliferación Celular , Femenino , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Modelos Animales , Embarazo , Ratas , Serotonina/biosíntesis , Vitamina B 6/metabolismo
2.
J Pharmacol Sci ; 147(3): 251-259, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34507634

RESUMEN

The effects of cyclophosphamide on 5-hydroxytryptamine (5-HT) synthesis in the intestinal tissue of rats were investigated. Rats received 120 mg/kg cyclophosphamide intraperitoneally as a single administration, and kaolin and food intake was measured by an automatic monitoring apparatus. Ileal tissues were collected at either 24 or 72 h after administration. Cyclophosphamide caused a significant increase in kaolin intake at the acute and the delayed phases and was associated with a decrease in food intake, and body weight. Cyclophosphamide had no significant effect on intestinal mucosal morphology, or inducible nitric oxide synthase and cyclooxygenase-2 expression in the intestine. Cyclophosphamide significantly increased tryptophan hydroxylase 1 (TPH1) mRNA expression, number of anti-TPH antibody-positive cells, and 5-HT content in the intestine. Cyclophosphamide also significantly increased the expression of Tac1 mRNA, encoding preprotachykinin-1, which is a preprotein of substance P, and the number of anti-substance P antibody-positive cells in the intestine. Cyclophosphamide significantly increased Lgr5, Bmi1, and Atoh1 mRNA levels, which are markers for the proliferation and differentiation of stem cells. This study demonstrated that cyclophosphamide induced pica in rats, and potentiated 5-HT synthesis associated with hyperplasia of substance P-containing enterochromaffin cells without causing severe intestinal injury.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ciclofosfamida/efectos adversos , Células Enterocromafines/patología , Intestinos/metabolismo , Pica/inducido químicamente , Serotonina/biosíntesis , Animales , Peso Corporal/efectos de los fármacos , Ciclofosfamida/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Hiperplasia/metabolismo , Infusiones Parenterales , Caolín/administración & dosificación , Masculino , Ratas Wistar , Sustancia P/metabolismo , Triptófano Hidroxilasa/metabolismo
3.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070942

RESUMEN

Among mammals, serotonin is predominantly found in the gastrointestinal tract, where it has been shown to participate in pathway-regulating satiation. For the stomach, vascular serotonin release induced by gastric distension is thought to chiefly contribute to satiation after food intake. However, little information is available on the capability of gastric cells to synthesize, release and respond to serotonin by functional changes of mechanisms regulating gastric acid secretion. We investigated whether human gastric cells are capable of serotonin synthesis and release. First, HGT-1 cells, derived from a human adenocarcinoma of the stomach, and human stomach specimens were immunostained positive for serotonin. In HGT-1 cells, incubation with the tryptophan hydroxylase inhibitor p-chlorophenylalanine reduced the mean serotonin-induced fluorescence signal intensity by 27%. Serotonin release of 147 ± 18%, compared to control HGT-1 cells (set to 100%) was demonstrated after treatment with 30 mM of the satiating amino acid L-Arg. Granisetron, a 5-HT3 receptor antagonist, reduced this L-Arg-induced serotonin release, as well as L-Arg-induced proton secretion. Similarly to the in vitro experiment, human antrum samples released serotonin upon incubation with 10 mM L-Arg. Overall, our data suggest that human parietal cells in culture, as well as from the gastric antrum, synthesize serotonin and release it after treatment with L-Arg via an HTR3-related mechanism. Moreover, we suggest not only gastric distension but also gastric acid secretion to result in peripheral serotonin release.


Asunto(s)
Arginina/farmacología , Ácido Gástrico/metabolismo , Células Parietales Gástricas/efectos de los fármacos , Protones , Serotonina/biosíntesis , Línea Celular Tumoral , Fenclonina/farmacología , Expresión Génica , Granisetrón/farmacología , Humanos , Concentración de Iones de Hidrógeno , Células Parietales Gástricas/citología , Células Parietales Gástricas/metabolismo , Inhibidores de Proteasas/farmacología , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas de la Serotonina/farmacología , Estómago/citología , Estómago/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Triptófano Hidroxilasa/antagonistas & inhibidores , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
4.
Can J Physiol Pharmacol ; 99(9): 983-988, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33517848

RESUMEN

Fluoxetine, a commonly prescribed selective serotonin reuptake inhibitor antidepressant, has been shown to increase hepatic lipid accumulation, a key factor in the development of nonalcoholic fatty liver disease. Interestingly, fluoxetine has also been reported to increase peripheral serotonin synthesis. As emerging evidence suggests that serotonin may be involved in the development of nonalcoholic fatty liver disease, we sought to determine if fluoxetine-induced hepatic lipid accumulation is mediated via altered serotonin production. Fluoxetine treatment increased lipid accumulation in association with increased mRNA expression of tryptophan hydroxylase 1 (Tph1, serotonin biosynthetic enzyme) and intracellular serotonin content. Serotonin alone had a similar effect to increase lipid accumulation. Moreover, blocking serotonin synthesis reversed the fluoxetine-induced increases in lipid accumulation. Collectively, these data suggest that fluoxetine-induced lipid accumulation can be mediated, in part, by elevated serotonin production. These results suggest a potential therapeutic target to ameliorate the adverse metabolic effects of fluoxetine exposure.


