Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Microbiol Spectr ; 11(6): e0282523, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37823634

RESUMO

IMPORTANCE: We screened 66 bacteriocins to see if they exhibited anti-gonococcal activity. We found 12 bacteriocins with anti-gonococcal effects, and 4 bacteriocins showed higher anti-gonococcal activity. Three bacteriocins, lacticin Z, lacticin Q, and Garvicin KS (ABC), showed in vitro anti-gonococcal activity but no in vivo inhibitory effects against the Neisseria gonorrhoeae (WHO-P) isolate. On the other hand, NAI-107 showed in vivo anti-gonococcal activity. The findings suggest that NAI-107 is a promising alternative to treat gonorrhea infections.


Assuntos
Bacteriocinas , Gonorreia , Humanos , Neisseria gonorrhoeae , Bacteriocinas/farmacologia , Gonorreia/tratamento farmacológico , Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana
2.
Cancer Discov ; 12(2): 522-541, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34615655

RESUMO

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive leukemia of plasmacytoid dendritic cells (pDC). BPDCN occurs at least three times more frequently in men than in women, but the reasons for this sex bias are unknown. Here, studying genomics of primary BPDCN and modeling disease-associated mutations, we link acquired alterations in RNA splicing to abnormal pDC development and inflammatory response through Toll-like receptors. Loss-of-function mutations in ZRSR2, an X chromosome gene encoding a splicing factor, are enriched in BPDCN, and nearly all mutations occur in males. ZRSR2 mutation impairs pDC activation and apoptosis after inflammatory stimuli, associated with intron retention and inability to upregulate the transcription factor IRF7. In vivo, BPDCN-associated mutations promote pDC expansion and signatures of decreased activation. These data support a model in which male-biased mutations in hematopoietic progenitors alter pDC function and confer protection from apoptosis, which may impair immunity and predispose to leukemic transformation. SIGNIFICANCE: Sex bias in cancer is well recognized, but the underlying mechanisms are incompletely defined. We connect X chromosome mutations in ZRSR2 to an extremely male-predominant leukemia. Aberrant RNA splicing induced by ZRSR2 mutation impairs dendritic cell inflammatory signaling, interferon production, and apoptosis, revealing a sex- and lineage-related tumor suppressor pathway.This article is highlighted in the In This Issue feature, p. 275.


Assuntos
Células Dendríticas/metabolismo , Transtornos Mieloproliferativos/genética , Ribonucleoproteínas/genética , Apoptose , Feminino , Identidade de Gênero , Humanos , Masculino , Mutação
3.
Sci Rep ; 11(1): 6509, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33753765

RESUMO

Transanal endoscopic microsurgery (TEM) is widely used for the excision of rectal adenomas and early rectal adenocarcinoma. Few recommendations currently exist for surveillance of lesions excised by TEM. The purpose of this study was to review the surveillance practices and the patterns of recurrence among TEM resected lesions at a tertiary care hospital. A retrospective chart review was performed on all patients who underwent TEM for rectal adenoma or adenocarcinoma before June 2017. In our study population of 114 patients, the final pathology included 78 (68%) adenomas and 36 (32%) adenocarcinomas. Of the adenocarcinomas 23, 9, and 4 were T1, T2, T3 lesions, respectively. Of those, 25 patients opted for surveillance instead of further treatment. The most commonly recommended endoscopic surveillance strategy by our group for both adenomas and adenocarcinomas excised by TEM was flexible sigmoidoscopy every 6 months for 2 years. Recurrences occurred in 4/78 (5.1%) adenoma patients, all found within 16.9 months of surgery, and in 4/25 (16%) adenocarcinoma patients, found between 7.4 and 38.5 months post-surgery. Our data highlights the fact that the timing of recurrences post TEM surgery is variable. Further studies looking at recurrence patterns are needed in order to create comprehensive guidelines for surveillance of these patients.


Assuntos
Adenocarcinoma/cirurgia , Recidiva Local de Neoplasia/epidemiologia , Complicações Pós-Operatórias/epidemiologia , Neoplasias Retais/cirurgia , Sigmoidoscopia/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Complicações Pós-Operatórias/diagnóstico , Sigmoidoscopia/efeitos adversos , Sigmoidoscopia/normas , Centros de Atenção Terciária/estatística & dados numéricos
4.
Methods Mol Biol ; 2235: 169-180, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33576977

RESUMO

Renal pericytes have a critical importance for angiogenesis and vascular remodeling, medullary blood flow regulation, and development of fibrosis. An emerging role for kidney pericytes is their ability to induce renin expression and synthesis. Here, we present methods for purification of human renal pericytes, their primary culture, and differentiation into renin-producing cells. Possible applications of these protocols include investigations into (1) renin cell recruitment mechanisms, (2) modulation of renin expression/secretion by small molecules, and (3) renin expression/secretion in nonrenal pericytes. A potential therapeutic application of this work is the identification of new players regulating the renin-angiotensin system.


Assuntos
Pericitos/metabolismo , Cultura Primária de Células/métodos , Sistema Renina-Angiotensina/fisiologia , Angiotensinas/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Humanos , Rim/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos
5.
6.
Nutrients ; 11(2)2019 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-30744113

RESUMO

Mechanisms to explain post-prandial increases in circulating glucocorticoids are not well understood and may involve increased adrenal secretion and/or altered steroid metabolism. We have compared salivary levels of cortisol and cortisone levels in healthy male and female volunteers fed either a low or cholesterol-rich midday meal. Urinary levels of steroids, bile acids and markers of lipid peroxidation were also measured. Males and females showed expected circadian changes in salivary steroids and postprandial peaks within 1h of feeding. After a high-cholesterol meal, postprandial cortisol increases were higher in males whereas post-prandial cortisone levels were higher in females. Urinary cortisol but not cortisone levels were higher on the day when males and females ate a high-cholesterol meal. Urinary bile acid excretion and anti-oxidant markers of lipid peroxidation, thiobarbituric acid reactive substances (TBARS), and total phenol content were not affected by dietary cholesterol but tended to be higher in males. Cross-tabulation of correlation coefficients indicated positive associations between urinary markers of peroxidation, bile acids, and cortisol:cortisone ratios. We conclude that dietary cholesterol (a substrate for steroidogenesis) does not have an acute effect on adrenal glucocorticoid synthesis and that gender but not a high-cholesterol meal may influence the interconversion of cortisol and cortisone. Longer term studies of the effects of dietary cholesterol are needed to analyze the associations between bile acids, steroid metabolism, and secretion and lipid peroxidation.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colesterol na Dieta/metabolismo , Glucocorticoides/análise , Período Pós-Prandial/fisiologia , Saliva/química , Adulto , Ácidos e Sais Biliares/urina , Colesterol na Dieta/administração & dosagem , Estudos Cross-Over , Dieta/estatística & dados numéricos , Feminino , Humanos , Masculino , Adulto Jovem
7.
Nat Commun ; 9(1): 4525, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30375380

RESUMO

Malaria reduces host fitness and survival by pathogen-mediated damage and inflammation. Disease tolerance mechanisms counter these negative effects without decreasing pathogen load. Here, we demonstrate that in four different mouse models of malaria, adrenal hormones confer disease tolerance and protect against early death, independently of parasitemia. Surprisingly, adrenalectomy differentially affects malaria-induced inflammation by increasing circulating cytokines and inflammation in the brain but not in the liver or lung. Furthermore, without affecting the transcription of hepatic gluconeogenic enzymes, adrenalectomy causes exhaustion of hepatic glycogen and insulin-independent lethal hypoglycemia upon infection. This hypoglycemia is not prevented by glucose administration or TNF-α neutralization. In contrast, treatment with a synthetic glucocorticoid (dexamethasone) prevents the hypoglycemia, lowers cerebral cytokine expression and increases survival rates. Overall, we conclude that in malaria, adrenal hormones do not protect against lung and liver inflammation. Instead, they prevent excessive systemic and brain inflammation and severe hypoglycemia, thereby contributing to tolerance.


Assuntos
Glândulas Suprarrenais/metabolismo , Encéfalo/imunologia , Citocinas/imunologia , Hormônios/imunologia , Hipoglicemia/imunologia , Fígado/imunologia , Pulmão/imunologia , Malária/imunologia , Glândulas Suprarrenais/imunologia , Adrenalectomia , Animais , Glicemia/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Corticosterona/imunologia , Corticosterona/metabolismo , Citocinas/efeitos dos fármacos , Dexametasona/farmacologia , Modelos Animais de Doenças , Epinefrina/imunologia , Epinefrina/metabolismo , Glucocorticoides/imunologia , Glucocorticoides/farmacologia , Glicogênio/metabolismo , Hidrocortisona/imunologia , Hidrocortisona/metabolismo , Inflamação , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Camundongos , Mineralocorticoides/imunologia , Mineralocorticoides/metabolismo , Norepinefrina/imunologia , Norepinefrina/metabolismo , Plasmodium berghei , Plasmodium chabaudi , Taxa de Sobrevida
8.
Front Physiol ; 9: 848, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30038578

RESUMO

Salt-sensitive hypertension is common in glucocorticoid excess. Glucocorticoid resistance also presents with hypercortisolemia and hypertension but the relationship between salt intake and blood pressure (BP) is not well defined. GRßgeo/+ mice have global glucocorticoid receptor (GR) haploinsufficiency and increased BP. Here we examined the effect of high salt diet on BP, salt excretion and renal blood flow in GRßgeo/+mice. Basal BP was ∼10 mmHg higher in male GRßgeo/+ mice than in GR+/+ littermates. This modest increase was amplified by ∼10 mmHg following a high-salt diet in GRßgeo/+ mice. High salt reduced urinary aldosterone excretion but increased renal mineralocorticoid receptor expression in both genotypes. Corticosterone, and to a lesser extent deoxycorticosterone, excretion was increased in GRßgeo/+ mice following a high-salt challenge, consistent with enhanced 24 h production. GR+/+ mice increased fractional sodium excretion and reduced renal vascular resistance during the high salt challenge, retaining neutral sodium balance. In contrast, sodium excretion and renal vascular resistance did not adapt to high salt in GRßgeo/+ mice, resulting in transient sodium retention and sustained hypertension. With high-salt diet, Slc12a3 and Scnn1a mRNAs were higher in GRßgeo/+ than controls, and this was reflected in an exaggerated natriuretic response to thiazide and benzamil, inhibitors of NCC and ENaC, respectively. Reduction in GR expression causes salt-sensitivity and an adaptive failure of the renal vasculature and tubule, most likely reflecting sustained mineralocorticoid receptor activation. This provides a mechanistic basis to understand the hypertension associated with loss-of-function polymorphisms in GR in the context of habitually high salt intake.

10.
Psychoneuroendocrinology ; 89: 13-22, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29306773

RESUMO

Chronic exposure to stress during midlife associates with subsequent age-related cognitive decline and may increase the vulnerability to develop psychiatric conditions. Increased hypothalamic-pituitary-adrenal (HPA) axis activity has been implicated in pathogenesis though any causative role for glucocorticoids is unestablished. This study investigated the contribution of local glucocorticoid regeneration by the intracellular enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), in persisting midlife stress-induced behavioral effects in mice. Middle-aged (10 months old) 11ß-HSD1-deficient mice and wild-type congenic controls were randomly assigned to 28 days of chronic unpredictable stress or left undisturbed (non-stressed). All mice underwent behavioral testing at the end of the stress/non-stress period and again 6-7 months later. Chronic stress impaired spatial memory in middle-aged wild-type mice. The effects, involving a wide spectrum of behavioral modalities, persisted for 6-7 months after cessation of stress into early senescence. Enduring effects after midlife stress included impaired spatial memory, enhanced contextual fear memory, impaired fear extinction, heightened anxiety, depressive-like behavior, as well as reduced hippocampal glucocorticoid receptor mRNA expression. In contrast, 11ß-HSD1 deficient mice resisted both immediate and enduring effects of chronic stress, despite similar stress-induced increases in systemic glucocorticoid activity during midlife stress. In conclusion, chronic stress in midlife exerts persisting effects leading to cognitive and affective dysfunction in old age via mechanisms that depend, at least in part, on brain glucocorticoids generated locally by 11ß-HSD1. This finding supports selective 11ß-HSD1 inhibition as a novel therapeutic target to ameliorate the long-term consequences of stress-related psychiatric disorders in midlife.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Afeto/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Corticosterona/metabolismo , Medo/fisiologia , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Memória/fisiologia , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Memória Espacial/fisiologia
11.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-29123029

RESUMO

Normal renin synthesis and secretion is important for the maintenance of juxtaglomerular apparatus architecture. Mice lacking a functional Ren1d gene are devoid of renal juxtaglomerular cell granules and exhibit an altered macula densa morphology. Due to the species-specificity of renin activity, transgenic mice are ideal models for experimentally investigating and manipulating expression patterns of the human renin gene in a native cellular environment without confounding renin-angiotensin system interactions. A 55-kb transgene encompassing the human renin locus was crossed onto the mouse Ren1d-null background, restoring granulation in juxtaglomerular cells. Correct processing of human renin in dense core granules was confirmed by immunogold labeling. After stimulation of the renin-angiotensin system, juxtaglomerular cells contained rhomboid protogranules with paracrystalline contents, dilated rough endoplasmic reticulum, and electron-lucent granular structures. However, complementation of Ren1d-/- mice with human renin was unable to rescue the abnormality seen in macula densa structure. The juxtaglomerular apparatus was still able to respond to tubuloglomerular feedback in isolated perfused juxtaglomerular apparatus preparations, although minor differences in glomerular tuft contractility and macula densa cell calcium handling were observed. This study reveals that the human renin protein is able to complement the mouse Ren1d-/- non-granulated defect and suggests that granulopoiesis requires a structural motif that is conserved between the mouse Ren1d and human renin proteins. It also suggests that the altered macula densa phenotype is related to the activity of the renin-1d enzyme in a local juxtaglomerular renin-angiotensin system.


Assuntos
Teste de Complementação Genética , Sistema Justaglomerular/enzimologia , Renina/biossíntese , Transgenes , Animais , Humanos , Sistema Justaglomerular/patologia , Camundongos , Camundongos Knockout , Renina/genética
12.
Sci Rep ; 7(1): 10633, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28878267

RESUMO

Carbonyl Reductase 1 (CBR1) is a ubiquitously expressed cytosolic enzyme important in exogenous drug metabolism but the physiological function of which is unknown. Here, we describe a role for CBR1 in metabolism of glucocorticoids. CBR1 catalyzes the NADPH- dependent production of 20ß-dihydrocortisol (20ß-DHF) from cortisol. CBR1 provides the major route of cortisol metabolism in horses and is up-regulated in adipose tissue in obesity in horses, humans and mice. We demonstrate that 20ß-DHF is a weak endogenous agonist of the human glucocorticoid receptor (GR). Pharmacological inhibition of CBR1 in diet-induced obesity in mice results in more marked glucose intolerance with evidence for enhanced hepatic GR signaling. These findings suggest that CBR1 generating 20ß-dihydrocortisol is a novel pathway modulating GR activation and providing enzymatic protection against excessive GR activation in obesity.


Assuntos
Carbonil Redutase (NADPH)/metabolismo , Metabolismo Energético , Glucocorticoides/metabolismo , Obesidade/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Carbonil Redutase (NADPH)/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Estudos de Associação Genética , Variação Genética , Glucocorticoides/química , Glucocorticoides/urina , Cavalos , Humanos , Hidrocortisona/metabolismo , Hidroxicorticosteroides/metabolismo , Hidroxicorticosteroides/urina , Fígado/metabolismo , Masculino , Camundongos , Modelos Moleculares , Conformação Molecular , Obesidade/genética , Fenótipo , Ligação Proteica , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/química , Relação Estrutura-Atividade
13.
Endocr Connect ; 6(7): 446-457, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28720595

RESUMO

Chronic ACTH exposure is associated with adrenal hypertrophy and steroidogenesis. The underlying molecular processes in mice have been analysed by microarray, histological and immunohistochemical techniques. Synacthen infused for 2 weeks markedly increased adrenal mass and plasma corticosterone levels. Microarray analysis found greater than 2-fold changes in expression of 928 genes (P < 0.001; 397 up, 531 down). These clustered in pathways involved in signalling, sterol/lipid metabolism, cell proliferation/hypertrophy and apoptosis. Signalling genes included some implicated in adrenal adenomas but also upregulated genes associated with cyclic AMP and downregulated genes associated with aldosterone synthesis. Sterol metabolism genes were those promoting cholesterol supply (Scarb1, Sqle, Apoa1) and disposal (Cyp27a1, Cyp7b1). Oil red O staining showed lipid depletion consistent with reduced expression of genes involved in lipid synthesis. Genes involved in steroidogenesis (Star, Cyp11a1, Cyp11b1) were modestly affected (P < 0.05; <1.3-fold). Increased Ki67, Ccna2, Ccnb2 and Tk1 expression complemented immunohistochemical evidence of a 3-fold change in cell proliferation. Growth arrest genes, Cdkn1a and Cdkn1c, which are known to be active in hypertrophied cells, were increased >4-fold and cross-sectional area of fasciculata cells was 2-fold greater. In contrast, genes associated with apoptosis (eg Casp12, Clu,) were downregulated and apoptotic cells (Tunel staining) were fewer (P < 0.001) and more widely distributed throughout the cortex. In summary, long-term steroidogenesis with ACTH excess is sustained by genes controlling cholesterol supply and adrenal mass. ACTH effects on adrenal morphology and genes controlling cell hypertrophy, proliferation and apoptosis suggest the involvement of different cell types and separate molecular pathways.

14.
J Endocrinol ; 233(3): 315-327, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28522730

RESUMO

We have previously demonstrated that neutrophil recruitment to the heart following myocardial infarction (MI) is enhanced in mice lacking 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) that regenerates active glucocorticoid within cells from intrinsically inert metabolites. The present study aimed to identify the mechanism of regulation. In a mouse model of MI, neutrophil mobilization to blood and recruitment to the heart were higher in 11ß-HSD1-deficient (Hsd11b1-/- ) relative to wild-type (WT) mice, despite similar initial injury and circulating glucocorticoid. In bone marrow chimeric mice, neutrophil mobilization was increased when 11ß-HSD1 was absent from host cells, but not when absent from donor bone marrow-derived cells. Consistent with a role for 11ß-HSD1 in 'host' myocardium, gene expression of a subset of neutrophil chemoattractants, including the chemokines Cxcl2 and Cxcl5, was selectively increased in the myocardium of Hsd11b1-/- mice relative to WT. SM22α-Cre directed disruption of Hsd11b1 in smooth muscle and cardiomyocytes had no effect on neutrophil recruitment. Expression of Cxcl2 and Cxcl5 was elevated in fibroblast fractions isolated from hearts of Hsd11b1-/- mice post MI and provision of either corticosterone or of the 11ß-HSD1 substrate, 11-dehydrocorticosterone, to cultured murine cardiac fibroblasts suppressed IL-1α-induced expression of Cxcl2 and Cxcl5 These data identify suppression of CXCL2 and CXCL5 chemoattractant expression by 11ß-HSD1 as a novel mechanism with potential for regulation of neutrophil recruitment to the injured myocardium, and cardiac fibroblasts as a key site for intracellular glucocorticoid regeneration during acute inflammation following myocardial injury.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Quimiocina CXCL2/metabolismo , Quimiocina CXCL5/metabolismo , Fibroblastos/fisiologia , Neutrófilos/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Animais , Células da Medula Óssea , Células Cultivadas , Quimiocina CXCL5/genética , Corticosterona/análogos & derivados , Corticosterona/farmacologia , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Infarto do Miocárdio
15.
J Endocrinol ; 232(3): 437-450, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28057868

RESUMO

Corticosteroids directly affect the heart and vasculature and are implicated in the pathogenesis of heart failure. Attention is focussed upon the role of the mineralocorticoid receptor (MR) in mediating pro-fibrotic and other adverse effects of corticosteroids upon the heart. In contrast, the role of the glucocorticoid receptor (GR) in the heart and vasculature is less well understood. We addressed this in mice with cardiomyocyte and vascular smooth muscle deletion of GR (SMGRKO mice). Survival of SMGRKO mice to weaning was reduced compared with that of littermate controls. Doppler measurements of blood flow across the mitral valve showed an elongated isovolumetric contraction time in surviving adult SMGRKO mice, indicating impairment of the initial left ventricular contractile phase. Although heart weight was elevated in both genders, only male SMGRKO mice showed evidence of pathological cardiomyocyte hypertrophy, associated with increased myosin heavy chain-ß expression. Left ventricular fibrosis, evident in both genders, was associated with elevated levels of mRNA encoding MR as well as proteins involved in cardiac remodelling and fibrosis. However, MR antagonism with spironolactone from birth only modestly attenuated the increase in pro-fibrotic gene expression in SMGRKO mice, suggesting that elevated MR signalling is not the primary driver of cardiac fibrosis in SMGRKO mice, and cardiac fibrosis can be dissociated from MR activation. Thus, GR contributes to systolic function and restrains normal cardiac growth, the latter through gender-specific mechanisms. Our findings suggest the GR:MR balance is critical in corticosteroid signalling in specific cardiac cell types.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Corticosterona/sangue , Feminino , Fibrose/metabolismo , Fibrose/patologia , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Contração Miocárdica/genética , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/metabolismo , Receptores de Glucocorticoides/genética , Fatores Sexuais , Espironolactona/farmacologia , Função Ventricular Esquerda/genética
16.
Kidney Int ; 90(6): 1251-1261, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27678158

RESUMO

Pericytes, perivascular cells embedded in the microvascular wall, are crucial for vascular homeostasis. These cells also play diverse roles in tissue development and regeneration as multi-lineage progenitors, immunomodulatory cells and as sources of trophic factors. Here, we establish that pericytes are renin producing cells in the human kidney. Renin was localized by immunohistochemistry in CD146 and NG2 expressing pericytes, surrounding juxtaglomerular and afferent arterioles. Similar to pericytes from other organs, CD146+CD34-CD45-CD56- renal fetal pericytes, sorted by flow cytometry, exhibited tri-lineage mesodermal differentiation potential in vitro. Additionally, renin expression was triggered in cultured kidney pericytes by cyclic AMP as confirmed by immuno-electron microscopy, and secretion of enzymatically functional renin, capable of generating angiotensin I. Pericytes derived from second trimester human placenta also expressed renin in an inducible fashion although the renin activity was much lower than in renal pericytes. Thus, our results confirm and extend the recently discovered developmental plasticity of microvascular pericytes, and may open new perspectives to the therapeutic regulation of the renin-angiotensin system.


Assuntos
Rim/ultraestrutura , Pericitos/metabolismo , Renina/metabolismo , Humanos , Rim/embriologia , Células-Tronco Mesenquimais , Cultura Primária de Células
17.
Nat Med ; 22(7): 771-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27270587

RESUMO

The discovery of genetic mechanisms for resistance to obesity and diabetes may illuminate new therapeutic strategies for the treatment of this global health challenge. We used the polygenic 'lean' mouse model, which has been selected for low adiposity over 60 generations, to identify mitochondrial thiosulfate sulfurtransferase (Tst; also known as rhodanese) as a candidate obesity-resistance gene with selectively increased expression in adipocytes. Elevated adipose Tst expression correlated with indices of metabolic health across diverse mouse strains. Transgenic overexpression of Tst in adipocytes protected mice from diet-induced obesity and insulin-resistant diabetes. Tst-deficient mice showed markedly exacerbated diabetes, whereas pharmacological activation of TST ameliorated diabetes in mice. Mechanistically, TST selectively augmented mitochondrial function combined with degradation of reactive oxygen species and sulfide. In humans, TST mRNA expression in adipose tissue correlated positively with insulin sensitivity in adipose tissue and negatively with fat mass. Thus, the genetic identification of Tst as a beneficial regulator of adipocyte mitochondrial function may have therapeutic significance for individuals with type 2 diabetes.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Resistência à Insulina/genética , Mitocôndrias/metabolismo , Obesidade/genética , Tiossulfato Sulfurtransferase/genética , Animais , Diferenciação Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Técnicas de Introdução de Genes , Técnica Clamp de Glucose , Teste de Tolerância a Glucose , Humanos , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Modelos Animais , Terapia de Alvo Molecular , Obesidade/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tiossulfato Sulfurtransferase/metabolismo
18.
Circulation ; 133(14): 1360-70, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26951843

RESUMO

BACKGROUND: The hypertensive syndrome of Apparent Mineralocorticoid Excess is caused by loss-of-function mutations in the gene encoding 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2), allowing inappropriate activation of the mineralocorticoid receptor by endogenous glucocorticoid. Hypertension is attributed to sodium retention in the distal nephron, but 11ßHSD2 is also expressed in the brain. However, the central contribution to Apparent Mineralocorticoid Excess and other hypertensive states is often overlooked and is unresolved. We therefore used a Cre-Lox strategy to generate 11ßHSD2 brain-specific knockout (Hsd11b2.BKO) mice, measuring blood pressure and salt appetite in adults. METHODS AND RESULTS: Basal blood pressure, electrolytes, and circulating corticosteroids were unaffected in Hsd11b2.BKO mice. When offered saline to drink, Hsd11b2.BKO mice consumed 3 times more sodium than controls and became hypertensive. Salt appetite was inhibited by spironolactone. Control mice fed the same daily sodium intake remained normotensive, showing the intrinsic salt resistance of the background strain. Dexamethasone suppressed endogenous glucocorticoid and abolished the salt-induced blood pressure differential between genotypes. Salt sensitivity in Hsd11b2.BKO mice was not caused by impaired renal sodium excretion or volume expansion; pressor responses to phenylephrine were enhanced and baroreflexes impaired in these animals. CONCLUSIONS: Reduced 11ßHSD2 activity in the brain does not intrinsically cause hypertension, but it promotes a hunger for salt and a transition from salt resistance to salt sensitivity. Our data suggest that 11ßHSD2-positive neurons integrate salt appetite and the blood pressure response to dietary sodium through a mineralocorticoid receptor-dependent pathway. Therefore, central mineralocorticoid receptor antagonism could increase compliance to low-sodium regimens and help blood pressure management in cardiovascular disease.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Fissura/fisiologia , Hipertensão/genética , Síndrome de Excesso Aparente de Minerolocorticoides/fisiopatologia , Proteínas do Tecido Nervoso/deficiência , Receptores de Mineralocorticoides/fisiologia , Cloreto de Sódio na Dieta/toxicidade , Núcleo Solitário/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Animais , Barorreflexo/efeitos dos fármacos , Corticosterona/sangue , Dexametasona/farmacologia , Comportamento de Ingestão de Líquido , Genes Sintéticos , Hipertensão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome de Excesso Aparente de Minerolocorticoides/tratamento farmacológico , Síndrome de Excesso Aparente de Minerolocorticoides/genética , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Néfrons/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Nestina/genética , Neurônios/fisiologia , Potássio/urina , RNA Mensageiro/biossíntese , Reflexo Anormal , Núcleo Solitário/fisiopatologia , Espironolactona/farmacologia
19.
Exp Physiol ; 100(11): 1362-78, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26337786

RESUMO

NEW FINDINGS: What is the central question of this study? Glucocorticoids act in the kidney to promote salt and water retention. Renal 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1), by increasing local concentrations of glucocorticoids, may exert an antinatriuretic effect. We hypothesized that global deletion of 11ßHSD1 in the mouse would give rise to a salt-wasting renal phenotype. What is the main finding and its importance? We subjected a mouse model of global 11ßHSD1 deletion to studies of water and electrolyte balance, renal clearance, urinary steroid excretion, renin-angiotensin system activation and renal sodium transporter expression. We found no significant effects on renal sodium or water excretion. Any effect of renal 11ßHSD1 on sodium homeostasis is subtle. Glucocorticoids act in the kidney to regulate glomerular haemodynamics and tubular sodium transport; the net effect favours sodium retention. 11ß-Hydroxysteroid dehydrogenase type 1 (11ßHSD1) is expressed in the renal tubules and the interstitial cells of the medulla, where it is likely to regenerate active glucocorticoids from inert 11-keto forms. The physiological function of renal 11ßHSD1 is largely unknown. We hypothesized that loss of renal 11ßHSD1 would result in salt wasting and tested this in a knockout mouse model in which 11ßHSD1 was deleted in all body tissues. In balance studies, 11ßHSD1 deletion had no effect on water, sodium or potassium metabolism; transition to a low-sodium diet did not reveal a natriuretic phenotype. Renal clearance studies demonstrated identical haemodynamic parameters (arterial blood pressure, renal blood flow and glomerular filtration rate) in knockout and wild-type mice, but revealed an augmented kaliuretic response to thiazides in 11ßHSD1 knockout animals. There was no effect on the natriuretic response to the amiloride analogue benzamil. Urinary excretion of deoxycorticosterone was higher in 11ßHSD1 knockout mice, and there was hypertrophy of cells in the zona fasciculata of the adrenal cortex. There was no difference in the activity of the renin-angiotensin and nitric oxide systems, no difference in renal histology and no difference in the abundance of key tubular transporter proteins. We conclude that any effect of 11ßHSD1 on renal sodium excretion is subtle.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Homeostase , Rim/fisiologia , Sódio/fisiologia , Animais , Glucocorticoides/fisiologia , Camundongos Knockout , Potássio/metabolismo , Sistema Renina-Angiotensina , Sódio/metabolismo , Equilíbrio Hidroeletrolítico
20.
Hypertension ; 66(3): 667-73, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077568

RESUMO

Obesity and hypertension are 2 major health issues of the 21st century. The syndrome of apparent mineralocorticoid excess is caused by deficiency of 11ß-hydroxysteroid dehydrogenase type 2 (Hsd11b2), which normally inactivates glucocorticoids, rendering the mineralocorticoid receptor aldosterone-specific. The metabolic consequences of Hsd11b2 knockout in the rat are investigated in parallel with electrolyte homeostasis. Hsd11b2 was knocked out, by pronuclear microinjection of targeted zinc-finger nuclease mRNAs, and 1 line was characterized for its response to renal and metabolic challenges. Plasma 11-dehydrocorticosterone was below detection thresholds, and Hsd11b2 protein was undetected by Western blot, indicating complete ablation. Homozygotes were 13% smaller than wild-type littermates, and were polydipsic and polyuric. Their kidneys, adrenals, and hearts were significantly enlarged, but mesenteric fat pads and liver were significantly smaller. On a 0.3% Na diet, mean arterial blood pressure was ≈65 mm Hg higher than controls but only 25 mm Hg higher on a 0.03% Na(+) diet. Urinary Na/K ratio of homozygotes was similar to controls on 0.3% Na(+) diet but urinary albumin and calcium were elevated. Corticosterone and aldosterone levels showed normal circadian variation on both a 0.3% and 0.03% Na(+) diet, but plasma renin was suppressed in homozygotes on both diets. Plasma glucose responses to an oral glucose challenge were reduced despite low circulating insulin, indicating much greater sensitivity to insulin in homozygotes. The rat model reveals mechanisms linking electrolyte homeostasis and metabolic control through the restriction of Hsd11b1 substrate availability.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Glândulas Suprarrenais/patologia , Aldosterona/sangue , Corticosterona/análogos & derivados , Rim/patologia , Miocárdio/patologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Glândulas Suprarrenais/metabolismo , Animais , Glicemia/metabolismo , Pressão Sanguínea/genética , Corticosterona/sangue , Rim/metabolismo , Miocárdio/metabolismo , Tamanho do Órgão/genética , Fenótipo , Ratos , Ratos Transgênicos , Renina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...