Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(2): e0296390, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38315701

RESUMO

Estradiol is an important regulator of bone accumulation and maintenance. Circulating estrogens are primarily produced by the gonads. Aromatase, the enzyme responsible for the conversion of androgens to estrogen, is expressed by bone marrow cells (BMCs) of both hematopoietic and nonhematopoietic origin. While the significance of gonad-derived estradiol to bone health has been investigated, there is limited understanding regarding the relative contribution of BMC derived estrogens to bone metabolism. To elucidate the role of BMC derived estrogens in male bone, irradiated wild-type C57BL/6J mice received bone marrow cells transplanted from either WT (WT(WT)) or aromatase-deficient (WT(ArKO)) mice. MicroCT was acquired on lumbar vertebra to assess bone quantity and quality. WT(ArKO) animals had greater trabecular bone volume (BV/TV p = 0.002), with a higher trabecular number (p = 0.008), connectivity density (p = 0.017), and bone mineral content (p = 0.004). In cortical bone, WT(ArKO) animals exhibited smaller cortical pores and lower cortical porosity (p = 0.02). Static histomorphometry revealed fewer osteoclasts per bone surface (Oc.S/BS%), osteoclasts on the erosion surface (ES(Oc+)/BS, p = 0.04) and low number of osteoclasts per bone perimeter (N.Oc/B.Pm, p = 0.01) in WT(ArKO). Osteoblast-associated parameters in WT(ArKO) were lower but not statistically different from WT(WT). Dynamic histomorphometry suggested similar bone formation indices' patterns with lower mean values in mineral apposition rate, label separation, and BFR/BS in WT(ArKO) animals. Ex vivo bone cell differentiation assays demonstrated relative decreased osteoblast differentiation and ability to form mineralized nodules. This study demonstrates a role of local 17ß-estradiol production by BMCs for regulating the quantity and quality of bone in male mice. Underlying in vivo cellular and molecular mechanisms require further study.


Assuntos
Transtornos 46, XX do Desenvolvimento Sexual , Aromatase , Transplante de Medula Óssea , Ginecomastia , Infertilidade Masculina , Erros Inatos do Metabolismo , Camundongos , Animais , Masculino , Aromatase/genética , Aromatase/metabolismo , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/metabolismo , Porosidade , Camundongos Endogâmicos C57BL , Estrogênios , Estradiol , Células da Medula Óssea/metabolismo , Coluna Vertebral/metabolismo , Camundongos Knockout
2.
Am J Physiol Endocrinol Metab ; 324(5): E461-E475, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37053049

RESUMO

Hypogonadism in males confers elevated cardiovascular disease (CVD) risk by unknown mechanisms. Recent radiological evidence suggests that low testosterone (T) is associated with mediobasal hypothalamic (MBH) gliosis, a central nervous system (CNS) cellular response linked to metabolic dysfunction. To address mechanisms linking CNS androgen action to CVD risk, we generated a hypogonadal, hyperlipidemic mouse model with orchiectomy (ORX) combined with hepatic PCSK9 overexpression. After 4 wk of high-fat, high-sucrose diet (HFHS) consumption, despite equal body weights and glucose tolerance, androgen-deficient ORX mice had a more atherogenic lipid profile and increased liver and leukocyte inflammatory signaling compared with sham-operated control mice. Along with these early CVD risk indicators, ORX markedly amplified HFHS-induced astrogliosis in the MBH. Transcriptomic analysis further revealed that ORX and high-fat diet feeding induced upregulation of inflammatory pathways and downregulation of metabolic pathways in hypothalamic astrocytes. To interrogate the role of sex steroid signaling in the CNS in cardiometabolic risk and MBH inflammation, central infusion of T and dihydrotestosterone (DHT) was performed on ORX mice. Central DHT prevented MBH astrogliosis and reduced the liver inflammatory signaling and monocytosis induced by HFHS and ORX; T had a partial protective effect. Finally, a cross-sectional study in 41 adult men demonstrated a positive correlation between radiological evidence of MBH gliosis and plasma lipids. These findings demonstrate that T deficiency in combination with a Western-style diet promotes hypothalamic gliosis concomitant with increased atherogenic risk factors and provide supportive evidence for regulation of lipid metabolism and cardiometabolic risk determinants by the CNS action of sex steroids.NEW & NOTEWORTHY This study provides evidence that hypothalamic gliosis is a key early event through which androgen deficiency in combination with a Western-style diet might lead to cardiometabolic dysregulation in males. Furthermore, this work provides the first evidence in humans of a positive association between hypothalamic gliosis and LDL-cholesterol, advancing our knowledge of CNS influences on CVD risk progression.


Assuntos
Androgênios , Doenças Cardiovasculares , Humanos , Camundongos , Masculino , Animais , Pró-Proteína Convertase 9 , Dieta Hiperlipídica/efeitos adversos , Gliose , Orquiectomia , Estudos Transversais , Fatores de Risco , Di-Hidrotestosterona
3.
J Mol Endocrinol ; 69(3): R95-R108, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35900842

RESUMO

Vitamin A (retinol) is an essential, fat-soluble vitamin that plays critical roles in embryonic development, vision, immunity, and reproduction. Severe vitamin A deficiency results in profound embryonic dysgenesis, blindness, and infertility. The roles of bioactive vitamin A metabolites in regulating cell proliferation, cellular differentiation, and immune cell function form the basis of their clinical use in the treatment of dermatologic conditions and hematologic malignancies. Increasingly, vitamin A also has been recognized to play important roles in cardiometabolic health, including the regulation of adipogenesis, energy partitioning, and lipoprotein metabolism. While these roles are strongly supported by animal and in vitro studies, they remain poorly understood in human physiology and disease. This review briefly introduces vitamin A biology and presents the key preclinical data that have generated interest in vitamin A as a mediator of cardiometabolic health. The review also summarizes clinical studies performed to date, highlighting the limitations of many of these studies and the ongoing controversies in the field. Finally, additional perspectives are suggested that may help position vitamin A metabolism within a broader biological context and thereby contribute to enhanced understanding of vitamin A's complex roles in clinical cardiometabolic disease.


Assuntos
Doenças Cardiovasculares , Deficiência de Vitamina A , Adipogenia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/etiologia , Feminino , Homeostase/fisiologia , Humanos , Gravidez , Vitamina A/metabolismo , Vitamina A/uso terapêutico , Deficiência de Vitamina A/tratamento farmacológico , Deficiência de Vitamina A/metabolismo
4.
Clin Transl Sci ; 15(6): 1460-1471, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35213790

RESUMO

The prevalence of obesity continues to rise, underscoring the need to better understand the pathways mediating adipose tissue (AT) expansion. All-trans-retinoic acid (atRA), a bioactive vitamin A metabolite, regulates adipogenesis and energy metabolism, and, in rodent studies, aberrant vitamin A metabolism appears a key facet of metabolic dysregulation. The relevance of these findings to human disease is unknown, as are the specific enzymes implicated in vitamin A metabolism within human AT. We hypothesized that in human AT, family 1A aldehyde dehydrogenase (ALDH1A) enzymes contribute to atRA biosynthesis in a depot-specific manner. To test this hypothesis, parallel samples of subcutaneous and omental AT from participants (n = 15) were collected during elective abdominal surgeries to quantify atRA biosynthesis and key atRA synthesizing enzymes. ALDH1A1 was the most abundant ALDH1A isoform in both AT depots with expression approximately twofold higher in omental than subcutaneous AT. ALDH1A2 was detected only in omental AT. Formation velocity of atRA was approximately threefold higher (p = 0.0001) in omental AT (9.8 [7.6, 11.2]) pmol/min/mg) than subcutaneous AT (3.2 [2.1, 4.0] pmol/min/mg) and correlated with ALDH1A2 expression in omental AT (ß-coefficient = 3.07, p = 0.0007) and with ALDH1A1 expression in subcutaneous AT (ß-coefficient = 0.13, p = 0.003). Despite a positive correlation between body mass index (BMI) and omental ALDH1A1 protein expression (Spearman r = 0.65, p = 0.01), BMI did not correlate with atRA formation. Our findings suggest that ALDH1A2 is the primary mediator of atRA formation in omental AT, whereas ALDH1A1 is the principal atRA-synthesizing enzyme in subcutaneous AT. These data highlight AT depot as a critical variable for defining the roles of retinoids in human AT biology.


Assuntos
Tecido Adiposo , Vitamina A , Tecido Adiposo/metabolismo , Humanos , Obesidade/metabolismo , Gordura Subcutânea , Tretinoína/metabolismo
5.
Clin Transl Sci ; 14(5): 1681-1688, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33742772

RESUMO

Alzheimer's disease (AD) is the most common form of dementia, and its prevalence is increasing rapidly. According to the Alzheimer's Association, over 5 million adults in the United States over the age of 65 years currently have AD, and this number is expected to exceed 13 million by 2050 in the absence of novel, preventative strategies. Epidemiologic studies have implicated the presence of type 2 diabetes mellitus (T2DM) specifically at midlife as a key modifiable risk factor for AD, and AD may increase risk of dysglycemia and T2DM. However, data have been inconsistent with regard to the magnitude of AD risk attributable to T2DM, and the pathways underlying this apparent relationship remain poorly understood. Elucidating the impact of T2DM on AD risk and progression requires greater attention to the myriad facets of T2DM pathophysiology, its comorbid conditions, and attendant treatment modalities, all of which may differentially impact the relationships among T2DM, cognitive decline, and AD. This mini-review will summarize the discrete facets of T2DM that may influence AD risk and highlight the importance of careful clinical phenotyping in both epidemiologic and interventional studies to better delineate the key pathways and mechanisms linking T2DM and AD.


Assuntos
Doença de Alzheimer/epidemiologia , Diabetes Mellitus Tipo 2/epidemiologia , Doença de Alzheimer/sangue , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Glicemia/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Progressão da Doença , Humanos , Prevalência , Fatores de Risco
7.
J Clin Lipidol ; 15(1): 151-161.e0, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33288437

RESUMO

BACKGROUND: The cardiovascular (CV) safety of estrogen replacement therapy (ERT) in perimenopausal women remains uncertain. Although exogenous estrogens increase HDL cholesterol (HDL-C), estrogen-mediated effects on alternative metrics of HDL that may better predict CV risk are unknown. OBJECTIVE: To determine the effects of transdermal ERT on HDL composition and cholesterol efflux capacity (CEC), as well as the relationships between these metrics and CV risk factors. METHODS: Fasting plasma samples were analyzed from 101 healthy, perimenopausal women randomized to receive either transdermal placebo or transdermal estradiol (100 µg/24 h) with intermittent micronized progesterone. At baseline and after 6 months of treatment, serum HDL CEC, HDL particle concentration, HDL protein composition, insulin resistance and brachial artery flow-mediated dilatation (FMD) were measured. RESULTS: No difference between groups was found for change in plasma HDL-C (p = 0.69). Between-group differences were found for changes in serum HDL total CEC [median change from baseline -5.4 (-17.3,+8.4)% ERT group versus +5.8 (-6.3,+16.9)% placebo group, p = 0.01] and ABCA1-specific CEC [median change from baseline -5.3 (-10.7,+6.7)% ERT group versus +7.4 (-1.5,+18.1)% placebo group, p = 0.0002]. Relative to placebo, transdermal ERT led to reductions in LDL-C (p < 0.0001) and insulin resistance (p = 0.0002). An inverse correlation was found between changes in serum HDL total CEC and FMD (ß = -0.26, p = 0.004). CONCLUSIONS: Natural menopause leads to an increase in serum HDL CEC, an effect that is abrogated by transdermal ERT. However, transdermal ERT leads to favorable changes in major CV risk factors.


Assuntos
Fatores de Risco de Doenças Cardíacas , Adulto , HDL-Colesterol , Estradiol , Feminino , Humanos , Pessoa de Meia-Idade
8.
Clin Endocrinol (Oxf) ; 93(5): 555-563, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32633813

RESUMO

OBJECTIVE: Ageing in male adults is typically accompanied by adiposity accumulation and changes in circulating sex hormone concentrations. We hypothesized that an ageing-associated increase in oestrogens and decrease in androgens would correlate with an increase in adiposity. DESIGN: 10-year prospective, observational study. STUDY SUBJECTS: A total of 190, community-dwelling men in the Japanese American Community Diabetes Study. MEASUREMENTS: At 0 and 10 years, CT scanning quantified intra-abdominal fat (IAF) and subcutaneous fat (SCF) areas while plasma concentrations of oestradiol, oestrone, testosterone and dihydrotestosterone were measured by liquid chromatography-tandem-mass spectrometry at each time point. Multivariate linear regression analyses assessed correlations between 10-year changes in hormone concentrations and IAF or SCF, adjusting for age and baseline fat depot area. RESULTS: Participants were middle-aged [median 54.8 years, interquartile range (IQR) 39.9-62.8] men and mostly overweight by Asian criterion (median BMI 24.9, IQR 23.3-27.1) and with few exceptions had normal sex-steroid concentrations. Median oestradiol and dihydrotestosterone did not change significantly between 0 and 10 years (P = .084 and P = .596, respectively) while median oestrone increased (P < .001) and testosterone decreased (P < .001). Median IAF and SCF increased from 0 to 10 years (both P < .001). In multivariate analyses, change in oestrone positively correlated (P = .019) while change in testosterone (P = .003) and dihydrotestosterone (P = .014) negatively correlated with change in IAF. Plasma oestradiol and oestrone positively correlated with change in SCF (P = .041 and P = .030, respectively) while testosterone (P = .031) negatively correlated in multivariate analysis. CONCLUSION: Among 190 community-dwelling, Japanese American men, increases in IAF were associated with decreases in plasma androgens and increases in plasma oestrone, but not oestradiol, at 10 years. Further research is necessary to understand whether changing hormone concentrations are causally related to changes in regional adiposity or whether the reverse is true.


Assuntos
Adiposidade , Asiático , Adulto , Estradiol , Estrona , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Testosterona , Tomografia Computadorizada por Raios X
9.
PLoS One ; 15(1): e0227830, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31971970

RESUMO

Estrogens are important for maintaining metabolic health in males. However, the key sources of local estrogen production for regulating energy metabolism have not been fully defined. Immune cells exhibit aromatase activity and are resident in metabolic tissues. To determine the relative contribution of immune cell-derived estrogens for metabolic health in males, C57BL6/J mice underwent bone marrow transplant with marrow from either wild-type (WT(WT)) or aromatase-deficient (WT(ArKO)) donors. Body weight, body composition, and glucose and insulin tolerance were assessed over 24 weeks with mice maintained on a regular chow diet. No differences were found in insulin sensitivity between groups, but WT(ArKO) mice were more glucose tolerant than WT(WT) mice 20 weeks after transplant, suggestive of enhanced glucose disposal (AUCglucose 6061±3349 in WT(WT) mice versus 3406±1367 in WT(ArKO) mice, p = 0.01). Consistent with this, skeletal muscle from WT(ArKO) mice showed higher expression of the mitochondrial genes Ppargc1a (p = 0.03) and Nrf1 (p = 0.01), as well as glucose transporter type 4 (GLUT4, Scl2a4; p = 0.02). Skeletal muscle from WT(ArKO) mice had a lower concentration of 17ß-estradiol (5489±2189 pg/gm in WT(WT) mice versus 3836±2160 pg/gm in WT(ArKO) mice, p = 0.08) but higher expression of estrogen receptor-α (ERα, Esr1), raising the possibility that aromatase deficiency in immune cells led to a compensatory increase in ERα signaling. No differences between groups were found with regard to body weight, adiposity, or gene expression within adipose tissue or liver. Immune cells are a key source of local 17ß-estradiol production and contribute to metabolic regulation in males, particularly within skeletal muscle. The respective intracrine and paracrine roles of immune cell-derived estrogens require further delineation, as do the pathways that regulate aromatase activity in immune cells specifically within metabolic tissues.


Assuntos
Aromatase/genética , Glucose/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Músculo Esquelético/metabolismo , Animais , Aromatase/metabolismo , Transplante de Medula Óssea , Células Cultivadas , Estrogênios/metabolismo , Deleção de Genes , Teste de Tolerância a Glucose , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
J Clin Lipidol ; 14(1): 66-76.e11, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31859127

RESUMO

BACKGROUND: Humans spend most of the time in the postprandial state, yet most knowledge about high-density lipoproteins (HDL) derives from the fasted state. HDL protein and lipid cargo mediate HDL's antiatherogenic effects, but whether these HDL constituents change in the postprandial state and are affected by dietary macronutrients remains unknown. OBJECTIVES: This study aimed to assess changes in HDL protein and lipid composition after the consumption of a high-carbohydrate or high saturated fat (HSF) meal. METHODS: We isolated HDL from plasma collected during a randomized, cross-over study of metabolically healthy subjects. Subjects consumed isocaloric meals consisting predominantly of either carbohydrate or fat. At baseline and at 3 and 6 hours postprandial, we quantified HDL protein and lipid composition by liquid chromatography-mass spectrometry. RESULTS: A total of 15 subjects were included (60% female, aged 34 ± 15 years, body mass index: 24.1 ± 2.7 kg/m2). Consumption of the HSF meal led to HDL enrichment in total lipid (P = .006), triglyceride (P = .02), and phospholipid (P = .008) content and a corresponding depletion in protein content. After the HSF meal, 16 of the 25 measured phosphatidylcholine species significantly increased in abundance (P values range from .027 to <.001), along with several sphingolipids including ceramides (P < .004), lactosylceramide (P = .023), and sphingomyelin-14 (P = .013). Enrichment in apolipoprotein A-I (P = .001) was the only significant change in HDL protein composition after the HSF meal. The high-carbohydrate meal conferred only minimal changes in HDL composition. CONCLUSION: Meal macronutrient content acutely affects HDL composition in the postprandial state, with the HSF meal resulting in enrichment of HDL phospholipid content with possible consequences for HDL function.


Assuntos
Carboidratos/administração & dosagem , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/sangue , Lipoproteínas HDL/sangue , Obesidade/sangue , Adulto , Glicemia/genética , Índice de Massa Corporal , Carboidratos/efeitos adversos , LDL-Colesterol/sangue , Gorduras na Dieta/efeitos adversos , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Jejum , Feminino , Humanos , Lipidômica/métodos , Masculino , Refeições , Obesidade/dietoterapia , Obesidade/genética , Obesidade/patologia , Período Pós-Prandial/genética , Triglicerídeos/sangue
11.
Obesity (Silver Spring) ; 26(12): 1898-1904, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30460775

RESUMO

OBJECTIVE: This study aimed to determine whether a relationship was evident between gliosis in the mediobasal hypothalamus (MBH) and plasma testosterone concentrations in men. METHODS: A total of 41 adult men (aged 18-50 years) from 23 twin pairs underwent fasting morning blood draw and brain magnetic resonance imaging. T2 relaxation time was used to quantify gliosis in the MBH and control areas in the putamen and amygdala. Plasma concentrations of testosterone and 17ß-estradiol were measured by liquid chromatography-tandem mass spectrometry. Body composition including visceral adiposity was measured by dual x-ray absorptiometry. RESULTS: A negative association was found between MBH T2 relaxation time and plasma concentrations of both free and total testosterone (r = -0.29, P < 0.05 and r = -0.37, P < 0.01, respectively). Visceral adiposity exhibited a negative correlation with plasma total testosterone concentration (r = -0.45, P = 0.001) but a positive correlation with MBH T2 relaxation time (r = 0.24, P = 0.03). The negative correlation between plasma total testosterone and MBH T2 relaxation time remained significant after adjustment for visceral adiposity, age, BMI, and insulin resistance. CONCLUSIONS: In healthy men across a range of BMIs, MBH gliosis was associated with higher visceral adiposity but lower endogenous testosterone. These findings suggest that MBH gliosis could provide novel mechanistic insights into gonadal dysfunction in men with obesity.


Assuntos
Gliose/diagnóstico por imagem , Hipotálamo/diagnóstico por imagem , Gordura Intra-Abdominal/anormalidades , Imageamento por Ressonância Magnética/métodos , Obesidade/sangue , Testosterona/sangue , Adolescente , Adulto , Gliose/patologia , Voluntários Saudáveis , Humanos , Hipotálamo/patologia , Gordura Intra-Abdominal/patologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
12.
J Clin Lipidol ; 12(4): 1072-1082, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29793828

RESUMO

BACKGROUND: Exogenous testosterone decreases serum concentrations of high-density lipoprotein cholesterol (HDL-C) in men, but whether this alters cardiovascular risk is uncertain. OBJECTIVE: To investigate the effects of testosterone and estradiol on HDL particle concentration (HDL-Pima) and metrics of HDL function. METHODS: We enrolled 53 healthy men, 19 to 55 years of age, in a double-blinded, placebo-controlled, randomized trial. Subjects were rendered medically castrate using the GnRH receptor antagonist acyline and administered either (1) placebo gel, (2) low-dose transdermal testosterone gel (1.62%, 1.25 g), (3) full replacement dose testosterone gel (1.62%, 5 g) or (4) full replacement dose testosterone gel together with an aromatase inhibitor for 4 weeks. At baseline and end of treatment, serum HDL total macrophage and ABCA1-specific cholesterol efflux capacity (CEC), HDL-Pima and size, and HDL protein composition were determined. RESULTS: Significant differences in serum HDL-C were observed with treatment across groups (P = .01 in overall repeated measures ANOVA), with increases in HDL-C seen after both complete and partial testosterone deprivation. Medical castration increased total HDL-Pima (median [interquartile range] 19.1 [1.8] nmol/L at baseline vs 21.3 [3.1] nmol/L at week 4, P = .006). However, corresponding changes in total macrophage CEC and ABCA1-specific CEC were not observed. Change in serum 17ß-estradiol concentration correlated with change in total macrophage CEC (ß = 0.33 per 10 pg/mL change in serum 17ß-estradiol, P = .03). CONCLUSIONS: Testosterone deprivation in healthy men leads to a dissociation between changes in serum HDL-C and HDL CEC. Changes in serum HDL-C specifically due to testosterone exposure may not reflect changes in HDL function.


Assuntos
HDL-Colesterol/sangue , Oligopeptídeos/administração & dosagem , Testosterona/administração & dosagem , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Adulto , Inibidores da Aromatase/administração & dosagem , Doenças Cardiovasculares/diagnóstico , Linhagem Celular , HDL-Colesterol/química , Método Duplo-Cego , Estradiol/sangue , Humanos , Injeções Subcutâneas , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Tamanho da Partícula , Efeito Placebo , Testosterona/sangue , Testosterona/farmacologia , Adulto Jovem
13.
Mol Metab ; 15: 92-103, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29551633

RESUMO

BACKGROUND: Over the past two decades, parallel recognition has grown of the importance of both sex steroids and immune activity in metabolic regulation. More recently, these discrete areas have been integrated in studies examining the metabolic effects of sex steroid immunomodulation. Implicit in these studies has been a traditional, endocrine model of sex steroid delivery from the gonads to target cells, including immune cells. Thus, research to date has focused on the metabolic effects of sex steroid receptor signaling in immune cells. This endocrine model, however, overlooks the extensive capacity of immune cells to generate and metabolize sex steroids, enabling the production of sex steroids for intracrine signaling - that is, sex steroid production for signaling within the cell of origin. Intracrine function allows highly cell-autonomous regulation of sex steroid exposure, and sex steroid secretion by immune cells could confer paracrine signaling effects in neighboring cells within metabolic tissues. In this review, immune cell intracrinology will denote sex steroid production within immune cells for either intracrine or paracrine signaling. This intracrine capacity of immune cells has been well established, and prior work has supported its importance in autoimmune disorders, trauma, and cancer. The potential relevance of immune cell intracrine function to the regulation of energy balance, body weight, body composition, and insulin sensitivity has yet to be explored. SCOPE OF REVIEW: The following review will detail findings to date regarding the steroidogenic and steroid metabolizing capacity of immune cells, the regulation of immune cell intracrine function, and the biological effects of immune-derived sex steroids, including the clinical relevance of immune cell intracrinology in fields other than metabolism. These findings will serve as the basis for a proposed model of immune cell intracrinology constituting a new frontier in metabolism research. MAJOR CONCLUSIONS: The development of highly sensitive mass spectrometric methods for sex steroid measurement and quantitation of metabolic flux now allows unprecedented ability to interrogate sex steroid production, metabolism and secretion by immune cells. Immune cell intracrinology could reveal key mechanisms underlying immune cell-mediated metabolic regulation.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Homeostase , Linfócitos/imunologia , Macrófagos/imunologia , Animais , Humanos , Transdução de Sinais
14.
Asian J Androl ; 20(3): 276-283, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29205180

RESUMO

Androgen deprivation in men leads to increased adiposity, but the mechanisms underlying androgen regulation of fat mass have not been fully defined. Androgen receptor (AR) is expressed in monocytes/macrophages, which are resident in key metabolic tissues and influence energy metabolism in surrounding cells. Male mice bearing a cell-specific knockout of the AR in monocytes/macrophages (M-ARKO) were generated to determine whether selective loss of androgen signaling in these cells would lead to altered body composition. Wild-type (WT) and M-ARKO mice (12-22 weeks of age, n = 12 per group) were maintained on a regular chow diet for 8 weeks and then switched to a high-fat diet for 8 additional weeks. At baseline and on both the regular chow and high-fat diets, no differences in lean mass or fat mass were observed between groups. Consistent with the absence of differential body weight or adiposity, no differences in food intake (3.0 ± 0.5 g per day for WT mice vs 2.8 ± 0.4 g per day for M-ARKO mice) or total energy expenditure (0.6 ± 0.1 Kcal h-1 for WT mice vs 0.5 ± 0.1 Kcal h-1 for M-ARKO mice) were evident between groups during high-fat feeding. Liver weight was greater in M-ARKO than that in WT mice (1.5 ± 0.1 g vs 1.3 ± 0.0 g, respectively, P = 0.02). Finally, M-ARKO mice did not exhibit impairments in glucose tolerance or insulin sensitivity relative to WT mice at any study time point. In aggregate, these findings suggest that AR signaling specifically in monocytes/macrophages does not contribute to the regulation of systemic energy balance, adiposity, or insulin sensitivity in male mice.


Assuntos
Adiposidade/genética , Fígado/anatomia & histologia , Macrófagos/metabolismo , Monócitos/metabolismo , Receptores Androgênicos/genética , Animais , Glicemia/genética , Glicemia/metabolismo , Metabolismo Energético/genética , Teste de Tolerância a Glucose , Homeostase/genética , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Receptores Androgênicos/metabolismo , Transdução de Sinais
15.
Adv Exp Med Biol ; 1043: 285-313, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224100

RESUMO

Our understanding of the metabolic roles of sex steroids in men has evolved substantially over recent decades. Whereas testosterone once was believed to contribute to metabolic risk in men, the importance of adequate androgen exposure for the maintenance of metabolic health has been demonstrated unequivocally. A growing body of evidence now also supports a critical role for estrogens in metabolic regulation in men. Recent data from clinical intervention studies indicate that estradiol may be a stronger determinant of adiposity than testosterone in men, and even short-term estradiol deprivation contributes to fat mass accrual. The following chapter will outline findings to date regarding the mechanisms, whereby estrogens contribute to the regulation of body weight and adiposity in men. It will present emergent clinical data as well as preclinical findings that reveal mechanistic insights into estrogen-mediated regulation of body composition. Findings in both males and females will be reviewed, to draw comparisons and to highlight knowledge gaps regarding estrogen action specifically in males. Finally, the clinical relevance of estrogen exposure in men will be discussed, particularly in the context of a rising global prevalence of obesity and expanding clinical use of sex steroid-based therapies in men.


Assuntos
Adiposidade , Peso Corporal , Metabolismo Energético , Estrogênios/metabolismo , Obesidade/metabolismo , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Animais , Fármacos Antiobesidade/uso terapêutico , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Estrogênios/uso terapêutico , Humanos , Masculino , Camundongos , Mutação , Obesidade/genética , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Fatores Sexuais
16.
Kidney Int ; 92(6): 1556, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29153142
17.
Kidney Int ; 92(6): 1526-1535, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28754556

RESUMO

Patients with chronic kidney disease (CKD) exhibit a myriad of metabolic derangements, including dyslipidemia characterized by low plasma concentrations of high-density lipoprotein (HDL)-associated cholesterol. However, the effects of kidney disease on HDL composition have not been comprehensively determined. Here we used a targeted mass spectrometric approach to quantify 38 proteins contained in the HDL particles within a CKD cohort of 509 participants with a broad range of estimated glomerular filtration rates (eGFRs) (CKD stages I-V, and a mean eGFR of 45.5 mL/min/1.73m2). After adjusting for multiple testing, demographics, comorbidities, medications, and other characteristics, eGFR was significantly associated with differences in four HDL proteins. Compared to participants with an eGFR of 60 mL/min/1.73m2 or more, those with an eGFR under 15 mL/min/1.73m2 exhibited 1.89-fold higher retinol-binding protein 4 (95% confidence interval 1.34-2.67), 1.52-fold higher apolipoprotein C-III (1.25-1.84), 0.70-fold lower apolipoprotein L1 (0.55-0.92), and 0.64-fold lower vitronectin (0.48-0.85). Although the HDL apolipoprotein L1 was slightly lower among African Americans than among Caucasian individuals, the relationship to eGFR did not differ by race. After adjustment, no HDL-associated proteins associated with albuminuria. Thus, modest changes in the HDL proteome provide preliminary evidence for an association between HDL proteins and declining kidney function, but this needs to be replicated. Future analyses will determine if HDL proteomics is indeed a clinical predictor of declining kidney function or cardiovascular outcomes.


Assuntos
Dislipidemias/sangue , Lipoproteínas HDL/sangue , Insuficiência Renal Crônica/sangue , Adulto , Negro ou Afro-Americano/estatística & dados numéricos , Idoso , Cromatografia Líquida/métodos , Estudos de Coortes , Dislipidemias/metabolismo , Feminino , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Proteômica/métodos , Insuficiência Renal Crônica/metabolismo , Fatores de Risco , Espectrometria de Massas em Tandem/métodos , População Branca/estatística & dados numéricos
18.
Am J Physiol Endocrinol Metab ; 313(5): E528-E539, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28698282

RESUMO

Male hypogonadism results in changes in body composition characterized by increases in fat mass. Resident immune cells influence energy metabolism in adipose tissue and could promote increased adiposity through paracrine effects. We hypothesized that manipulation of circulating sex steroid levels in healthy men would alter adipose tissue immune cell populations. Subjects (n = 44 men, 19-55 yr of age) received 4 wk of treatment with the gonadotropin-releasing hormone receptor antagonist acyline with daily administration of 1) placebo gel, 2) 1.25 g testosterone gel (1.62%), 3) 5 g testosterone gel, or 4) 5 g testosterone gel with an aromatase inhibitor. Subcutaneous adipose tissue biopsies were performed at baseline and end-of-treatment, and adipose tissue immune cells, gene expression, and intra-adipose estrogen levels were quantified. Change in serum total testosterone level correlated inversely with change in the number of CD3+ (ß = -0.36, P = 0.04), CD4+ (ß = -0.34, P = 0.04), and CD8+ (ß = -0.33, P = 0.05) T cells within adipose tissue. Change in serum 17ß-estradiol level correlated inversely with change in the number of adipose tissue macrophages (ATMs) (ß = -0.34, P = 0.05). A negative association also was found between change in serum testosterone and change in CD11c+ ATMs (ß = -0.41, P = 0.01). Overall, sex steroid deprivation was associated with increases in adipose tissue T cells and ATMs. No associations were found between changes in serum sex steroid levels and changes in adipose tissue gene expression. Circulating sex steroid levels may regulate adipose tissue immune cell populations. These exploratory findings highlight a possible novel mechanism that could contribute to increased metabolic risk in hypogonadal men.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Hormônios Esteroides Gonadais/fisiologia , Imunidade Celular/fisiologia , Adulto , Inibidores da Aromatase/farmacologia , Antígeno CD11c/metabolismo , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Estradiol/farmacologia , Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/sangue , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Receptores LHRH/antagonistas & inibidores , Linfócitos T/imunologia , Testosterona/sangue , Testosterona/farmacologia , Adulto Jovem
19.
Dialogues Clin Neurosci ; 19(1): 19-26, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28566944

RESUMO

The recognition of altered immune system function in many chronic disease states has proven to be a pivotal advance in biomedical research over the past decade. For many metabolic and mood disorders, this altered immune activity has been characterized as inflammation, with the attendant assumption that the immune response is aberrant. However, accumulating evidence challenges this assumption and suggests that the immune system may be mounting adaptive responses to chronic stressors. Further, the inordinate complexity of immune function renders a simplistic, binary model incapable of capturing critical mechanistic insights. In this perspective article, we propose alternative paradigms for understanding the role of the immune system in chronic disease. By invoking allostasis or systems biology rather than inflammation, we can ascribe greater functional significance to immune mediators, gain newfound appreciation of the adaptive facets of altered immune activity, and better avoid the potentially disastrous effects of translating erroneous assumptions into novel therapeutic strategies.


Durante la última década, el reconocimiento de la alteración de la función del sistema inmune en muchas enfermedades crónicas ha demostrado ser un avance central en la investigación biomédica. En diversos trastornos metabólicos y anímicos, esta actividad inmune alterada ha sido caracterizada como inflamación, asumiendo concomitantemente que la respuesta inmune es aberrante. Sin embargo, la evidencia acumulada desafía esta suposición y sugiere que el sistema inmune puede estar estructurando respuestas adaptativas a estresores crónicos. Además, un modelo simplista y binario no es capaz de dar cuenta de la excesiva complejidad de la función inmune ni hacer comprensibles los mecanismos internos esenciales. En este contexto, este artículo de perspectiva se propone entregar paradigmas alternativos para la comprensión del papel del sistema inmune en la enfermedad crónica. Al invocar la alostasis o la biología de sistemas antes que la inflamación, se puede atribuir un mayor significado funcional a los mediadores inmunes, obtener una nueva apreciación de los aspectos adaptativos de la actividad inmune alterada, y evitar mejor los efectos potencialmente desastrosos de traducir suposiciones erróneas en nuevas estrategias terapéuticas.


Ces 10 dernières années, l'identification d'une altération du système immunitaire dans de nombreuses maladies chroniques est au centre des avancées de la recherche biomédicale. Dans de nombreux troubles métaboliques ou de l'humeur, cette activité immunitaire modifiée se caractérise par une inflammation, ce qui suppose une réponse immunitaire anormale. Cette hypothèse est néanmoins mise en défaut car un nombre croissant d'arguments suggère que le système immunitaire développerait des réponses adaptatives aux facteurs de stress chroniques. De plus, un modèle simpliste, binaire, ne peut rendre compte de l'extraordinaire complexité de la fonction immunitaire et n'est pas suffisant pour en restituer les mécanismes fondamentaux. Dans ce contexte, nous proposons dans cet article d'autres modèles pour comprendre le rôle du système immunitaire dans la maladie chronique. En invoquant l'allostase ou la biologie de systèmes plutôt que l'inflammation, nous pouvons accorder une plus grande importance fonctionnelle aux médiateurs immunitaires, réviser notre conception des capacités d'adaptation d'une activité immunitaire altérée et mieux éviter les effets potentiellement désastreux d'une traduction d'hypothèses erronées en nouveaux traitements.


Assuntos
Adaptação Fisiológica/imunologia , Sistema Imunitário/imunologia , Doenças Metabólicas/imunologia , Transtornos do Humor/imunologia , Adaptação Fisiológica/fisiologia , Alostase/imunologia , Alostase/fisiologia , Doença Crônica , Humanos , Sistema Imunitário/fisiopatologia , Inflamação/imunologia , Inflamação/fisiopatologia , Doenças Metabólicas/fisiopatologia , Transtornos do Humor/fisiopatologia
20.
Metabolism ; 70: 12-22, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28403936

RESUMO

CONTEXT: The mechanisms mediating the short- and long-term improvements in glucose homeostasis following bariatric/metabolic surgery remain incompletely understood. OBJECTIVE: To investigate whether a reduction in adipose tissue inflammation plays a role in the metabolic improvements seen after bariatric/metabolic surgery, both in the short-term and longer-term. DESIGN: Fasting blood and subcutaneous abdominal adipose tissue were obtained before (n=14), at one month (n=9), and 6-12months (n=14) after bariatric/metabolic surgery from individuals with obesity who were not on insulin or anti-diabetes medication. Adipose tissue inflammation was assessed by a combination of whole-tissue gene expression and flow cytometry-based quantification of tissue leukocytes. RESULTS: One month after surgery, body weight was reduced by 13.5±4.4kg (p<0.001), with improvements in glucose tolerance reflected by a decrease in area-under-the-curve (AUC) glucose in 3-h oral glucose tolerance tests (-105±98mmol/L * min; p=0.009) and enhanced pancreatic ß-cell function (insulinogenic index: +0.8±0.9pmol/mmol; p=0.032), but no change in estimated insulin sensitivity (Matsuda insulin sensitivity index [ISI]; p=0.720). Furthermore, although biomarkers of systemic inflammation and pro-inflammatory gene expression in adipose tissue remained unchanged, the number of neutrophils increased in adipose tissue 15-20 fold (p<0.001), with less substantial increases in other leukocyte populations. By the 6-12month follow-up visit, body weight was reduced by 34.8±10.8kg (p<0.001) relative to baseline, and glucose tolerance was further improved (AUC glucose -276±229; p<0.001) along with estimated insulin sensitivity (Matsuda ISI: +4.6±3.2; p<0.001). In addition, improvements in systemic inflammation were reflected by reductions in circulating C-reactive protein (CRP; -2.0±5.3mg/dL; p=0.002), and increased serum adiponectin (+1358±1406pg/mL; p=0.003). However, leukocyte infiltration of adipose tissue remained elevated relative to baseline, with pro-inflammatory cytokine mRNA expression unchanged, while adiponectin mRNA expression trended downward (p=0.069). CONCLUSION: Both the short- and longer-term metabolic improvements following bariatric/metabolic surgery occur without significant reductions in measures of adipose tissue inflammation, as assessed by measuring the expression of genes encoding key mediators of inflammation and by flow cytometric immunophenotyping and quantification of adipose tissue leukocytes.


Assuntos
Cirurgia Bariátrica/métodos , Inflamação/cirurgia , Gordura Subcutânea/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Resistência à Insulina , Contagem de Leucócitos , Masculino , Metabolismo , Gordura Subcutânea/cirurgia , Fatores de Tempo , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...