Asunto(s)
Fluoxetina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Serotonina/biosíntesis , Línea Celular Tumoral , Humanos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Triptófano Hidroxilasa/genética
5.
BMJ Case Rep ; 13(11)2020 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-33168531

RESUMEN

A 27-year-old otherwise healthy man of African descent presented to the hospital with initial symptoms of carcinoid syndrome that later evolved into symptoms of hyperinsulinemic hypoglycaemia. Investigations revealed a metastatic neuroendocrine tumour (NET), co-secreting both serotonin and insulin. Management involved a multimodal approach in an attempt to reduce tumour burden and achieve euglycaemia, which proved to be a significant challenge in the face of refractory hypoglycaemia despite the administration of multiple prohyperglycaemic agents in combination. Unfortunately, given the burden of metastatic disease and multiple medical complications that ensued, the patient passed away. This case highlights the clinical history of a NET co-secreting serotonin and insulin, the use of combination therapy in the treatment of refractory hypoglycaemia in a metastatic insulin-producing tumour and emerging therapeutic modalities in the treatment of these rare malignancies.


Asunto(s)
Manejo de la Enfermedad , Hipoglucemia/terapia , Insulina/biosíntesis , Tumores Neuroendocrinos/complicaciones , Octreótido/uso terapéutico , Neoplasias Pancreáticas/complicaciones , Serotonina/biosíntesis , Adulto , Biopsia , Fármacos Gastrointestinales/uso terapéutico , Humanos , Hipoglucemia/diagnóstico , Hipoglucemia/etiología , Masculino , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/metabolismo , Tomografía de Emisión de Positrones , Tomografía Computarizada por Rayos X
6.
Theranostics ; 10(16): 7351-7368, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32641996

RESUMEN

Rationale: Protein acetylation is tightly linked to transcriptional control and energy metabolism. However, the role of protein acetylation in islet function remains enigmatic. This study aims to determine how protein acetylation controls ß-cell function and explore the underlying mechanism. Methods: The gene-expression profiles were analyzed for rat islets in response to two histone deacetylase (HDAC) inhibitors. Insulin secretion, tryptophan hydroxylase 1 (Tph1) expression, and serotonin synthesis of rat islets were detected after HDAC inhibitor treatment both in vivo and ex vivo. ß-cell-specific Tph1-overexpressing transgenic rats and ß-cell-specific Tph1 knockout mice were constructed to evaluate the role of Tph1 in ß-cell function. The deacetylation of PKA in ß-cells by HDAC1 was investigated by adenoviral infection, immunoprecipitation, and western blot. Results: Inhibition of HDACs greatly potentiated pancreatic ß-cell function and reprogrammed transcriptional landscape of islets. Among the commonly up-regulated genes by two pan-HDAC inhibitors, Tph1 displayed the most prominent change. Specifically, inhibition of HDAC1 and HDAC3 by MS-275 strongly promoted Tph1 expression and endogenous serotonin synthesis in rat islets, concomitantly with enhanced insulin secretory capacity in vivo and ex vivo. ß-cell-specific Tph1-overexpressing transgenic rats exhibited improved glucose tolerance and amplified glucose-stimulated insulin secretion. On the contrary, ß-cell-specific Tph1 knockout mice displayed glucose intolerance and impaired insulin secretion with aging. Moreover, depletion of Tph1 in ß-cells abrogated MS-275-induced insulin hypersecretion. Overexpression of HDAC1, not HDAC3, inhibited Tph1 transcriptional activity and decreased MS-275-stimulated Tph1 expression. Mechanistically, HDAC1 deacetylated PKA catalytic subunit and decreased its activity, resulting in Tph1 transcriptional repression. The acetylation mimetic K62Q mutant of PKA increased its catalytic activity. HDAC1 inhibition exerted a synergistic effect with cAMP/PKA signal on Tph1 expression. Conclusions: The present findings highlight a novel role of HDAC1-PKA-Tph1 signaling in governing ß-cell functional compensation by derepressing serotonin synthesis.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Células Secretoras de Insulina/metabolismo , Serotonina/biosíntesis , Triptófano Hidroxilasa/genética , Acetilación/efectos de los fármacos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Metabolismo Energético/efectos de los fármacos , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Mutación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Transgénicas , Activación Transcripcional/efectos de los fármacos , Triptófano Hidroxilasa/metabolismo
7.
Neurosci Lett ; 735: 135232, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32621948

RESUMEN

Growing evidence suggested that energy deficiency might be involved in the pathophysiological mechanism of depression. Energy deficiency, mainly results from mitochondrial damage, can lead to the dysfunction of synaptic neurotransmission, and further cause depressive-like behavior. The antidepressant effect of resveratrol had been widely demonstrated in previous studies; however, the underlying mechanism remains poorly understood. The present study aimed to investigate whether the antidepressant effects of resveratrol involved in the energy levels and neurotransmission in the hippocampus. We found that resveratrol and fluoxetine significantly attenuated depressive-like behaviors induced by chronic unpredictable mild stress (CUMS), which evidenced by the increased sucrose preference and the reduced immobility time in a forced swimming test. In addition, resveratrol increased hippocampal ATP levels, decreased Na+-K+-ATPase and pyruvate levels, and upregulated the levels of mitochondrial DNA (mtDNA), mRNA expression of sirtuin (SIRT)1 and peroxisome proliferator-activated receptor γ coactivator (PGC)1α. Furthermore, resveratrol and fluoxetine increased serotonin (5-HT) levels and downregulated the mRNA expression of 5-HT transporter (SERT) in the hippocampus. The decreased protein expression of growth-associated protein (GAP)-43 induced by CUMS was also ameliorated by resveratrol and fluoxetine. These findings demonstrated the antidepressant effects of resveratrol and suggested that resveratrol was able to promote mitochondrial biogenesis, enhance ATP and 5-HT levels, as well as upregulate GAP-43 expression in the hippocampus.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Proteína GAP-43/biosíntesis , Hipocampo/metabolismo , Resveratrol/uso terapéutico , Serotonina/biosíntesis , Estrés Psicológico/metabolismo , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Enfermedad Crónica , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Resveratrol/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/psicología , Resultado del Tratamiento
8.
Pharmacol Ther ; 205: 107423, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31629717

RESUMEN

The rate-limiting enzyme in serotonin synthesis is tryptophan hydroxylase (TPH). There are two independent serotonin systems in the body characterized by two isoforms of TPH, TPH1 and TPH2. While TPH2 synthesizes serotonin in the brain, TPH1 is expressed in the gut and in other peripheral tissues and supplies platelets in the circulation with serotonin. This duality of the serotonin system is enforced by the blood-brain barrier which is impermeable for serotonin. In the brain serotonin acts as neurotransmitter and is a main target for the treatment of psychiatric disorders. In the periphery it is released by platelets at the site of activation and elicits numerous physiological effects. TPH1 deficient mice were shown to be protected from diverse diseases including hemostatic, inflammatory, fibrotic, gastrointestinal, and metabolic disorders and therefore serotonin synthesis inhibition emerged as a reasonable therapeutic paradigm. Recently the first TPH inhibitor, telotristat ethyl, came on the market for the treatment of carcinoid syndrome. This review summarizes the state of development and the therapeutic opportunities of such compounds.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Serotonina/biosíntesis , Triptófano Hidroxilasa/antagonistas & inhibidores , Animales , Encéfalo/metabolismo , Desarrollo de Medicamentos , Humanos , Isoenzimas , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/fisiopatología , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Pirimidinas/farmacología , Triptófano Hidroxilasa/metabolismo
9.
J Pathol ; 249(1): 102-113, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31038736

RESUMEN

Serotonin (5-HT) signaling pathways are thought to be involved in colorectal tumorigenesis (CRT), but the role of 5-HT synthesis in the early steps of this process is presently unknown. In this study, we used carcinogen treatment in the tryptophan hydroxylase 1 knockout (Tph1KO) and transgenic (Tph1fl/fl VillinCre ) mouse models defective in 5-HT synthesis to investigate the early mutagenic events associated with CRT. Our observations of the colonic crypt post-treatment followed a timeline designed to understand how disruption of 5-HT synthesis affects the initial steps leading to CRT. We found Tph1KO mice had decreased development of both allograft tumors and colitis-related CRT. Interestingly, carcinogenic exposure alone induced multiple colon tumors and increased cyclooxygenase-2 (Ptgs2) expression in Tph1KO mice. Deletion of interleukin 6 (Il6) in Tph1KO mice confirmed that inflammation was a part of the process. 5-HT deficiency increased colonic DNA damage but inhibited genetic repair of specific carcinogen-related damage, leading to CRT-related inflammatory reactions and dysplasia. To validate a secondary effect of 5-HT deficiency on another DNA repair pathway, we exposed Tph1KO mice to ionizing radiation and found an increase in DNA damage associated with reduced levels of ataxia telangiectasia and Rad3 related (Atr) gene expression in colonocytes. Restoring 5-HT levels with 5-hydroxytryptophan treatment decreased levels of DNA damage and increased Atr expression. Analysis of Tph1fl/fl VillinCre mice with intestine-specific loss of 5-HT synthesis confirmed that DNA repair was tissue specific. In this study, we report a novel protective role for 5-HT synthesis that promotes DNA repair activity during the early stages of colorectal carcinogenesis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Colon/metabolismo , Neoplasias Colorrectales/prevención & control , Daño del ADN , Reparación del ADN , Lesiones Precancerosas/prevención & control , Serotonina/biosíntesis , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Interleucina-6/deficiencia , Interleucina-6/genética , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Transducción de Señal , Factores de Tiempo , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/genética
10.
Drug Des Devel Ther ; 13: 817-824, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30880915

RESUMEN

Gene knockout has been a powerful technique to evaluate the physiologic role of selected gene products. Lexicon pioneered high-throughput gene knockout technology and went further in designing agents to inhibit products of gene expression. Two agents have entered late-stage development. Telotristat is an inhibitor of tryptophan hydroxylase (TPH), preventing the production of serotonin. Although this agent blocks the two isoforms of TPH, it does not cross the blood-brain barrier, thus avoiding central neurologic manifestations. It inhibits the peripheral production of serotonin, and in particular prevents serotonin action in the intestines, resulting in decreased peristaltic action. Lexicon successfully developed telotristat to treat carcinoid syndrome not responding adequately to somatostatin inhibitors. Sotagliflozin development proceeded from the observation that dual inhibition of SGLT2 in the kidneys and SGLT1 in the intestines resulted in increased renal glucose excretion, reduced early-phase glucose absorption, as well as increased blood levels of GLP-1 and PYY. Initial development efforts focused on type 1 diabetes and have shown reduced postprandial glucose levels, less tendency to hypoglycemia, and lower HbA1c. Several other SGLT2 inhibitors have been associated with increased frequency of diabetic ketoacidosis (DKA). In the type 1 trials, sotagliflozin-treated individuals experienced DKA at a higher rate than placebo-treated patients. The sotagliflozin development program has now been extended to trials on type 2 diabetes. Long-term clinical trials will determine the benefits and risks of the agent in comparison to other currently marketed SGLT2 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Técnicas de Inactivación de Genes , Glicósidos/farmacología , Hipoglucemiantes/farmacología , Fenilalanina/análogos & derivados , Pirimidinas/farmacología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Transportador 2 de Sodio-Glucosa/metabolismo , Triptófano Hidroxilasa/antagonistas & inhibidores , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Humanos , Fenilalanina/farmacología , Serotonina/biosíntesis , Serotonina/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/genética , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
11.
Aquat Toxicol ; 200: 206-216, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29775928

RESUMEN

Tributyltin (TBT), an organotin acting as aromatase (Cyp19a1) inhibitor, has been found to disrupt gametogenesis and reproductive behaviors in several fish species. However, few studies addressing the mechanisms underlying the impaired gametogenesis and reproduction have been reported. In this study, female adults of zebrafish (Danio rerio) were continuously exposed to two nominal concentrations of TBT (100 and 500 ng/L, actual concentrations: 90.8 ±â€¯1.3 ng/L and 470.3 ±â€¯2.7 ng/L, respectively) for 28 days. After exposures, TBT decreased the total egg number, reduced the hatchability and elevated the mortality of the larvae. Decreased gonadosomatic index (GSI) and altered percentages of follicles in different developmental stages (increased early-stage follicles and reduced mid/late-stage follicles) were also observed in the ovary of TBT-treated fish. TBT also lowered the plasma level of 17ß-estradiol and suppressed the expressions of cyp19a1a in the ovary. In treated fish, up-regulated expressions of aldhla2, sycp3 and dmc1 were present in the ovary, indicating an enhanced level of meiosis. The mRNA level of vtg1 was dramatically suppressed in the liver of TBT-treated fish, suggesting an insufficient synthesis of Vtg protein, consistent with the decreased percentage of mid/late-stage follicles in the ovaries. Moreover, TBT significantly suppressed the reproductive behaviors of the female fish (duration of both sexes simultaneously in spawning area, the frequency of meeting and the visit in spawning area) and down-regulated the mRNA levels of genes involved in the regulation of reproductive behaviors (cyp19a1b, gnrh-3 and kiss 2) in the brain. In addition, TBT significantly suppressed the expressions of serotonin-related genes, such as tph2 (encoding serotonin synthase), pet1 (marker of serotonin neuron) and kiss 1 (the modulator of serotonin synthesis), suggesting that TBT might disrupt the non-reproductive behaviors of zebrafish. The present study demonstrated that TBT may impair the reproductive success of zebrafish females probably through disrupting oogenesis, disturbing reproductive behaviors and altering serotonin synthesis. The present study greatly extends our understanding on the reproductive toxicity of TBT on fish.


Asunto(s)
Oogénesis/efectos de los fármacos , Reproducción/efectos de los fármacos , Serotonina/biosíntesis , Compuestos de Trialquiltina/toxicidad , Pez Cebra/fisiología , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Estradiol/sangre , Femenino , Larva/efectos de los fármacos , Larva/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Meiosis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serotonina/metabolismo , Vitelogeninas/genética , Vitelogeninas/metabolismo , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/sangre
12.
Trends Pharmacol Sci ; 39(6): 560-572, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29628275

RESUMEN

The first step in serotonin (5-HT) biosynthesis is catalyzed by tryptophan hydroxylase (TPH). There are two independent sources of the monoamine that have distinct functions: first, the TPH1-expressing enterochromaffin cells (ECs) of the gut; second, TPH2-expressing serotonergic neurons. TPH1-deficient mice revealed that peripheral 5-HT plays important roles in platelet function and in inflammatory and fibrotic diseases of gut, pancreas, lung, and liver. Therefore, TPH inhibitors were developed which cannot pass the blood-brain barrier to specifically block peripheral 5-HT synthesis. They showed therapeutic efficacy in several rodent disease models, and telotristat ethyl is the first TPH inhibitor to be approved for the treatment of carcinoid syndrome. We review this development and discuss further therapeutic options for these compounds.


Asunto(s)
Diseño de Fármacos , Células Enterocromafines/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Serotonina/biosíntesis , Triptófano Hidroxilasa/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Células Enterocromafines/enzimología , Humanos , Terapia Molecular Dirigida
13.
Dig Dis Sci ; 63(6): 1473-1484, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29569002

RESUMEN

BACKGROUND: Trypanosoma cruzi (T. cruzi) infects millions of Latin Americans each year and can induce chagasic megacolon. Little is known about how serotonin (5-HT) modulates this condition. Aim We investigated whether 5-HT synthesis alters T. cruzi infection in the colon. MATERIALS AND METHODS: Forty-eight paraffin-embedded samples from normal colon and chagasic megacolon were histopathologically analyzed (173/2009). Tryptophan hydroxylase 1 (Tph1) knockout (KO) mice and c-KitW-sh mice underwent T. cruzi infection together with their wild-type counterparts. Also, mice underwent different drug treatments (16.1.1064.60.3). RESULTS: In both humans and experimental mouse models, the serotonergic system was activated by T. cruzi infection (p < 0.05). While treating Tph1KO mice with 5-HT did not significantly increase parasitemia in the colon (p > 0.05), rescuing its synthesis promoted trypanosomiasis (p < 0.01). T. cruzi-related 5-HT release (p < 0.05) seemed not only to increase inflammatory signaling, but also to enlarge the pericryptal macrophage and mast cell populations (p < 0.01). Knocking out mast cells reduced trypanosomiasis (p < 0.01), although it did not further alter the neuroendocrine cell number and Tph1 expression (p > 0.05). Further experimentation revealed that pharmacologically inhibiting mast cell activity reduced colonic infection (p < 0.01). A similar finding was achieved when 5-HT synthesis was blocked in c-KitW-sh mice (p > 0.05). However, inhibiting mast cell activity in Tph1KO mice increased colonic trypanosomiasis (p < 0.01). CONCLUSION: We show that mast cells may modulate the T. cruzi-related increase of 5-HT synthesis in the intestinal colon.


Asunto(s)
Enfermedad de Chagas/metabolismo , Colon/metabolismo , Parasitosis Intestinales/metabolismo , Mastocitos/metabolismo , Megacolon/metabolismo , Serotonina/biosíntesis , Trypanosoma cruzi/patogenicidad , Adulto , Anciano , Animales , Estudios de Casos y Controles , Enfermedad de Chagas/genética , Enfermedad de Chagas/parasitología , Colon/parasitología , Interacciones Huésped-Patógeno , Humanos , Parasitosis Intestinales/genética , Parasitosis Intestinales/parasitología , Masculino , Mastocitos/parasitología , Megacolon/genética , Megacolon/parasitología , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Tiempo , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
14.
Am J Surg Pathol ; 41(9): 1238-1246, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28719461

RESUMEN

Differential diagnosis based on morphology and immunohistochemistry between a clinically nonfunctioning pituitary neuroendocrine tumor (NET)/pituitary adenoma and a primary or secondary NET of nonpituitary origin in the sellar region may be difficult. Serotonin, a frequently expressed marker in the NETs, has not been systematically evaluated in pituitary NETs. Although mutations in ATRX or DAXX have been reported in a significant proportion of pancreatic NETs, the mutational status of ATRX and DAXX and their possible pathogenetic role in pituitary NETs are unknown. Facing a difficult diagnostic case of an invasive serotonin and adrenocorticotroph hormone immunoreactive NET in the sellar region, we explored the immunohistochemical expression of serotonin, ATRX, and DAXX in a large series of pituitary endocrine tumors of different types from 246 patients and in 2 corticotroph carcinomas. None of the pituitary tumors expressed serotonin, suggesting that serotonin immunoreactive sellar tumors represent primary or secondary NETs of nonpituitary origin. Normal expression of ATRX and DAXX in pituitary tumors suggests that ATRX and DAXX do not play a role in the pathogenesis of pituitary endocrine tumors that remain localized to the sellar and perisellar region. A lack of ATRX or DAXX in a sellar NET suggests a nonpituitary NET, probably of pancreatic origin. One of the 2 examined corticotroph carcinomas, however, demonstrated negative ATRX immunolabeling due to an ATRX gene mutation. Further studies on a larger cohort of pituitary carcinomas are needed to clarify whether ATRX mutations may contribute to the metastatic potential in a subset of pituitary NETs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Adenoma/metabolismo , Biomarcadores de Tumor/biosíntesis , ADN Helicasas/biosíntesis , Tumores Neuroendocrinos/diagnóstico , Proteínas Nucleares/biosíntesis , Neoplasias Hipofisarias/metabolismo , Silla Turca , Serotonina/biosíntesis , Neoplasias Craneales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Adenoma/diagnóstico , Adulto , Anciano , Biomarcadores de Tumor/análisis , Proteínas Co-Represoras , ADN Helicasas/análisis , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Masculino , Chaperonas Moleculares , Proteínas Nucleares/análisis , Neoplasias Hipofisarias/diagnóstico , Serotonina/análisis , Neoplasias Craneales/diagnóstico , Proteína Nuclear Ligada al Cromosoma X
15.
Chemosphere ; 181: 518-529, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28463726

RESUMEN

Amitraz is a formamidine insecticide/acaricide that alters different neurotransmitters levels, among other neurotoxic effects. Oral amitraz exposure (20, 50 and 80 mg/kg bw, 5 days) has been reported to increase serotonin (5-HT), norepinephrine (NE) and dopamine (DA) content and to decrease their metabolites and turnover rates in the male rat brain, particularly in the striatum, prefrontal cortex, and hippocampus. However, the mechanisms by which these alterations are produced are not completely understood. One possibility is that amitraz monoamine oxidase (MAO) inhibition could mediate these effects. Alternatively, it alters serum concentrations of sex steroids that regulate the enzymes responsible for these neurotransmitters synthesis and metabolism. Thus, alterations in sex steroids in the brain could also mediate the observed effects. To test these hypothesis regarding possible mechanisms, we treated male rats with 20, 50 and 80 mg/kg bw for 5 days and then isolated tissue from striatum, prefrontal cortex, and hippocampus. We then measured tissue levels of expression and/or activity of MAO, catechol-O-metyltransferase (COMT), dopamine-ß-hydroxylase (DBH), tyrosine hydroxylase (TH) and tryptophan hydroxylase (TRH) as well as estradiol levels in these regions. Our results show that amitraz did not inhibit MAO activity at these doses, but altered MAO, COMT, DBH, TH and TRH gene expression, as well as TH and TRH activity and estradiol levels. The alteration of these enzymes was partially mediated by dysregulation of estradiol levels. Our present results provide new understanding of the mechanisms contributing to the harmful effects of amitraz.


Asunto(s)
Sistema Nervioso Central/química , Dopamina/metabolismo , Estradiol/metabolismo , Insecticidas/toxicidad , Norepinefrina/metabolismo , Serotonina/metabolismo , Toluidinas/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Cuerpo Estriado/metabolismo , Dopamina/biosíntesis , Estradiol/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Insecticidas/farmacología , Masculino , Neostriado/efectos de los fármacos , Neostriado/enzimología , Neurotransmisores/metabolismo , Norepinefrina/biosíntesis , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/enzimología , Ratas , Serotonina/biosíntesis , Tirosina 3-Monooxigenasa/metabolismo
16.
Am J Dermatopathol ; 39(3): 189-194, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27759693

RESUMEN

BACKGROUND: Giant basal cell carcinomas (GBCCs), (BCC ≥ 5 cm), are often painless, destructive tumors resulting from poorly understood patient neglect. OBJECTIVES: To elucidate etiopathogenic factors distinguishing GBCC from basal cell carcinoma (BCC) and identify predictors for disease-specific death (DSD). METHODS: Case-control study examining clinicopathologic and neuroactive factors (ß-endorphin, met-enkephalin, serotonin, adrenocorticotropic hormone, and neurofilament expression) in GBCC and BCC. Systematic literature review to determine DSD predictors. RESULTS: Thirteen GBCCs (11 patients) were compared with 26 BCCs (25 patients). GBCC significantly differed in size, disease duration, and outcomes; patients were significantly more likely to live alone, lack concern, and have alcoholism. GBCC significantly exhibited infiltrative/morpheic phenotypes, perineural invasion, ulceration, and faster growth. All neuromediators were similarly expressed. Adenoid phenotype was significantly more common in GBCC. Adenoid tumors expressed significantly more ß-endorphin (60% vs. 18%, P = 0.01) and serotonin (30% vs. 4%, P = 0.02). In meta-analysis (n ≤ 311: median age 68 years, disease duration 90 months, tumor diameter 8 cm, 18.4% disease-specific mortality), independent DSD predictors included tumor diameter (cm) (hazard ratio (HR): 1.12, P = 0.003), bone invasion (HR: 4.19, P = 0.015), brain invasion (HR: 8.23, P = 0.001), and distant metastases (HR: 14.48, P = 0.000). CONCLUSIONS: GBCC etiopathogenesis is multifactorial (ie, tumor biology, psychosocial factors). BCC production of paracrine neuromediators deserves further study.


Asunto(s)
Carcinoma Basocelular/patología , Serotonina/biosíntesis , Neoplasias Cutáneas/patología , betaendorfina/biosíntesis , Hormona Adrenocorticotrópica/análisis , Hormona Adrenocorticotrópica/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/psicología , Estudios de Casos y Controles , Encefalina Metionina/análisis , Encefalina Metionina/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Serotonina/análisis , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/psicología , Adulto Joven , betaendorfina/análisis
17.
Cell Rep ; 17(12): 3281-3291, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009296

RESUMEN

In the pancreatic islet, serotonin is an autocrine signal increasing beta cell mass during metabolic challenges such as those associated with pregnancy or high-fat diet. It is still unclear whether serotonin is relevant for regular islet physiology and hormone secretion. Here, we show that human beta cells produce and secrete serotonin when stimulated with increases in glucose concentration. Serotonin secretion from beta cells decreases cyclic AMP (cAMP) levels in neighboring alpha cells via 5-HT1F receptors and inhibits glucagon secretion. Without serotonergic input, alpha cells lose their ability to regulate glucagon secretion in response to changes in glucose concentration, suggesting that diminished serotonergic control of alpha cells can cause glucose blindness and the uncontrolled glucagon secretion associated with diabetes. Supporting this model, pharmacological activation of 5-HT1F receptors reduces glucagon secretion and has hypoglycemic effects in diabetic mice. Thus, modulation of serotonin signaling in the islet represents a drug intervention opportunity.


Asunto(s)
Diabetes Mellitus/metabolismo , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Animales , AMP Cíclico/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/patología , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Serotonina/biosíntesis , Transducción de Señal , Receptor de Serotonina 5-HT1F
18.
Microb Ecol ; 72(3): 730-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27488594

RESUMEN

Listeria monocytogenes is a Gram-positive bacterium that can cause a serious infection. Intestinal microorganisms have been demonstrated to contribute to intestinal physiology not only through immunological responses but also by modulating the intestinal serotonergic system. Serotonin (5-HT) is a neuromodulator that is synthesized in the intestinal epithelium and regulates the whole intestinal physiology. The serotonin transporter (SERT), located in enterocytes, controls intestinal 5-HT availability and therefore serotonin's effects. Infections caused by L. monocytogenes are well described as being due to the invasion of intestinal epithelial cells; however, the effect of L. monocytogenes on the intestinal epithelium remains unknown. The main aim of this work, therefore, was to study the effect of L. monocytogenes on SERT. Caco2/TC7 cell line was used as an enterocyte-like in vitro model, and SERT functional and molecular expression assays were performed. Our results demonstrate that living L. monocytogenes inhibits serotonin uptake by reducing SERT expression at the brush border membrane. However, neither inactivated L. monocytogenes nor soluble metabolites were able to affect SERT. The results also demonstrate that L. monocytogenes yields TLR2 and TLR10 transcriptional changes in intestinal epithelial cells and suggest that TLR10 is potentially involved in the inhibitory effect observed on SERT. Therefore, L. monocytogenes, through TLR10-mediated SERT inhibition, may induce increased intestinal serotonin availability and potentially contributing to intestinal physiological changes and the initiation of the inflammatory response.


