Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pituitary ; 24(3): 351-358, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33433890

RESUMEN

CONTEXT: Somatostatin (SST) and dopamine (DA) inhibit growth hormone (GH) secretion and proliferation of GH-secreting pituitary adenomas (GHomas) through binding to SSTR2 and D2R receptors. Chimeric SST-DA compounds (Dopastatins) display increased potency in inhibiting GH secretion, as compared with individual SST or DA analogs (alone or combined). OBJECTIVE: To assess the efficacy of a second-generation dopastatin, TBR-065, in suppressing GH secretion from human GH- and GH/prolactin(PRL)-omas. DESIGN: We compared the ability of TBR-065 to inhibit GH secretion from primary cultures of human GH- or GH/PRLoma cells to that of the first generation dopastatin, TBR-760 (formerly BIM-23A760), octreotide (OCT) and cabergoline (CAB), the later either alone or combined. We investigated whether there was any impact of BIM-133, the metabolite of TBR-065, on the ability of TBR-065 to inhibit GH in these cultures. METHODS: 17 GH- and GH/PRLomas were included in this study. Inhibition of GH secretion by TBR-065, TBR-760, OCT and CAB (0.1 pM to 0.1 µM) was assessed over a period of 8 h. RESULTS: All tumors expressed SSTR2 and D2R mRNAs. GH suppression was higher with TBR-065 as compared with TBR-760 (Emax = 57 ± 5.6% vs. 41.1 ± 12.5%, respectively, p < 0.001) or with OCT + CAB (Emax = 56.8 ± 7.2% vs. 44.4 ± 9.4%, p < 0.001). BIM-133 did not have any impact on the activity of TBR-065. CONCLUSION: TBR-065 has significantly improved efficacy in suppressing GH secretion as compared to current available therapies and may represent a new promising option for the treatment of acromegaly.


Asunto(s)
Adenoma , Hormona de Crecimiento Humana , Neoplasias Hipofisarias , Adenoma/tratamiento farmacológico , Cabergolina , Dopamina , Humanos , Octreótido/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Receptores de Dopamina D2 , Receptores de Somatostatina/genética , Somatostatina/farmacología , Células Tumorales Cultivadas
2.
Neuroendocrinology ; 110(1-2): 70-82, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31272096

RESUMEN

BACKGROUND: Pituitary neuroendocrine tumors (PitNETs) represent approximately 15% of all intracranial tumors and usually are associated with severe comorbidities. Unfortunately, a relevant number of patients do not respond to currently available pharmacological treatments, that is, somatostatin analogs (SSAs) or dopamine-agonists (DA). Thus, novel, chimeric somatostatin/dopamine compounds (dopastatins) that could improve medical treatment of PitNETs have been designed. OBJECTIVE: This study aims to determine the direct therapeutic effects of a new-generation dopastatin, BIM-065, on primary cell cultures from different PitNETs subtypes. METHODS: Thirty-one PitNET-derived cell cultures (9 corticotropinomas, 9 somatotropinomas, 11 nonfunctioning pituitary adenomas [NFPAs], and 2 prolactinomas), were treated with BIM-065, and key functional endpoints were assessed (cell viability, apoptosis, hormone secretion, expression levels of key genes, free cytosolic [Ca2+]i dynamics, etc.). AtT-20 cell line was used to evaluate signaling pathways in response to BIM-065. RESULTS: This chimeric compound decreased cell viability in all corticotropinomas and somatotropinomas tested, but not in NFPAs. BIM-065 reduced ACTH, GH, chromogranin-A and PRL secretion, and increased apoptosis in corticotropinomas, somatotropinomas, and NFPAs. These effects were possibly mediated through modulation of pivotal signaling cascades like [Ca2+]i kinetic and Akt- or ERK1/2-phosphorylation. CONCLUSIONS: Our results unveil a robust antitumoral effect in vitro of the novel chimeric compound BIM-065 on the main PitNET subtypes, inform on the mechanisms involved, and suggest that BIM-065 could be an efficacious therapeutic option to be considered in the treatment of PitNETs.


Asunto(s)
Dopaminérgicos/farmacología , Dopamina/análogos & derivados , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Hipofisarias/tratamiento farmacológico , Somatostatina/análogos & derivados , Somatostatina/farmacología , Dopamina/farmacología , Humanos , Somatostatina/análisis , Células Tumorales Cultivadas
3.
Carcinogenesis ; 39(3): 447-457, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29272342

RESUMEN

Ghrelin gene generates several variants that regulate multiple pathophysiological functions, including tumor-related processes. In1-ghrelin is a splicing variant that was previously shown to be overexpressed in breast cancer (BCa), where it correlated with proliferation markers; however, its possible association with clinical outcome of BCa patients and underlying mechanisms are still unknown. To address this issue, expression levels and clinical associations of In1-ghrelin were analyzed in a cohort of 117 BCa samples. Additionally, a battery of cellular and molecular assays was implemented using two BCa cell lines (MCF-7 and MDA-MB-231), wherein the role of In1-ghrelin on proliferation, migration, dedifferentiation and signaling pathways was explored. The results generated revealed that high expression of In1-ghrelin in BCa samples was associated with lymph node metastasis and reduced disease-free survival. Indeed, In1-ghrelin overexpression stimulated proliferation and migration in MCF-7 and MDA-MB-231 cells. Similar results were found by treating MDA-MB-231 and MCF-7 with In1-ghrelin-derived peptides. Conversely, In1-ghrelin silencing decreased proliferation and migration capacities of MDA-MB-231. Furthermore, In1-ghrelin (but not ghrelin) overexpression increased the capacity to form mammospheres in both cell lines. These effects could be associated with activation of MAPK-ERK, Jag1/Notch, Wnt/ß-catenin and/or TGF-ß1 pathways. Altogether, our data indicate that In1-ghrelin could play relevant functional roles in the regulation of BCa development and progression and may provide insights to identify novel biomarkers and new therapeutic approaches for this pathology.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Ghrelina/genética , Adulto , Anciano , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/mortalidad , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Ghrelina/metabolismo , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Isoformas de Proteínas
4.
FASEB J ; 31(11): 4682-4696, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28705809

RESUMEN

sst5TMD4, a splice variant of the sst5 gene, is overexpressed and associated with aggressiveness in various endocrine-related tumors, but its presence, functional role, and mechanisms of actions in prostate cancer (PCa)-the most common cancer type in males-is completely unexplored. In this study, formalin-fixed, paraffin-embedded prostate pieces from patients with localized PCa, which included tumoral and nontumoral adjacent regions (n = 45), fresh biopsies from patients with high-risk PCa (n = 52), and healthy fresh prostates from cystoprostatectomies (n = 14) were examined. In addition, PCa cell lines and xenograft models were used to determine the presence and functional role of sst5TMD4. Results demonstrated that sst5TMD4 is overexpressed (mRNA/protein) in PCa samples, and this is especially drastic in metastatic and/or high Gleason score tumor samples. Remarkably, sst5TMD4 expression was associated with an altered frequency of 2 single-nucleotide polymorphisms: rs197055 and rs12599155. In addition, PCa cell lines and xenograft models were used to demonstrate that sst5TMD4 overexpression increases cell proliferation and migration in PCa cells and induces larger tumors in nude mice, whereas its silencing decreased proliferation and migration. Remarkably, sst5TMD4 overexpression activated multiple intracellular pathways (ERK/JNK, MYC/MAX, WNT, retinoblastoma), altered oncogenes and tumor suppressor gene expression, and disrupted the normal response to somatostatin analogs in PCa cells. Altogether, we demonstrate that sst5TMD4 is overexpressed in PCa, especially in those patients with a worse prognosis, and plays an important pathophysiologic role in PCa, which suggesting its potential as a biomarker and/or therapeutic target.-Hormaechea-Agulla, D., Jiménez-Vacas, J. M., Gómez-Gómez, E., L.-López, F., Carrasco-Valiente, J., Valero-Rosa, J., Moreno, M. M., Sánchez-Sánchez, R., Ortega-Salas, R., Gracia-Navarro, F., Culler, M. D., Ibáñez-Costa, A., Gahete, M. D., Requena, M. J., Castaño, J. P., Luque, R. M. The oncogenic role of the spliced somatostatin receptor sst5TMD4 variant in prostate cancer.


Asunto(s)
Empalme Alternativo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas , Neoplasias de la Próstata , Receptores de Somatostatina , Vía de Señalización Wnt , Anciano , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Somatostatina/biosíntesis , Receptores de Somatostatina/genética
5.
Mol Cancer ; 16(1): 146, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851363

RESUMEN

BACKGROUND: The Ghrelin-system is a complex, pleiotropic family composed of several peptides, including native-ghrelin and its In1-ghrelin splicing variant, and receptors (GHSR 1a/b), which are dysregulated in various endocrine-related tumors, where they associate to pathophysiological features, but the presence, functional role, and mechanisms of actions of In1-ghrelin splicing variant in prostate-cancer (PCa), is completely unexplored. Herein, we aimed to determine the presence of key ghrelin-system components (native-ghrelin, In1-ghrelin, GHSR1a/1b) and their potential pathophysiological role in prostate cancer (PCa). METHODS: In1-ghrelin and native-ghrelin expression was evaluated by qPCR in prostate tissues from patients with high PCa-risk (n = 52; fresh-tumoral biopsies), and healthy-prostates (n = 12; from cystoprostatectomies) and correlated with clinical parameters using Spearman-test. In addition, In1-ghrelin and native-ghrelin was measured in plasma from an additional cohort of PCa-patients with different risk levels (n = 30) and control-healthy patients (n = 20). In vivo functional (proliferation/migration) and mechanistic (gene expression/signaling-pathways) assays were performed in PCa-cell lines in response to In1-ghrelin and native-ghrelin treatment, overexpression and/or silencing. Finally, tumor progression was monitored in nude-mice injected with PCa-cells overexpressing In1-ghrelin, native-ghrelin and empty vector (control). RESULTS: In1-ghrelin, but not native-ghrelin, was overexpressed in high-risk PCa-samples compared to normal-prostate (NP), and this expression correlated with that of PSA. Conversely, GHSR1a/1b expression was virtually absent. Remarkably, plasmatic In1-ghrelin, but not native-ghrelin, levels were also higher in PCa-patients compared to healthy-controls. Furthermore, In1-ghrelin treatment/overexpression, and to a much lesser extent native-ghrelin, increased aggressiveness features (cell-proliferation, migration and PSA secretion) of NP and PCa cells. Consistently, nude-mice injected with PC-3-cells stably-transfected with In1-ghrelin, but not native-ghrelin, presented larger tumors. These effects were likely mediated by ERK1/2-signaling activation and involved altered expression of key oncogenes/tumor suppressor genes. Finally, In1-ghrelin silencing reduced cell-proliferation and PSA secretion from PCa cells. CONCLUSIONS: Altogether, our results indicate that In1-ghrelin levels (in tissue) and circulating levels (in plasma) are increased in PCa where it can regulate key pathophysiological processes, thus suggesting that In1-ghrelin may represent a novel biomarker and a new therapeutic target in PCa.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Ghrelina/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Anciano , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular , Ghrelina/análisis , Ghrelina/química , Ghrelina/genética , Xenoinjertos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Próstata/química , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/química , Neoplasias de la Próstata/epidemiología , Isoformas de Proteínas
6.
Circ Res ; 112(11): 1444-55, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23595952

RESUMEN

RATIONALE: Proliferation and migration of smooth muscle cells (SMCs) are key steps for the progression of atherosclerosis and restenosis. Cortistatin is a multifunctional neuropeptide belonging to the somatostatin family that exerts unique functions in the nervous and immune systems. Cortistatin is elevated in plasma of patients experiencing coronary heart disease and attenuates vascular calcification. OBJECTIVE: To investigate the occurrence of vascular cortistatin and its effects on the proliferation and migration of SMCs in vitro and in vivo and to delimitate the receptors and signal transduction pathways governing its actions. METHODS AND RESULTS: SMCs from mouse carotid and human aortic arteries and from human atherosclerotic plaques highly expressed cortistatin. Cortistatin expression positively correlated with the progression of arterial intima hyperplasia. Cortistatin inhibited platelet-derived growth factor-stimulated proliferation of human aortic SMCs via binding to somatostatin receptors (sst2 and sst5) and ghrelin receptor, induction of cAMP and p38-mitogen-activated protein kinase, and inhibition of Akt activity. Moreover, cortistatin impaired lamellipodia formation and migration of human aortic SMCs toward platelet-derived growth factor by inhibiting, in a ghrelin-receptor-dependent manner, Rac1 activation and cytosolic calcium increases. These effects on SMC proliferation and migration correlated with an inhibitory action of cortistatin on the neointimal formation in 2 models of carotid arterial ligation. Endogenous cortistatin seems to play a critical role in regulating SMC function because cortistatin-deficient mice developed higher neointimal hyperplasic lesions than wild-type mice. CONCLUSIONS: Cortistatin emerges as a natural endogenous regulator of SMCs under pathological conditions and an attractive candidate for the pharmacological management of vascular diseases that course with neointimal lesion formation.


Asunto(s)
Aterosclerosis/patología , Traumatismos de las Arterias Carótidas/patología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Neointima/patología , Neuropéptidos/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Humanos , Ligadura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/tratamiento farmacológico , Neointima/metabolismo , Neuropéptidos/genética , Neuropéptidos/farmacología , ARN Mensajero/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Somatostatina/metabolismo
7.
Neurobiol Dis ; 63: 141-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333694

RESUMEN

Clinical pain, as a consequence of inflammation or injury of peripheral organs (inflammatory pain) or nerve injury (neuropathic pain), represents a serious public health issue. Treatment of pain-related suffering requires knowledge of how pain signals are initially interpreted and subsequently transmitted and perpetuated. To limit duration and intensity of pain, inhibitory signals participate in pain perception. Cortistatin is a cyclic-neuropeptide that exerts potent inhibitory actions on cortical neurons and immune cells. Here, we found that cortistatin is a natural analgesic component of the peripheral nociceptive system produced by peptidergic nociceptive neurons of the dorsal root ganglia in response to inflammatory and noxious stimuli. Moreover, cortistatin is produced by GABAergic interneurons of deep layers of dorsal horn of spinal cord. By using cortistatin-deficient mice, we demonstrated that endogenous cortistatin critically tunes pain perception in physiological and pathological states. Furthermore, peripheral and spinal injection of cortistatin potently reduced nocifensive behavior, heat hyperalgesia and tactile allodynia in experimental models of clinical pain evoked by chronic inflammation, surgery and arthritis. The analgesic effects of cortistatin were independent of its anti-inflammatory activity and directly exerted on peripheral and central nociceptive terminals via Gαi-coupled somatostatin-receptors (mainly sstr2) and blocking intracellular signaling that drives neuronal plasticity including protein kinase A-, calcium- and Akt/ERK-mediated release of nociceptive peptides. Moreover, cortistatin could modulate, through its binding to ghrelin-receptor (GHSR1), pain-induced sensitization of secondary neurons in spinal cord. Therefore, cortistatin emerges as an anti-inflammatory factor with potent analgesic effects that offers a new approach to clinical pain therapy, especially in inflammatory states.


Asunto(s)
Analgésicos/uso terapéutico , Neuropéptidos/metabolismo , Dolor/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Femenino , Ganglios Espinales/citología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/deficiencia , Nitrobencenos/uso terapéutico , Dolor/etiología , Dolor/genética , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Médula Espinal/citología , Sulfonamidas/uso terapéutico , Factores de Tiempo
8.
Orphanet J Rare Dis ; 19(1): 164, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637809

RESUMEN

BACKGROUND: Hypoparathyroidism (HP) is a rare endocrine disease commonly caused by the removal or damage of parathyroid glands during surgery and resulting in transient (tHP) or chronic (cHP) disease. cHP is associated with multiple complications and comorbid conditions; however, the economic burden has not been well characterized. The objective of this study was to evaluate the healthcare resource utilization (HCRU) and costs associated with post-surgical cHP, using tHP as a reference. METHODS: This analysis of a US claims database included patients with both an insurance claim for HP and thyroid/neck surgery between October 2014 and December 2019. cHP was defined as an HP claim ≥ 6 months following surgery and tHP was defined as only one HP claim < 6 months following surgery. The cHP index date was the first HP diagnosis claim following their qualifying surgery claim, whereas the tHP index date was the last HP diagnosis claim following the qualifying surgery claim. Patients were continuously enrolled at least 1 year pre- and post-index. Patients' demographic and clinical characteristics, all-cause HCRU, and costs were descriptively analyzed. Total all-cause costs were calculated as the sum of payments for hospitalizations, emergency department, office/clinic visits, and pharmacy. RESULTS: A total of 1,406 cHP and 773 tHP patients met inclusion criteria. The average age (52.1 years cHP, 53.5 years tHP) and representation of females (83.2% cHP, 81.2% tHP) were similar for both groups. Neck dissection surgery was more prevalent in cHP patients (23.6%) than tHP patients (5.3%). During the 1-2 year follow-up period, cHP patients had a higher prevalence of inpatient admissions (17.4%), and emergency visits (26.0%) than the reference group -tHP patients (14.4% and 21.4% respectively). Among those with a hospitalization, the average number of hospitalizations was 1.5-fold higher for cHP patients. cHP patients also saw more specialists, including endocrinologists (28.7% cHP, 15.8% tHP), cardiologists (16.7% cHP, 9.7% tHP), and nephrologists (4.6% cHP, 3.3% tHP). CONCLUSION: This study demonstrates the increased healthcare burden of cHP on the healthcare system in contrast to patients with tHP. Effective treatment options are needed to minimize the additional resources utilized by patients whose HP becomes chronic.


Asunto(s)
Hipoparatiroidismo , Seguro , Femenino , Humanos , Estados Unidos , Persona de Mediana Edad , Estrés Financiero , Estudios Retrospectivos , Atención a la Salud , Hipoparatiroidismo/epidemiología , Costos de la Atención en Salud
9.
Cancer Metastasis Rev ; 30 Suppl 1: 9-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21369878

RESUMEN

Somatostatin is an important regulator of endocrine and exocrine secretion, affecting the release of many hormones. The effects of somatostatin are mediated through its interaction with one of five somatostatin receptors. Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) express multiple somatostatin receptors, making them excellent potential therapeutic targets. Many trials have shown that treatment with somatostatin analogs is associated with disease stabilization and prolonged survival. More recently, somatostatin analogs have been shown to have antiproliferative effects, thus broadening the scope of their uses. In this review, we update the current data on the treatment of GEP-NETs with somatostatin analogs, with particular emphasis on the results of the PROMID study. In addition, we discuss the current state of knowledge of novel therapies against GEP-NETs, including the use of somatostatin analogs with broader receptor binding profiles, chimeric somatostatin-dopamine molecules, combinations of somatostatin analogs with other active chemotherapy agents, and peptide receptor-targeted radionuclide therapy.


Asunto(s)
Hormonas/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Antineoplásicos/uso terapéutico , Dopamina/metabolismo , Dopamina/uso terapéutico , Humanos , Tumores Neuroendocrinos/radioterapia , Radiofármacos/uso terapéutico , Receptores de Péptidos/metabolismo
10.
Clin Endocrinol (Oxf) ; 76(3): 407-14, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21848909

RESUMEN

CONTEXT: First-line therapy for thyrotropin-secreting pituitary adenomas (TSHomas) is neurosurgery, while medical treatment rests mainly on somatostatin analogues. Clinically available sst(2) -preferring analogues, octreotide and lanreotide, induce normalization of hormone levels in approximately 90% of patients and tumour shrinkage in 45%. OBJECTIVE: We evaluated somatostatin 1, 2, 3 and 5 and dopamine D2 receptor expression in tumour samples from three TSHomas, and the relationships between receptor expression, in vitro antiproliferative response and clinical data, including octreotide test and three months of therapy with octreotide long-acting repeatable (LAR). TSHoma cell proliferation was tested in vitro using octreotide, cabergoline and two chimeric compounds, BIM-23A760 and BIM-23A387. RESULTS: All patients showed significant TSH lowering to acute octreotide test, but a hormonal response to long-term treatment was observed in only two patients, showing a high sst(5) /sst(2) ratio. Patient 2, characterized by high expression of sst(2) and sst(1) and a relative lower expression of sst(5) , experienced tachyphylaxis after prolonged octreotide treatment. In vitro, the somatostatin/dopamine receptor agonist BIM-23A760 caused the highest antiproliferative effect among those tested. Combined treatment with octreotide and cabergoline displayed an additive effect of magnitude comparable to that of the other chimeric compound (BIM-23A387). Octreotide resistance was confirmed in cells isolated from the nonresponder patient, although it could be overcome by treatment with the chimeric compounds. CONCLUSIONS: A high sst(5) /sst(2) ratio might be predictive of a positive outcome to long-term treatment with somatostatin analogues in TSHomas. Moreover, combined somatostatin and D(2) receptor targeting might be considered as a potential tool to improve the response rate in octreotide-resistant tumours.


Asunto(s)
Adenoma/tratamiento farmacológico , Agonistas de Dopamina/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética , Somatostatina/uso terapéutico , Adenoma/genética , Adenoma/metabolismo , Adulto , Cabergolina , Proliferación Celular/efectos de los fármacos , Dopamina/análogos & derivados , Dopamina/farmacología , Dopamina/uso terapéutico , Agonistas de Dopamina/farmacología , Sinergismo Farmacológico , Ergolinas/farmacología , Ergolinas/uso terapéutico , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Masculino , Octreótido/farmacología , Octreótido/uso terapéutico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Somatostatina/análogos & derivados , Somatostatina/farmacología , Tirotropina/sangre , Tirotropina/metabolismo , Tiroxina/sangre , Resultado del Tratamiento , Triyodotironina/sangre , Células Tumorales Cultivadas
11.
Bioorg Med Chem ; 20(21): 6394-402, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23040895

RESUMEN

The modulation of 11ß-HSD1 activity with selective inhibitors has beneficial effects on various metabolic disorders including insulin resistance, dyslipidemia and obesity. Here we report the discovery of a series of novel adamantyl carboxamide and acetamide derivatives as selective inhibitors of human 11ß-HSD1 in HEK-293 cells transfected with the HSD11B1 gene. Optimization based on an initially identified 11ß-HSD1 inhibitor (3) led to the discovery of potent inhibitors with IC(50) values in the 100 nM range. These compounds are also highly selective 11ß-HSD1 inhibitors with no activity against 11ß-HSD2 and 17ß-HSD1. Compound 15 (IC(50)=114 nM) with weak inhibitory activity against the key human cytochrome P450 enzymes and moderate stability in incubation with human liver microsomes is worthy of further development. Importantly, compound 41 (IC(50)=280 nM) provides a new lead that incorporates an adamantyl group surrogate and should enable further series diversification.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Acetamidas/farmacología , Adamantano/análogos & derivados , Adamantano/farmacología , Inhibidores Enzimáticos/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Acetamidas/síntesis química , Acetamidas/química , Adamantano/síntesis química , Adamantano/química , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
12.
Elife ; 102021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128468

RESUMEN

Human reproduction is controlled by ~2000 hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Here, we report the discovery and characterization of additional ~150,000-200,000 GnRH-synthesizing cells in the human basal ganglia and basal forebrain. Nearly all extrahypothalamic GnRH neurons expressed the cholinergic marker enzyme choline acetyltransferase. Similarly, hypothalamic GnRH neurons were also cholinergic both in embryonic and adult human brains. Whole-transcriptome analysis of cholinergic interneurons and medium spiny projection neurons laser-microdissected from the human putamen showed selective expression of GNRH1 and GNRHR1 autoreceptors in the cholinergic cell population and uncovered the detailed transcriptome profile and molecular connectome of these two cell types. Higher-order non-reproductive functions regulated by GnRH under physiological conditions in the human basal ganglia and basal forebrain require clarification. The role and changes of GnRH/GnRHR1 signaling in neurodegenerative disorders affecting cholinergic neurocircuitries, including Parkinson's and Alzheimer's diseases, need to be explored.


Asunto(s)
Ganglios Basales , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas , Adulto , Prosencéfalo Basal/citología , Ganglios Basales/citología , Ganglios Basales/metabolismo , Ganglios Basales/fisiología , Células Cultivadas , Colina O-Acetiltransferasa , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología , Putamen/citología , Transcriptoma
13.
Kidney Int ; 77(1): 23-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19890275

RESUMEN

Skeletal muscle mitochondrial dysfunction and insulin resistance occur in chronic kidney disease. Ghrelin is a gastric hormone previously shown to enhance muscle mitochondrial enzyme activities and AKT-mediated insulin signaling independent of food intake in healthy rats. Here we determined the impact of ghrelin treatment on anorexia, skeletal muscle mitochondrial oxidative capacity, AKT phosphorylation as a measure of insulin signaling, and lean body mass in a rat model of chronic kidney disease. Ghrelin infusion promoted higher food intake and lean body mass. Further, although muscle mitochondrial enzyme activities were low in the rats with CKD (chronic kidney disease), they normalized with ghrelin treatment, a change that was consistent with the increase in the transcript levels of regulators of mitochondrial biogenesis and lipid metabolism. This was associated with a lower muscle triglyceride content and higher AKT phosphorylation. Pair-feeding showed that mitochondrial effects of ghrelin are independent of changes in food intake, whereas combined ghrelin treatment and higher food intake were needed to enhance AKT phosphorylation. Thus, ghrelin-induced muscle mitochondrial changes and lower tissue triglycerides could favor insulin action and muscle anabolism in the presence of improvement in food intake. Our study shows that combined effects of ghrelin on appetite and muscle mitochondria improve muscle metabolic and nutritional alterations in chronic kidney disease. This could have potential beneficial impact on patient morbidity and survival.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Insulina , Fallo Renal Crónico , Mitocondrias Musculares/enzimología , Músculo Esquelético/ultraestructura , Estado Nutricional , Oxidación-Reducción , Fosforilación , Ratas , Triglicéridos
14.
Mol Endocrinol ; 23(3): 337-48, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19131507

RESUMEN

Somatostatin (SRIF) binds G protein-coupled SRIF receptor subtypes (SST1, -2, -3, -4, and -5) to regulate cell secretion and proliferation. Hypothalamic SRIF inhibits pituitary growth hormone, thyroid stimulating hormone, and ACTH secretion. We tested SRIF-independent constitutive SST activity in AtT20 mouse pituitary corticotroph cells in which ACTH secretion is highly sensitive to SRIF action. Stable transfectants expressing SST2 or SST5 were sensitized to selective agonist action, and constitutive SST receptor activity was demonstrated by forskolin and pertussis toxin cAMP cell responses. Persistent constitutive SST activity decreased cell ACTH responses to CRH through decreased expression of CRH receptor subtype 1. Decreased dopamine receptor type 1 expression was associated with attenuated dopamine agonist action, whereas responses to isoproterenol were enhanced through increased beta2-adrenoreceptor expression. Thus, integrated pituitary cell ACTH regulation is determined both by phasic SRIF action, as well as by tonic constitutive SST activity, independently of SRIF.


Asunto(s)
Células Endocrinas/fisiología , Hipófisis/fisiología , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/fisiología , Hormona Adrenocorticotrópica/metabolismo , Células Cultivadas , Células Endocrinas/efectos de los fármacos , Células Endocrinas/metabolismo , Humanos , Ligandos , Octreótido/farmacología , Péptidos/farmacología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/genética , Somatostatina/metabolismo , Somatostatina/farmacología , Especificidad por Sustrato , Transfección
15.
Endocrinol Metab (Seoul) ; 35(1): 177-187, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32207278

RESUMEN

BACKGROUND: Acromegaly is a rare disease primarily caused by growth hormone (GH)-secreting pituitary adenomas, and its treatment is costly. Moreover, some patients are unresponsive to treatment. Hence, there are increasing efforts to develop new drugs with improved effectiveness for this disease. BIM23B065 is a novel chimeric molecule that acts on both somatostatin and dopamine receptors. This study aimed to investigate the effects of BIM23B065 compared with those of a somatostatin receptor analog and a dopamine agonist. METHODS: The effects of BIM23B065 on the proliferation, GH and insulin-like growth factor-1 (IGF-1) levels, and extracellular signal-regulated kinase (ERK) 1/2 and cyclic AMP response element binding (CREB) phosphorylation of GH3 cells were investigated with MTS assay, enzyme-linked immunosorbent assay, and Western blotting, respectively. The dosage and treatment duration of BIM23B065 were tested in animal models of GH-secreting pituitary adenoma. The effect of BIM23B065 (3 mg/kg/day) on changes in IGF-1 levels before and after treatment was further investigated. RESULTS: In vitro, BIM23B065 treatment decreased GH release in the culture media and downregulated ERK 1/2 and CREB phosphorylation to 22% and 26%, respectively. In vivo, IGF-1 expression decreased to 50 % after 4 weeks of treatment with BIM23B065 using an osmotic pump implant. Moreover, magnetic resonance imaging results showed that the tumor size decreased significantly following treatment with BIM23B065 for 4 weeks. CONCLUSION: The novel chimeric molecule was effective in decreasing IGF-1 and GH levels and may serve as an effective therapeutic agent for acromegaly.


Asunto(s)
Adenoma/tratamiento farmacológico , Dopamina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/fisiología , Receptores Dopaminérgicos/química , Receptores de Somatostatina/antagonistas & inhibidores , Somatostatina/farmacología , Adenoma/metabolismo , Adenoma/patología , Animales , Proliferación Celular , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Células Tumorales Cultivadas
16.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591776

RESUMEN

TBR-760 (formerly BIM-23A760) is a chimeric dopamine (DA)-somatostatin (SST) compound with potent agonist activity at both DA type 2 (D2R) and SST type 2 (SSTR2) receptors. Studies have shown that chimeric DA-SST compounds are more efficacious than individual DA and/or SST analogues, either alone or combined, in inhibiting secretion from primary cultures of human somatotroph and lactotroph tumor cells. Nonfunctioning pituitary adenomas (NFPAs) express both D2R and SSTR2 and, consequently, may respond to TBR-760. We used a mouse model with the pro-opiomelanocortin (POMC) gene knocked out that spontaneously develops aggressive NFPAs. Genomic microarray and DA and SST receptor messenger RNA expression analysis indicate that POMC KO mouse tumors and human NFPAs have similar expression profiles, despite arising from different cell lineages, establishing POMC KO mice as a model for study of NFPAs. Treatment with TBR-760 for 8 weeks resulted in nearly complete inhibition of established tumor growth, whereas tumors from vehicle-treated mice increased in size by 890 ± 0.7%. Comparing TBR-760 with its individual DA and SST components, TBR-760 arrested tumor growth. Treatment with equimolar or 10×-higher doses of the individual SST or DA agonists, either alone or in combination, had no significant effect. One exception was the lower dose of DA agonist that induced modest suppression of tumor growth. Only the chimeric compound TBR-760 arrested tumor growth in this mouse model of NFPA. Further, significant tumor shrinkage was observed in 20% of the mice treated with TBR-760. These results support the development of TBR-760 as a therapy for patients with NFPA.


Asunto(s)
Adenoma/tratamiento farmacológico , Adenoma/patología , Proliferación Celular/efectos de los fármacos , Dopamina/análogos & derivados , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/patología , Somatostatina/análogos & derivados , Adenoma/genética , Animales , Proliferación Celular/genética , Modelos Animales de Enfermedad , Dopamina/farmacología , Dopamina/uso terapéutico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Noqueados , Análisis por Micromatrices , Invasividad Neoplásica , Neoplasias Hipofisarias/genética , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Somatostatina/farmacología , Somatostatina/uso terapéutico
17.
Clin Cancer Res ; 26(4): 957-969, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31624102

RESUMEN

PURPOSE: Somatostatin analogues (SSA) are efficacious and safe treatments for a variety of neuroendocrine tumors, especially pituitary neuroendocrine tumors (PitNET). Their therapeutic effects are mainly mediated by somatostatin receptors SST2 and SST5. Most SSAs, such as octreotide/lanreotide/pasireotide, are either nonselective or activate mainly SST2. However, nonfunctioning pituitary tumors (NFPTs), the most common PitNET type, mainly express SST3 and finding peptides that activate this particular somatostatin receptor has been very challenging. Therefore, the main objective of this study was to identify SST3-agonists and characterize their effects on experimental NFPT models. EXPERIMENTAL DESIGN: Binding to SSTs and cAMP level determinations were used to screen a peptide library and identify SST3-agonists. Key functional parameters (cell viability/caspase activity/chromogranin-A secretion/mRNA expression/intracellular signaling pathways) were assessed on NFPT primary cell cultures in response to SST3-agonists. Tumor growth was assessed in a preclinical PitNET mouse model treated with a SST3-agonist. RESULTS: We successfully identified the first SST3-agonist peptides. SST3-agonists lowered cell viability and chromogranin-A secretion, increased apoptosis in vitro, and reduced tumor growth in a preclinical PitNET model. As expected, inhibition of cell viability in response to SST3-agonists defined two NFPT populations: responsive and unresponsive, wherein responsive NFPTs expressed more SST3 than unresponsive NFPTs and exhibited a profound reduction of MAPK, PI3K-AKT/mTOR, and JAK/STAT signaling pathways upon SST3-agonist treatments. Concurrently, SSTR3 silencing increased cell viability in a subset of NFPTs. CONCLUSIONS: This study demonstrates that SST3-agonists activate signaling mechanisms that reduce NFPT cell viability and inhibit pituitary tumor growth in experimental models that expresses SST3, suggesting that targeting this receptor could be an efficacious treatment for NFPTs.


Asunto(s)
Tumores Neuroendocrinos/tratamiento farmacológico , Péptidos/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Receptores de Somatostatina/agonistas , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Péptidos/química , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Transducción de Señal , Células Tumorales Cultivadas , Adulto Joven
18.
Am J Physiol Endocrinol Metab ; 297(5): E1078-88, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19706788

RESUMEN

Somatostatin receptors (SSTR1-5) mediate antiproliferative effects. In C6 rat glioma cells, somatostatin is cytostatic in vitro via phosphotyrosine phosphatase-dependent inhibition of ERK1/2 activity mediated by SSTR1, -2, and -5. Here we analyzed the effects of SSTR activation on C6 glioma growth in vivo and the intracellular mechanisms involved, comparing somatostatin effects with selective agonists for SSTR1, -2, and -5 (BIM-23745, BIM-23120, BIM-23206) or receptor biselective compounds (SSTR1 and -2, BIM-23704; and SSTR2 and -5, BIM-23190). Nude mice subcutaneously xenografted with C6 cells were treated with somatostatin, SSTR agonists (50 µg, twice/day), or vehicle. Tumor growth was evaluated every 3 days for 19 days. The intracellular pathways responsible of SSTR effects in vivo were evaluated measuring Ki-67, phospho-ERK1/2, and p27(kip1) expression by immunohistochemistry in sections from explanted tumors. Somatostatin and SSTR1, -2, and -5 agonists strongly inhibited in vivo C6 tumor growth, intratumoral neovessel formation, Ki-67 expression, and ERK1/2 phosphorylation and induced upregulation of p27(Kip1), whereas only a modest activation of caspase-3 was observed. Somatostatin (acting on SSTR1, -2, and -5) displayed the highest efficacy; SSTR5 selective agonist showed a stronger effect than SSTR1 agonist, and SSTR2 agonist was less effective. On the other hand, SSTR1 and -2 agonists maximally reduced tumor neovascularization. The combined activation of SSTR1 and -2 showed a synergistic activity, reaching a higher efficacy than BIM-23206, whereas the simultaneous activation of SSTR2 and -5 resulted in a response resembling SSTR5 effects. Thus the simultaneous activation of different SSTRs inhibits glioma cell proliferation in vivo through both direct cytotostatic and antiangiogenic effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Receptores de Somatostatina/agonistas , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Glioma/irrigación sanguínea , Glioma/patología , Inmunohistoquímica , Ratones , Ratones Desnudos , Microcirculación/efectos de los fármacos , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Oligopéptidos/farmacología , Piperazinas/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Somatostatina/análogos & derivados , Somatostatina/farmacología
19.
Mol Cell Endocrinol ; 301(1-2): 169-73, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18775471

RESUMEN

11beta-Hydroxysteroid dehydrogenases (11beta-HSDs) are key enzymes regulating the pre-receptor metabolism of glucocorticoid hormones, which play essential roles in various vital physiological processes. The modulation of 11beta-HSD type 1 activity with selective inhibitors has beneficial effects on various conditions including insulin resistance, dyslipidemia and obesity. Therefore, inhibition of tissue-specific glucocorticoid action by regulating 11beta-HSD1 constitutes a promising treatment for metabolic and cardiovascular diseases. Here we report the discovery of a series of novel adamantyl carboxamides as selective inhibitors of human 11beta-HSD1 in HEK-293 cells transfected with the HSD11B1 gene. Compounds 9 and 14 show inhibitory activity against 11beta-HSD1 with IC(50) values in 100nM range. Docking studies with the potent compound 8 into the crystal structure of human 11beta-HSD1 (1XU9) reveals how the molecule may interact with the enzyme and cofactor.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Línea Celular , Cortisona/química , Cortisona/metabolismo , Inhibidores Enzimáticos/química , Humanos , Hidrocortisona/química , Hidrocortisona/metabolismo , Concentración 50 Inhibidora , Modelos Moleculares , NADP/metabolismo
20.
Endocr Relat Cancer ; 26(6): 585-599, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30939452

RESUMEN

Control of symptoms related to hormonal hypersecretion by functioning neuroendocrine tumors (NETs) is challenging. New therapeutic options are required. Since novel in vitro tumor models seem to better mimic the tumor in vivo conditions, we aimed to study the effect of somatostatin and dopamine receptor agonists (octreotide and cabergoline, respectively) and novel somatostatin-dopamine chimeric multi-receptor drugs (BIM-065, BIM-23A760) using 2D (monolayer) and 3D (spheroids) cultures. Dose-response studies in 2D and 3D human pancreatic NET cell cultures (BON-1 and QGP-1) were performed under serum-containing and serum-deprived conditions. Cell proliferation, somatostatin and dopamine receptor expression (SSTs and D2R), apoptosis, lactate dehydrogenase, as well as serotonin and chromogranin A (CgA) release were assessed. The following results were obtained. 3D cultures of BON-1/QGP-1 allowed better cell survival than 2D cultures in serum-deprived conditions. SSTs and D2R mRNA levels were higher in the 3D model vs 2D model. Octreotide/cabergoline/BIM-065/BIM-23A760 treatment did not affect cell growth or spheroid size. In BON-1 2D-cultures, only BIM-23A760 significantly inhibited CgA release -this effect being more pronounced in 3D cultures. In BON-1 2D cultures, cabergoline/BIM-065/BIM-23A760 treatment decreased serotonin release (maximal effect up to 40%), being this effect again more potent in 3D cultures (up to 67% inhibition; with BIM-23A760 having the most potent effects). In QGP-1, cabergoline/BIM-065 treatment decreased serotonin release only in the 3D model. In conclusion, cultures of NET 3D spheroids represent a promising method for evaluating cell proliferation and secretion in NET cell-line models. Compared to 2D models, 3D models grow relatively serum independent. In 3D model, SST-D2R multi-receptor targeting drugs inhibit CgA and serotonin secretion, but not NET cell growth.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular/efectos de los fármacos , Cromogranina A/metabolismo , Tumores Neuroendocrinos/patología , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/metabolismo , Serotonina/metabolismo , Antineoplásicos Hormonales/farmacología , Cabergolina/farmacología , Dopamina/análogos & derivados , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Humanos , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Octreótido/farmacología , Receptores Dopaminérgicos/química , Receptores de Somatostatina/química , Somatostatina/análogos & derivados , Somatostatina/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA