Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638926

RESUMEN

Myo-inositol (myo-Ins) and D-chiro-inositol (D-chiro-Ins) are natural compounds involved in many biological pathways. Since the discovery of their involvement in endocrine signal transduction, myo-Ins and D-chiro-Ins supplementation has contributed to clinical approaches in ameliorating many gynecological and endocrinological diseases. Currently both myo-Ins and D-chiro-Ins are well-tolerated, effective alternative candidates to the classical insulin sensitizers, and are useful treatments in preventing and treating metabolic and reproductive disorders such as polycystic ovary syndrome (PCOS), gestational diabetes mellitus (GDM), and male fertility disturbances, like sperm abnormalities. Moreover, besides metabolic activity, myo-Ins and D-chiro-Ins deeply influence steroidogenesis, regulating the pools of androgens and estrogens, likely in opposite ways. Given the complexity of inositol-related mechanisms of action, many of their beneficial effects are still under scrutiny. Therefore, continuing research aims to discover new emerging roles and mechanisms that can allow clinicians to tailor inositol therapy and to use it in other medical areas, hitherto unexplored. The present paper outlines the established evidence on inositols and updates on recent research, namely concerning D-chiro-Ins involvement into steroidogenesis. In particular, D-chiro-Ins mediates insulin-induced testosterone biosynthesis from ovarian thecal cells and directly affects synthesis of estrogens by modulating the expression of the aromatase enzyme. Ovaries, as well as other organs and tissues, are characterized by a specific ratio of myo-Ins to D-chiro-Ins, which ensures their healthy state and proper functionality. Altered inositol ratios may account for pathological conditions, causing an imbalance in sex hormones. Such situations usually occur in association with medical conditions, such as PCOS, or as a consequence of some pharmacological treatments. Based on the physiological role of inositols and the pathological implications of altered myo-Ins to D-chiro-Ins ratios, inositol therapy may be designed with two different aims: (1) restoring the inositol physiological ratio; (2) altering the ratio in a controlled way to achieve specific effects.


Asunto(s)
Diabetes Gestacional/tratamiento farmacológico , Inositol/farmacología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Testosterona/metabolismo , Células Tecales/efectos de los fármacos , Diabetes Gestacional/metabolismo , Femenino , Humanos , Inositol/química , Inositol/metabolismo , Estructura Molecular , Síndrome del Ovario Poliquístico/metabolismo , Embarazo , Transducción de Señal/efectos de los fármacos , Células Tecales/metabolismo
2.
Expert Rev Endocrinol Metab ; 16(1): 9-18, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33382003

RESUMEN

Introduction: Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies in reproductive-aged women. Hyperandrogenism, polycystic ovaries, chronic anovulation, and metabolic aberrations are its common features. The treatment approach focuses on the main aberrations, which characterize the different phenotypes. Areas covered: Management strategies targeting the metabolic phenotype include lifestyle modifications for weight loss and improvement of dietary habits, as well as medication, such as insulin-sensitizers. The treatment of hyperandrogenic phenotype includes cosmetic procedures and the combined oral contraceptives with or without antiandrogens. The therapeutic approach to reproductive phenotype includes diet and lifestyle modifications, clomiphene citrate, and aromatase inhibitors. Alternative treatments include dietary supplements, herbs, resveratrol, myo-inositol, and acupuncture. Expert opinion: New studies have shown that higher anti-Müllerian hormone levels, gut microbiome composition, and plasma metabolomics are new parameters that are related to the most severe phenotypes. The clinical phenotypes can change over the lifespan with weight gain and can coexist in the same individual. Individualized treatment remains the main approach but grouping the phenotypes and following therapeutic recommendations may prove to be also clinically appropriate.


Asunto(s)
Anovulación , Hiperandrogenismo , Síndrome del Ovario Poliquístico , Adulto , Hormona Antimülleriana , Femenino , Humanos , Fenotipo , Síndrome del Ovario Poliquístico/tratamiento farmacológico
3.
Trends Endocrinol Metab ; 31(6): 435-447, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32396844

RESUMEN

This review details the physiologic roles of two insulin sensitizers, myo-inositol (MI) and d-chiro-inositol (DCI). In the human ovary, MI is a second messenger of follicle-stimulating hormone (FSH) and DCI is an aromatase inhibitor. These activities allow a treatment for polycystic ovary syndrome (PCOS) to be defined based on the combined administration of MI and DCI, where the best MI:DCI ratio is 40:1. Moreover, MI enhances the effect of metformin and clomiphene on the fertility of PCOS women seeking pregnancy. As impaired intestinal transport may lead to unsuccessful inositol treatment, we also discuss new data on the use of alpha-lactalbumin to boost inositol absorption. Overall, the physiological activities of MI and DCI dictate the dosages and timing of inositol supplementation in the treatment of PCOS.


Asunto(s)
Inositol/farmacología , Inositol/fisiología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Complejo Vitamínico B/farmacología , Animales , Femenino , Humanos , Inositol/administración & dosificación , Complejo Vitamínico B/administración & dosificación
5.
Expert Opin Drug Metab Toxicol ; 16(3): 255-274, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32129111

RESUMEN

Introduction: This Experts' opinion provides an updated scientific support to gynecologists, obstetricians, endocrinologists, nutritionists, neurologists and general practitioners on the use of Inositols in the therapy of Polycystic Ovary Syndrome (PCOS) and non-insulin dependent (type 2) diabetes mellitus (NIDDM).Areas covered: This paper summarizes the physiology of Myo-Inositol (MI) and D-Chiro-Inositol (DCI), two important molecules present in human organisms, and their therapeutic role, also for treating infertility. Some deep differences between the physiological functions of MI and DCI, as well as their safety and intestinal absorption are discussed. Updates include new evidence on the efficacy exerted in PCOS by the 40:1 MI/DCI ratio, and the innovative approach based on alpha-lactalbumin to overcome the decreased therapeutic efficacy of Inositols in some patients.Expert opinion: The evidence suggests that MI, alone or with DCI in the 40:1 ratio, offers a promising treatment for PCOS and NIDDM. However, additional studies need to evaluate some still unresolved issues, such as the best MI/DCI ratio for treating NIDDM, the potential cost-effectiveness of reduced gonadotropins administration in IVF due to MI treatment, or the benefit of MI supplementation in ovulation induction with clomiphene citrate in PCOS patients.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Testimonio de Experto , Inositol/uso terapéutico , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Reproducción/efectos de los fármacos , Complejo Vitamínico B/uso terapéutico , Animales , Diabetes Mellitus Tipo 2/metabolismo , Testimonio de Experto/tendencias , Femenino , Humanos , Inositol/farmacocinética , Síndrome del Ovario Poliquístico/metabolismo , Reproducción/fisiología , Complejo Vitamínico B/farmacocinética
6.
Front Horm Res ; 53: 50-64, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31499502

RESUMEN

Sex steroids, except for their primary reproductive role, exert key effects on metabolic target tissues. Androgen receptors have been detected in various tissues, participating in both central and peripheral regulation of metabolism and insulin action. The physiological role of androgens in regulating multiple aspects of female insulin signaling and energy metabolism becomes evident early in utero, thus programming how insulin-targeted tissues will behave in later life. Across lifespan, distinct effects of androgens in all insulin-targeted tissues are controlled by their circulating serum levels, within a narrow window, outside of which disturbances in metabolism are observed. Thus, androgen excess in women, as documented in those with polycystic ovary syndrome, can adversely affect insulin sensitivity, promoting visceral adiposity, adipose tissue dysfunction, and, ultimately, insulin resistance.


Asunto(s)
Adiposidad , Andrógenos/metabolismo , Hiperandrogenismo/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Andrógenos/sangre , Femenino , Humanos , Hiperandrogenismo/sangre , Insulina/sangre , Síndrome del Ovario Poliquístico/sangre
7.
Artículo en Inglés | MEDLINE | ID: mdl-31275245

RESUMEN

Background: Twenty-one-hydroxylase-deficient non-classic adrenal hyperplasia (NC-CAH) is a very common autosomal recessive syndrome with prevalence between 1:1,000 and 1:2,000 individuals and the frequency varies according to ethnicity. On the other hand, polycystic ovary syndrome has a familial basis and it is inherited under a complex hereditary trait. This syndrome affects 6 to 10% of women in reproductive age and it is the most common endocrine disorder in young women. Our aim was to investigate, through a systematic review, the distinct characteristics and common findings of these syndromes. Methods: The search period covered January 1970 to November 2018, using the scientific databases PubMed. Inclusion criteria were adult women patients with PCOS or NC-CAH. Search terms were "polycystic ovary syndrome," "PCOS," "non-classical adrenal hyperplasia," "NC-CAH," "21-hydroxylase deficiency." From an initial 16,255 titles, the evaluations led to the final inclusion of 97 papers. Results: The clinical features of NC-CAH are hirsutism and ovulatory and menstrual dysfunction therefore; differentiation between these two syndromes is difficult based on clinical grounds only. Additionally, NC-CAH and PCOS are both associated with obesity, insulin resistance, and dyslipidaemia. Reproductive abnormalities are also common between these hyperandrogenemic disorders since in patients with NC-CAH polycystic ovarian morphology and subfertility are present as they are in women with PCOS. The diagnosis of PCOS, is confirmed once other disorders that mimic PCOS have been excluded e.g., conditions that are related to oligoovulation or anovulation and/or hyperandrogenism, such as hyperprolactinaemia, thyroid disorders, non-classic congenital adrenal hyperplasia, and androgen-producing neoplasms. Conclusions: The screening tool to distinguish non-classic adrenal hyperplasia from PCOS is the measurement of 17-hydroxyprogesterone levels. The basal levels of 17-hydroxyprogesterone may overlap, but ACTH stimulation testing can distinguish the two entities. In this review these two common endocrine disorders are discussed in an effort to unveil their commonalities and to illuminate their shadowed distinctive characteristics.

8.
Artículo en Inglés | MEDLINE | ID: mdl-30881345

RESUMEN

Endocrine disrupting chemicals (EDCs), a heterogeneous group of exogenous chemicals that can interfere with any aspect of endogenous hormones, represent an emerging global threat for human metabolism. There is now considerable evidence that the observed upsurge of metabolic disease cannot be fully attributed to increased caloric intake, physical inactivity, sleep deficit, and ageing. Among environmental factors implicated in the global deterioration of metabolic health, EDCs have drawn the biggest attention of scientific community, and not unjustifiably. EDCs unleash a coordinated attack toward multiple components of human metabolism, including crucial, metabolically-active organs such as hypothalamus, adipose tissue, pancreatic beta cells, skeletal muscle, and liver. Specifically, EDCs' impact during critical developmental windows can promote the disruption of individual or multiple systems involved in metabolism, via inducing epigenetic changes that can permanently alter the epigenome in the germline, enabling changes to be transmitted to the subsequent generations. The clear effect of this multifaceted attack is the manifestation of metabolic disease, clinically expressed as obesity, metabolic syndrome, diabetes mellitus, and non-alcoholic fatty liver disease. Although limitations of EDCs research do exist, there is no doubt that EDCs constitute a crucial parameter of the global deterioration of metabolic health we currently encounter.

9.
Trends Endocrinol Metab ; 30(3): 150-162, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30712978

RESUMEN

Human, animal, and in vitro studies provide evidence that advanced glycation end-products (AGEs) may contribute to the pathogenesis of polycystic ovary syndrome (PCOS) and its metabolic and reproductive consequences. AGEs are able to induce, via activation of key intracellular signaling pathways, the generation of oxidative stress and proinflammatory cytokines, thus contributing to the adverse health impact of PCOS. This review presents the implications of AGEs in several disease pathophysiologies, including PCOS, as well as the cellular and systemic effects of AGEs on insulin resistance (IR), hyperandrogenemia, endoplasmic reticulum (ER) stress, hypoxia, and ovarian function. The gaps in our knowledge will serve as launching pad for future developments ranging from dietary and lifestyle changes to pharmaceutical interventions aiming at potential applications in women with PCOS.


Asunto(s)
Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/fisiopatología , Animales , Femenino , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Resistencia a la Insulina/fisiología , Receptor para Productos Finales de Glicación Avanzada/metabolismo
10.
Exp Biol Med (Maywood) ; 243(1): 29-33, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28914097

RESUMEN

Advanced glycation end products accumulate in the ovarian granulosa-cell layer of women with polycystic ovarian syndrome. Taken that the MAPK/ERK-pathway is a key regulator of oocyte maturation and function, consisting the main pathway used by the gonadotrophic hormones (luteinizing hormone, follicle stimulating hormone) to control ovulation, the present study aims to assess advanced glycation end products' interference into luteinizing hormone-and follicle stimulating hormone-signaling via the MAPK/ERK-pathway in the human granulosa KGN cell line. KGN cells were treated with luteinizing hormone or follicle stimulating hormone in the absence or presence of human glycated albumin. The specific activation of the main components of the MAPK/ERK1/2-pathway (namely c-Raf, MEK and ERK1/2) was assessed. Treatment of KGN cells with an MEK1/2-inhibitor or a blocking anti-RAGE-antibody was also performed to shed further light on the mechanism of the involvement of advanced glycation end products in luteinizing hormone and/or follicle stimulating hormone-related signaling pathways. Luteinizing hormone treatment increased p-ERK1/2 levels in human granulosa cells, while the combined treatment of luteinizing hormone and human glycated albumin provoked a decrease of p-ERK1/2 levels. A similar reducing effect was also observed for the upstream molecule phospho-cRaf upon combined treatment, while treatment with an MEK-inhibitor confirmed that the phenomenon is MAPK/ERK-pathway-dependent. Similarly, follicle stimulating hormone treatment increased p-ERK1/2 and p-MEK1/2 levels, while the combined treatment of follicle stimulating hormone and human glycated albumin downregulated their levels. Advanced glycation end products reduce the luteinizing hormone- and follicle stimulating hormone-induced ERK1/2 activation that is critical for granulosa cell mitogenesis and proliferation. Inappropriate activation of ERK1/2 in granulosa cells may block the granulosa cell differentiation pathway and/or impair follicular responses to hormones, potentially leading to ovulation failure that characterizes polycystic ovarian syndrome.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Hormona Luteinizante/metabolismo , Línea Celular , Femenino , Humanos , Transducción de Señal
11.
Eur J Endocrinol ; 176(6): R283-R308, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28264815

RESUMEN

Aging and its underlying pathophysiological background has always attracted the attention of the scientific society. Defined as the gradual, time-dependent, heterogeneous decline of physiological functions, aging is orchestrated by a plethora of molecular mechanisms, which vividly interact to alter body homeostasis. The ability of an organism to adjust to these alterations, in conjunction with the dynamic effect of various environmental stimuli across lifespan, promotes longevity, frailty or disease. Endocrine function undergoes major changes during aging, as well. Specifically, alterations in hormonal networks and concomitant hormonal deficits/excess, augmented by poor sensitivity of tissues to their action, take place. As hypothalamic-pituitary unit is the central regulator of crucial body functions, these alterations can be translated in significant clinical sequelae that can impair the quality of life and promote frailty and disease. Delineating the hormonal signaling alterations that occur across lifespan and exploring possible remedial interventions could possibly help us improve the quality of life of the elderly and promote longevity.


Asunto(s)
Envejecimiento/metabolismo , Sistema Endocrino/metabolismo , Estrés Oxidativo , Adyuvantes Inmunológicos/uso terapéutico , Andrógenos/uso terapéutico , Antioxidantes/uso terapéutico , Ritmo Circadiano , Deshidroepiandrosterona/uso terapéutico , Diabetes Mellitus Tipo 2/metabolismo , Dietoterapia , Terapia de Reemplazo de Estrógeno , Retroalimentación Fisiológica , Femenino , Preservación de la Fertilidad , Gonadotropinas/metabolismo , Terapia de Reemplazo de Hormonas , Humanos , Hiperandrogenismo/metabolismo , Hipertiroidismo/metabolismo , Hipertiroidismo/terapia , Hipoglucemiantes/uso terapéutico , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Menopausia/metabolismo , Reserva Ovárica , Medicina de Precisión , Calidad de Vida , Trasplante de Células Madre , Células Madre , Testosterona/uso terapéutico , Glándula Tiroides , Equilibrio Hidroelectrolítico
12.
Arch Endocrinol Metab ; 61(1): 54-61, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28273204

RESUMEN

OBJECTIVE: The aim of this study was to evaluate the association between thyroid function abnormalities and breast cancer and, in particular, the prognostic markers of breast cancer.. SUBJECTS AND METHODS: Baseline levels of thyrotropin, free triiodothyronine, free thyroxine and thyroid autoantibodies were measured in 97 women with primary breast cancer, 27 women with benign breast disease, and 4 women with atypical ductal hyperplasia. Their baseline levels were compared with those in 48 healthy women with a normal mammography in the last 2 years. RESULTS: There were no significant associations between history of thyroid disease and breast cancer (p = 0.33). The mean baseline levels of triiodothyronine and thyrotropin did not differ significantly between the compared groups. The mean baseline levels of free thyroxine were found to be significantly higher in the breast cancer group, even after adjusting for thyroid replacement therapy. The presence of thyroid antibodies did not differ significantly between the compared groups. In a subgroup analysis, breast cancer cases with thyroid disease and particularly hypothyroidism had a significantly lower incidence of lymph node metastases compared with breast cancer cases without thyroid disease. CONCLUSIONS: Our data confirmed the proliferative effect of thyroid hormones on breast cells, which had previously been shown in vitro. Additionally, thyroid disease and particularly hypothyroid function appeared to be associated with a lower incidence of lymph node metastases. Further studies to determine the prognostic role of thyroid hormones in breast cancer are warranted.


Asunto(s)
Biomarcadores/sangre , Neoplasias de la Mama/complicaciones , Glándula Tiroides/fisiopatología , Autoanticuerpos/sangre , Neoplasias de la Mama/sangre , Neoplasias de la Mama/fisiopatología , Estudios de Casos y Controles , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Pronóstico , Glándula Tiroides/irrigación sanguínea , Tirotropina/sangre , Tiroxina/sangre , Triyodotironina/sangre
13.
Minerva Endocrinol ; 42(4): 340-355, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28146139

RESUMEN

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive aged women. PCOS incorporates not only symptoms related to the reproductive system but also a clustering of systemic metabolic abnormalities that are linked with increased risk for cardiovascular disease (CVD). More specifically, metabolic aberrations such as impaired glucose and lipid metabolism, accompanied by increased low-grade inflammation as well as elevated coagulation factors appear to contribute to the increased cardiovascular risk. Even though many studies have indicated a rise in surrogate biomarkers of CVD in women with PCOS, it is still doubtful to what extent and magnitude this elevation can be translated to real cardiovascular events. Furthermore, the cardiovascular risk factors appear to vary significantly in the different phenotypes of the syndrome. Women with PCOS have the potential for early atherosclerosis, myocardial and endothelial dysfunction. Whether PCOS women are at real cardiovascular risk compared to controls remains between the verge of theoretical and real threat for the PCOS women at any age but particularly in the post-menopausal state. Interestingly, although the presence of the CVD risk factors is well documented in PCOS women, their combination on different phenotypes may play a role, which eventually results in a spectrum of clinical manifestations of CVD with variable degree of severity. The present manuscript aims to review the interaction between PCOS and the combination of several cardiovascular risk factors.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Síndrome del Ovario Poliquístico/complicaciones , Enfermedades Cardiovasculares/epidemiología , Femenino , Humanos , Síndrome del Ovario Poliquístico/epidemiología , Factores de Riesgo
14.
Arch. endocrinol. metab. (Online) ; 61(1): 54-61, Jan.-Feb. 2017. tab
Artículo en Inglés | LILACS | ID: biblio-838410

RESUMEN

ABSTRACT Objective The aim of this study was to evaluate the association between thyroid function abnormalities and breast cancer and, in particular, the prognostic markers of breast cancer.. Subjects and methods Baseline levels of thyrotropin, free triiodothyronine, free thyroxine and thyroid autoantibodies were measured in 97 women with primary breast cancer, 27 women with benign breast disease, and 4 women with atypical ductal hyperplasia. Their baseline levels were compared with those in 48 healthy women with a normal mammography in the last 2 years. Results There were no significant associations between history of thyroid disease and breast cancer (p = 0.33). The mean baseline levels of triiodothyronine and thyrotropin did not differ significantly between the compared groups. The mean baseline levels of free thyroxine were found to be significantly higher in the breast cancer group, even after adjusting for thyroid replacement therapy. The presence of thyroid antibodies did not differ significantly between the compared groups. In a subgroup analysis, breast cancer cases with thyroid disease and particularly hypothyroidism had a significantly lower incidence of lymph node metastases compared with breast cancer cases without thyroid disease. Conclusions Our data confirmed the proliferative effect of thyroid hormones on breast cells, which had previously been shown in vitro. Additionally, thyroid disease and particularly hypothyroid function appeared to be associated with a lower incidence of lymph node metastases. Further studies to determine the prognostic role of thyroid hormones in breast cancer are warranted.


Asunto(s)
Humanos , Femenino , Persona de Mediana Edad , Glándula Tiroides/fisiopatología , Neoplasias de la Mama/complicaciones , Biomarcadores/sangre , Pronóstico , Autoanticuerpos/sangre , Glándula Tiroides/irrigación sanguínea , Tiroxina/sangre , Triyodotironina/sangre , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/sangre , Tirotropina/sangre , Inmunohistoquímica , Estudios de Casos y Controles
15.
Expert Rev Endocrinol Metab ; 12(1): 87-95, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30058880

RESUMEN

INTRODUCTION: Polycystic ovary syndrome (PCOS) is a complex endocrinopathy affecting a remarkable proportion of premenopausal women. Different studies have shown that stress is widely encountered in women with PCOS. Areas covered: As PCOS is a multifaceted disorder, 'stress' incorporates different translations. We performed a literature review, focusing on the most recent data, regarding the multipotent role of stress in the syndrome. Expert commentary: Stress is believed to be an important component of PCOS. It encompasses different definitions that are all equivalent, like metabolic, inflammatory, oxidative and emotional stress. However, the type of stress that distinguishes PCOS is metabolic stress. It becomes evident early in life and constitutes the pathophysiological heart of the syndrome. Metabolic stress along with the other types of stress are the progenitors of severe long-term health implications, which exacerbate further the reproductive, metabolic and psychological derangements of the syndrome, leading to an endless cycle of chronic illness.

16.
Eur J Endocrinol ; 176(2): R79-R99, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27678478

RESUMEN

Nutrition can generate oxidative stress and trigger a cascade of molecular events that can disrupt oxidative and hormonal balance. Nutrient ingestion promotes a major inflammatory and oxidative response at the cellular level in the postprandial state, altering the metabolic state of tissues. A domino of unfavorable metabolic changes is orchestrated in the main metabolic organs, including adipose tissue, skeletal muscle, liver and pancreas, where subclinical inflammation, endothelial dysfunction, mitochondrial deregulation and impaired insulin response and secretion take place. Simultaneously, in reproductive tissues, nutrition-induced oxidative stress can potentially violate delicate oxidative balance that is mandatory to secure normal reproductive function. Taken all the above into account, nutrition and its accompanying postprandial oxidative stress, in the unique context of female hormonal background, can potentially compromise normal metabolic and reproductive functions in women and may act as an active mediator of various metabolic and reproductive disorders.


Asunto(s)
Endocrinología , Enfermedades Metabólicas/fisiopatología , Fenómenos Fisiológicos de la Nutrición , Estrés Oxidativo/fisiología , Reproducción/fisiología , Tejido Adiposo/metabolismo , Femenino , Humanos , Músculo Esquelético/metabolismo
17.
Menopause ; 24(3): 345-351, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27749734

RESUMEN

OBJECTIVE: The hypothalamic gonadotropin-releasing hormone pulse generator, the pituitary gonadotropes, the ovaries, and the uterus play a crucial role in female fertility. A decline in reproductive performance represents a complex interplay of actions at all levels of the hypothalamic-pituitary-ovarian axis. Recently, in the field of female reproductive aging attention is drawn to the carbonyl stress theory. Advanced glycation end products (AGEs) contribute directly to protein damage, induce a chain of oxidative stress (OS) reactions, and increase inflammatory reactions. Here, we highlight some of the mechanisms underlying glycation damage in the ovary. METHODS: Searches of electronic databases were performed. Articles relevant to possible role of OS, AGEs, and receptor for AGE (RAGE) in aging ovary were summarized in this interpretive literature review. RESULTS: Follicular microenvironment undergoes an increase in OS with aging. Data support the role of OS in ovulatory dysfunction because AGEs are well-recognized mediators of increased OS. RAGE and AGE-modified proteins with activated nuclear factor-kappa B are expressed in human ovarian tissue. It was suggested that accumulation of AGEs products at the level of the ovarian follicle might trigger early ovarian aging or could be responsible for reduced glucose uptake by granulosa cells, potentially altering follicular growth. Moreover, impaired methylglyoxal detoxification causing relevant damage to the ovarian proteome might be one of the mechanisms underlying reproductive aging. CONCLUSIONS: Further investigation of the role for the AGE-RAGE axis in the ovarian follicular environment is needed, and results could relate to assisted reproduction technology outcomes and new measures of ovarian reserve.


Asunto(s)
Envejecimiento/fisiología , Productos Finales de Glicación Avanzada/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Ovario/fisiología , Adulto , Femenino , Humanos , Inflamación/fisiopatología , Persona de Mediana Edad , Estrés Oxidativo/fisiología
18.
Hormones (Athens) ; 15(3): 321-344, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27838603

RESUMEN

Postprandial dysmetabolism is a postprandial state characterized by abnormal metabolism of glucose and lipids and, more specifically, of elevated levels of glucose and triglyceride (TG) containing lipoproteins. Since there is evidence that postprandial dysmetabolism is associated with increased cardiovascular mortality and morbidity, due to macro- and microvascular complications, as well as with conditions such as polycystic ovary syndrome (PCOS) and non-alcoholic fatty liver disease (NAFLD), it is recommended that clinicians be alert for early detection and management of this condition. Management consists of a holistic approach including dietary modification, exercise and use of hypoglycemic and hypolipidemic medication aiming to decrease the postprandial values of circulating glucose and triglycerides. This review aims to explain glucose and lipid homeostasis and the impact of postprandial dysmetabolism on the cardiovascular system as well as to offer suggestions with regard to the therapeutic approach for this entity. However, more trials are required to prevent or reverse early and not too late the actual tissue damage due to postprandial dysmetabolism.


Asunto(s)
Glucemia/metabolismo , Hiperglucemia/sangre , Hipertrigliceridemia/sangre , Periodo Posprandial , Triglicéridos/sangre , Animales , Biomarcadores/sangre , Enfermedades Cardiovasculares/etiología , Humanos , Hiperglucemia/diagnóstico , Hiperglucemia/etiología , Hiperglucemia/terapia , Hipertrigliceridemia/diagnóstico , Hipertrigliceridemia/etiología , Hipertrigliceridemia/terapia , Lipoproteínas/sangre , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo
19.
J BUON ; 21(4): 989-993, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27685924

RESUMEN

PURPOSE: Several studies have suggested that patients with acromegaly have an increased risk of thyroid, colorectal, breast and prostate cancers. In this study we determined the prevalence of malignant neoplasms in patients with acromegaly. METHODS: Cancer risk was evaluated in a cohort of 110 patients (M/F 48/62, age 58.63±13.8 years, range 30-86) with acromegaly. Mean age at diagnosis of acromegaly was 46.37±13.11 years. Mean period of time since diagnosis of acromegaly was 12.26+9.6 years. RESULTS: From 110 patients, cancer was diagnosed in 26 (23.6%) patients. Thyroid cancer was the most common cancer and was diagnosed in 13 patients (11.8%); other cancers encountered were gastric cancer (N=2), endometrial cancer (N-2), and breast cancer, colon cancer, prostate cancer (N-2), myelodysplastic syndrome, renal cell carcinoma, lung cancer and pancreatic carcinoma, one case each. Age, gender, age at the time of diagnosis of acromegaly, tumor size of pituitary adenoma and duration of disease were not associated with cancer development. CONCLUSIONS: This study suggests that patients with acromegaly have an increased risk of thyroid cancer and therefore they should undergo regular screening with hormonal and ultrasound evaluation of the thyroid and FNAB when required.


Asunto(s)
Acromegalia/complicaciones , Neoplasias de la Tiroides/etiología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Prevalencia , Riesgo , Glándula Tiroides/patología , Neoplasias de la Tiroides/patología
20.
Fertil Steril ; 106(4): 948-58, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27559705

RESUMEN

Polycystic ovary syndrome (PCOS) is the most common, heterogeneous, and multifactorial endocrine disorder in premenopausal women. The pathophysiology of this endocrinopathy is still unclear; however, the heterogeneity of its features within ethnic races, geographic location, and families suggests that environment and lifestyle are of prime importance. This work is mainly focused on the possible role of the most common and studied environmental toxins for this syndrome in the pathogenesis of PCOS. Plasticizers, such as bisphenol A (BPA) or phthalates, which belong to the categories of endocrine disrupting chemicals (EDCs) and advanced glycation end products (AGEs), affect humans' health in everyday, industrialized life; therefore special attention should be paid to such exposure. Timing of exposure to EDCs is crucial for the intensity of adverse health effects. It is now evident that fetuses, infants, and/or young children are the most susceptible groups, especially in the early development periods. Prenatal exposure to EDCs that mimic endogenous hormones may contribute to the altered fetal programming and in consequence lead to PCOS and other adverse health effects, potentially transgenerationally. Acute or prolonged exposure to EDCs and AGEs through different life cycle stages may result in destabilization of the hormonal homeostasis and lead to disruption of reproductive functions. They may also interfere with metabolic alterations such as obesity, insulin resistance, and compensatory hyperinsulinemia that can exacerbate the PCOS phenotype and contribute to PCOS consequences such as type 2 diabetes and cardiovascular disease. Since wide exposure to environmental toxins and their role in the pathophysiology of PCOS are supported by extensive data derived from diverse scientific models, protective strategies and strong recommendations should be considered to reduce human exposure to protect present and future generations from their adverse health effects.


Asunto(s)
Disruptores Endocrinos/efectos adversos , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/efectos adversos , Ovario/efectos de los fármacos , Síndrome del Ovario Poliquístico/inducido químicamente , Animales , Femenino , Productos Finales de Glicación Avanzada/metabolismo , Hormonas Esteroides Gonadales/biosíntesis , Humanos , Resistencia a la Insulina , Obesidad/complicaciones , Ovario/metabolismo , Ovario/patología , Ovario/fisiopatología , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Síndrome del Ovario Poliquístico/fisiopatología , Medición de Riesgo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...