Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(4)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38396713

RESUMEN

Carcinoid heart disease (CHD) is a frequent and life-threatening complication in patients with carcinoid tumors. Its clinical management is challenging is some cases since serotonin-induced valve fibrosis leads to heart failure. Telotristat is an inhibitor of tryptophan-hydroxylase (TPH), a key enzyme in serotonin production. Telotristat use in patients with carcinoid syndrome and uncontrollable diarrhea under somatostatin analogs is approved, but its specific role in patients with CHD is still not clear. IN this context, we aimed to explore the effect of telotristat in heart fibrosis using a mouse model of serotonin-secreting metastasized neuroendocrine neoplasm (NEN). To this aim, four treatment groups (n = 10/group) were evaluated: control, monthly octreotide, telotristat alone, and telotristat combined with octreotide. Plasma serotonin and NT-proBNP levels were determined. Heart fibrosis was histologically evaluated after 6 weeks of treatment or when an individual mouse's condition was close to being terminal. Heart fibrosis was observed in all groups. Non-significant reductions in primary tumor growth were observed in all of the treated groups. Feces volume was increased in all groups. A non-significant decrease in feces volume was observed in the octreotide or telotristat-treated groups, while it was significantly reduced with the combined treatment at the end of the study compared with octreotide (52 g reduction; p < 0.01) and the control (44.5 g reduction; p = 0.05). Additionally, plasma NT-proBNP decreased in a non-significant, but clinically relevant, manner in the octreotide (28.2% reduction), telotristat (45.9% reduction), and the octreotide + telotristat (54.1% reduction) treatment groups. No significant changes were observed in plasma serotonin levels. A similar non-significant decrease in heart valve fibrosis was observed in the three treated groups. In conclusion, Telotristat alone and especially in combination with octreotide decreases NT-proBNP levels in a mouse model of serotonin-secreting metastasized NEN, when compared with the control and octreotide, but its effect on heart valve fibrosis (alone and in combination) was not superior to octreotide in monotherapy.


Asunto(s)
Cardiopatía Carcinoide , Tumores Neuroendocrinos , Fenilalanina/análogos & derivados , Pirimidinas , Humanos , Octreótido/farmacología , Octreótido/uso terapéutico , Cardiopatía Carcinoide/tratamiento farmacológico , Serotonina , Tumores Neuroendocrinos/tratamiento farmacológico , Fibrosis
2.
J Transl Med ; 21(1): 879, 2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-38049848

RESUMEN

BACKGROUND: Lung neuroendocrine neoplasms (LungNENs) comprise a heterogeneous group of tumors ranging from indolent lesions with good prognosis to highly aggressive cancers. Carcinoids are the rarest LungNENs, display low to intermediate malignancy and may be surgically managed, but show resistance to radiotherapy/chemotherapy in case of metastasis. Molecular profiling is providing new information to understand lung carcinoids, but its clinical value is still limited. Altered alternative splicing is emerging as a novel cancer hallmark unveiling a highly informative layer. METHODS: We primarily examined the status of the splicing machinery in lung carcinoids, by assessing the expression profile of the core spliceosome components and selected splicing factors in a cohort of 25 carcinoids using a microfluidic array. Results were validated in an external set of 51 samples. Dysregulation of splicing variants was further explored in silico in a separate set of 18 atypical carcinoids. Selected altered factors were tested by immunohistochemistry, their associations with clinical features were assessed and their putative functional roles were evaluated in vitro in two lung carcinoid-derived cell lines. RESULTS: The expression profile of the splicing machinery was profoundly dysregulated. Clustering and classification analyses highlighted five splicing factors: NOVA1, SRSF1, SRSF10, SRSF9 and PRPF8. Anatomopathological analysis showed protein differences in the presence of NOVA1, PRPF8 and SRSF10 in tumor versus non-tumor tissue. Expression levels of each of these factors were differentially related to distinct number and profiles of splicing events, and were associated to both common and disparate functional pathways. Accordingly, modulating the expression of NOVA1, PRPF8 and SRSF10 in vitro predictably influenced cell proliferation and colony formation, supporting their functional relevance and potential as actionable targets. CONCLUSIONS: These results provide primary evidence for dysregulation of the splicing machinery in lung carcinoids and suggest a plausible functional role and therapeutic targetability of NOVA1, PRPF8 and SRSF10.


Asunto(s)
Tumor Carcinoide , Neoplasias Pulmonares , Humanos , Tumor Carcinoide/genética , Tumor Carcinoide/metabolismo , Tumor Carcinoide/patología , Neoplasias Pulmonares/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Empalme Alternativo/genética , Factores de Empalme de ARN/genética , Biomarcadores/metabolismo , Biología , Pulmón/patología , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Antígeno Ventral Neuro-Oncológico
3.
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
4.
Neuroendocrinology ; 110(11-12): 1028-1041, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31940630

RESUMEN

INTRODUCTION: Pituitary neuroendocrine tumors (PitNETs), the most abundant of all intracranial tumors, entail severe comorbidities. First-line therapy is transsphenoidal surgery, but subsequent pharmacological therapy is often required. Unfortunately, many patients are/become unresponsive to available drugs (somatostatin analogues [SSAs]/dopamine agonists), underscoring the need for new therapies. Statins are well-known drugs commonly prescribed to treat hyperlipidemia/cardiovascular diseases, but can convey additional beneficial effects, including antitumor actions. The direct effects of statins on normal human pituitary or PitNETs are poorly known. Thus, we aimed to explore the direct effects of statins, especially simvastatin, on key functional parameters in normal and tumoral pituitary cells, and to evaluate the combined effects of simvastatin with metformin (MF) or SSAs. METHODS: Effects of statins in cell proliferation/viability, hormone secretion, and signaling pathways were evaluated in normal pituitary cells from a primate model (Papio anubis), tumor cells from corticotropinomas, somatotropinomas, nonfunctioning pituitary tumors, and PitNET cell-lines (AtT20/GH3-cells). RESULTS: All statins decreased AtT20-cell proliferation, simvastatin showing stronger effects. Indeed, simvastatin reduced cell viability and/or hormone secretion in all PitNETs subtypes and cell-lines, and ACTH/GH/PRL/FSH/LH secretion (but not expression), in primate cell cultures, by modulating MAPK/PI3K/mTOR pathways and expression of key receptors (GH-releasing hormone-receptor/ghrelin-R/Kiss1-R) regulating pituitary function. Addition of MF or SSAs did not enhance simvastatin antitumor effects. CONCLUSION: Our data reveal direct antitumor effects of simvastatin on PitNET-cells, paving the way to explore these compounds as a possible tool to treat PitNETs.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Hipófisis/efectos de los fármacos , Neoplasias Hipofisarias/tratamiento farmacológico , Simvastatina/farmacología , Adulto , Anciano , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Hipoglucemiantes/farmacología , Masculino , Metformina/farmacología , Ratones , Persona de Mediana Edad , Papio anubis , Ratas , Somatostatina/farmacología , Adulto Joven
5.
Clin Endocrinol (Oxf) ; 86(1): 97-107, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27327536

RESUMEN

OBJECTIVE: The management of patients with differentiated thyroid cancer (DTC) has changed in recent years, and monitoring depends on the risk of persistent/recurrent disease. The objective was to assess the prognostic value of a single stimulated thyroglobulin (Tg) measured at the time of the first radioiodine therapy (Stim-Tg1), and the utility of a second stimulated Tg measurement performed 6-12 months later (Stim-Tg2). We also examined the role of neck ultrasound (US) in the early diagnosis of recurrence. DESIGN: This was a retrospective observational cohort study conducted in a tertiary referral hospital. Of 213 evaluated patients with DTC, 169 were finally included. METHODS: Measurement of Stim-Tg1, Stim-Tg2 and neck US. RESULTS: Stim-Tg1 was undetectable in 71 of 169 patients (42%). All of them (71/71) continued to have negative Stim-Tg2. Seventy of 71 had an excellent response to the first treatment. Sixty-eight of 71 had no evidence of disease after an average follow-up of 7·2 years. In patients with detectable Stim-Tg1 (98/169; 58%), Stim-Tg2 became negative in 40. The negative predictive value (NPV) of Stim-Tg1 was 0·96. The optimal Stim-Tg1 cut-off level for identifying persistence was 3·65 ng/ml. Recurrence was detected in 14 patients. Neck US was useful for identifying local recurrence (13/14; 92·85%). CONCLUSIONS: Stim-Tg1 is a reliable marker with a high NPV. A second stimulation test should be avoided in patients with negative Stim-Tg1. In patients with biochemical persistence, Stim-Tg2 is useful for confirming/ruling out final status. Neck US plays a valuable role in the early diagnosis of recurrence.


Asunto(s)
Recurrencia Local de Neoplasia/diagnóstico , Tiroglobulina/sangre , Neoplasias de la Tiroides/diagnóstico , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia/sangre , Curva ROC , Estudios Retrospectivos , Neoplasias de la Tiroides/sangre , Neoplasias de la Tiroides/terapia , Tirotropina
6.
BMC Endocr Disord ; 17(1): 51, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810850

RESUMEN

BACKGROUND: There are multiple criteria to define remission of type 2 diabetes (DM2) after bariatric surgery but there is not a specific one widely accepted. Our objectives were to compare diagnostic criteria for DM2 remission after bariatric surgery: Criteria from Spanish scientific associations (SEEN/SEEDO/SED) and from the American Diabetes Association (ADA). We also aim to analyse the degree of correlation between these sets of criteria. METHODS: Retrospective observational study in 127 patients undergoing bariatric surgery in a single centre (Hospital Universitario Reina Sofía, Córdoba, Spain) between January 2001 and December 2009. We analysed DM2 remission following bariatric surgery comparing DM2 diagnostic criteria approved by Spanish scientific associations and ADA criteria. RESULTS: In total, 62.2% of patients were women; mean age was 47.1 years. Following surgery, 52% achieved complete remission according to ADA criteria, and 63.8% following the criteria approved by Spanish associations (p = 0.001);18.9 and 8.7%, respectively, showed partial remission (p = 0.007), and 29.1 and 27.6% no remission, according to the criteria approved by each association (p = 0.003). There was good correlation between both sets of criteria (Rho 0.781; p < 0.001). CONCLUSIONS: In our series, using more stringent criteria for defining DM2 remission (ADA criteria) results in a lower rate of remission, although we found a a high degree of correlation between both sets of criteria.


Asunto(s)
Cirugía Bariátrica , Diabetes Mellitus Tipo 2/cirugía , Adulto , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/fisiopatología , Obesidad/cirugía , Inducción de Remisión , Estudios Retrospectivos , España/epidemiología , Resultado del Tratamiento
7.
Q J Nucl Med Mol Imaging ; 60(2): 163-71, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26563902

RESUMEN

BACKGROUND: Most publications to date compare outcomes after post-surgical thyroid remnant ablation stimulated by recombinant human thyrotropin (rhTSH) versus thyroid hormone withholding/withdrawal (THW) in low-recurrence risk differentiated thyroid carcinoma (DTC) patients. We sought to perform this comparison in high-risk patients. METHODS: We retrospectively analyzed ~9-year single-center experience in 70 consecutive adults with initial UICC (Union for International Cancer Control) stage III/IV, M0 DTC undergoing rhTSH-aided (N.=54) or THW-aided (N.=16) high-activity ablation. Endpoints included ablation success and DTC outcome. Assessed ≥1 year post-ablation, ablation success comprised a) no visible scintigraphic thyroid bed uptake or pathological extra-thyroidal uptake; b) undetectable stimulated serum thyroglobulin (Tg) without interfering autoantibodies; c) both criteria. DTC outcome, determined at the latest visit, comprised either 1) "no evidence of disease" (NED): undetectable Tg, negative Tg autoantibodies, negative most recent whole-body scan, no suspicious findings clinically, on neck ultrasonography, or on other imaging; 2) persistent disease: failure to attain NED; or 3) recurrence: loss of NED. RESULTS: After the first ablative activity, ablation success by scintigraphic plus biochemical criteria was 64.8% in rhTSH patients, 56.3% in THW patients (P=NS). After 3.5-year versus 6.2-year median follow-up (P<0.05), DTC outcomes were NED, 85.2%, persistent disease, 13.0%, recurrence, 1.9%, in the rhTSH group and NED, 87.5%, persistent or recurrent disease, 6.3% each, in the THW group (P=NS). CONCLUSION: In patients with initial stage III/IV, M0 DTC, rhTSH-aided and THW-assisted ablation were associated with comparable remnant eradication or DTC cure rates.


Asunto(s)
Técnicas de Ablación/métodos , Proteínas Recombinantes/farmacología , Glándula Tiroides/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/patología , Tirotropina/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Determinación de Punto Final , Femenino , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Recombinantes/uso terapéutico , Estudios Retrospectivos , Neoplasias de la Tiroides/metabolismo , Tirotropina/uso terapéutico , Resultado del Tratamiento , Adulto Joven
8.
Front Endocrinol (Lausanne) ; 14: 1243906, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37867510

RESUMEN

Background: Obesity (OB) is a chronic metabolic disease with important associated comorbidities and mortality. Vitamin D supplementation is frequently administered after bariatric surgery (BS), so as to reduce OB-related complications, maybe including chronic inflammation. Aim: This study aimed to explore relations between vitamin D metabolites and components of the inflammasome machinery in OB before and after BS and their relations with the improvement of metabolic comorbidities. Patients and methods: Epidemiological/clinical/anthropometric/biochemical evaluation was performed in patients with OB at baseline and 6 months after BS. Evaluation of i) vitamin-D metabolites in plasma and ii) components of the inflammasome machinery and inflammatory-associated factors [NOD-like-receptors (NLRs), inflammasome-activation-components, cytokines and inflammation/apoptosis-related components, and cell-cycle and DNA-damage regulators] in peripheral blood mononuclear cells (PBMCs) was performed at baseline and 6 months after BS. Clinical and molecular correlations/associations were analyzed. Results: Significant correlations between vitamin D metabolites and inflammasome-machinery components were observed at baseline, and these correlations were significantly reduced 6 months after BS in parallel to a decrease in inflammation markers, fat mass, and body weight. Treatment with calcifediol remarkably increased 25OHD levels, despite 24,25(OH)2D3 remained stable after BS. Several inflammasome-machinery components were associated with improvement in metabolic comorbidities, especially hypertension and dyslipidemia. Conclusion: The beneficial effects of vitamin D on OB-related comorbidities after BS patients are associated with significant changes in the molecular expression of key inflammasome-machinery components. The expression profile of these inflammasome components can be dynamically modulated in PBMCs after BS and vitamin D supplementation, suggesting that this profile could likely serve as a sensor and early predictor of the reversal of OB-related complications after BS.


Asunto(s)
Cirugía Bariátrica , Obesidad Mórbida , Humanos , Calcifediol , Inflamasomas , Leucocitos Mononucleares , Obesidad/complicaciones , Obesidad/cirugía , Obesidad Mórbida/cirugía , Vitamina D , Inflamación
9.
Transl Res ; 251: 63-73, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35882361

RESUMEN

Dysregulation of the splicing machinery is emerging as a hallmark in cancer due to its association with multiple dysfunctions in tumor cells. Inappropriate function of this machinery can generate tumor-driving splicing variants and trigger oncogenic actions. However, its role in pancreatic neuroendocrine tumors (PanNETs) is poorly defined. In this study we aimed to characterize the expression pattern of a set of splicing machinery components in PanNETs, and their relationship with aggressiveness features. A qPCR-based array was first deployed to determine the expression levels of components of the major (n = 13) and minor spliceosome (n = 4) and associated splicing factors (n = 27), using a microfluidic technology in 20 PanNETs and non-tumoral adjacent samples. Subsequently, in vivo and in vitro models were applied to explore the pathophysiological role of NOVA1. Expression analysis revealed that a substantial proportion of splicing machinery components was altered in tumors. Notably, key splicing factors were overexpressed in PanNETs samples, wherein their levels correlated with clinical and malignancy features. Using in vivo and in vitro assays, we demonstrate that one of those altered factors, NOVA1, is tightly related to cell proliferation, alters pivotal signaling pathways and interferes with responsiveness to drug treatment in PanNETs, suggesting a role for this factor in the aggressiveness of these tumors and its suitability as therapeutic target. Altogether, our results unveil a severe alteration of the splicing machinery in PanNETs and identify the putative relevance of NOVA1 in tumor development/progression, which could provide novel avenues to develop diagnostic biomarkers and therapeutic tools for this pathology.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Pancreáticas , Humanos , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/terapia , Proteínas de Unión al ARN/genética , Proliferación Celular/genética , Factores de Empalme de ARN/genética , Neoplasias Pancreáticas/patología , Antígeno Ventral Neuro-Oncológico
10.
Mol Oncol ; 16(3): 764-779, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34601790

RESUMEN

Somatostatin receptor subtype 5 (SST5 ) is an emerging biomarker and actionable target in pituitary (PitNETs) and pancreatic (PanNETs) neuroendocrine tumors. Transcriptional and epigenetic regulation of SSTR5 gene expression and mRNA biogenesis is poorly understood. Recently, an overlapping natural antisense transcript, SSTR5-AS1, potentially regulating SSTR5 expression, was identified. We aimed to elucidate whether epigenetic processes contribute to the regulation of SSTR5 expression in PitNETs (somatotropinomas) and PanNETs. We analyzed the SSTR5/SSTR5-AS1 human locus in silico to identify CpG islands. SSTR5 and SSTR5-AS1 expression was assessed by quantitative real-time PCR (qPCR) in 27 somatotropinomas, 11 normal pituitaries (NPs), and 15 PanNETs/paired adjacent (control) samples. We evaluated methylation grade in four CpG islands in the SSTR5/SSTR5-AS1 genes. Results revealed that SSTR5 and SSTR5-AS1 were directly correlated in NP, somatotropinoma, and PanNET samples. Interestingly, selected CpG islands were differentially methylated in somatotropinomas compared with NPs. In PanNETs cell lines, SSTR5-AS1 silencing downregulated SSTR5 expression, altered aggressiveness features, and influenced pasireotide response. These results provide evidence that SSTR5 expression in PitNETs and PanNETs can be epigenetically regulated by the SSTR5-AS1 antisense transcript and, indirectly, by DNA methylation, which may thereby impact tumor behavior and treatment response.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Pancreáticas , Neoplasias Hipofisarias , Receptores de Somatostatina , Metilación de ADN , Epigénesis Genética , Humanos , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo
11.
Front Endocrinol (Lausanne) ; 12: 693004, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566886

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents in some cases with hemostatic and thrombotic complications. Pheochromocytomas are unusual, though potentially lethal tumors. Herein we describe the first case of hemorrhage in a pheochromocytoma related to SARS-CoV-2 infection. A 62-year-old man consulted for syncope, fever, and palpitations. He was diagnosed with SARS-CoV-2 pneumonia and presented with a hemorrhage in a previously unknown adrenal mass, which resulted in a catecholaminergic crisis. Medical treatment and surgery were required for symptom control and stabilization. We hereby alert clinicians to watch for additional/unreported clinical manifestations in COVID-19 infection.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/complicaciones , COVID-19/complicaciones , Hemorragia/complicaciones , Feocromocitoma/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Neumonía/complicaciones
12.
J Clin Endocrinol Metab ; 106(12): e4917-e4934, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34363480

RESUMEN

BACKGROUND: Obesity is a metabolic chronic disease with important associated morbidities and mortality. Bariatric surgery is the most effective treatment for maintaining long-term weight loss in severe obesity and, consequently, for decreasing obesity-related complications, including chronic inflammation. AIM: To explore changes in components of the inflammasome machinery after bariatric surgery and their relation with clinical/biochemical parameters at baseline and 6 months after bariatric surgery. PATIENTS AND METHODS: Twenty-two patients with morbid-obesity that underwent bariatric surgery (sleeve gastrectomy and Roux-en-Y gastric bypass) were included. Epidemiological/clinical/anthropometric/biochemical evaluation was performed at baseline and 6 months after bariatric surgery. Inflammasome components and inflammatory-associated factors [nucleotide-binding oligomerization domain-like receptors (NLRs), inflammasome activation components, cytokines and inflammation/apoptosis-related components, and cell-cycle and DNA-damage regulators) were evaluated in peripheral blood mononuclear cells (PBMCs) at baseline and 6 months after bariatric surgery. Clinical molecular correlations/associations were analyzed. Functional parameters (lipid accumulation/viability/apoptosis) were analyzed in response to specific inflammasome components silencing in liver HepG2 cells). RESULTS: A profound dysregulation of inflammasome components after bariatric surgery was found, especially in NLRs and cell-cycle and DNA damage regulators. Several components were associated with baseline metabolic comorbidities including type 2 diabetes (C-C motif chemokine ligand 2/C-X-C motif chemokine receptor 1/sirtuin 1), hypertension (absent in melanoma 2/ASC/purinergic receptor P2X 7), and dyslipidemia [C-X-C motif chemokine ligand 3 (CXCL3)/NLR family pyrin domain containing (NLRP) 7) and displayed changes in their molecular profile 6 months after bariatric surgery. The gene expression fingerprint of certain factors NLR family CARD domain containing 4 (NLRC4)/NLRP12/CXCL3)/C-C motif chemokine ligand 8/toll-like receptor 4) accurately differentiated pre- and postoperative PBMCs. Most changes were independent of the performed surgical technique. Silencing of NLRC4/NLRP12 resulted in altered lipid accumulation, apoptosis rate, and cell viability in HepG2 cells. CONCLUSION: Bariatric surgery induces a profound alteration in the gene expression pattern of components of the inflammasome machinery in PBMCs. Expression and changes of certain inflammasome components are associated to baseline metabolic comorbidities, including type 2 diabetes, and may be related to the improvement and reversion of some obesity-related comorbidities after bariatric surgery.


Asunto(s)
Cirugía Bariátrica/efectos adversos , Diabetes Mellitus Tipo 2/patología , Dislipidemias/patología , Mediadores de Inflamación/metabolismo , Inflamación/patología , Leucocitos Mononucleares/patología , Obesidad Mórbida/cirugía , Adulto , Biomarcadores/metabolismo , Enfermedad Crónica , Estudios de Cohortes , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/etiología , Dislipidemias/metabolismo , Femenino , Estudios de Seguimiento , Gastrectomía/efectos adversos , Humanos , Inflamasomas/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Obesidad Mórbida/patología , Pronóstico
13.
J Clin Endocrinol Metab ; 104(1): 57-73, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30265346

RESUMEN

Context: Biguanides and statins exert beneficial effects on various cancer types. Their precise effects and underlying molecular mechanisms are poorly understood. Materials and Methods: We analyzed the relationship between metabolic syndrome and histological, epidemiological, and prognosis variables in two cohorts of patients with neuroendocrine tumors (NETs): those with lung carcinoids (LCs; n = 81) and those with gastroenteropancreatic NET (GEP-NET; n = 100). Biguanide and statin antitumor effects were investigated by evaluating proliferation, migration, secretion, gene expression, and involved molecular pathways in BON1/QGP1 cell cultures. Results: Pleura invasion was higher (LCs group; P < 0.05) and tumor diameter tended to be increased (GEP-NET group) in patients with type 2 diabetes (T2DM) than in those without. Somatostatin and ghrelin systems mRNA levels differed in tumor tissue of patients with T2DM taking metformin or not. Biguanides decreased proliferation rate in BON1/QGP1 cells; the effects of statins on proliferation rate depended on the statin and cell types, and time. Specifically, only simvastatin and atorvastatin decreased proliferation in BON1 cells, whereas all statins decreased proliferation rate in QGP1 cells. Metformin and simvastatin decreased migration capacity in BON1 cells; biguanides decreased serotonin secretion in BON1 cells. Phenformin increased apoptosis in BON1/QGP1 cells; simvastatin increased apoptosis in QGP1 cells. These antitumor effects likely involved altered expression of key genes related to cancer aggressiveness. Conclusion: A clear inhibitory effect of biguanides and statins was seen on NET-cell aggressiveness. Our results invite additional exploration of the potential therapeutic role of these drugs in treatment of patients with NETs.


Asunto(s)
Biguanidas/uso terapéutico , Diabetes Mellitus Tipo 2/etiología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Tumores Neuroendocrinos/complicaciones , Adulto , Anciano , Apoptosis/efectos de los fármacos , Tumor Carcinoide/tratamiento farmacológico , Tumor Carcinoide/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pleurales/patología , Neoplasias Pleurales/secundario , Pronóstico , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo
14.
Cancers (Basel) ; 11(10)2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31561558

RESUMEN

Pituitary neuroendocrine tumors (PitNETs) constitute approximately 15% of all brain tumors, and most have a sporadic origin. Recent studies suggest that altered alternative splicing and, consequently, appearance of aberrant splicing variants, is a common feature of most tumor pathologies. Moreover, spliceosome is considered an attractive therapeutic target in tumor pathologies, and the inhibition of SF3B1 (e.g., using pladienolide-B) has been shown to exert antitumor effects. Therefore, we aimed to analyze the expression levels of selected splicing-machinery components in 261 PitNETs (somatotropinomas/non-functioning PitNETS/corticotropinomas/prolactinomas) and evaluated the direct effects of pladienolide-B in cell proliferation/viability/hormone secretion in human PitNETs cell cultures and pituitary cell lines (AtT-20/GH3). Results revealed a severe dysregulation of splicing-machinery components in all the PitNET subtypes compared to normal pituitaries and a unique fingerprint of splicing-machinery components that accurately discriminate between normal and tumor tissue in each PitNET subtype. Moreover, expression of specific components was associated with key clinical parameters. Interestingly, certain components were commonly dysregulated throughout all PitNET subtypes. Finally, pladienolide-B reduced cell proliferation/viability/hormone secretion in PitNET cell cultures and cell lines. Altogether, our data demonstrate a drastic dysregulation of the splicing-machinery in PitNETs that might be associated to their tumorigenesis, paving the way to explore the use of specific splicing-machinery components as novel diagnostic/prognostic and therapeutic targets in PitNETs.

15.
J Clin Endocrinol Metab ; 104(8): 3501-3513, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30860580

RESUMEN

CONTEXT: Pituitary neuroendocrine tumors (PitNETs) are a commonly underestimated pathology in terms of incidence and associated morbimortality. Currently, an appreciable subset of patients are resistant or poorly responsive to the main current medical treatments [i.e., synthetic somatostatin analogs (SSAs) and dopamine agonists]. Thus, development and optimization of novel and available medical therapies is necessary. Biguanides (metformin, buformin, and phenformin) are antidiabetic drugs that exert antitumoral actions in several tumor types, but their pharmacological effects on PitNETs are poorly known. OBJECTIVE: We aimed to explore the direct effects of biguanides on key functions (cell viability, hormone release, apoptosis, and signaling pathways) in primary cell cultures from human PitNETs and cell lines. Additionally, we evaluated the effect of combined metformin with SSAs on cell viability and hormone secretion. DESIGN: A total of 13 corticotropinomas, 13 somatotropinomas, 13 nonfunctioning PitNETs, 3 prolactinomas, and 2 tumoral pituitary cell lines (AtT-20 and GH3) were used to evaluate the direct effects of biguanides on cell viability, hormone release, apoptosis, and signaling pathways. RESULTS: Biguanides reduced cell viability in all PitNETs and cell lines (with phenformin being the most effective biguanide) and increased apoptosis in somatotropinomas. Moreover, buformin and phenformin, but not metformin, reduced hormone secretion in a cell type-specific manner. Combination metformin/SSA therapy did not increase SSA monotherapy effectiveness. Effects of biguanides on PitNETs could involve the modulation of AMP-activated protein kinase-dependent ([Ca2+]i, PI3K/Akt) and independent (MAPK) mechanisms. CONCLUSION: Altogether, our data unveil clear antitumoral effects of biguanides on PitNET cells, opening avenues to explore their potential as drugs to treat these pathologies.


Asunto(s)
Antineoplásicos/farmacología , Biguanidas/farmacología , Hipoglucemiantes/farmacología , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Hipofisarias/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
16.
Endocr Relat Cancer ; 26(3): R157-R179, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30615596

RESUMEN

Some biomarkers for functioning and non-functioning neuroendocrine neoplasms (NENs) are currently available. Despite their application in clinical practice, results should be interpreted cautiously. Considering the variable sensitivity and specificity of these parameters, there is an unmet need for novel biomarkers to improve diagnosis and predict patient outcome. Nowadays, several new biomarkers are being evaluated and may become future tools for the management of NENs. These biomarkers include (1) peptides and growth factors; (2) DNA and RNA markers based on genomics analysis, for example, the so-called NET test, which has been developed for analyzing gene transcripts in circulating blood; (3) circulating tumor/endothelial/progenitor cells or cell-free tumor DNA, which represent minimally invasive methods that would provide additional information for monitoring treatment response and (4) improved imaging techniques with novel radiolabeled somatostatin analogs or peptides. Below we summarize some future directions in the development of novel diagnostic and predictive/prognostic biomarkers in NENs. This review is focused on circulating and selected tissue markers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/normas , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/terapia , Biomarcadores de Tumor/genética , Ácidos Nucleicos Libres de Células/metabolismo , Humanos , Células Neoplásicas Circulantes/metabolismo , Tumores Neuroendocrinos/patología , Péptidos/metabolismo , Pronóstico
17.
Drugs ; 79(1): 21-42, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30560479

RESUMEN

Neuroendocrine tumors (NETs) originate from the neuroendocrine cell system in the bronchial and gastrointestinal tract and can produce hormones leading to distinct clinical syndromes. Systemic treatment of patients with unresectable NETs aims to control symptoms related to hormonal overproduction and tumor growth. In the last decades prognosis has improved as a result of increased detection of early stage disease and the introduction of somatostatin analogs (SSAs) as well as several new therapeutic options. SSAs are the first-line medical treatment of NETs and can control hormonal production and tumor growth. The development of next-generation multireceptor targeted and radiolabelled somatostatin analogs, as well as target-directed therapies (as second-line treatment options) further improve progression-free survival in NET patients. To date, however, a significant prolongation of overall survival with systemic treatment in NET has not been convincingly demonstrated. Several new medical options and treatment combinations will become available in the upcoming years, and although preliminary results of preclinical and clinical trials are encouraging, large, preferrably randomized clinical studies are required to provide definitive evidence of their effect on survival and symptom control.


Asunto(s)
Antineoplásicos , Tumores Neuroendocrinos/tratamiento farmacológico , Somatostatina , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Terapia Combinada , Humanos , Terapia Molecular Dirigida/tendencias , Tumores Neuroendocrinos/radioterapia , Supervivencia sin Progresión , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico
18.
J Clin Endocrinol Metab ; 93(6): 2269-76, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18349058

RESUMEN

CONTEXT: Rab proteins regulate the sequential steps of intracellular membrane transport. Alterations of these GTPases and their associated proteins are emerging as the underlying cause for several human diseases involving dysregulated secretory activities. OBJECTIVE: Herein we investigated the role of Rab18, which negatively regulates hormone secretion by interacting with secretory granules, in relation to the altered functioning of tumoral pituitary somatotropes causing acromegaly. PATIENTS: A total of 18 patients diagnosed with pituitary tumors causing acromegaly (nine patients) or nonfunctioning adenomas (nine patients) underwent endoscopic transsphenoidal surgery. Adenomas were subsequently processed to evaluate Rab18 production in relation to GH secretion. RESULTS: We found that somatotropinoma cells are characterized by a high secretory activity concomitantly with a remarkably reduced Rab18 expression (15%) and protein content levels (30%), as compared with cells from nonfunctioning pituitary adenomas derived from patients with normal or reduced GH plasma levels (100%). Furthermore, immunoelectron microscopy revealed that Rab18 association with the surface of GH-containing secretory granules was significantly lower in somatotropes from acromegalies than nonfunctioning pituitary adenomas. Finally, we provide evidence that modulation of Rab18 gene expression can revert substantially the hypersecretory activity of cells because Rab18 overexpression reduced by 40% the capacity of cells from acromegalies to respond to GHRH stimulation. CONCLUSION: These results suggest that molecular alterations affecting individual components of the secretory granule traffic machinery can contribute to maintain a high level of GH in plasma. Accordingly, Rab18 constitutes a valuable target as a diagnostic, prognostic, and/or therapeutic tool for human acromegaly.


Asunto(s)
Acromegalia/genética , Adenoma/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Hormona de Crecimiento Humana/metabolismo , Proteínas de Unión al GTP rab/genética , Acromegalia/etiología , Adenoma/metabolismo , Membrana Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Humanos , ARN Mensajero/metabolismo , Vesículas Secretoras/metabolismo , Somatotrofos/metabolismo , Distribución Tisular , Transfección , Células Tumorales Cultivadas , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/fisiología
19.
Nutr Hosp ; 35(3): 698-706, 2018 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-29974782

RESUMEN

INTRODUCTION: lipid profile suffers adaptive changes during pregnancy due to estrogen stimulation and insulin resistance. Several relations have been suggested between maternal lipid profile, glucose tolerance, endothelial cell dysfunction and long-term cardiovascular risk; the effects of maternal lipid profile metabolism in fetal growth are also inconclusive. Since a regular evaluation and follow-up of lipid profile during pregnancy has not been established yet, we aimed to evaluate the incidence of dyslipidemia in patients with gestational diabetes (GDM) and analyze some putative relations with pregnancy, offspring complications and maternal metabolic syndrome parameters determined three and twelve months after delivery. PATIENTS AND METHODS: two hundred and fifty patients with GDM were included. Full medical history, offspring characteristics, lipid profile and maternal variables of metabolic syndrome were evaluated during pregnancy and three- and twelve-months after delivery. The incidence of dyslipidemia during pregnancy was determined using two different classifications. RESULTS: lower plasma HDL and hypertriglyceridemia were the most current disorders; prematurity or birth weight were not correlated with dyslipidemia. During pregnancy, the lipid-related parameter that better predicted the risk of offspring macrosomia was triglycerides (TG). High TG three months after delivery were correlated to macrosomia and metabolic syndrome variables before and after pregnancy (three and twelve months). CONCLUSIONS: TG during pregnancy is the parameter that best predicts the risk of macrosomia and is related to increased metabolic risk after delivery. The evaluation of lipid profile and other metabolic variables during pregnancy and after delivery is required to early diagnose cardiovascular risk factors, especially in high risk population.


Asunto(s)
Diabetes Gestacional/sangre , Hiperlipidemias/etiología , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/epidemiología , Adulto , Femenino , Macrosomía Fetal , Humanos , Hiperlipidemias/sangre , Recién Nacido , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/etiología , Lípidos/sangre , Valor Predictivo de las Pruebas , Embarazo
20.
Endocrine ; 59(2): 426-437, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29196939

RESUMEN

PURPOSE: Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) comprise a heterogeneous group of malignancies often presenting with metastasis at diagnosis and whose clinical outcome is difficult to predict. Somatostatin (SST) analogs (SSAs) provide a valuable pharmacological tool to palliate hormonal symptoms, and control progression in some NETs. However, many patients do not respond to SSAs or develop resistance, and there are many uncertainties regarding pathophysiology of SST and its receptors (sst1-sst5) in GEP-NETs. METHODS: The expression of SST system components in GEP-NETs was determined, compared with that of non-tumor adjacent and normal tissues and correlated with clinical and histological characteristics. Specifically, 58 patients with GEP-NETs and 14 normal samples were included. Cell viability in NET cell lines was determined in response to specific SSAs. RESULTS: Normal samples and non-tumor adjacent tissues presented a similar expression profile, with appreciable expression of sst2 and sst3, and a lower expression of the other receptors. In contrast, cortistatin, sst1, sst4, and sst5 were overexpressed in tumors, while sst3 and sst4 seemed overexpressed in less differentiated tumors. Some SST system components were related to vascular/nerve invasion and metastasis. In vitro, sst1 and sst3 agonists reduced viability in BON-1 cells, while they, similar to octreotide and pasireotide, increased viability in QGP-1 cells. CONCLUSIONS: These results provide novel information on SST system pathophysiology in GEP-NETs, including relevant associations with clinical-histological parameters, which might help to better understand the intrinsic heterogeneity of NETs and to identify novel biomarkers and/or targets with potential prognostic and/or therapeutic value for GEP-NETs patients.


Asunto(s)
Neoplasias Intestinales/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Intestinales/patología , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA