Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38396713

RESUMO

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.


Assuntos
Doença Cardíaca Carcinoide , Tumores Neuroendócrinos , Fenilalanina/análogos & derivados , Pirimidinas , Humanos , Octreotida/farmacologia , Octreotida/uso terapêutico , Doença Cardíaca Carcinoide/tratamento farmacológico , Serotonina , Tumores Neuroendócrinos/tratamento farmacológico , Fibrose
2.
J Transl Med ; 21(1): 879, 2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-38049848

RESUMO

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.


Assuntos
Tumor Carcinoide , Neoplasias Pulmonares , Humanos , Tumor Carcinoide/genética , Tumor Carcinoide/metabolismo , Tumor Carcinoide/patologia , Neoplasias Pulmonares/patologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo/genética , Fatores de Processamento de RNA/genética , Biomarcadores/metabolismo , Biologia , Pulmão/patologia , Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Antígeno Neuro-Oncológico Ventral
3.
Front Endocrinol (Lausanne) ; 14: 1243906, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37867510

RESUMO

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.


Assuntos
Cirurgia Bariátrica , Obesidade Mórbida , Humanos , Calcifediol , Inflamassomos , Leucócitos Mononucleares , Obesidade/complicações , Obesidade/cirurgia , Obesidade Mórbida/cirurgia , Vitamina D , Inflamação
4.
Transl Res ; 251: 63-73, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35882361

RESUMO

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.


Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/terapia , Proteínas de Ligação a RNA/genética , Proliferação de Células/genética , Fatores de Processamento de RNA/genética , Neoplasias Pancreáticas/patologia , Antígeno Neuro-Oncológico Ventral
6.
Mol Oncol ; 16(3): 764-779, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34601790

RESUMO

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.


Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Hipofisárias , Receptores de Somatostatina , Metilação de DNA , Epigênese Genética , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo
7.
Front Endocrinol (Lausanne) ; 12: 693004, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34566886

RESUMO

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.


Assuntos
Neoplasias das Glândulas Suprarrenais/complicações , COVID-19/complicações , Hemorragia/complicações , Feocromocitoma/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Pneumonia/complicações
8.
J Clin Endocrinol Metab ; 106(12): e4917-e4934, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34363480

RESUMO

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.


Assuntos
Cirurgia Bariátrica/efeitos adversos , Diabetes Mellitus Tipo 2/patologia , Dislipidemias/patologia , Mediadores da Inflamação/metabolismo , Inflamação/patologia , Leucócitos Mononucleares/patologia , Obesidade Mórbida/cirurgia , Adulto , Biomarcadores/metabolismo , Doença Crônica , Estudos de Coortes , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/etiologia , Dislipidemias/metabolismo , Feminino , Seguimentos , Gastrectomia/efeitos adversos , Humanos , Inflamassomos/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade Mórbida/patologia , Prognóstico
10.
Neuroendocrinology ; 110(11-12): 1028-1041, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31940630

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Hipófise/efeitos dos fármacos , Neoplasias Hipofisárias/tratamento farmacológico , Sinvastatina/farmacologia , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Humanos , Hipoglicemiantes/farmacologia , Masculino , Metformina/farmacologia , Camundongos , Pessoa de Meia-Idade , Papio anubis , Ratos , Somatostatina/farmacologia , Adulto Jovem
11.
Neuroendocrinology ; 110(1-2): 70-82, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31272096

RESUMO

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.


Assuntos
Dopaminérgicos/farmacologia , Dopamina/análogos & derivados , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Hipofisárias/tratamento farmacológico , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Dopamina/farmacologia , Humanos , Somatostatina/análise , Células Tumorais Cultivadas
12.
Cancers (Basel) ; 11(10)2019 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-31561558

RESUMO

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.

13.
J Clin Endocrinol Metab ; 104(8): 3501-3513, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30860580

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Biguanidas/farmacologia , Hipoglicemiantes/farmacologia , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Hipofisárias/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
14.
Endocr Relat Cancer ; 26(3): R157-R179, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30615596

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/normas , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/terapia , Biomarcadores Tumorais/genética , Ácidos Nucleicos Livres/metabolismo , Humanos , Células Neoplásicas Circulantes/metabolismo , Tumores Neuroendócrinos/patologia , Peptídeos/metabolismo , Prognóstico
15.
Drugs ; 79(1): 21-42, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30560479

RESUMO

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.


Assuntos
Antineoplásicos , Tumores Neuroendócrinos/tratamento farmacológico , Somatostatina , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Terapia Combinada , Humanos , Terapia de Alvo Molecular/tendências , Tumores Neuroendócrinos/radioterapia , Intervalo Livre de Progressão , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico
16.
J Clin Endocrinol Metab ; 104(1): 57-73, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30265346

RESUMO

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.


Assuntos
Biguanidas/uso terapêutico , Diabetes Mellitus Tipo 2/etiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Tumores Neuroendócrinos/complicações , Adulto , Idoso , Apoptose/efeitos dos fármacos , Tumor Carcinoide/tratamento farmacológico , Tumor Carcinoide/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Neoplasias Intestinais/tratamento farmacológico , Neoplasias Intestinais/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pleurais/patologia , Neoplasias Pleurais/secundário , Prognóstico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo
17.
Clin Transl Gastroenterol ; 9(10): 196, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30297816

RESUMO

OBJECTIVES: The association between the presence and alterations of the components of the ghrelin system and the development and progression of neuroendocrine tumors (NETs) is still controversial and remains unclear. METHODS: Here, we systematically evaluated the expression levels (by quantitative-PCR) of key ghrelin system components of in gastroenteropancreatic (GEP)-NETs, as compared to non-tumor adjacent (NTA; n = 42) and normal tissues (NT; n = 14). Then, we analyzed their putative associations with clinical-histological characteristics. RESULTS: The results indicate that ghrelin and its receptor GHSR1a are present in a high proportion of normal tissues, while the enzyme ghrelin-O-acyltransferase (GOAT) and the splicing variants In1-ghrelin and GHSR1b were present in a lower proportion of normal tissues. In contrast, all ghrelin system components were present in a high proportion of tumor and NTA tissues. GOAT was significantly overexpressed (by quantitative-PCR (qPCR)) in tumor samples compared to NTA, while a trend was found for ghrelin, In1-ghrelin and GHSR1a. In addition, expression of these components displayed significant correlations with key clinical parameters. The marked overexpression of GOAT in tumor samples compared to NTA regions was confirmed by IHC, revealing that this enzyme is particularly overexpressed in gastrointestinal NETs, where it is directly correlated with tumor diameter. CONCLUSIONS: These results provide novel information on the presence and potential pathophysiological implications of the ghrelin system components in GEP-NETs, wherein GOAT might represent a novel diagnostic biomarker.


Assuntos
Aciltransferases/metabolismo , Biomarcadores Tumorais/metabolismo , Grelina/metabolismo , Neoplasias Intestinais/metabolismo , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Grelina/metabolismo , Neoplasias Gástricas/metabolismo , Aciltransferases/genética , Adulto , Idoso , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Feminino , Grelina/genética , Humanos , Neoplasias Intestinais/genética , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Tumores Neuroendócrinos/genética , Neoplasias Pancreáticas/genética , Valor Preditivo dos Testes , Receptores de Grelina/genética , Neoplasias Gástricas/genética
18.
Nutr Hosp ; 35(3): 698-706, 2018 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-29974782

RESUMO

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.


Assuntos
Diabetes Gestacional/sangue , Hiperlipidemias/etiologia , Complicações na Gravidez/sangue , Complicações na Gravidez/epidemiologia , Adulto , Feminino , Macrossomia Fetal , Humanos , Hiperlipidemias/sangue , Recém-Nascido , Doenças do Recém-Nascido/epidemiologia , Doenças do Recém-Nascido/etiologia , Lipídeos/sangue , Valor Preditivo dos Testes , Gravidez
19.
Nutr. hosp ; 35(3): 698-706, mayo-jun. 2018. tab, graf
Artigo em Inglês | IBECS | ID: ibc-180130

RESUMO

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


Introducción: los cambios del perfil lipídico durante el embarazo se relacionan probablemente con la estimulación estrogénica y la resistencia a la insulina. Diversas relaciones se han planteado entre el perfil lipídico gestacional, la intolerancia a la glucosa, la disfunción endotelial y el riesgo cardiovascular a largo plazo; sus efectos sobre el crecimiento fetal no son concluyentes. Dado que no existe un protocolo de diagnóstico y seguimiento de la dislipemia durante el embarazo, el objetivo del presente estudio fue evaluar la incidencia de dislipemia en pacientes con diabetes gestacional (DMG) y analizar su relación con complicaciones maternas, fetales y variables de síndrome metabólico 3 y 12 meses tras el parto. Pacientes y métodos: fueron incluidos 250 pacientes con DMG. Se analizaron variables clínicas maternas y del recién nacido y se determinó el perfil lipídico durante el embarazo, 3 y 12 meses tras el parto. La incidencia de dislipemia se realizó utilizando dos clasificaciones diferentes. Resultados: las alteraciones más frecuentes fueron bajos niveles de HDL y altos de triglicéridos (TG). La prematuridad o el peso al nacer no se relacionaron con la presencia de dislipemia. El parámetro lipídico que mejor predijo el riesgo de macrosomía fueron los TG. Altos niveles de TG tres meses después del parto se relacionaron con macrosomía y variables de síndrome metabólico pregestacional, así como 3 y 12 meses después del parto. Conclusiones: los niveles de TG durante el embarazo representan el parámetro que mejor predice el riesgo de macrosomía y se relacionan con un mayor riesgo metabólico después del parto. La evaluación del perfil lipídico durante el embarazo y después del parto permite un diagnóstico precoz de factores de riesgo cardiovascular, especialmente en poblaciones de alto riesgo


Assuntos
Humanos , Masculino , Feminino , Gravidez , Recém-Nascido , Adulto , Diabetes Gestacional/sangue , Hiperlipidemias/etiologia , Complicações na Gravidez/sangue , Complicações na Gravidez/epidemiologia , Macrossomia Fetal , Hiperlipidemias/sangue , Doenças do Recém-Nascido/epidemiologia , Doenças do Recém-Nascido/etiologia , Lipídeos/sangue , Valor Preditivo dos Testes
20.
Endocrine ; 59(2): 426-437, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29196939

RESUMO

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.


Assuntos
Neoplasias Intestinais/metabolismo , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Idoso , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Intestinais/patologia , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Neoplasias Gástricas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...