Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Curr Issues Mol Biol ; 45(11): 9084-9102, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37998747

RESUMEN

The central mechanism involved in the pathogenesis of MAFLD is insulin resistance with hyperinsulinemia, which stimulates triglyceride synthesis and accumulation in the liver. On the other side, triglyceride and free fatty acid accumulation in hepatocytes promotes insulin resistance via oxidative stress, endoplasmic reticulum stress, lipotoxicity, and the increased secretion of hepatokines. Cytokines and adipokines cause insulin resistance, thus promoting lipolysis in adipose tissue and ectopic fat deposition in the muscles and liver. Free fatty acids along with cytokines and adipokines contribute to insulin resistance in the liver via the activation of numerous signaling pathways. The secretion of hepatokines, hormone-like proteins, primarily by hepatocytes is disturbed and impairs signaling pathways, causing metabolic dysregulation in the liver. ER stress and unfolded protein response play significant roles in insulin resistance aggravation through the activation of apoptosis, inflammatory response, and insulin signaling impairment mediated via IRE1/PERK/ATF6 signaling pathways and the upregulation of SREBP 1c. Circadian rhythm derangement and biological clock desynchronization are related to metabolic disorders, insulin resistance, and NAFLD, suggesting clock genes as a potential target for new therapeutic strategies. This review aims to summarize the mechanisms of hepatic insulin resistance involved in NAFLD development and progression.

2.
Horm Behav ; 153: 105392, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37295324

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex disorder characterized by endocrine and metabolic abnormalities such as obesity and insulin resistance. PCOS is also associated with psychiatric disorders and cognitive impairment. The animal model of PCOS was induced by treating rats with 5α-dihydrotestosterone (5α-DHT) and additionally modified to induce adiposity by litter size reduction (LSR). Spatial learning and memory were assessed using the Barnes Maze test, and striatal markers of synaptic plasticity were analyzed. Striatal insulin signaling was estimated by the levels of insulin receptor substrate 1 (IRS1), its inhibitory phosphorylation at Ser307, and glycogen synthase kinase-3α/ß (GSK3α/ß) activity. Both LSR and DHT treatment significantly decreased striatal protein levels of IRS1, followed by increased GSK3α/ß activity in small litters. Results of the behavioral study showed that LSR had a negative effect on learning rate and memory retention, whereas DHT treatment did not induce impairment in memory formation. While protein levels of synaptophysin, GAP43, and postsynaptic density protein 95 (PSD-95) were not altered by the treatments, DHT treatment induced an increase in phosphorylation of PSD-95 at Ser295 in both normal and small litters. This study revealed that LSR and DHT treatment suppressed insulin signaling by downregulating IRS1 in the striatum. However, DHT treatment did not have an adverse effect on learning and memory, probably due to compensatory elevation in pPSD-95-Ser295, which had a positive effect on synaptic strength. This implies that hyperandrogenemia in this setting does not represent a threat to spatial learning and memory, opposite to the effect of overnutrition-related adiposity.


Asunto(s)
Hiperandrogenismo , Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Femenino , Humanos , Ratas , Animales , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/metabolismo , Hiperandrogenismo/complicaciones , Hiperandrogenismo/metabolismo , Aprendizaje Espacial , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Dihidrotestosterona/farmacología , Obesidad/complicaciones , Modelos Animales de Enfermedad
3.
Rev Endocr Metab Disord ; 23(3): 431-447, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34529221

RESUMEN

The complex mechanisms, which are related to the pathophysiology and the development of autoimmune thyroid diseases, involve transforming growth factor beta (TGF-ß) and its interplay with the immune system. The aim of this review is to examine the role of TGF-ß regarding thyroid autoimmunity and explore the potent role of this molecule either as a diagnostic or prognostic marker or a therapeutic target regarding autoimmune thyroid diseases. TGF-ß is clearly a master regulator of the immune response, exerting either inhibitory or facilitatory effects on cells of the immune system. Thus, this molecule is involved in the pathogenesis and development of autoimmune thyroid diseases. Recent research has revealed the involvement of TGF-ß in the pathophysiology of autoimmune thyroid diseases. The role of TGF-ß in the development of autoimmune thyroid diseases varies, depending on its concentrations, the type of the activated TGF-ß signalling pathway, the genetic predisposition of the patient and the pathophysiologic stage of the disease. TGF-ß could emerge as a useful diagnostic or prognostic marker for the evolution of thyroid autoimmunity. Promising perspectives for the effective therapeutic use of TGF-ß regarding thyroid autoimmunity exist. The main treatment approaches incorporate either enhancement of the immunosuppressive role of TGF-ß or inhibition of its facilitatory role in the autoimmune thyroid diseases. Further research towards deeper understanding of TGF-ß physiology and clinical application of its possible therapeutic role regarding thyroid autoimmunity is needed.


Asunto(s)
Autoinmunidad , Enfermedad de Hashimoto , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
4.
Br J Nutr ; 128(4): 604-612, 2022 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-34511137

RESUMEN

Polycystic ovary syndrome (PCOS) is associated with altered lipid profile and increased small, dense LDL particles (sdLDL). Considering that paraoxonase 1 (PON1) is an antioxidative enzyme located on HDL particles, the aim of this study was to investigate the connection between oxidative stress (OS) and PON1 activity with lipoprotein subclasses in PCOS depending on obesity. In 115 PCOS patients, lipoprotein subclasses distributions were determined by gradient gel electrophoresis. OS status was assessed by total oxidative status (TOS), advanced oxidation protein products, malondialdehyde (MDA), prooxidant-antioxidant balance (PAB), total antioxidative status (TAS) and superoxide dismutase (SOD) and PON1 activity. Overweight/obese PCOS patients (n 55) had increased OS compared with normal weight patients (n 60). In addition, overweight/obese group had lower HDL size and higher proportion of HDL 3a subclasses (P < 0·05). PAB was in negative correlation with HDL 2a (P < 0·001), whereas MDA and SOD correlated positively with HDL 3 subclasses (P < 0·05). Serum PON1 activity was positively associated with proportions of PON1 activity on HDL 2b (P < 0·05) and 2a (P < 0·01), but negatively with the proportion on HDL 3 particles (P < 0·01). LDL B phenotype patients had increased TAS, SOD and PON1 activity on HDL 2b, but decreased PON1 activity on HDL 3 subclasses. OS is associated with altered lipoprotein subclasses distribution in PCOS patients. Obesity in PCOS affects the profile of HDL subclasses, reflected through the reduced proportion of PON1 activity on HDL 3 subclasses in the presence of sdLDL particles.


Asunto(s)
Dislipidemias , Síndrome del Ovario Poliquístico , Humanos , Femenino , Sobrepeso , Lipoproteínas HDL3/metabolismo , Estrés Oxidativo , Antioxidantes/metabolismo , Inflamación , Obesidad , Superóxido Dismutasa/metabolismo , Arildialquilfosfatasa/metabolismo
5.
Int J Mol Sci ; 23(16)2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-36012206

RESUMEN

Polycystic ovary syndrome (PCOS) is a well-known reproductive syndrome usually associated with obesity, insulin resistance, and hyperinsulinemia. Although the first signs of PCOS begin early in adolescence, it is underexplored whether peripubertal obesity predisposes women to PCOS metabolic disturbances. To highlight that, we examined the impact of postnatal overfeeding-induced obesity, achieved by litter size reduction during the suckling period, on metabolic disturbances associated with visceral and subcutaneous adipose tissue (VAT and SAT) function in the 5α-dihydrotestosterone (5α-DHT)-induced animal model of PCOS. We analyzed markers of insulin signaling, lipid metabolism, and energy sensing in the VAT and SAT. Our results showed that postnatally overfed DHT-treated Wistar rats had increased VAT mass with hypertrophic adipocytes, together with hyperinsulinemia and increased HOMA index. In the VAT of these animals, insulin signaling remained unchanged while lipogenic markers decreased, which was accompanied by increased AMPK activation. In the SAT of the same animals, markers of lipogenesis and lipolysis increased, while the activity of AMPK decreased. Taken together, obtained results showed that postnatal overfeeding predisposes development of PCOS systemic insulin resistance, most likely as a result of worsened metabolic function of SAT, while VAT preserved its tissue insulin sensitivity through increased activity of AMPK.


Asunto(s)
Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo/metabolismo , Animales , Femenino , Humanos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Ratas , Ratas Wistar , Grasa Subcutánea/metabolismo
6.
Horm Metab Res ; 52(3): 179-185, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32074632

RESUMEN

If circulating adrenal androgens levels rise before the age of 8 years in girls, this phenomenon is termed premature adrenarche (PA), while the concomitant appearance of pubic hair is called premature pubarche (PP). Girls with PA-PP display an unfavorable hormonal profile compared to their normal peers and have an increased risk of developing polycystic ovary syndrome (PCOS) features peripubertally. However, the sequelae of premature adrenarche remains unclear. We assessed metabolic, hormonal, psychologic profiles, and ovarian morphology in 21 women of mean age (±SD) 21.3±3.3 years, BMI: 23.6±4.4 kg/m2 with PA-PP, 45 women with PCOS and 26 controls, matched for age and BMI. PA-PP women displayed a favorable lipid profile compared to PCOS and controls. Insulin resistance index (HOMA-IR), however, were similar in PA-PP and PCOS women (2.09±1.42, 2.08±0.83) and higher than controls (1.13±0.49, p <0.05). Circulating androstenedione levels did not differ between PA-PP and PCOS women (0.11±0.05 vs. 0.12±0.03), but was higher than that of controls (0.02±0.0 nmol/l, p <0,05). Ovarian volume was increased in PA-PP and PCOS (11.14±3.3 vs. 10.99±4.61) compared to controls (6.74±1.83 cm3). PA-PP women had a higher score of state/trait anxiety and depressive and eating disorder symptoms than controls, with a pattern that matched that of PCOS women. Only 14% of the PA-PP group fulfilled the Rotterdam PCOS criteria. Some women with a history of PA-PP displayed hormonal and psychologic profile similar to PCOS, and accordingly a regular monitoring of these girls during adulthood is advised.


Asunto(s)
Hormonas/sangre , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/psicología , Pubertad Precoz/sangre , Pubertad Precoz/psicología , Adolescente , Adrenarquia/sangre , Adrenarquia/psicología , Andrógenos/sangre , Índice de Masa Corporal , Estudios Transversales , Femenino , Humanos , Ovario/diagnóstico por imagen , Ovario/crecimiento & desarrollo , Síndrome del Ovario Poliquístico/diagnóstico por imagen , Síndrome del Ovario Poliquístico/etiología , Pubertad Precoz/complicaciones , Pubertad Precoz/diagnóstico por imagen , Ultrasonografía , Adulto Joven
7.
Women Health ; 59(9): 1015-1025, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30870132

RESUMEN

This study aimed to analyze the measurement properties of the Health-related quality of life questionnaire for polycystic ovary syndrome (PCOSQ-50) in a sample of Serbian women with polycystic ovary syndrome (PCOS). Seventy-six women with PCOS from an endocrinology clinic and 28 healthy women participated between October 2016 and March 2017. The measure was rigorously translated and culturally adapted into Serbian. Psychometric evaluation included descriptive analysis, internal consistency (Cronbach's alpha coefficient), test-retest reliability (intraclass-correlation coefficient - ICC) and construct validity testing. Cronbach's alpha coefficient ranged from 0.67 to 0.96 for domain scales of PCOSQ-50 scores, while the ICCs for test-retest reliability for these domains ranged from 0.66 to 0.89. Women with PCOS had significantly lower scores than healthy women for hirsutism, obesity and menstrual disorders and the total PCOSQ-50 scale score (p ≤ 0.03), but not for the psychosocial and emotional, fertility, sexual function, and coping scales. These results show that the Serbian PCOSQ-50 measure is acceptable and could produce reliable and valid assessments of PCOS-related quality of life for at least four out of seven domains. Considering that validity testing is an iterative process, additional work is needed before the whole measure is used in routine clinical practice.


Asunto(s)
Síndrome del Ovario Poliquístico/psicología , Psicometría/estadística & datos numéricos , Calidad de Vida/psicología , Encuestas y Cuestionarios/normas , Adaptación Psicológica , Adulto , Emociones , Femenino , Humanos , Infertilidad Femenina/complicaciones , Trastornos de la Menstruación/complicaciones , Cuestionario de Salud del Paciente , Síndrome del Ovario Poliquístico/complicaciones , Reproducibilidad de los Resultados , Serbia
9.
Mol Cell Biochem ; 396(1-2): 99-105, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25052005

RESUMEN

Methionine is the only endogenous precursor of homocysteine, sulfur-containing amino acid and well known as risk factor for various brain disorders. Acetylcholinesterase is a serine protease that rapidly hydrolyzes neurotransmitter acetylcholine. It is widely distributed in different brain regions. The aim of this study was to elucidate the effects of methionine nutritional overload on acetylcholinesterase activity in the rat brain. Males of Wistar rats were randomly divided into control and experimental group, fed from 30th to 60th postnatal day with standard or methionine-enriched diet (double content comparing to standard, 7.7 g/kg), respectively. On the 61st postnatal day, total homocysteine concentration was determined and showed that animals fed with methionine-enriched diet had significantly higher serum total homocysteine concentrations comparing to control rats (p < 0.01). Acetylcholinesterase activity has been determined spectrophotometrically in homogenates of the cerebral cortex, hippocampus, thalamus, and nc. caudatus. Acetylcholinesterase activity showed tendency to decrease in all examined brain structures in experimental comparing to control rats, while statistical significance of this reduction was achieved in the cerebral cortex (p < 0.05). Brain slices were stained with haematoxylin and eosin (H&E) and observed under light microscopy. Histological analysis of H&E-stained brain slices showed that there were no changes in the brain tissue of rats which were on methionine-enriched diet compared to control rats. Results of this study showed selective vulnerability of different brain regions on reduction of acetylcholinesterase activity induced by methionine-enriched diet and consecutive hyperhomocysteinemia.


Asunto(s)
Acetilcolinesterasa/metabolismo , Encéfalo/metabolismo , Hiperhomocisteinemia/inducido químicamente , Metionina/efectos adversos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Dieta , Homocisteína/sangre , Hiperhomocisteinemia/metabolismo , Masculino , Ratas Wistar
10.
Gynecol Endocrinol ; 30(2): 100-2, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24308767

RESUMEN

INTRODUCTION: Although numerous studies indicated a link between antithyroid antibodies and recurrent spontaneous abortions (RSA), consensus on the treatment of this condition is still lacking. CASE REPORT: We present a case of a 35-year-old pregnant woman (gestation week 4) with primary hypothyroidism, total alopecia, high level of positive antithyroid antibodies, and history of two recurrent spontaneous abortions in early pregnancy. Along with L-thyroxin substitution, intravenous human immunoglobulin (IVIg) combined with anticoagulation and antiaggregation therapy was introduced. During pregnancy her scalp hair completely re-grew, and following gestation week 39 she delivered healthy female child. CONCLUSION: Thyroid antibodies could contribute to previous recurrent abortions in our patient. It is suggested that in older primiparas with Hashimoto thyroiditis and history of RSA, a combined treatment with IVIg, anticoagulation and antiaggregation therapy should be considered.


Asunto(s)
Aborto Habitual/tratamiento farmacológico , Alopecia/tratamiento farmacológico , Anticoagulantes/uso terapéutico , Enfermedad de Hashimoto/tratamiento farmacológico , Inmunoglobulinas Intravenosas/uso terapéutico , Tiroxina/uso terapéutico , Aborto Habitual/etiología , Adulto , Alopecia/complicaciones , Femenino , Enfermedad de Hashimoto/complicaciones , Humanos , Embarazo , Resultado del Embarazo , Resultado del Tratamiento
11.
Endocrine ; 86(1): 58-69, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38954374

RESUMEN

INTRODUCTION: Over the recent years, scientific community has increased its interest on solving problems of female fertility pathology. Many factors acting separately or in combination affect significantly the reproductive life of a woman. This review summarizes current evidence regarding the direct and/or indirect action of environmental factors and endocrine disrupting chemicals (EDCs; i.e. heavy metals, plasticizers, parabens, industrial chemicals, pesticides, or medications, by-products, anti-bacterial agents, perfluorochemicals) upon assisted and non-assisted female fertility, extracted from in vivo and in vitro animal and human published data. Transgenerational effects which could have been caused epigenetically by the action of EDCs have been raised. METHODS: This narrative review englobes and describes data from in vitro and in vivo animal and human studies with regard to the action of environmental factors, which include EDCs, on female fertility following the questions for narrative reviews of the SANRA (a scale for the quality assessment of narrative review articles). The identification of the studies was done: through the PubMed Central and the PubMed of the MEDLINE, the Google Scholar database and the Cochrane Library database until December 2023 combining appropriate keywords ("specific environmental factors" including "EDCs" AND "specific negative fertility outcomes"); by manual scanning of references from selected articles and reviews focusing on these subjects. It includes references to EDCs-induced transgenerational effects. RESULTS: From the reported evidence emerge negative or positive associations between specific environmental factors or EDCs and infertility outcomes such as infertility indices, disrupted maturation of the oocytes, anovulation, deranged transportation of the embryo and failure of implantation. CONCLUSION: The revealed adverse outcomes related to female fertility could be attributed to exposure to specific environmental factors such as temperature, climate, radiation, air pollutants, nutrition, toxic substances and EDCs. The recognition of fertility hazards related to the environment will permit the limitation of exposure to them, will improve female fertility and protect the health potential of future generations.


Asunto(s)
Disruptores Endocrinos , Exposición a Riesgos Ambientales , Contaminantes Ambientales , Fertilidad , Infertilidad Femenina , Humanos , Femenino , Disruptores Endocrinos/efectos adversos , Exposición a Riesgos Ambientales/efectos adversos , Fertilidad/efectos de los fármacos , Fertilidad/fisiología , Infertilidad Femenina/inducido químicamente , Animales , Contaminantes Ambientales/toxicidad , Contaminantes Ambientales/efectos adversos
12.
Front Endocrinol (Lausanne) ; 15: 1402905, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39268230

RESUMEN

Background: Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and is closely associated with chronic low-grade inflammation and insulin resistance. To clarify the contribution of prepubertal weight gain to the development of insulin resistance in PCOS, we investigated the effects of early postnatal overfeeding on inflammatory and energy-sensing pathways as well as on markers of insulin signaling in the liver of the PCOS rat model. Methods: Obesity induced by overfeeding was achieved by reducing litter size, while the PCOS-like condition was developed by treatment with 5α-dihydrotestosterone (DHT). Western blot and qPCR were used to analyze the expression of pro-inflammatory transcription factors and cytokines, as well as markers of the energy sensing and insulin signaling pathways. Results: The results showed that hepatic insulin sensitivity was impaired only in DHT-treated rats raised in small litters, as evidenced by increased phosphorylation of IRS1 on Ser307 and decreased expression of total IRS1. Postnatal overfeeding stimulated JNK1 activation independent of hyperandrogenemia; nevertheless, the synergistic effect of both factors triggered NLRP3 activation and increased IL1ß expression in the small litter DHT-treated group. This pro-inflammatory state was accompanied by decreased activatory phosphorylation of AMPK and reduced levels of its protein targets. Conclusions: Overfeeding in the early postnatal period leads to a decrease in hepatic insulin sensitivity in the rat model of PCOS, which is associated with decreased activation of AMPK and stimulation of the hepatic NLRP3-IL1ß signaling pathway. Accordingly, the inhibition of NLRP3 activation could provide a basis for the development of new therapeutic strategies for the treatment of insulin resistance in women with PCOS.


Asunto(s)
Dihidrotestosterona , Modelos Animales de Enfermedad , Inflamación , Resistencia a la Insulina , Hígado , Hipernutrición , Síndrome del Ovario Poliquístico , Animales , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/patología , Femenino , Ratas , Dihidrotestosterona/farmacología , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Inflamación/metabolismo , Inflamación/patología , Hipernutrición/metabolismo , Hipernutrición/complicaciones , Ratas Wistar , Obesidad/metabolismo , Animales Recién Nacidos , Transducción de Señal/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo
13.
Endocrine ; 85(1): 18-34, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38285412

RESUMEN

Obesity is the best described risk factor for the development of non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction associated steatotic liver disease (MASLD) and polycystic ovary syndrome (PCOS) while the major pathogenic mechanism linking these entities is insulin resistance (IR). IR is primarily caused by increased secretion of proinflammatory cytokines, adipokines, and lipids from visceral adipose tissue. Increased fatty acid mobilization results in ectopic fat deposition in the liver which causes endoplasmic reticulum stress, mitochondrial dysfunction, and oxidative stress resulting in increased cytokine production and subsequent inflammation. Similarly, IR with hyperinsulinemia cause hyperandrogenism, the hallmark of PCOS, and inflammation in the ovaries. Proinflammatory cytokines from both liver and ovaries aggravate IR thus providing a complex interaction between adipose tissue, liver, and ovaries in inducing metabolic abnormalities in obese subjects. Although many pathogenic mechanisms of IR, NAFLD/MASLD, and PCOS are known, there is still no effective therapy for these entities suggesting the need for further evaluation of their pathogenesis. Extracellular vesicles (EVs) represent a novel cross-talk mechanism between organs and include membrane-bound vesicles containing proteins, lipids, and nucleic acids that may change the phenotype and function of target cells. Adipose tissue releases EVs that promote IR, the development of all stages of NAFLD/MASLD and PCOS, while mesenchymal stem cell-derived AVs may alleviate metabolic abnormalities and may represent a novel therapeutic device in NAFLD/MASLD, and PCOS. The purpose of this review is to summarize the current knowledge on the role of adipose tissue-derived EVs in the pathogenesis of IR, NAFLD/MASLD, and PCOS.


Asunto(s)
Tejido Adiposo , Vesículas Extracelulares , Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Síndrome del Ovario Poliquístico , Humanos , Síndrome del Ovario Poliquístico/metabolismo , Femenino , Resistencia a la Insulina/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Vesículas Extracelulares/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Obesidad/metabolismo , Obesidad/complicaciones
14.
Endocr Connect ; 13(11)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39158582

RESUMEN

Introduction: To investigate whether synthetic (s) glucocorticoids (GCs) administered between the 24th and the 34th gestational weeks in pre-term labor might precipitate labor, studies on sGCs administration were reviewed. The physiology of endogenous glucocorticoid-related increase in fetal-maternal circulation and its association with labor, followed by a scoping review of studies on exogenous sGCs administered for fetal lung maturation and the timing of labor, were included. Materials and methods: The methodology of systematic reviews was followed. MEDLINE, Cochrane Library, and Google Scholar databases were searched until October 2023, for original studies investigating the administration of sGCs in pregnancies risking pre-term labor. Duplicates were removed, and 1867 abstracts were excluded as irrelevant. Six controlled and four non-controlled studies were included. The index group consisted of 6001 subjects and 7691 controls in the former, while in the latter, the index group consisted of 2069 subjects. Results: In three out of the six controlled studies, gestational age at labor was significantly lower in sGC-treated women than in controls, while in three studies, gestational age at labor was lower in sGC-treated women than in controls, with a trend toward statistical significance. In one study, gestational age at labor was significantly lower in controls than in sGC-treated women. In the non-controlled studies, the majority of women delivered less than 1 week from the day of sGC administration. Conclusions: In this scoping review, studies lack homogeneity. However, in the controlled studies, a pattern of earlier labor emerges among sGC-treated pregnant women. The use of multiple courses of antenatal sGCs appears to be associated with precipitated labor. Their use should be carefully weighed. Carefully designed trials should examine this ongoing scientific query.

15.
Hormones (Athens) ; 23(1): 49-58, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37792213

RESUMEN

PURPOSE: Polycystic ovary syndrome (PCOS) is an endocrine, metabolic, and reproductive disorder which, according to the Rotterdam criteria, affects up to 24% of women of childbearing age. Although the prevalence of infertility in this subpopulation of women is high, the optimal treatment has not been fully established yet. Insulin resistance is considered to be an important mechanism involved in the development of PCOS; hence, the aim of this narrative review is to present an overview of the current pharmacological insulin-sensitizing treatment modalities for infertile women with PCOS. METHODS: A MEDLINE and PubMed search for the years 1990-2023 was performed using a combination of keywords. Clinical trials with insulin sensitizers used for infertility treatment as well as analyses of systematic reviews and meta-analyses were evaluated. When deemed necessary, additional articles referenced in the retrieved papers were included in this narrative review. RESULTS: Several insulin-sensitizing compounds and various therapeutical protocols are available for infertility treatment of women with PCOS. Metformin is the most common adjuvant medication to induce ovulation in infertile women with PCOS and is more frequently administered in combination with clomiphene citrate than on its own. Recently, inositol and glucagon-like peptide-1 (GLP-1) receptor agonists have emerged as possible options for infertility treatment in PCOS. CONCLUSION: The future of medical treatment of PCOS women with infertility lies in a personalized pharmacological approach, which involves various compounds with different mechanisms of action that could modify ovarian function and endometrial receptivity, ultimately leading to better overall reproductive outcomes in these women.


Asunto(s)
Infertilidad Femenina , Metformina , Síndrome del Ovario Poliquístico , Femenino , Humanos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Infertilidad Femenina/tratamiento farmacológico , Insulina , Inducción de la Ovulación/métodos , Revisiones Sistemáticas como Asunto , Clomifeno/uso terapéutico , Metformina/uso terapéutico , Hipoglucemiantes/uso terapéutico
16.
Eur J Endocrinol ; 191(2): 134-143, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39099229

RESUMEN

BACKGROUND: Although polycystic ovary syndrome (PCOS) is a very common endocrinopathy, there are several issues related to this disorder which perplex clinicians in their everyday practice. OBJECTIVE: To determine the current state of knowledge among European endocrinologists concerning the full spectrum of PCOS. METHODS: An online survey comprising 41 items covering various aspects of PCOS diagnosis and management was distributed to members of the European Society of Endocrinology. RESULTS: A total of 505 European endocrinologists (64% females), with a mean age of 47 ± 11.6 years, participated in the survey. The Rotterdam criteria were the primary diagnostic tool for 85% of respondents. Most referrals (87.1%) occurred between ages 20 and 40 years. Twenty-five percent of physicians have access to mass spectrometry for the evaluation of androgen levels. While an extended metabolic profile was commonly employed as part of the workup, there was uncertainty regarding chronic anovulation diagnosis. Diabetes, including gestational or type 2, was recognized as a significant risk factor with universal screening irrespective of BMI status. Lifestyle modification and metformin were considered as standard interventions by all participants alongside oral contraceptives, though there was significant discrepancy in treatment duration. CONCLUSIONS: The Rotterdam diagnostic criteria are widely adopted for PCOS diagnosis among European endocrinologists. The current updated survey shows an emphasis on steroid profiling as an important part of diagnostic workup and a strong position held for recognition of PCOS as a metabolic condition with potentially serious implications. Current therapy thus shifted to the demand for prioritizing lifestyle interventions and metabolic therapies, either as monotherapy or in combination with standard hormone compounds.


Asunto(s)
Síndrome del Ovario Poliquístico , Humanos , Síndrome del Ovario Poliquístico/diagnóstico , Síndrome del Ovario Poliquístico/terapia , Femenino , Adulto , Europa (Continente)/epidemiología , Encuestas y Cuestionarios , Persona de Mediana Edad , Masculino , Adulto Joven , Endocrinólogos , Endocrinología/métodos , Metformina/uso terapéutico
17.
Hormones (Athens) ; 23(2): 183-204, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38619812

RESUMEN

EndoBridge 2023 took place on October 20-22, 2023, in Antalya, Turkey. Accredited by the European Council, the 3-day scientific program of the 11th Annual Meeting of EndoBridge included state-of-the-art lectures and interactive small group discussion sessions incorporating interesting and challenging clinical cases led by globally recognized leaders in the field and was well attended by a highly diverse audience. Following its established format over the years, the program provided a comprehensive update across all aspects of endocrinology and metabolism, including topics in pituitary, thyroid, bone, and adrenal disorders, neuroendocrine tumors, diabetes mellitus, obesity, nutrition, and lipid disorders. As usual, the meeting was held in English with simultaneous translation into Russian, Arabic, and Turkish. The abstracts of clinical cases presented by the delegates during oral and poster sessions have been published in JCEM Case Reports. Herein, we provide a paper on highlights and pearls of the meeting sessions covering a wide range of subjects, from thyroid nodule stratification to secondary osteoporosis and from glycemic challenges in post-bariatric surgery to male hypogonadism. This report emphasizes the latest developments in the field, along with clinical approaches to common endocrine issues. The 12th annual meeting of EndoBridge will be held on October 17-20, 2024 in Antalya, Turkey.


Asunto(s)
Enfermedades del Sistema Endocrino , Humanos , Enfermedades del Sistema Endocrino/terapia , Endocrinología/historia , Osteoporosis/terapia
18.
Clin Endocrinol (Oxf) ; 78(4): 586-92, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22888988

RESUMEN

OBJECTIVE: The polycystic ovary syndrome (PCOS) and the metabolic syndrome (MetS) are common disorders that share many characteristics, particularly abdominal obesity and insulin resistance. Our objective was to compare the prevalence of MetS between a large cohort of patients with PCOS and body mass index -matched controls. DESIGN: Cross-sectional study. PATIENTS: We studied 1223 patients with PCOS and 277 healthy women. Diagnosis of PCOS was based on the revised Rotterdam criteria. Women with PCOS were divided into those who fulfilled both the Rotterdam criteria and the diagnostic criteria of the 1990 National Institutes of Health definition of PCOS (group 1, n = 905) and into those with the additional phenotypes introduced by the Rotterdam criteria (group 2, n = 318). Diagnosis of MetS was based on four different definitions. MEASUREMENTS: Anthropometric, metabolic, hormonal and ultrasonographic features of PCOS. RESULTS: The prevalence of metabolic syndrome (MetS) was higher in women with PCOS than in controls when the National Cholesterol Education Program Adult Treatment Panel III definition of MetS was applied (15·8% and 10·1%, respectively; P = 0·021) but not with the three more recent MetS definitions. The prevalence of MetS was higher in group 1 than in controls regardless of the applied MetS definition. In contrast, the prevalence of MetS was similar in group 2 and in controls regardless of the applied MetS definition. In logistic regression analysis, PCOS did not predict the presence of MetS. CONCLUSIONS: Polycystic ovary syndrome per se does not appear to increase the risk of MetS independent of abdominal obesity.


Asunto(s)
Síndrome Metabólico/epidemiología , Síndrome del Ovario Poliquístico/epidemiología , Adulto , Pesos y Medidas Corporales , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Síndrome Metabólico/diagnóstico por imagen , Obesidad Abdominal/complicaciones , Obesidad Abdominal/diagnóstico por imagen , Obesidad Abdominal/epidemiología , Síndrome del Ovario Poliquístico/diagnóstico por imagen , Prevalencia , Pronóstico , Factores de Riesgo , Ultrasonografía , Adulto Joven
19.
Hum Reprod ; 28(3): 785-93, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23315058

RESUMEN

STUDY QUESTION: Do women with the polycystic ovary syndrome (PCOS) differ from those with the metabolic syndrome (MetS) in markers of insulin resistance (IR) and circulating androgens? SUMMARY ANSWER: Women with MetS have more pronounced IR than those with PCOS whereas only the latter have elevated circulating androgens. WHAT IS KNOWN ALREADY: PCOS and MetS share many similarities, including abdominal obesity and IR, and PCOS is regarded as the ovarian manifestation of MetS. However, there are limited data on the differences between markers of IR and circulating androgens between women with these two syndromes. STUDY DESIGN, SIZE, DURATION: A prospective study in 1223 Caucasian women with PCOS and 277 women without PCOS, matched for BMI, was performed between May 2004 and December 2011. The presence/absence of MetS in PCOS+ and PCOS- women was recorded and comparisons among the resulting four groups were performed. PARTICIPANTS/MATERIALS, SETTING, METHODS: This study was performed in a university department of obstetrics and gynecology. The following markers of IR were determined: serum glucose and insulin levels, glucose/insulin ratio, area under the oral glucose tolerance test, homeostasis model assessment of IR index and quantitative insulin sensitivity check index. MAIN RESULTS AND THE ROLE OF CHANCE: PCOS+MetS+ women (n = 361) were more insulin-resistant than PCOS+MetS- women (n = 862) (P < 0.001 for the comparisons in all markers of IR). Similarly, PCOS-MetS+ women (n = 66) were more insulin-resistant than PCOS-MetS- women (n = 211) (P < 0.001 for the comparisons in all markers of IR). In contrast, PCOS+MetS+ showed only borderline significant differences in some markers of IR compared with PCOS-MetS+ women (P < 0.05). Similarly, PCOS+MetS- women showed only borderline significant differences in some markers of IR compared with PCOS-MetS- women (P = 0.037). Moreover, PCOS-MetS+ women were more insulin-resistant than PCOS + MetS- women (P < 0.001 for the comparisons in all markers of IR). Regarding circulating androgens, PCOS+MetS+ women had higher levels of circulating androgens than PCOS-MetS+ women (P < 0.001 for the comparisons in all circulating androgens). Similarly, PCOS+MetS- women had higher levels of circulating androgens than PCOS-MetS- women (P < 0.001 for the comparisons in all circulating androgens). In contrast, circulating androgens did not differ between PCOS+MetS+ women and PCOS+MetS- women. Similarly, circulating androgens did not differ between PCOS-MetS+ women and PCOS-MetS- women. LIMITATIONS, REASONS FOR CAUTION: Only Caucasian women were included in the study. IR was not assessed with the euglycemic hyperinsulinemic clamp. WIDER IMPLICATIONS OF THE FINDINGS: Even though MetS and PCOS have many similarities, they are distinct disorders. PCOS does not appear to simply represent the ovarian manifestation of MetS. Further studies are required to assess the contribution of hyperandrogenism to the pathogenesis of IR in PCOS. STUDY FUNDING/COMPETING INTEREST(S): No external funding was either sought or obtained for this study.


Asunto(s)
Andrógenos/sangre , Resistencia a la Insulina , Síndrome Metabólico/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Adolescente , Adulto , Biomarcadores/sangre , Índice de Masa Corporal , Estudios de Cohortes , Femenino , Grecia , Hospitales Universitarios , Humanos , Síndrome Metabólico/sangre , Síndrome Metabólico/complicaciones , Síndrome Metabólico/diagnóstico , Obesidad/complicaciones , Servicio Ambulatorio en Hospital , Sobrepeso/complicaciones , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/diagnóstico , Guías de Práctica Clínica como Asunto , Estudios Prospectivos , Adulto Joven
20.
Gynecol Endocrinol ; 29(10): 926-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23885694

RESUMEN

AIM: To compare the prevalence of metabolic syndrome (MetS) between women with polycystic ovary syndrome (PCOS) and controls across different age (≤20, 21-30 and 31-39 years old) and body mass index (BMI) (normal weight, overweight and obese) groups. METHODS: We studied 1223 women with PCOS and 277 BMI-matched controls. The prevalence of MetS in women with PCOS and controls was estimated according to four different MetS definitions. RESULTS: In subjects ≤20 and 21-30 years old, the prevalence of MetS did not differ between women with PCOS and controls regardless of the MetS definition, even though women with PCOS were more obese than controls in the ≤20 years old group. In subjects 31-39 years old, the prevalence of MetS was higher in women with PCOS than in controls but the former were more obese than controls. The prevalence of MetS did not differ significantly between women with PCOS and controls in any of the BMI groups (normal weight, overweight or obese) regardless of the MetS definition. CONCLUSION: The prevalence of Mets appears to be primarily determined by obesity and age whereas PCOS per se appears to be a less important contributing factor.


Asunto(s)
Índice de Masa Corporal , Síndrome Metabólico/epidemiología , Síndrome del Ovario Poliquístico/epidemiología , Adolescente , Adulto , Factores de Edad , Estudios de Casos y Controles , Femenino , Humanos , Resistencia a la Insulina , Síndrome Metabólico/complicaciones , Síndrome del Ovario Poliquístico/complicaciones , Prevalencia , Circunferencia de la Cintura , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA