Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Actas Esp Psiquiatr ; 45(5): 227-47, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29044447

ABSTRACT

Aging and Alzheimer’s disease (AD) are associated with a declination of cognition and memory, whose severity increases in AD. Recent investigations point to a greater participation of neurofibrillary tangles (NFTs) than that of senile plaques, as responsible for cognitive impairment in AD and normal aging. On the other hand, aging is related with reduced levels of dehydroepiandrosterone (DHEA) and its sulfate (DHEA-S) as well as testosterone (T). Basic and clinical studies give evidence that hypoandrogenism is associated with memory impairment. Accordingly, some animal studies show that the administration of these hormones improves the performance of cognitive tasks. However, effects of DHEA, DHEA-S, and T in the clinical setting, are not clear in part because of the balance between the benefits and risks of hormone therapy in aging subjects and because the cellular mechanism underlying its effects on memory in old age and related pathologies are unknown. The objective of this review is to analyze the role of DHEA, DHEA-S, and T, on memory in normal aging and in AD, and to determine whether these hormones modulate the hyperphosphorylation of tau protein, a molecular marker in AD pathology. The method used in the review included articles from the PubMed database, using the following search terms: DHEA, DHEA-S, T, memory, androgen deprivation therapy, tau protein, aging, and AD. Finally, we analyze the use of these steroids as an adjunct in the treatment of memory deficits in aging subjects and AD patients.


Subject(s)
Alzheimer Disease/etiology , Dehydroepiandrosterone/deficiency , Memory Disorders/etiology , Testosterone/deficiency , Aged , Aging , Humans
2.
Actas esp. psiquiatr ; 45(5): 227-247, sept.-oct. 2017. tab, graf
Article in Spanish | IBECS | ID: ibc-167714

ABSTRACT

El envejecimiento y la enfermedad de Alzheimer (EA) se asocian con una declinación de la cognición y la memoria, cuya gravedad aumenta en la EA. Varias investigaciones apuntan a una mayor participación de los ovillos neurofbrilares respecto a las placas seniles, como responsables del deterioro cognitivo en la EA y en el envejecimiento normal. Por otro lado, el envejecimiento se relaciona con una reducción en los niveles de dehidroepiandrosterona (DHEA) y su sulfato (DHEA-S), así como de testosterona (T); algunas evidencias básicas y clínicas indican que esta condición se asocia con deterioro en la memoria. Varios estudios en animales revelan que la administración de DHEA, DHEA-S y T mejoran la ejecución de tareas cognitivas. Sin embargo, el efecto de estas hormonas en el ámbito clínico no es claro, en parte por el balance entre los beneficios y los riesgos de una terapia hormonal en pacientes ancianos, así como por el desconocimiento de los mecanismos celulares que subyacen a sus efectos sobre la memoria en la vejez y en patologías relacionadas. El objetivo de esta revisión narrativa es analizar el papel de los esteroides DHEA, DHEA-S y T en la memoria en el envejecimiento normal y en la EA, así como la modulación en la hiperfosforilación de la proteína tau, un marcador molecular de la patología de la EA, por estas hormonas. El método empleado en esta revisión fue una búsqueda en la base de datos de Pubmed con los siguientes términos: DHEA, DHEA-S, T, memoria, terapia de privación de andrógenos, proteína tau, envejecimiento y EA. Finalmente, se analizará el empleo de estos esteroides como un coadyuvante en el tratamiento de las alteraciones de memoria en sujetos envejecidos y en pacientes con EA (AU)


Aging and Alzheimer's disease (AD) are associated with a declination of cognition and memory, whose severity increases in AD. Recent investigations point to a greater participation of neurofibrillary tangles (NFTs) than that of senile plaques, as responsible for cognitive impairment in AD and normal aging. On the other hand, aging is related with reduced levels of dehydroepiandrosterone (DHEA) and its sulfate (DHEA-S) as well as testosterone (T). Basic and clinical studies give evidence that hypoandrogenism is associated with memory impairment. Accordingly, some animal studies show that the administration of these hormones improves the performance of cognitive tasks. However, effects of DHEA, DHEA-S, and T in the clinical setting, are not clear in part because of the balance between the benefits and risks of hormone therapy in aging subjects and because the cellular mechanism underlying its effects on memory in old age and related pathologies are unknown. The objective of this review is to analyze the role of DHEA, DHEA-S, and T, on memory in normal aging and in AD, and to determine whether these hormones modulate the hyperphosphorylation of tau protein, a molecular marker in AD pathology. The method used in the review included articles from the PubMed database, using the following search terms: DHEA, DHEA-S, T, memory, androgen deprivation therapy, tau protein, aging, and AD. Finally, we analyze the use of these steroids as an adjunct in the treatment of memory deficits in aging subjects and AD patients (AU)


Subject(s)
Humans , Androgens/deficiency , Aging , Alzheimer Disease/complications , Memory Disorders/epidemiology , Cognition Disorders/epidemiology , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone Sulfate/analysis , Testosterone/deficiency
3.
Rev Med Suisse ; 10(439): 1555-6, 1558-61, 2014 Aug 27.
Article in French | MEDLINE | ID: mdl-25272673

ABSTRACT

Biological aging is characterized by a progressive loss of the secretion of various hormones, a phenomenon that leads some physicians to propose an anti-aging hormonal therapy. It is mandatory to differentiate: 1) the physiological functional loss, which is a natural phenomenon without clear deleterious consequences on health and should not be compensated by the administration of hormones only to restore plasma levels similar to those measured in young people and 2) a pathological defect that deserves a replacement therapy to correct the endocrine deficiency and improve the health status of older individuals. This article considers the deficiencies in insulin, thyroid hormones, growth hormone, dehydroepiandrosterone (DHEA) and testosterone. For each hormone, a benefit/risk ratio of a so-called replacement therapy will be analyzed.


Subject(s)
Aging/metabolism , Hormone Replacement Therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/deficiency , Aged , Androgens/administration & dosage , Androgens/deficiency , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/deficiency , Diabetes Mellitus, Type 2/drug therapy , Female , Glucagon-Like Peptide 1/agonists , Growth Hormone/deficiency , Growth Hormone/therapeutic use , Health Status , Hormone Replacement Therapy/adverse effects , Hormone Replacement Therapy/methods , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Male , Metformin/therapeutic use , Testosterone/administration & dosage , Testosterone/deficiency , Thyroid Hormones/administration & dosage , Thyroid Hormones/deficiency
4.
Pain Physician ; 17(4): E543-8, 2014.
Article in English | MEDLINE | ID: mdl-25054405

ABSTRACT

BACKGROUND: Intrathecal drug delivery of opioids is an efficient and effective treatment option for pain management in the chronic nonmalignant pain population. As with all treatments, in addition to the benefits, risks and side effects exist. One such risk in intrathecal opioids is opioid-induced androgen deficiency. OBJECTIVE: This study evaluates opioid-induced androgen deficiency in long-term intrathecal opioid administration in chronic nonmalignant pain. STUDY DESIGN: Case series. Sixteen consecutive patients with intrathecal drug delivery with opioids were screened for androgen deficiency. SETTING: Academic university-based pain management center. METHOD: All the subjects were seen in a 2 month period, during a scheduled maintenance refill visit. Eight consecutive men and eight consecutive women receiving intrathecal drug delivery therapy for non-malignant chronic pain were ordered blood work and asked to complete a questionnaire. Patient and patient-related data were also collected. RESULTS: Ten of the 16 (62.5%) patients were found to have androgen deficiency, 4 of 8 men based on free testosterone levels and 6 of 8 women based on DHEA levels. In men, erectile dysfunction correlated with endocrine dysfunction (P = 0.02) while depressive symptoms correlated in women (P = .03). Overall, 2 of the 16 patients had hydromorphone as the opioid in the intrathecal system. Both patients had normal endocrine functions. Both patients with hydromorphone were men and the use of hydromorphone showed an insignificant trend (P = 0.06). Three of the 4 men with normal endocrine functions had in addition to an opioid, bupivacaine, in the intrathecal system. The presence of bupivicaine in men was significant (P = 0.02). No women had bupivicaine while one of the 8 women had clonidine in addition to the opioid. Presence of another substance in addition to the opioid showed an insignificant trend (P = 0.08). LIMITATIONS: Study limitations include the small sample size and case series nature. Additionally the symptoms data was solely based on subjective patient reports. CONCLUSIONS: Androgen deficiency is common in patients treated with intrathecal opioids for chronic nonmalignant pain. Patients experience numerous and wide ranging symptoms. Erectile dysfunction may be more suggestive for androgen deficiency in men while complaints of depressed mood may be correlative in women. Additionally, combining bupivicaine with the intrathecal opioid may provide a protective role.


Subject(s)
Analgesics, Opioid/administration & dosage , Chronic Pain/drug therapy , Dehydroepiandrosterone/deficiency , Testosterone/deficiency , Aged , Aged, 80 and over , Analgesics, Opioid/adverse effects , Androgens/blood , Androgens/deficiency , Dehydroepiandrosterone/blood , Female , Humans , Injections, Spinal , Male , Middle Aged , Testosterone/blood
5.
Med Sci Monit ; 18(4): CR225-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22460094

ABSTRACT

BACKGROUND: Cigarette smoking is a major risk factor of atherosclerosis. The aim of this study was to assess the relationship between smoking and arterial hypertension as well as endothelial dysfunction in postmenopausal women without clinically manifested symptoms of atherosclerosis. MATERIAL/METHODS: The study groups consisted of 35 current smokers and 45 nonsmokers. The thickness of intima-media complex (IMT), a marker of atherosclerosis, was measured in carotid arteries. Plasma concentrations of fasting glucose, insulin, lipoproteins, inflammatory markers (tumor necrosis factor-alpha, intercellular adhesion molecule-1), matrix metalloproteinases (metalloproteinase-9, tissue inhibitor of metalloproteinase-1), insulin, and dehydroepiandrosterone sulfate (DHEA-S) were measured. RESULTS: Smokers compared with nonsmokers showed lower fasting glucose levels in blood (87.0±10.9 and 93.2±13.6 mg/dl, p<0.05), higher mean systolic (131.1±15.9 vs. 123.0±10.9 mm Hg, p<0.05) and diastolic (81.7±11.4 vs. 75.2±9.2 mm Hg, p<0.05) blood pressure during daytime, and higher average heart rate during the daytime (78.2±9.3/min vs. 71.5±9.5/min, p<0.01) and at night (67.2±10.6/min vs. 61.7±7.7/min, p<0.05), respectively. The IMT in the right carotid artery was significantly higher in smokers than in nonsmokers (0.96±0.16 mm vs. 0.82±0.21, p<0.05) and was positively correlated with smoking intensity (R=0.36) and habit duration (R=0.35). The comparison of inflammatory markers, metalloproteinases, and DHEA-S concentrations in plasma did not reveal significant differences between the 2 groups. A significant negative correlation between DHEA-S concentration in plasma and IMT in right carotid artery was found in smokers. CONCLUSIONS: Smoking in hypertensive postmenopausal women is associated with lower fasting blood glucose and BMI values, but higher arterial pressure and heart rate, and increases in IMT in right carotid artery.


Subject(s)
Carotid Intima-Media Thickness , Dehydroepiandrosterone/deficiency , Endothelium, Vascular/physiopathology , Hypertension/physiopathology , Metabolic Syndrome/physiopathology , Postmenopause/physiology , Smoking/adverse effects , Anthropometry , Biomarkers/blood , Blood Pressure/physiology , Dehydroepiandrosterone/blood , Diastole/physiology , Female , Heart Rate/physiology , Humans , Hypertension/blood , Hypertension/complications , Inflammation Mediators/blood , Matrix Metalloproteinases/blood , Metabolic Syndrome/blood , Metabolic Syndrome/complications , Middle Aged , Postmenopause/blood , Systole/physiology
6.
Am J Cardiol ; 108(6): 862-6, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21752346

ABSTRACT

The purpose of this study was to establish the role of hormonal anabolic deficiencies in exercise intolerance in patients with chronic heart failure One hundred four consecutive men (mean age 53.1 ± 10.6 years) with established diagnoses of chronic heart failure were included. At enrollment, blood samples were taken, and echocardiography and cardiopulmonary exercise testing were carried out. Exercise capacity was expressed as peak oxygen consumption (Vo2), predicted peak Vo2, and the ventilatory response to exercise (VE/Vco2) slope. The mean left ventricular ejection fraction was 29.7 ± 11.9%, and most patients (86%) were in New York Heart Association class I or II, with a mean peak Vo2 of 18 ml/min/kg. According to the age-adjusted reference values, hormonal deficiencies were present in 29% for total testosterone, 39% for estimated free testosterone, 34% for insulin-like growth factor-1, and 61% for dehydroepiandrosterone sulfate. Dehydroepiandrosterone sulfate showed a significant correlation with peak Vo2 (r = 0.29, p = 0.007), predicted peak Vo2 (r = 0.28, p = 0.006), and VE/Vco2 slope (r = -0.39, p <0.001), whereas total testosterone, estimated free testosterone, and insulin-like growth factor-1 were not significantly correlated. After adjusting in a multivariable model, dehydroepiandrosterone sulfate remained an independent predictor of each exercise parameter. In conclusion, in a cohort of patients with mild chronic heart failure, exercise capacity objectively measured using cardiopulmonary exercise testing was related to anabolic impairment of the adrenal rather than the somatotropic or peripheral axis.


Subject(s)
Dehydroepiandrosterone/deficiency , Exercise Test , Heart Failure/metabolism , Heart Failure/physiopathology , Insulin-Like Growth Factor I/deficiency , Testosterone/deficiency , Chronic Disease , Echocardiography , Electrocardiography , Exercise Tolerance , Humans , Male , Middle Aged , Multivariate Analysis , Oxygen Consumption , Predictive Value of Tests , Prognosis , Respiratory Function Tests
7.
Eur J Heart Fail ; 12(9): 966-73, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20595194

ABSTRACT

AIMS: Elderly men with androgen deficiencies are prone to develop late-onset depression. We investigated links between circulating androgens and depression, and their combined impact on outcome in men with chronic heart failure (CHF). METHODS AND RESULTS: Serum total testosterone (TT) and dehydroepiandrosterone sulphate (DHEAS) were measured using immunoassays in 163 men with stable systolic CHF [age: 60 +/- 10 years, NYHA class (I/II/III/IV): 27/84/46/6] and 316 healthy men. Depression was assessed using Beck Depression Inventory (BDI) and defined as BDI > or =16 points. In men with CHF, reduced TT and DHEAS, advanced NYHA class, elevated N-terminal pro-B type natriuretic peptide (NT-proBNP), reduced glomerular filtration rate, and reduced haemoglobin independently predicted severity of depressive symptoms (all P < 0.05). Depression was present in 20, 37 and 77% of men with no androgen deficiency, either TT or DHEAS deficiency, and both androgen deficiencies, respectively (P < 0.0001). During follow-up (median: 28 months), there were 87 (53%) cardiovascular deaths or unplanned hospitalizations. TT and DHEAS deficiencies (defined as < or = the 10th percentile of serum androgen levels in healthy controls) and BDI > or =16 points independently predicted unfavourable outcome (all P < 0.05). CONCLUSION: TT and DHEAS deficiencies predict severity of depression in men with CHF. Depression and combined androgen deficiencies are independently related to poor outcome in these patients.


Subject(s)
Dehydroepiandrosterone/deficiency , Depression/blood , Heart Failure, Systolic/blood , Testosterone/deficiency , Adult , Biomarkers/blood , Dehydroepiandrosterone/blood , Depression/complications , Disease Progression , Follow-Up Studies , Heart Failure, Systolic/complications , Heart Failure, Systolic/mortality , Humans , Immunoassay , Male , Middle Aged , Poland/epidemiology , Prognosis , Retrospective Studies , Risk Factors , Survival Rate/trends , Testosterone/blood
8.
Climacteric ; 13(4): 314-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20540592

ABSTRACT

The marked age-related decline in serum dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) has suggested that a deficiency of these steroids may be causally related to the development of a series of diseases that are generally associated with aging. Postulated consequences of low DHEA levels include insulin resistance, obesity, cardiovascular disease, cancer, reduction of the immune defence system as well as psychosocial problems such as depression and a general deterioration in the sensation of well-being and cognitive function. Clinically, the spectrum of women that would benefit from DHEA therapy is not clearly defined and nor is the dosage of hormone treatment. Whether DHEA therapy could be prescribed as a general anti-aging therapy or could be an alternative treatment for women suffering from androgen deficiency syndrome remains uncertain across studies. The lack of definitive evidence for biological mechanisms and the presence of only a few studies that address these emerging issues of DHEA therapy in postmenopausal women might encourage a new critical analysis of the available literature, evidencing current limits and incongruities.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Hormone Replacement Therapy , Postmenopause , Aging/physiology , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/physiology , Dehydroepiandrosterone Sulfate/blood , Female , Hormone Replacement Therapy/methods , Humans , Postmenopause/blood , Sexual Behavior/physiology
9.
Curr Opin Endocrinol Diabetes Obes ; 17(3): 217-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20375886

ABSTRACT

PURPOSE OF REVIEW: Adrenal insufficiency, first codified in 1855 by Thomas Addison, remains relevant in 2010 because of its lethal nature. RECENT FINDINGS: Reports illuminate features of adrenal insufficiency cause, diagnosis and treatment, and the role of glucocorticoids in critical illness. SUMMARY: Progress has been made in identifying human leukocyte antigen and major histocompatability complex alleles that predispose to the development of adrenal insufficiency in patients with antibodies to 21-hydroxylase, but their role in clinical care is not established. Reports of HIV-associated infections and medication-induced hypocortisolism are reminders that autoimmune adrenal destruction does not underlie all cases. The diagnosis is adequately established by the 250 microg adrenocortocotropin hormone stimulation test in most patients; the 1 microg test carries the risk of misdiagnosis of healthy individuals as adrenally insufficient. Glucocorticoids provide life-saving treatment, but long-term quality of life is impaired, perhaps because therapy is not given in a physiologic way. The current recommended total daily dose is lower than that often prescribed. Dehydroepiandrosterone replacement may be useful in pubertal girls with hypopituitarism, but not in adults. Supraphysiologic hydrocortisone doses may aid in the reversal of septic shock independent of underlying adrenal function.


Subject(s)
Adrenal Insufficiency , Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/drug therapy , Adrenal Insufficiency/etiology , Adrenocorticotropic Hormone/metabolism , Dehydroepiandrosterone/deficiency , Glucocorticoids/therapeutic use , Humans , Hydrocortisone/metabolism , Quality of Life
10.
Arthritis Care Res (Hoboken) ; 62(1): 118-24, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20191499

ABSTRACT

OBJECTIVE: Primary Sjögren's syndrome (SS) is characterized by fatigue and low levels of serum dehydroepiandrosterone/dehydroepiandrosterone sulfate (DHEA/DHEAS). Our aim was to study whether SS patients with severe fatigue and low serum DHEAS values benefit from DHEA substitution (50 mg/day). METHODS: A multicenter, investigator-based, powered, randomized controlled clinical trial (crossover, washout design) using fatigue as the primary outcome measure was performed on patients with primary SS (n = 107) who had a general fatigue score > or =14 on the 20-item Multiple Fatigue Inventory (MFI-20), combined with age- and sex-adjusted serum DHEAS values below the mean. Fatigue was assessed using MFI-20 subscales, i.e., general fatigue, physical fatigue, mental fatigue, reduced motivation, and activity (scale 4-20), and with a visual analog scale (VAS; scale 0-100). RESULTS: In an intent-to-treat analysis, a 50-mg DHEA substitution dose and placebo similarly improved fatigue. All of the MFI-20 subscales and the fatigue VAS improved from the baseline levels as a result of treatment (P < 0.001), but with negligible differences between these 2 treatments. The mean between-treatment difference was -0.1 for general fatigue (the primary outcome measure), 0.0 for physical fatigue, 0.0 for mental fatigue, 0.0 for reduced motivation, 0.3 for reduced activity, and 2.2 for the fatigue VAS. None of these differences was statistically significant. CONCLUSION: Similar to earlier results using pharmacologic doses, substitution treatment with 50 mg of DHEA in DHEA-deficient and severely tired primary SS patients does not help against fatigue better than placebo. This may relate to the prohormone nature of DHEA and its recently described defective intracrine tissue-specific conversion to active sex steroids in SS.


Subject(s)
Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/therapeutic use , Fatigue/drug therapy , Fatigue/etiology , Sjogren's Syndrome/complications , Sjogren's Syndrome/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Dehydroepiandrosterone/blood , Fatigue/blood , Female , Humans , Middle Aged , Sjogren's Syndrome/blood , Young Adult
11.
Arch Gerontol Geriatr ; 51(1): e16-8, 2010.
Article in English | MEDLINE | ID: mdl-19665809

ABSTRACT

DHEA is secreted by the adrenal cortex and is also a neurosteroid. Its sulfate (DHEAS) is the most abundant steroid in circulation. The levels of both are seen to decline in concentration with age. Evidence is available for altered levels of DHEA and DHEAS in AD but is limited to relatively few studies assessing small cohorts. This study assessed plasma DHEA and DHEAS levels in AD sufferers (n=72) and compared them to age-matched controls (n=72). Plasma DHEA concentrations were significantly lower in AD patients compared to control (4.24+/-0.4 ng/ml for AD; 3.38+/-0.3 ng/ml for control, p=0.027, Mann-Whitney 1-tailed) and DHEA levels were significantly correlated to DHEAS levels in both control and AD conditions (Spearman's rho correlation coefficient=0.635 in controls and 0.467 in AD, p

Subject(s)
Alzheimer Disease/blood , Dehydroepiandrosterone Sulfate/blood , Dehydroepiandrosterone/deficiency , Aged , Dehydroepiandrosterone/blood , Female , Humans , Male , Plasma
13.
Menopause ; 16(5): 923-31, 2009.
Article in English | MEDLINE | ID: mdl-19424093

ABSTRACT

OBJECTIVE: The objective of this study was to provide evidence that the transformation of DHEA into both androgens and/or estrogens locally in cells of the three layers of the vagina (epithelium, lamina propria, and muscularis) would have effects of greater impact, including effects on sexual function, than only effects on superficial epithelial cells as achieved with estrogens. METHODS: This prospective, randomized, double-blind, and placebo-controlled phase III clinical trial has evaluated the effect of daily local intravaginal application of Prasterone (dehydroepiandrosterone; DHEA) for 12 weeks on the domains of sexual dysfunction, namely, desire/interest, arousal, orgasm, and pain at sexual activity, in 216 postmenopausal women with moderate to severe symptoms of vaginal atrophy. RESULTS: A time- and dose-dependent improvement of the four domains of sexual function was observed. At the 12-week time interval, the 1.0% DHEA dose led, compared with placebo, to 49% (P = 0.0061) and 23% (P = 0.0257) improvements of the desire domains in the Menopause Specific Quality of Life and Abbreviated Sex Function questionnaires, respectively. Compared with placebo, the Abbreviated Sex Function arousal/sensation domain was improved by 68% (P = 0.006), the arousal/lubrication domain by 39% (P = 0.0014), orgasm by 75% (P = 0.047), and dryness during intercourse by 57% (P = 0.0001). CONCLUSIONS: By a local action in the vagina, DHEA applied daily at doses at which serum steroids remain well within normal postmenopausal values exerts relatively potent beneficial effects on all four aspects of sexual dysfunction. Such data indicate that combined androgenic/estrogenic stimulation in the three layers of the vagina exerts important beneficial effects on sexual function in women without systemic action on the brain and other extravaginal tissues.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Libido/drug effects , Postmenopause/drug effects , Sexual Dysfunction, Physiological/drug therapy , Sexual Dysfunctions, Psychological/drug therapy , Vagina/drug effects , Administration, Intravaginal , Adult , Aged , Atrophy , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Libido/physiology , Middle Aged , Postmenopause/physiology , Postmenopause/psychology , Sexual Dysfunction, Physiological/etiology , Sexual Dysfunction, Physiological/psychology , Sexual Dysfunctions, Psychological/etiology , Sexual Dysfunctions, Psychological/psychology , Surveys and Questionnaires , Vagina/pathology
14.
Menopause ; 16(5): 897-906, 2009.
Article in English | MEDLINE | ID: mdl-19436226

ABSTRACT

OBJECTIVE: Because a previous 1-week study has shown no or minimal changes in the serum levels of dehydroepiandrosterone (DHEA) and its metabolites after up to daily 1.8% (23.4 mg) intravaginal DHEA, the objective of the present study was to investigate the serum steroid levels during a 12-week daily intravaginal administration of 0%, 0.25%, 0.5%, and 1.0% DHEA (Prasterone) 1.3 mL ovules. METHODS: In a double-blind, placebo-controlled phase III study, 218 postmenopausal women (age range, 42-74 y) were randomized to receive daily one of four DHEA concentrations intravaginally. Serum steroids were measured by a Good Laboratory Practice-validated mass spectrometry technology in samples obtained at time of visit. RESULTS: The serum levels of DHEA and 11 of its metabolites measured at screening, day 1, and weeks 2, 4, 8, and 12 in women showed no or minimal changes during the whole observation period, with all values remaining well within the limits of normal postmenopausal women. No accumulation of the steroid metabolites nor change in DHEA bioavailability was detected. CONCLUSIONS: The present data show that local daily intravaginal DHEA administration at DHEA doses of 3.25-13 mg was able to rapidly and efficiently achieve correction of all the signs and symptoms of vaginal atrophy and improve sexual function and caused no or minimal changes in serum sex steroid levels, which all remain within the normal postmenopausal range, thus avoiding the risks of all estrogen formulations.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Estradiol/blood , Hormone Replacement Therapy/methods , Postmenopause , Sexual Dysfunction, Physiological/drug therapy , Vagina/drug effects , Administration, Intravaginal , Aged , Atrophy , Biological Availability , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Drug Monitoring/methods , Female , Humans , Mass Spectrometry , Middle Aged , Postmenopause/drug effects , Postmenopause/physiology , Prospective Studies , Sexual Dysfunction, Physiological/etiology , Time Factors , Vagina/pathology
15.
Menopause ; 16(5): 907-22, 2009.
Article in English | MEDLINE | ID: mdl-19436225

ABSTRACT

OBJECTIVE: Because the secretion of dehydroepiandrosterone (DHEA), the exclusive source of sex steroids in postmenopausal women, is already decreased by 60% and continues to decline at the time of menopause, the objective of this study was to examine the effect of intravaginal DHEA on the symptoms and signs of vaginal atrophy. METHODS: This prospective, randomized, double-blind and placebo-controlled phase III clinical trial studied the effect of Prasterone (DHEA) applied locally in the vagina on the signs and symptoms of vaginal atrophy in 216 postmenopausal women. RESULTS: All three doses (0.25%, 0.5%, and 1.0%) of DHEA ovules applied daily intravaginally induced a highly significant beneficial change in the percentage of vaginal parabasal and superficial cells and pH as well as in the most bothersome symptom at 2 weeks. At the standard 12-week time interval, 0.5% DHEA caused a 45.9 +/- 5.31 (P < 0.0001 vs placebo) decrease in the percentage of parabasal cells, a 6.8 +/- 1.29% (P < 0.0001) increase in superficial cells, a 1.3 +/- 0.13 unit (P < 0.0001) decrease in vaginal pH, and a 1.5 +/- 0.14 score unit (P < 0.0001) decrease in the severity of the most bothersome symptom. Similar changes were seen on vaginal secretions, color, epithelial surface thickness, and epithelial integrity. Comparable effects were observed at the 0.25% and 1.0% DHEA doses. CONCLUSIONS: Local Prasterone, through local androgen and estrogen formation, causes a rapid and efficient reversal of all the symptoms and signs of vaginal atrophy with no or minimal changes in serum steroids, which remain well within the normal postmenopausal range. This approach avoids the fear of systemic effects common to all presently available estrogen formulations and adds a novel physiological androgenic component to therapy.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/deficiency , Hormone Replacement Therapy/methods , Postmenopause/drug effects , Vagina , Administration, Intravaginal , Adult , Analysis of Variance , Atrophy , Dehydroepiandrosterone/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hydrogen-Ion Concentration , Middle Aged , Postmenopause/physiology , Prospective Studies , Severity of Illness Index , Treatment Outcome , Vagina/drug effects , Vagina/pathology , Vaginal Smears
16.
J Rheumatol ; 35(11): 2229-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18843777

ABSTRACT

OBJECTIVE: .We hypothesized that in addition to dehydroepiandrosterone (DHEA) depletion, Sjögren's syndrome (SS) is characterized by local androgen deficiency in salivary glands and defects in local processing of DHEA. METHODS: Sex steroid levels in serum and saliva were measured using enzyme immunoassays. Androgen effects on salivary gland cells were analyzed using the cysteine-rich secretory protein-3 (CRISP-3) androgen biomarker. RESULTS: Serum and salivary concentrations of androgens were low in SS. Substrate to end-product ratios and correlations suggest that in SS salivary glands DHEA is effectively converted to testosterone, but that there are defects in converting testosterone further to dihydrotestosterone (DHT). In healthy controls no such phenomenon was seen, but testosterone is effectively converted to DHT. Salivary glands contained type I 5-alpha-reductase, and its inhibition with dutasteride completely blocked the upregulating effect of DHEA, but not of DHT, on CRISP-3 in human salivary gland acinar cells. CONCLUSION: DHEA and DHT upregulate CRISP-3, which is reportedly low in SS. The effect of DHEA on CRISP-3 is indirect and is inhibited by dutasteride, showing that there is intracrine processing of DHEA in salivary glands. In healthy glands, but not in SS, DHEA is effectively taken up and converted to DHT. Sex steroid concentrations in saliva in part reflect glandular uptake of DHEA-sulfate and local intracrine DHEA metabolism, which seem to be defective in SS. Our study demonstrates a prominent androgen deficiency and a defect in intracrine production of active androgens in SS salivary glands, also suggesting that salivary DHT cannot be maintained at a normal level in this female-dominant autoimmune exocrinopathy.


Subject(s)
Androgens/deficiency , Dehydroepiandrosterone/deficiency , Salivary Glands/metabolism , Sjogren's Syndrome/metabolism , Testosterone/deficiency , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , 5-alpha Reductase Inhibitors , Adult , Aged , Androgens/blood , Biopsy , Dehydroepiandrosterone/blood , Dehydroepiandrosterone Sulfate/blood , Fatigue/metabolism , Female , Humans , Middle Aged , RNA, Messenger/metabolism , Saliva/metabolism , Salivary Glands/cytology , Salivary Proteins and Peptides/metabolism , Seminal Plasma Proteins/metabolism , Testosterone/blood
18.
Hepatology ; 47(2): 484-92, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18220286

ABSTRACT

UNLABELLED: The biological basis of variability in histological progression of nonalcoholic fatty liver disease (NAFLD) is unknown. Dehydroepiandrosterone (DHEA) is the most abundant steroid hormone and has been shown to influence sensitivity to oxidative stress, insulin sensitivity, and expression of peroxisome proliferator-activated receptor alpha and procollagen messenger RNA. Our aim was to determine whether more histologically advanced NAFLD is associated with low circulating levels of DHEA. Serum samples were obtained prospectively at the time of liver biopsy in 439 patients with NAFLD (78 in an initial and 361 in validation cohorts) and in controls with cholestatic liver disease (n = 44). NAFLD was characterized as mild [simple steatosis or nonalcoholic steatohepatitis (NASH) with fibrosis stage 0-2] or advanced (NASH with fibrosis stage 3-4). Serum levels of sulfated DHEA (DHEA-S) were measured by enzyme-linked immunosorbent assay. Patients with advanced NAFLD had lower plasma levels of DHEA-S than patients with mild NAFLD in both the initial (0.25 +/- 0.07 versus 1.1 +/- 0.09 microg/mL, P < 0.001) and validation cohorts (0.47 +/- 0.06 versus 0.99 +/- 0.04 microg/mL, P < 0.001). A "dose effect" of decreasing DHEA-S and incremental fibrosis stage was observed with a mean DHEA-S of 1.03 +/- 0.05, 0.96 +/- 0.07, 0.83 +/- 0.11, 0.66 +/- 0.11, and 0.35 +/- 0.06 microg/mL for fibrosis stages 0, 1, 2, 3, and 4, respectively. All patients in both cohorts in the advanced NAFLD group had low DHEA-S levels, with the majority in the hypoadrenal range. The association between DHEA-S and severity of NAFLD persisted after adjusting for age. A relationship between disease/fibrosis severity and DHEA-S levels was not seen in patients with cholestatic liver diseases. CONCLUSION: More advanced NAFLD, as indicated by the presence of NASH with advanced fibrosis stage, is strongly associated with low circulating DHEA-S. These data provide novel evidence for relative DHEA-S deficiency in patients with histologically advanced NASH.


Subject(s)
Dehydroepiandrosterone/blood , Fatty Liver/blood , Fatty Liver/pathology , Adiponectin/blood , Adult , Biomarkers/blood , Biopsy , Blood Glucose/analysis , C-Reactive Protein/analysis , Cohort Studies , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone Sulfate/blood , Female , Humans , Leptin/blood , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Middle Aged , Reference Values , Resistin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...