Asunto(s)
Células CACO-2/efectos de los fármacos , Intestinos/microbiología , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Inhibidores Selectivos de la Recaptación de Serotonina/antagonistas & inhibidores , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Técnicas de Cultivo de Célula , Células Epiteliales/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/crecimiento & desarrollo , Listeriosis , Técnicas Microbiológicas , Factor 88 de Diferenciación Mieloide , Serotonina/biosíntesis , Serotonina/metabolismo , Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/biosíntesis , Receptor Toll-Like 10/antagonistas & inhibidores , Receptor Toll-Like 10/metabolismo , Receptor Toll-Like 2/metabolismo
19.
Sci Rep ; 6: 30059, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27444653

RESUMEN

The biogenic amine serotonin (5-HT) is a multi-faceted hormone that is synthesized from dietary tryptophan with the rate limiting step being catalyzed by the enzyme tryptophan hydroxylase (TPH). The therapeutic potential of peripheral 5-HT synthesis inhibitors has been demonstrated in a number of clinical and pre-clinical studies in diseases including carcinoid syndrome, lung fibrosis, ulcerative colitis and obesity. Due to the long half-life of 5-HT in blood and lung, changes in steady-state levels are slow to manifest themselves. Here, the administration of stable isotope labeled tryptophan (heavy "h-Trp") and resultant in vivo conversion to h-5-HT is used to monitor 5-HT synthesis in rats. Dose responses for the blockade of h-5-HT appearance in blood with the TPH inhibitors L-para-chlorophenylalanine (30 and 100 mg/kg) and telotristat etiprate (6, 20 and 60 mg/kg), demonstrated that the method enables robust quantification of pharmacodynamic effects on a short time-scale, opening the possibility for rapid screening of TPH1 inhibitors in vivo. In the bleomycin-induced lung fibrosis rat model, the mechanism of lung 5-HT increase was investigated using a combination of synthesis and steady state 5-HT measurement. Elevated 5-HT synthesis measured in the injured lungs was an early predictor of disease induced increases in total 5-HT.


Asunto(s)
Agonistas de Receptores de Serotonina/farmacocinética , Serotonina/biosíntesis , Animales , Modelos Animales de Enfermedad , Fibrosis/patología , Marcaje Isotópico , Enfermedades Pulmonares Intersticiales/patología , Ratas , Triptófano/administración & dosificación , Triptófano Hidroxilasa/antagonistas & inhibidores
20.
Development ; 143(14): 2616-28, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27287799

RESUMEN

Intestinal hormone-producing cells represent the largest endocrine system in the body, but remarkably little is known about enteroendocrine cell type specification in the embryo and adult. We analyzed stage- and cell type-specific deletions of Nkx2.2 and its functional domains in order to characterize its role in the development and maintenance of enteroendocrine cell lineages in the mouse duodenum and colon. Although Nkx2.2 regulates enteroendocrine cell specification in the duodenum at all stages examined, it controls the differentiation of progressively fewer enteroendocrine cell populations when deleted from Ngn3(+) progenitor cells or in the adult duodenum. During embryonic development Nkx2.2 regulates all enteroendocrine cell types, except gastrin and preproglucagon. In developing Ngn3(+) enteroendocrine progenitor cells, Nkx2.2 is not required for the specification of neuropeptide Y and vasoactive intestinal polypeptide, indicating that a subset of these cell populations derive from an Nkx2.2-independent lineage. In adult duodenum, Nkx2.2 becomes dispensable for cholecystokinin and secretin production. In all stages and Nkx2.2 mutant conditions, serotonin-producing enterochromaffin cells were the most severely reduced enteroendocrine lineage in the duodenum and colon. We determined that the transcription factor Lmx1a is expressed in enterochromaffin cells and functions downstream of Nkx2.2. Lmx1a-deficient mice have reduced expression of Tph1, the rate-limiting enzyme for serotonin biosynthesis. These data clarify the function of Nkx2.2 in the specification and homeostatic maintenance of enteroendocrine populations, and identify Lmx1a as a novel enterochromaffin cell marker that is also essential for the production of the serotonin biosynthetic enzyme Tph1.


Asunto(s)
Linaje de la Célula , Células Enterocromafines/citología , Células Enteroendocrinas/citología , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Serotonina/biosíntesis , Factores de Transcripción/metabolismo , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Linaje de la Célula/genética , Colon/metabolismo , Duodeno/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/química , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Reacción en Cadena de la Polimerasa , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Células Madre/citología , Factores de Transcripción/química , Proteínas de Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA