Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Endocr Relat Cancer ; 30(10)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37493200

RESUMO

Pheochromocytomas/paragangliomas (PPGLs) are neuroendocrine tumours, mostly resulting from mutations in predisposing genes. Mutations of succinate dehydrogenase (SDH) subunit B (SDHB) are associated with high probability of metastatic disease. Since bioelectrical properties and signalling in cancer are an emerging field, we investigated the metabolic, functional and electrophysiological characteristics in human succinate dehydrogenase subunit B (SDHB)-deficient pheochromocytoma cells. These cells exhibited reduced SDH function with elevated succinate-to-fumarate ratio and reduced intracellular ATP levels. The analysis of membrane passive properties revealed a more hyperpolarized membrane potential and a lower cell capacitance of SDHB-deficient cells compared to the parental ones. These bioelectrical changes were associated with reduced proliferation and adhesion capacity of SDHB-deficient cells. Only in SDHB-deficient cells, we also observed an increased amplitude of potassium currents suggesting an activation of ATP-sensitive potassium channels (KATP). Indeed, exposure of the SDHB-deficient cells to glibenclamide, a specific KATP inhibitor, or to ATP caused normalization of potassium current features and altered proliferation and adhesion. In this work, we show for the first time that reduced intracellular ATP levels in SDHB-deficient chromaffin cells impaired cell bioelectrical properties, which, in turn, are associated with an increased cell aggressiveness. Moreover, we first ever demonstrated that glibenclamide not only reduced the outward potassium currents in SDHB-deficient cells but increased their growth capacity, reduced their ability to migrate and shifted their phenotype towards one more similar to that of parental one.


Assuntos
Neoplasias das Glândulas Suprarrenais , Células Cromafins , Paraganglioma , Feocromocitoma , Humanos , Succinato Desidrogenase/genética , Glibureto/farmacologia , Paraganglioma/genética , Feocromocitoma/genética , Neoplasias das Glândulas Suprarrenais/genética , Células Cromafins/metabolismo , Células Cromafins/patologia , Trifosfato de Adenosina
2.
Neurobiol Dis ; 179: 106046, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36806818

RESUMO

From a pathogenic perspective, Huntington's disease (HD) is being considered as a synaptopathy. As such, alterations in brain neurotransmitter release occur. As the activity of the sympathoadrenal axis is centrally controlled, deficits in the exocytotic release of catecholamine release may also occur. In fact, in chromaffin cells (CCs) of the adrenal medulla of the R6/1 model of HD, decrease of secretion and altered kinetics of the exocytotic fusion pore have been reported. Those alterations could be linked to mitochondrial deficits occurring in peripheral CCs, similar to those described in brain mitochondria. Here we have inquired about alterations in mitochondrial structure and function and their impact on exocytosis and calcium channel currents (ICa). We have monitored various parameters linked to those events, in wild type (WT) and the R6/1 mouse model of HD at a pre-disease stage (2 months age, 2 m), and when motor deficits are present (7 months age, 7 m). In isolated CCs from 7 m and in the adrenal medulla of R6/1 mice, we found the following alterations (with respect 7 m WT mice): (i) augmented fragmented mitochondria and oxidative stress with increased oxidized glutathione; (ii) decreased basal and maximal respiration; (iii) diminution of ATP cell levels; (iv) mitochondrial depolarization; (v) drastic decrease of catecholamine release with poorer potentiation by protonophore FCCP; (vi) decreased ICa inhibition by FCCP; and (vii) lesser potentiation by BayK8644 of ICa and smaller prolongation of current deactivation. Of note was the fact several of these alterations were already manifested in CCs from 2 m R6/1 mice at pre-disease stages. Based on those results, a plausible hypothesis can be raised in the sense that altered mitochondrial function seems to be an early primary event in HD pathogenesis. This is in line with an increasing number of mitochondrial, metabolic, and inflammatory alterations being recently reported in various HD peripheral tissues.


Assuntos
Células Cromafins , Doença de Huntington , Camundongos , Animais , Doença de Huntington/metabolismo , Cálcio/metabolismo , Camundongos Transgênicos , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/metabolismo , Células Cromafins/metabolismo , Células Cromafins/patologia , Catecolaminas , Mitocôndrias/metabolismo , Exocitose/fisiologia , Modelos Animais de Doenças
3.
J Histochem Cytochem ; 70(5): 335-356, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35400231

RESUMO

Secretogranin II (SgII) and III (SgIII) function within peptide hormone-producing cells and are involved in secretory granule formation. However, their function in active amine-producing cells is not fully understood. In this study, we analyzed the expression profiles of SgII and SgIII in canine adrenal medulla and pheochromocytomas by immunohistochemical staining. In normal adrenal tissues, the intensity of coexpression of these two secretogranins (Sgs) differed from each chromaffin cell, although a complete match was not observed. The coexpression of vesicular monoamine transporter 2 (VMAT2) with SgIII was similar to that with chromogranin A, but there was a subpopulation of VMAT2-expressing cells that were negative or hardly detectable for SgII. These results are the first to indicate that there are distinct expression patterns for SgII and SgIII in adrenal chromaffin cells. Furthermore, the expression of these two Sgs varied in intensity among pheochromocytomas and did not necessarily correlate with clinical plasma catecholamine levels in patients. However, compared with SgIII, the expression of SgII was shown to be strong at the single-cell level in some tumor tissues. These findings provide a fundamental understanding of the expression differences between SgII and SgIII in normal adrenal chromaffin cells and pheochromocytomas.


Assuntos
Neoplasias das Glândulas Suprarrenais , Células Cromafins , Feocromocitoma , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Neoplasias das Glândulas Suprarrenais/veterinária , Animais , Células Cromafins/metabolismo , Células Cromafins/patologia , Cromograninas/metabolismo , Cães , Humanos , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Feocromocitoma/veterinária , Secretogranina II/metabolismo
4.
Am J Pathol ; 192(1): 121-129, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34626576

RESUMO

The pathogenesis of Ebola virus disease (EVD) is still incomplete, in spite of the availability of a nonhuman primate modelfor more than 4 decades. To further investigate EVD pathogenesis, a natural history study was conducted using 27 Chinese-origin rhesus macaques. Of these, 24 macaques were exposed intramuscularly to Kikwit Ebola virus and euthanized at predetermined time points or when end-stage clinical disease criteria were met, and 3 sham-exposed macaques were euthanized on study day 0. This study showed for the first time that Ebola virus causes uterine cervicitis, vaginitis, posthitis, and medullary adrenalitis. Not only was Ebola virus detected in the interstitial stromal cells of the genital tract, but it was also present in the epididymal and seminal vesicular tubular epithelial cells, ectocervical and vaginal squamous epithelial cells, and seminal fluid. Furthermore, as early as day 3 after exposure, Ebola virus replicative intermediate RNA was detected in Kupffer cells and hepatocytes. These findings in the nonhuman model provide additional insight into potential sexual transmission, possible disruption of sympathetic hormone production, and early virus replication sites in human EVD patients.


Assuntos
Ebolavirus/fisiologia , Hormônios/metabolismo , Fígado/virologia , Tropismo/fisiologia , Replicação Viral/fisiologia , Animais , Células Cromafins/patologia , Células Cromafins/virologia , Modelos Animais de Doenças , Epididimo/patologia , Epididimo/virologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Hepatócitos/patologia , Hepatócitos/virologia , Células de Kupffer/patologia , Células de Kupffer/virologia , Macaca mulatta , Masculino , Cervicite Uterina/patologia , Cervicite Uterina/virologia , Vaginite/patologia , Vaginite/virologia
5.
Nat Commun ; 12(1): 5309, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493726

RESUMO

Childhood neuroblastoma has a remarkable variability in outcome. Age at diagnosis is one of the most important prognostic factors, with children less than 1 year old having favorable outcomes. Here we study single-cell and single-nuclei transcriptomes of neuroblastoma with different clinical risk groups and stages, including healthy adrenal gland. We compare tumor cell populations with embryonic mouse sympatho-adrenal derivatives, and post-natal human adrenal gland. We provide evidence that low and high-risk neuroblastoma have different cell identities, representing two disease entities. Low-risk neuroblastoma presents a transcriptome that resembles sympatho- and chromaffin cells, whereas malignant cells enriched in high-risk neuroblastoma resembles a subtype of TRKB+ cholinergic progenitor population identified in human post-natal gland. Analyses of these populations reveal different gene expression programs for worst and better survival in correlation with age at diagnosis. Our findings reveal two cellular identities and a composition of human neuroblastoma tumors reflecting clinical heterogeneity and outcome.


Assuntos
Neoplasias das Glândulas Suprarrenais/genética , Glândulas Suprarrenais/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Receptor trkB/genética , Transcriptoma , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/mortalidade , Neoplasias das Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/patologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Pré-Escolar , Células Cromafins/metabolismo , Células Cromafins/patologia , Diagnóstico Precoce , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas de Neoplasias/classificação , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Receptor trkB/metabolismo , Medição de Risco , Análise de Célula Única , Especificidade da Espécie , Análise de Sobrevida
6.
Nat Genet ; 53(5): 694-706, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33833454

RESUMO

Characterization of the progression of cellular states during human embryogenesis can provide insights into the origin of pediatric diseases. We examined the transcriptional states of neural crest- and mesoderm-derived lineages differentiating into adrenal glands, kidneys, endothelium and hematopoietic tissue between post-conception weeks 6 and 14 of human development. Our results reveal transitions connecting the intermediate mesoderm and progenitors of organ primordia, the hematopoietic system and endothelial subtypes. Unexpectedly, by using a combination of single-cell transcriptomics and lineage tracing, we found that intra-adrenal sympathoblasts at that stage are directly derived from nerve-associated Schwann cell precursors, similarly to local chromaffin cells, whereas the majority of extra-adrenal sympathoblasts arise from the migratory neural crest. In humans, this process persists during several weeks of development within the large intra-adrenal ganglia-like structures, which may also serve as reservoirs of originating cells in neuroblastoma.


Assuntos
Linhagem da Célula , Embrião de Mamíferos/metabolismo , Neuroblastoma/embriologia , Neuroblastoma/genética , Análise de Célula Única , Sistema Simpático-Suprarrenal/embriologia , Transcriptoma/genética , Animais , Células Cromafins/metabolismo , Células Cromafins/patologia , Análise por Conglomerados , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Camundongos , Células-Tronco Neurais/metabolismo , Neuroblastoma/patologia , Células de Schwann/metabolismo , Células de Schwann/patologia , Microambiente Tumoral
7.
Cell Signal ; 76: 109811, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33075487

RESUMO

Resveratrol (RESV) is one of the most abundant polyphenol-stilbene compounds found in red wine with well-established cardioprotective and antihypertensive effects. Hyperactivity of the sympathoadrenal axis seems to be one of the major contributing factors in the pathogenesis of human essential hypertension. Alterations in outward voltage-dependent potassium currents (IK) and inward voltage-dependent sodium (INa), calcium (ICa) and nicotinic (IACh) currents, CCs excitability, Ca2+ homeostasis, and catecholamine exocytosis were previously related to the hypertensive state. This raised the issue of whether in vivo long-term RESV treatment can directly act as a modulator of Ca2+ influx or a regulator of ion channel permeability in CCs. We monitored outward and inward currents, and cytosolic Ca2+ concentrations ([Ca2+]c) using different pharmacological approaches in CCs from normotensive (WKY) and hypertensive (SHR) animals chronically exposed to trans-RESV (50 mg/L/v.o, 28 days). The long-term RESV treatment prevented the increase of the systolic blood pressure (SBP) in SHR, without reversion of cardiac hypertrophy. We also found an increase of the outward IK, reduction in inward INa,ICa, and IACh, and the mitigation of [Ca2+]c overload in CCs from SHR at the end of RESV treatment. Our data revealed that electrophysiological alterations of the CCs and in its Ca2+ homeostasis are potential new targets related to the antihypertensive effects of long-term RESV treatment.


Assuntos
Anti-Hipertensivos/farmacologia , Células Cromafins/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Resveratrol/farmacologia , Animais , Cálcio/metabolismo , Células Cromafins/patologia , Masculino , Cultura Primária de Células , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
8.
Cell Rep ; 32(12): 108171, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32966799

RESUMO

High-risk neuroblastomas typically display an undifferentiated or poorly differentiated morphology. It is therefore vital to understand molecular mechanisms that block the differentiation process. We identify an important role for oncogenic ALK-ERK1/2-SP1 signaling in the maintenance of undifferentiated neural crest-derived progenitors through the repression of DLG2, a candidate tumor suppressor gene in neuroblastoma. DLG2 is expressed in the murine "bridge signature" that represents the transcriptional transition state when neural crest cells or Schwann cell precursors differentiate to chromaffin cells of the adrenal gland. We show that the restoration of DLG2 expression spontaneously drives neuroblastoma cell differentiation, highlighting the importance of DLG2 in this process. These findings are supported by genetic analyses of high-risk 11q deletion neuroblastomas, which identified genetic lesions in the DLG2 gene. Our data also suggest that further exploration of other bridge genes may help elucidate the mechanisms underlying the differentiation of NC-derived progenitors and their contribution to neuroblastomas.


Assuntos
Quinase do Linfoma Anaplásico/genética , Diferenciação Celular , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Guanilato Quinases/genética , Neuroblastoma/genética , Neuroblastoma/patologia , Proteínas Supressoras de Tumor/genética , Adrenérgicos/metabolismo , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cromafins/efeitos dos fármacos , Células Cromafins/metabolismo , Células Cromafins/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Guanilato Quinases/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fator de Crescimento Neural/farmacologia , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Prognóstico , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Células de Schwann/patologia , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Resultado do Tratamento , Tretinoína/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
FASEB J ; 34(1): 303-315, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914648

RESUMO

Mutations in succinate dehydrogenase (SDH) lead to the development of tumors in a restricted subset of cell types, including chromaffin cells and paraganglia. The molecular basis for this specificity is currently unknown. We show that loss of SDH activity in a chromaffin cell model does not perturb complex I function, retaining the ability to oxidize NADH within the electron transport chain. This activity supports continued oxidation of substrates within the tricarboxylic acid (TCA) cycle. However, due to the block in the TCA cycle at SDH, the high glutamine oxidation activity is only maintained through an efflux of succinate. We also show that although the mitochondria of SDH-deficient cells are less active per se, their higher mass per cell results in an overall respiratory rate that is comparable with wild-type cells. Finally, we observed that when their mitochondria are uncoupled, SDH-deficient cells are unable to preserve their viability, suggesting that the mitochondrial metabolic network is unable to compensate when exposed to additional stress. We therefore show that in contrast to models of SDH deficiency based on epithelial cells, a chromaffin cell model retains aspects of metabolic "health," which could form the basis of cell specificity of this rare tumor type.


Assuntos
Células Cromafins/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Neoplasias/metabolismo , Succinato Desidrogenase/fisiologia , Animais , Células Cromafins/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Doenças Mitocondriais/patologia , Mutação , NAD/metabolismo , Neoplasias/patologia , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Transcriptoma
10.
Int J Obes (Lond) ; 43(2): 263-275, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29717268

RESUMO

BACKGROUND: Obesity-associated activation of sympathetic nervous outflow is well documented, whereas involvement of dysregulated adrenomedullary hormonal function in obesity is less clear. This study assessed relationships of sympathoadrenal function with indices of obesity and influences of circulating catecholamines on body mass. METHODS: Anthropometric and clinical data along with plasma and 24-h urine samples were collected from 590 volunteers and 1368 patients tested for phaeochromocytoma and paraganglioma (PPGL), among whom tumours were diagnosed in 210 individuals. RESULTS: Among patients tested for PPGL, those with tumours less often had a body mass index (BMI) above 30 kg/m2 (12 vs. 31%) and more often a BMI under 25 kg/m2 (56 vs. 32%) than those without tumours (P < 0.0001). Urinary outputs of catecholamines in patients with PPGL were negatively related to BMI (r = -0.175, P = 0.0133). Post-operative weight gain (P < 0.0001) after resection of PPGL was positively related to presurgical tumoural catecholamine output (r = 0.257, P = 0.0101). Higher BMI in men and women and percent body fat in women of the volunteer group were associated with lower plasma concentrations and urinary outputs of adrenaline and metanephrine, the former indicating obesity-related reduced adrenaline secretion and the latter obesity-related reduced adrenomedullary adrenaline stores. Daytime activity was associated with substantial increases in urinary adrenaline and noradrenaline excretion, with blunted responses in obese subjects. CONCLUSIONS: The findings in patients with PPGL support an influence of high circulating catecholamines on body weight. Additional associations of adrenomedullary dysfunction with obesity raise the possibility of a permissive influence of the adrenal medulla on the regulation of body weight.


Assuntos
Neoplasias das Glândulas Suprarrenais , Peso Corporal/fisiologia , Catecolaminas , Obesidade , Adolescente , Neoplasias das Glândulas Suprarrenais/complicações , Neoplasias das Glândulas Suprarrenais/epidemiologia , Medula Suprarrenal/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Catecolaminas/sangue , Catecolaminas/urina , Células Cromafins/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/epidemiologia , Feocromocitoma , Estudos Prospectivos , Adulto Jovem
11.
J Physiol Biochem ; 74(4): 667-677, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30367392

RESUMO

The adrenomedullary chromaffin cells' hormonal pathway has been related to the pathophysiology of diabetes mellitus. In mice, the deletion of insulin receptor substrate type 2 (Irs2) causes peripheral insulin resistance and reduction in ß-cell mass, leading to overt diabetes, with gender differences on adrenergic signaling. To further unravel the relevance of Irs2 on glycemic control, we analyzed in adult Irs2 deficient (Irs2-/-) mice, of both sexes but still normoglycemic, dopamine effects on insulin secretion and glycerol release, as well as their adrenal medulla by an immunohistochemical and morphologic approach. In isolated islets, 10 µM dopamine significantly inhibited insulin release in wild-type (WT) and female Irs2-/- mice; however, male Irs2-/- islets were insensitive to that catecholamine. Similarly, on isolated adipocytes, gender differences were observed between WT and Irs2-/- mice in basal and evoked glycerol release with crescent concentrations of dopamine. By immunohistochemistry, reactivity to tyrosine hydroxylase (TH) in female mice was significantly higher in the adrenal medulla of Irs2-/- compared to WT; although no differences for TH-immunopositivity were observed between the male groups of mice. However, compared to their corresponding WT animals, adrenomedullary chromaffin cells of Irs2-/- mice showed a significant decrease in the cellular and nuclear areas, and even in their percentage of apoptosis. Therefore, our observations suggest that, together with gender differences on dopamine responses in Irs2-/- mice, disturbances in adrenomedullary chromaffin cells could be related to deficiency of Irs2. Accordingly, Irs2 could be necessary for adequate glucose homeostasis and maintenance of the population of the adrenomedullary chromaffin cells.


Assuntos
Medula Suprarrenal/metabolismo , Dopamina/metabolismo , Hiperinsulinismo/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Estado Pré-Diabético/metabolismo , Adipócitos Brancos/metabolismo , Adipócitos Brancos/patologia , Medula Suprarrenal/enzimologia , Medula Suprarrenal/patologia , Animais , Apoptose , Células Cultivadas , Células Cromafins/enzimologia , Células Cromafins/metabolismo , Células Cromafins/patologia , Feminino , Hiperinsulinismo/sangue , Hiperinsulinismo/patologia , Técnicas In Vitro , Proteínas Substratos do Receptor de Insulina/genética , Ilhotas Pancreáticas/patologia , Lipólise , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Estado Pré-Diabético/sangue , Estado Pré-Diabético/patologia , Caracteres Sexuais , Técnicas de Cultura de Tecidos , Tirosina 3-Mono-Oxigenase/metabolismo
12.
J Neurochem ; 147(4): 454-476, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30182387

RESUMO

As the peripheral sympathoadrenal axis is tightly controlled by the cortex via hypothalamus and brain stem, the central pathological features of Hunting's disease, (HD) that is, deposition of mutated huntingtin and synaptic dysfunctions, could also be expressed in adrenal chromaffin cells. To test this hypothesis we here present a thorough investigation on the pathological and functional changes undergone by chromaffin cells (CCs) from 2-month (2 m) to 7-month (7 m) aged wild-type (WT) and R6/1 mouse model of Huntington's disease (HD), stimulated with acetylcholine (ACh) or high [K+ ] (K+ ). In order to do this, we used different techniques such as inmunohistochemistry, patch-clamp, and amperometric recording. With respect to WT cells, some of the changes next summarized were already observed in HD mice at a pre-disease stage (2 m); however, they were more pronounced at 7 m when motor deficits were clearly established, as follows: (i) huntingtin over-expression as nuclear aggregates in CCs; (ii) smaller CC size with decreased dopamine ß-hydroxylase expression, indicating lesser number of chromaffin secretory vesicles; (iii) reduced adrenal tissue catecholamine content; (iv) reduced Na+ currents with (v) membrane hyperpolarization and reduced ACh-evoked action potentials; (v) reduced [Ca2+ ]c transients with faster Ca2+ clearance; (vi) diminished quantal secretion with smaller vesicle quantal size; (vii) faster kinetics of the exocytotic fusion pore, pore expansion, and closure. On the basis of these data, the hypothesis is here raised in the sense that nuclear deposition of mutated huntingtin in adrenal CCs of R6/1 mice could be primarily responsible for poorer Na+ channel expression and function, giving rise to profound depression of cell excitability, altered Ca2+ handling and exocytosis. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14201.


Assuntos
Células Cromafins/metabolismo , Células Cromafins/patologia , Exocitose , Proteína Huntingtina/biossíntese , Proteína Huntingtina/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Medula Suprarrenal/metabolismo , Medula Suprarrenal/patologia , Animais , Catecolaminas/metabolismo , Humanos , Doença de Huntington/psicologia , Cinética , Masculino , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/fisiopatologia , Mutação/genética , Desempenho Psicomotor , Canais de Sódio/biossíntese , Vesículas Sinápticas/patologia
14.
Cell Tissue Res ; 372(2): 339-346, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29450724

RESUMO

Pheochromocytomas and paragangliomas (PCC/PGLs) are rare neuroendocrine tumors that are unusually diverse in metabolic profiles, in classes of molecular alterations and across a large number of altered genes. The Cancer Genome Atlas (TCGA) comprehensively profiled the molecular landscape of PCC/PGLs and identified novel genomic alterations and a new molecular classification of PCC/PGLs. In this review, we discuss the significant clinico-molecular findings of this integrated profiling study. We then review the molecular data of the TCGA cohort centering around known markers of sympathoadrenal cell lineage to better understand chromaffin cell biology. This analysis adds a new layer, that of chromaffin cell type, onto the published molecular classifications and in doing so provides inferences about underlying chromaffin cell biology and diversity.


Assuntos
Células Cromafins/patologia , Genoma Humano , Neoplasias/genética , Neoplasias/patologia , Estudos de Coortes , Humanos , Modelos Biológicos
15.
Cell Tissue Res ; 372(2): 211-221, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29445860

RESUMO

Neuroblastoma is an embryonal malignancy that affects normal development of the adrenal medulla and paravertebral sympathetic ganglia in early childhood. Extensive studies have revealed the molecular characteristics of human neuroblastomas, including abnormalities at genome, epigenome and transcriptome levels. However, neuroblastoma initiation mechanisms and even its origin are long-standing mysteries. In this review article, we summarize the current knowledge about normal development of putative neuroblastoma sources, namely sympathoadrenal lineage of neural crest cells and Schwann cell precursors that were recently identified as the source of adrenal chromaffin cells. A plausible origin of enigmatic stage 4S neuroblastoma is also discussed. With regard to the initiation mechanisms, we review genetic abnormalities in neuroblastomas and their possible association to initiation mechanisms. We also summarize evidences of neuroblastoma initiation observed in genetically engineered animal models, in which epigenetic alterations were involved, including transcriptomic upregulation by N-Myc and downregulation by polycomb repressive complex 2. Finally, several in vitro experimental methods are proposed that hopefully will accelerate our comprehension of neuroblastoma initiation. Thus, this review summarizes the state-of-the-art knowledge about the mechanisms of neuroblastoma initiation, which is critical for developing new strategies to cure children with neuroblastoma.


Assuntos
Carcinogênese/patologia , Neuroblastoma/patologia , Medula Suprarrenal/patologia , Animais , Células Cromafins/patologia , Modelos Animais de Doenças , Epigênese Genética , Humanos , Neuroblastoma/genética
16.
Eur J Pharmacol ; 815: 416-426, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28970016

RESUMO

Comorbidity of diabetes and hypertension is frequent. Here, we have performed a comparative study in three animal models namely, normotensive Wistar Kyoto (WKY) rats, streptozotocin-induced diabetic rats (STZ), and spontaneously hypertensive rats (SHR). With respect WKY rats, we have found the following alterations in adrenal chromaffin cells from STZ and SHR rats: (1) diminished Ca2+ currents; (2) augmented [Ca2+]c elevations and catecholamine release in cells stimulated with angiotensin II or high K+; (3) unchanged expression of angiotensin II receptors AT1 and AT2; (4) higher density of secretory vesicles at subplasmalemmal sites; (5) mitochondria with lower cristae density that were partially depolarized; and (6) lower whole cell ATP content. These alterations may have their origin in (i) an augmented capacity of the endoplasmic reticulum [Ca2+] store likely due to (ii) impaired mitochondrial Ca2+ uptake; (iii) augmented high-[Ca2+]c microdomains at subplasmalemmal sites secondary to augmented calcium-induce calcium release and to inositol tris-phosphate receptor mediated enhanced Ca2+ mobilization from the endoplasmic reticulum; and (iv) augmented vesicle pool. These alterations seem to be common to the two models of human hypertension here explored, STZ diabetic rats and SHR hypertensive rats.


Assuntos
Sinalização do Cálcio , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Células Cromafins/patologia , Diabetes Mellitus Experimental/patologia , Mitocôndrias/patologia , Animais , Contagem de Células , Masculino , Ratos , Ratos Endogâmicos SHR , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo
18.
Eur J Pharmacol ; 803: 65-77, 2017 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-28322841

RESUMO

It is known that chronic ethanol (EtOH) consumption leads to hypertension development and has been associated with deleterious effects on the cardiovascular system. Whether this condition alters calcium (Ca2+) signaling and exocytosis in adrenal chromaffin cells (CCs) as the case is for genetic hypertension, is unknown. We explored this question in four randomized experimental groups, male Wistar Kyoto (WKY/EtOH) and Spontaneously Hypertensive (SHR/EtOH) rats were subjected to the intake of increasing EtOH concentrations (5-20%, for 30 days) and their respective controls (WKY/Control and SHR/Control) received water. WKY/EtOH developed hypertension and cardiac hypertrophy; blood aldehyde dehydrogenase (ALDH) and H2O2 were also augmented. In comparison with WKY/Control, CCs from WKY/EtOH had the following features: (i) depolarization and higher frequency of spontaneous action potentials; (ii) decreased Ca2+ currents with slower inactivation; (iii) decreased K+ currents; (iv) augmented K+-elicited cytosolic Ca2+ transients ([Ca2+]c); (v) enhanced K+-elicited catecholamine release. These cardiovascular, blood and CCs changes were qualitatively similar to those undergone by SHR/Control and SHR/EtOH. The results suggest that the hypertension elicited by chronic EtOH has pathogenic features common to genetic hypertension namely, augmented [Ca2+]c transients and catecholamine release from their CCs.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Catecolaminas/metabolismo , Células Cromafins/efeitos dos fármacos , Células Cromafins/metabolismo , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertensão/patologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Células Cromafins/patologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Etanol/farmacologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Potássio/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Fatores de Tempo
19.
Oncotarget ; 6(32): 32955-65, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26460615

RESUMO

Metastatic pheochromocytomas and paragangliomas (PPGL) are malignant neuroendocrine tumors frequently associated with germline mutations in the SDHB gene. SDHB-mutated PPGL display a hypermethylator phenotype associated with hallmarks of epithelial-to-mesenchymal transition (EMT). In the present study, we report the characterization of a unique model of Sdhb knockout in mouse chromaffin cells. Sdhb deficient cells exhibit a metastatic phenotype as highlighted by increased individual cell migration (characterized by faster motility and increased persistence) as well as high invasive and adhesion abilities. This phenotype is associated with the modulation of Twist1, Twist2, Tcf3, Snai1, N-cadherin or Krt19 expression, reflecting an EMT-like reprogramming of cells. Krt19 is epigenetically silenced in Sdhb-deficient cells and re-expressed after treatment by the demethylating agent decitabine. Krt19 rescue by lentiviral transduction in Sdhb-deficient cells and Krt19 inhibition by RNA interference in wild-type cells were performed. Both studies revealed the involvement of KRT19 in the invasive phenotype by modulating collective and individual migration and cell/extra-cellular matrix adhesion properties. These findings underline the role of hypermethylation and EMT in the in vitro acquisition of metastatic properties, following SDHB loss of function.


Assuntos
Succinato Desidrogenase/deficiência , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Células Cromafins/metabolismo , Células Cromafins/patologia , Transição Epitelial-Mesenquimal , Humanos , Camundongos , Camundongos Knockout , Invasividade Neoplásica , Metástase Neoplásica , Paraganglioma/genética , Paraganglioma/metabolismo , Feocromocitoma/genética , Feocromocitoma/metabolismo , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Transcriptoma
20.
BMJ Case Rep ; 20152015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26243750

RESUMO

Pheochromocytomas are rare tumours originating in chromaffin cells, representing 0.1-1% of all secondary hypertension (HT) cases. The majority are benign and unilateral, characterised by the production of catecholamines and other neuropeptides. Mainly located in the adrenal gland, they are more frequent between the 3rd and 5th decades of life; however, 10-25% can be associated with genetic familial syndromes (multiple endocrine neoplasia type 2 (MEN 2), type 1 neurofibromatosis and Von-Hippel-Landau disease in younger ages. The authors present a rare case of secondary HT due to a pheochromocytoma in a 15-year-old patient, whose metanephrine assay confirmed the diagnosis, and abdominal ultrasound and CT localised the tumour in the adrenal gland. HT was controlled with α and ß blockers, with posterior retroperitoneal laparoscopic surgical intervention and subsequent resolution of HT. Age and concomitant hyperparathyroidism compelled genetic testing for the exclusion of MEN 2, which was negative.


Assuntos
Neoplasias das Glândulas Suprarrenais/diagnóstico , Glândulas Suprarrenais/patologia , Pressão Sanguínea , Células Cromafins/patologia , Hipertensão/diagnóstico , Metanefrina/metabolismo , Feocromocitoma/diagnóstico , Adolescente , Neoplasias das Glândulas Suprarrenais/complicações , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/cirurgia , Glândulas Suprarrenais/cirurgia , Antagonistas Adrenérgicos/uso terapêutico , Feminino , Humanos , Hipertensão/etiologia , Hipertensão/metabolismo , Hipertensão/cirurgia , Neoplasia Endócrina Múltipla Tipo 2a , Feocromocitoma/complicações , Feocromocitoma/metabolismo , Feocromocitoma/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...