Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 121: 109597, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31726369

ABSTRACT

In the present study, the therapeutic effects of 5-androstenediol on radiation-induced myeloid suppression and tissue damage in mice and the possible mechanism were explored. The mice were subjected to whole-body irradiation, and 5-androstenediol was administered subcutaneously at different times and doses. The evaluation of the survival rate showed that the administration of 5-androstenediol every three days post-irradiation was the most effective in decreasing the death of the mice. Additionally, 5-androstenediol dose-dependently reduced the death caused by 9 Gy radiation. The pharmacological mechanism was investigated by blood analysis, western blot analysis, immunofluorescence and immunohistochemistry. 5-Androstenediol significantly ameliorated myeloid suppression, as demonstrated by elevated levels of total white blood cells, including neutrophils and platelets, in the peripheral blood. By H&E staining, we found that radiation-induced myeloid suppression in the bone marrow and spleen, as well as tissue damage in the lung and colon, was significantly ameliorated by treatment with 5-androstenediol. Immunohistochemistry showed elevated phosphorylation of p65 in the bone marrow and spleen, indicating the activation of NF-κB signaling. Moreover, 5-androstenediol markedly hampered the radiation-induced activation of caspase-1 and GSDMD in the colon by decreasing the interaction between AIM2 and ASC. Taken together, our results suggest that, by promoting NF-κB signaling and inhibiting inflammasome-mediated pyroptosis, 5-androstenediol can be used as a radioprotective drug.


Subject(s)
Androstenediol/therapeutic use , DNA-Binding Proteins/metabolism , NF-kappa B/metabolism , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Radiation-Protective Agents/therapeutic use , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Androstenediol/pharmacology , Animals , DNA-Binding Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Radiation Injuries/pathology , Radiation-Protective Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
2.
Am J Mens Health ; 10(6): NP109-NP117, 2016 11.
Article in English | MEDLINE | ID: mdl-26272885

ABSTRACT

An increasing number of men are being diagnosed with hypogonadism. While many benefit from testosterone supplementation therapy, others who do not meet the criteria for hormone supplementation have turned to dietary adjuncts as a way or gaining improvements in libido, energy, and physical performance. These oral adjunct medications include controlled substances such as androstenedione, androstenediol as well as other "over-the-counter" options like DHEA (dehydroepiandrosterone) and herbal remedies like Tribulus terrestris This review will focus on the use of these adjunct medications in isolation, or in combination with testosterone supplementation therapy as well as the biochemical nature of the supplements, the results of scientific trials as well as the side effects that limit their use. At the end of this review, physicians will have an improved understanding of the popular testosterone adjuncts being used currently as well as the availability of these substances and how they are used.


Subject(s)
Androstenediol/therapeutic use , Androstenedione/therapeutic use , Dehydroepiandrosterone/therapeutic use , Dietary Supplements , Hypogonadism/drug therapy , Testosterone/blood , Hormone Replacement Therapy , Humans , Male , Quality of Life , Time Factors
3.
Neuropathol Appl Neurobiol ; 41(7): 964-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25786683

ABSTRACT

AIMS: Experimental evidence has shown that the adrenal steroid hormone, androstenediol, dampens the symptoms of demyelination. However, the cellular and molecular effects of androstenediol are not yet known. In the present study, we investigated the cellular and subcellular effects of this hormone in a gliotoxin-induced demyelination model. METHODS: Male Sprague Dawley rats received 2 µl of either saline or the gliotoxin ethidium bromide (EB, 0.04%) into the corpus callosum. These rats received daily subcutaneous injections of either oil or androstenediol (5 mg/kg). Their brains were collected at 2, 7, 14 and 28 days post-EB injection. Demyelinated lesions were assessed using Luxol fast blue staining. Immunofluorescent staining was used to investigate the number of oligodendrocyte progenitor cells, their maturation and microglial activation at the lesion site. Remyelination was further explored using transmission electron microscopy. The expression levels of total and phosphorylated MBP isoforms and CNPase were explored using western blot. RESULTS: Androstenediol decreased the size of demyelinated lesions in the corpus callosum at 7 and 14 days post-EB injection. It enhanced the number of oligodendrocyte precursor cells, promoted an increase in the number of mature oligodendrocytes and reduced microglial activation. Androstenediol also stimulated the phosphorylation of MBP at the site of the lesion and promoted remyelination of the affected axons. CONCLUSIONS: These data strongly suggest that androstenediol is endowed with promyelinating properties in a model of focal gliotoxin-induced demyelination. It induces its promyelinating effects by enhancing the number of oligodendrocyte precursor cells and their maturation at the lesion site.


Subject(s)
Androstenediol/therapeutic use , Corpus Callosum/drug effects , Demyelinating Diseases/drug therapy , Myelin Sheath/drug effects , Androstenediol/pharmacology , Animals , Corpus Callosum/metabolism , Corpus Callosum/pathology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Gliotoxin , Male , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
4.
Article in English | MEDLINE | ID: mdl-21158733

ABSTRACT

Due to improved treatment strategies mortality in multiple trauma patients has been decreased over the last decades. However, posttraumatic complications like sepsis and subsequent multiple organ dysfunction syndrome (MODS) remain a major problem on intensive care units following major trauma. The clinical course after multiple trauma depends on the balance or imbalance of the pro- and anti-inflammatory immune response. The predominance of the proinflammatory response leads to the "Systemic Inflammatory Response Syndrome" (SIRS), whereas the "Compensatory Anti-inflammatory Response Syndrome" (CARS) might result in immune suppression with an enhanced risk for infectious complications. Both, SIRS and CARS, play a pivotal role in the development of sepsis and the "Multiple Organ Dysfunction Syndrome" (MODS). A gender dimorphism in the host response after multiple trauma and sepsis has already been described. In experimental as well as clinical studies, a protective effect of female sex hormones and precursors like androstenediol has been revealed. Moreover, blockade of androgen receptors and the inhibition of dihydrotestosterone (DHT) synthesis were shown to provide beneficial effects on the immune response. Beside sex hormones, modulation of the Toll Like Receptor (TLR) pathway by macrophage-activating lipopeptide-2 (MALP-2) has sufficiently been described. Furthermore, hydrogen sulfide (H2S) and substance P have recently been revealed important for proinflammatory action in animal models of inflammation. Thus, these agents might be potential candidates for new treatment strategies in septic patients in order to improve the still unsatisfactory outcome of multiple trauma patients. If applicable, patents of each described agent are provided within the text.


Subject(s)
Immunomodulation , Multiple Trauma/drug therapy , Sepsis/drug therapy , Androstenediol/therapeutic use , Animals , Dehydroepiandrosterone/therapeutic use , Finasteride/therapeutic use , Flutamide/therapeutic use , Humans , Lipopeptides/therapeutic use , Multiple Trauma/immunology , Sepsis/immunology
5.
J Radiol Prot ; 30(4): 687-98, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21149931

ABSTRACT

5-androstenediol (5-AED) has been advanced as a possible countermeasure for treating the haematological component of acute radiation syndrome (ARS). It has been used in animal models to stimulate both innate and adaptive immunity and treat infection and radiation-induced immune suppression. We here report on the safety, tolerability and haematologic activity of 5-AED in four double-blinded, randomized, placebo-controlled studies on healthy adults including elderly subjects. A 5-AED injectable suspension formulation (NEUMUNE) or placebo was administered intramuscularly as either a single injection, or once daily for five consecutive days at doses of 50, 100, 200 or 400 mg. Subjects (n = 129) were randomized to receive NEUMUNE (n = 95) or the placebo (n = 34). NEUMUNE was generally well-tolerated; the most frequent adverse events were local injection site reactions (n = 104, 81%) that were transient, dose-volume dependent, mild to moderate in severity, and that resolved over the course of the study. Blood chemistries revealed a transient increase (up to 28%) in creatine phosphokinase and C-reactive protein levels consistent with intramuscular injection and injection site irritation. The blood concentration profile of 5-AED is consistent with a depot formulation that increases in disproportionate increments following each dose. NEUMUNE significantly increased circulating neutrophils (p < 0.001) and platelets (p < 0.001) in the peripheral blood of adult and elderly subjects. A dose-response relationship was identified. Findings suggest that parenteral administration of 5-AED in aqueous suspension may be a safe and effective means to stimulate innate immunity and alleviate neutropenia and thrombocytopenia associated with ARS.


Subject(s)
Acute Radiation Syndrome/drug therapy , Androstenediol/therapeutic use , Adult , Aged , Androstenediol/administration & dosage , Androstenediol/adverse effects , Androstenediol/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged
6.
Ann N Y Acad Sci ; 1051: 730-42, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16127013

ABSTRACT

5-Androsten-3beta, 17beta-diol (HE2100), and a synthetic analogue HE3204 are regarded as immune-regulating hormones, because both induce changes in the reporter antigen-popliteal lymph node assay (RA-PLNA). Mice were injected in the footpad with either HE2100 or HE3204 (0.01-3 mg), and a nonsensitizing dose of trinitrophenyl ovalbumin (TNP-OVA) was used as bystander reporter antigen. Seven days later, nodes were removed and numbers of cells (CD3, CD4, CD8, CD19; flow cytometry), TNP-specific IgM, IgG1, and IgG2a antibody-forming cells (AFCs; ELISPOT assay), and cytokines (interleukin-4 [IL-4], interferon-gamma [IFN-gamma]; ELISA) were measured. HE2100 and HE3204 increased cell numbers in a dose-dependent fashion. T (helper and suppressor) cells and B cells were increased (>5-fold). HE3204 was apparently twice as potent as HE2100. Both increased the B/T ratio (fivefold), increased TNP-specific IgM and IgG1 ( approximately 50-fold), and induced IgG2a AFCs. Both increased IL-4 and IFN-gamma secretion (up to threefold). Both displayed anti-inflammatory activity in the murine model of carrageenan-induced pleurisy, as evidenced by reduced neutrophil numbers and exudate volumes. Our observations suggest that both HE2100 and HE3204 are immune-regulating steroid hormones that exhibit anti-inflammatory properties. HE2100 (1 mg/mouse per day) provided significant benefit when given at disease onset in the SJL/J female mouse model of experimental autoimmune encephalomyelitis. These compounds and their analogues are candidates for further testing in autoimmune diseases.


Subject(s)
Androstenediol/therapeutic use , Androstenols/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Amino Acid Sequence , Androstenediol/analogs & derivatives , Androstenediol/pharmacology , Androstenols/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pleurisy/drug therapy , Shock, Septic/drug therapy
7.
Inflamm Res ; 53(5): 189-98, 2004 May.
Article in English | MEDLINE | ID: mdl-15105968

ABSTRACT

OBJECTIVE AND DESIGN: Sex hormones have immunomodulatory properties and may play an important role in the pathogenesis of autoimmune diseases like rheumatoid arthritis (RA). This study sought to examine the effects of the natural weak androgen dehydroepiandrosterone (DHEA) and its metabolite androstenediol (AED) on the development of murine antigen-induced arthritis (AIA). METHODS: DHEA and AED were administered orally, approximately 10 mg/day, from the time of AIA induction (i.e., 3 weeks after start of immunization) in young male as well as young and old female C57BL/6 mice. The effects were assessed in terms of joint swelling, histological changes, and cell-mediated and humoral immunity. RESULTS: Compared to untreated AIA animals, continuous administration of DHEA decreased knee joint swelling during acute and chronic AIA, as well as histological signs of inflammation and joint destruction during chronic AIA. These effects were age- and gender-independent. Delayed-type hypersensitivity (DTH) to the specific antigen methylated bovine serum albumin (mBSA) was significantly reduced, but there were no changes in the balance of the T helper (Th) cell subsets Th1/Th2, as tested by the ratio of IgG isotypes in the sera. Whereas serum levels of IgG antibodies to mBSA were not influenced, the formation of IgG autoantibodies to the matrix constituents collagen type I, collagen type II, and cartilage proteoglycans was significantly inhibited. In all experiments, the effects of AED were not significantly stronger than those of DHEA. CONCLUSIONS: Administration of exogenous DHEA ameliorates the severity of acute and chronic AIA, presumably by suppressing cell-mediated immunity against mBSA (the inducing antigen) and formation of autoantibodies. However, because of the fundamentally different DHEA physiology in rodents, the role of such a replacement therapy in human RA deserves further elucidation.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Androstenediol/therapeutic use , Antibodies, Anti-Idiotypic/immunology , Arthritis, Experimental/drug therapy , Dehydroepiandrosterone/therapeutic use , Animals , Arthritis, Experimental/immunology , Collagen Type I/immunology , Collagen Type II/immunology , Female , Hypersensitivity, Delayed/etiology , Hypersensitivity, Delayed/prevention & control , Immunity, Cellular/drug effects , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Male , Mice , Mice, Inbred C57BL , Proteoglycans/immunology , Serum Albumin, Bovine , Severity of Illness Index
8.
Biol Pharm Bull ; 25(12): 1634-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12499654

ABSTRACT

We examined the serum concentrations of delta(5)-3beta-hydroxysteroids, pregnenolone (Preg), 17-hydroxypregnenolone (17-OH-Preg), dehydroepiandrosterone (DHEA), androstenediol (ADIOL) and their sulfates in 30 well controlled (Group I: HbA1c<7.0%) and 15 poorly controlled (Group II: HbA1c>7.1%) type 2 diabetic patients, and 30 normal controls. These patients were treated with diet therapy or anti-diabetic agent. The distribution of gender and age of the subjects were matched between the groups. The serum levels of sulfo-conjugated and unconjugated steroids described above were measured by GC-MS and enzyme immunoassay (EIA), respectively. The serum levels of the entire sulfo-conjugated steroid measured in this study were significantly lower in Groups I and II than in controls. On the other hand, Preg levels in both Groups I and II were significantly higher than those in controls, whereas the serum levels of the downstream unconjugated steroids were not different from controls. To investigate the effect of sulfonylurea (SU) on the serum levels of steroids, the serum concentrations of steroids between the patients who were treated with diet therapy and SU agent were compared in Group I. No significant differences were observed between both groups. These results suggest that (1) since increased Preg levels did not cause any changes in the downstream delta(5)-3beta-hydroxysteroid levels, the metabolic pathway of delta(4)-3-ketosteroids may be accelerated in type 2 diabetes; (2) serum steroid levels were not affected by SU treatment; (3) sulfo-conjugated steroid catabolism was altered in type 2 diabetes; (4) the decreased sulfo-conjugated steroids especially ADIOLS may contribute to the alteration of sex steroid levels and onset or exacerbate infectious diseases in diabetes.


Subject(s)
17-alpha-Hydroxypregnenolone/blood , Androstenediol/blood , Dehydroepiandrosterone/blood , Diabetes Mellitus, Type 2/blood , Pregnenolone/blood , 17-alpha-Hydroxypregnenolone/therapeutic use , Adult , Androstenediol/therapeutic use , Dehydroepiandrosterone/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Female , Humans , Male , Middle Aged , Pregnenolone/therapeutic use , Statistics, Nonparametric
11.
Arch Intern Med ; 160(20): 3093-104, 2000 Nov 13.
Article in English | MEDLINE | ID: mdl-11074738

ABSTRACT

BACKGROUND: Since the passage of The Dietary Supplement Health and Education Act in 1994, there has been a flood of new "dietary" supplements promoting anti-aging benefits such as the enhancement of growth hormone or testosterone levels. Androstenediol and androstenedione are such products. This study's purpose was to elucidate the physiological and hormonal effects of 200 mg/d of oral androstenediol and androstenedione supplementation in men aged 35 to 65 years while participating in a 12-week high-intensity resistance training program. METHODS: Fifty men not consuming any androgenic-enhancing substances and with normal total testosterone levels, prostate-specific antigen, hemoglobin, and hematocrit, and with no sign of cardiovascular or metabolic diseases participated. Subjects were randomly assigned to a placebo, androstenediol (diol), or androstenedione (dione) group using a double-blind study design. Main outcomes included serum sex hormone profile, body composition assessment, muscular strength, and blood lipid profiles. RESULTS: During the 12 weeks of androstenedione or androstenediol use, a significant increase in the aromatization by-products estrone and estradiol was observed in both groups (P =.03). In the dione group, total testosterone levels significantly increased 16% after 1 month of use, but by the end of 12 weeks, they returned to pretreatment levels. This return to baseline levels resulted from increases in aromatization and down-regulation in endogenous testosterone synthesis based on the fact that luteinizing hormone was attenuated 18% to 33% during the treatment period. Neither androstenediol nor androstenedione enhanced the adaptations to resistance training compared with placebo for body composition or muscular strength. However, both androstenediol and androstenedione supplementation adversely affected high-density lipoprotein cholesterol (HDL-C) levels, coronary heart disease risk (representing a 6.5% increase), and each group's respective (low-density lipoprotein cholesterol [LDL-C]/HDL-C)/(apolipoprotein A/apolipoprotein B) lipid ratio (diol: +5.2%; dione: +10.5%; P =.05). In contrast, the placebo group's HDL-C levels increased 5.1%, with a 12.3% decline in the (LDL-C/HDL-C)/(apolipoprotein A/apolipoprotein B) lipid ratio. These negative and positive lipid effects occurred despite no significant alterations in body composition or dietary intakes in the supplemental groups or placebo group, respectively. CONCLUSIONS: Testosterone precursors do not enhance adaptations to resistance training when consumed in dosages recommended by manufacturers. Testosterone precursor supplementation does result in significant increases in estrogen-related compounds, dehydroepiandrosterone sulfate concentrations, down-regulation in testosterone synthesis, and unfavorable alterations in blood lipid and coronary heart disease risk profiles of men aged 35 to 65 years.


Subject(s)
Androstenediol/therapeutic use , Androstenedione/therapeutic use , Dietary Supplements , Exercise , Adult , Aged , Body Composition , Diet , Estradiol/blood , Humans , Lipids/blood , Male , Middle Aged , Testosterone/blood
12.
J Gerontol A Biol Sci Med Sci ; 55(9): B418-24, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10995038

ABSTRACT

Androstenediol (AED), a metabolite of dehydroepiandrosterone (DHEA) regulates innate and adaptive immune responses. To examine whether AED could effectively reverse the age-associated decline of antiviral immunity, 3-, 10-, and 22-month-old mice were treated with AED-sulfate (AED-S) for 45 days beginning 10 days prior to vaccination. Subsequently, mice were primed and boosted with suboptimal doses of a commercially-available trivalent influenza vaccine. Treatment of 10-month-old animals with AED-S during vaccination increased the titer of circulating antiviral immunoglobulin G to levels comparable with those in 3-month-old mice. Furthermore, AED-S treatment protected 10-month-old animals from intranasal challenge with a lethal dose of influenza virus 21 days after secondary vaccination. Although AED-S treatment of 22-month-old mice did not enhance vaccine responses and failed to protect against lethal challenge, the data from the 10-month-old animals suggest that treatment with AED-S will prevent the early manifestations of immunosenescence.


Subject(s)
Aging/immunology , Anabolic Agents/therapeutic use , Androstenediol/therapeutic use , Immunocompetence/drug effects , Influenza Vaccines , Vaccination , Age Factors , Animals , Chi-Square Distribution , Germ-Free Life , Immunity, Active/drug effects , Immunity, Active/immunology , Immunization, Secondary , Immunoglobulin G/blood , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Time Factors , Treatment Outcome
13.
Immunology ; 100(3): 391-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10929063

ABSTRACT

Problems of logistics, compliance and drug resistance point to an urgent need for immunotherapeutic strategies capable of shortening the current 6-month chemotherapy regimens used to treat tuberculosis, or of supplementing ineffective therapy. In this study we sought to define the mechanism of action of two immunotherapies, both of which have previously been shown to prolong survival. Secondly, we wished to identify any clinically useful synergy between these therapies. In BALB/c mice infected via the trachea with Mycobacterium tuberculosis H37Rv there is an initial phase of partial resistance dominated by type 1 cytokines plus tumour necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1), followed by a phase of progressive disease. This progressive phase is accompanied by increasing expression of IL-4, and diminished expression of IL-1 and TNF-alpha. Animals in this late progressive phase of the disease (day 60) were treated with two injections (day 60 and day 90) of 0.1 or 1.0 mg of heat-killed Mycobacterium vaccae, or with 3beta, 17beta-androstenediol (AED; 25 microg subcutaneously three times/week), or with both therapies. We show here using four techniques in parallel (morphometry, immunohistochemistry with automated cell counting, semiquantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assays of cytokines in lung extracts) that treatment with M. vaccae causes a switch back towards a type 1 cytokine profile, restoration of expression of IL-1alpha and TNF-alpha, and a switch from pneumonia to granuloma. This is very similar to the changes previously seen after treatment with AED. However, there was no evidence for synergy between M. vaccae and AED.


Subject(s)
Androstenediol/therapeutic use , Immunotherapy/methods , Tuberculosis, Pulmonary/therapy , Animals , Antigens, Bacterial/immunology , Colony Count, Microbial , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Hypersensitivity, Delayed/immunology , Immunotherapy, Active , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology
14.
Ann N Y Acad Sci ; 917: 860-7, 2000.
Article in English | MEDLINE | ID: mdl-11268417

ABSTRACT

Androstenetriol (AET) and Androstenediol (AED) upregulate host immunity, leading to increased resistance against infections. AET augments IL-2, IL-3, IFN gamma levels, and counteracts hydrocortisone immune suppression. AET and AED at a dose of 0.75 mg/- and 8.0 mg/25-g mouse, protected 60 and 70%, respectively, of C57/BL/6J mice irradiated with a lethal dose. These hormones also protected mice irradiated with 6 Gy and infected with a coxsackievirus B4 LD50. AET significantly increased spleen lymphocyte numbers at 7, 14, and 21 days after a 6-Gy exposure. Fluorescent activated cell-sorter analysis of irradiated mice, spleen, and bone marrow showed that AET significantly augmented the myeloid precursor markers, CD11b/Mac-1, and B220 (pan B), as well as the absolute numbers of CD4+/CD8+ cells over the 21 days of testing. Overall, the data are consistent with AET/AED inducing a more rapid recovery of all hematopoietic precursors from the small number of surviving stem cells.


Subject(s)
Anabolic Agents/pharmacology , Androstenediol/pharmacology , Immunity/drug effects , Radiation Injuries/immunology , Radiation Injuries/prevention & control , Anabolic Agents/therapeutic use , Androstenediol/immunology , Androstenediol/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation
15.
Horm Res ; 54(5-6): 316-7, 2000.
Article in English | MEDLINE | ID: mdl-11595825

ABSTRACT

We previously reported a patient with generalized vitiligo improved by oral administration of the drug for menopausal syndrome (sex hormone-thyroid powder mixture). In this study, we reevaluated the efficiency of this drug for vitiligo, and examined its pharmacological action in melanogenesis.


Subject(s)
Androstenediol/therapeutic use , Androstenedione/therapeutic use , Estrone/therapeutic use , Pregnenolone/therapeutic use , Testosterone/therapeutic use , Thyroid (USP)/therapeutic use , Vitiligo/drug therapy , Adult , Aged , Androstenediol/adverse effects , Androstenedione/adverse effects , Drug Combinations , Drug Resistance , Estrone/adverse effects , Female , Humans , Male , Melanocytes/metabolism , Middle Aged , Pregnenolone/adverse effects , Sex Characteristics , Skin Pigmentation , Testosterone/adverse effects , Thyroid (USP)/adverse effects , Time Factors , Vitiligo/physiopathology , alpha-MSH/metabolism
16.
J Dermatol ; 26(8): 502-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10487004

ABSTRACT

We previously reported that a sex steroid-thyroid hormone (Metharmon-F; MF, 2 tablets daily) was a potent drug for treatment of vitiligo. Using five patients with generalized vitiligo who were successfully treated with oral administration of MF, we performed an immunohistochemical analysis to elucidate its action mechanism at the cellular level. Histopathologically, the repigmented skin after the treatment showed increased numbers of melanocytes and melanin granules. Immunohistochemically, there was little significant difference between the depigmented lesions before treatment and the repigmented lesion after treatment in terms of the reactivity to adrenocorticotropic hormone (ACTH) and alpha-melanocyte-stimulating hormone (alpha-MSH) in keratinocytes. The immunoreactivity to ACTH in melanocytes both before and after the treatment was minimal, but alpha-MSH in melanocytes became much stronger after the treatment, than before the treatment. The efficacy of MF in treatment of vitiligo was proven to be due to the stimulatory effect of melanocyte proliferation and melanin production via alpha-MSH.


Subject(s)
Adrenocorticotropic Hormone/analysis , Androstenediol/therapeutic use , Androstenedione/therapeutic use , Estrone/therapeutic use , Pregnenolone/therapeutic use , Testosterone/therapeutic use , Thyroid (USP)/therapeutic use , Vitiligo/drug therapy , alpha-MSH/analysis , Adrenocorticotropic Hormone/immunology , Aged , Drug Combinations , Female , Humans , Immunohistochemistry , Keratinocytes/chemistry , Male , Melanocytes/chemistry , Middle Aged , Vitiligo/metabolism , Vitiligo/pathology , alpha-MSH/immunology
17.
J Med Microbiol ; 48(5): 425-431, 1999 May.
Article in English | MEDLINE | ID: mdl-10229539

ABSTRACT

The protective effects of the hormones androstenediol (androstene-3beta, 17beta,-diol; AED) and dehydroepiandrosterone (5-androsten-3beta-ol-17-one; DHEA) on the pathophysiology of two lethal bacterial infections and endotoxin shock were examined. The infections included a gram-positive organism (Enterococcus faecalis) and a gram-negative organism (Pseudomonas aeruginosa). Both hormones protected mice from the lethal bacterial infections and from lipopolysaccharide (LPS) challenge. Treatment of animals lethally infected with P. aeruginosa with DHEA resulted in a 43% protection whereas treatment with AED gave a 67% protection. Both hormones also protected completely animals infected with an LD50 dose of E. faecalis. Similarly, the 88% mortality rate seen in LPS challenge was reduced to 17% and 8.5%, by treatment with DHEA and AED, respectively. The protective influences of both steroids were shown not to be directly antibacterial, but primarily an indirect antitoxin reaction. DHEA appears to mediate its protective effect by a mechanism that blocks the toxin-induced production of pathophysiological levels of tumour necrosis factor-alpha (TNF-alpha) and interleukin-1. AED usually had greater protective effects than DHEA; however, the AED effect was independent of TNF-alpha suppression, both in vivo and in vitro. The data suggest that both DHEA and AED may have a role in the neuro-endocrine regulation of antibacterial immune resistance.


Subject(s)
Androstenediol/therapeutic use , Dehydroepiandrosterone/therapeutic use , Gram-Positive Bacterial Infections/prevention & control , Lipopolysaccharides/toxicity , Pseudomonas Infections/prevention & control , Shock, Septic/prevention & control , Animals , Enterococcus faecalis , Female , Interleukin-1/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Tumor Necrosis Factor-alpha/biosynthesis
18.
Immunology ; 95(2): 234-41, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9824481

ABSTRACT

Immunity to Mycobacterium tuberculosis requires a T helper 1 (Th1) cytokine balance accompanied by tumour necrosis factor-alpha (TNF-alpha), and activated macrophages. These facets of the immune response are sensitive to suppression by glucocorticoids (GC), which can reactivate and exacerbate tuberculosis in man and animals. Dehydroepiandrosterone (DHEA) and its derivative, 3beta,17beta androstenediol (AED), are reported to have antiglucocorticoid properties in vivo. We therefore investigated the effects of predetermined optimal doses of these compounds, on the course of pulmonary tuberculosis in an established model in BALB/c mice in which an early phase of Th1-mediated response accompanied by adrenal hyperplasia, is followed by a switch to Th2, progressive loss of TNF-alpha expression and disease progression. Both compounds were protective, particularly AED which caused a fall in bacterial counts and prolonged survival. These effects correlated with the appearance within 3 days of cellular infiltrates rich in cells expressing interleukin-2 (IL-2), IL-1alpha and TNF-alpha, and with partial suppression of the switch to IL-4 producing cells that occurred in controls. AED also caused enhanced development of granulomas at 14 days, and persistence of granuloma formation to 120 days, with a corresponding suppression of areas affected by pneumonia. Much of the therapeutic effect of AED and DHEA was obtained by treating for only the first 3 weeks, which is the phase of adrenal hyperplasia. These results suggest that the ratio of GC to anti-GC steroids may play a role in the pathogenesis of tuberculosis, and further investigation could lead to novel treatment strategies.


Subject(s)
Androstenediol/therapeutic use , Cytokines/metabolism , Dehydroepiandrosterone/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Animals , Colony Count, Microbial , Cytokines/analysis , Immunohistochemistry , Interleukin-1/analysis , Interleukin-1/metabolism , Interleukin-2/analysis , Interleukin-2/metabolism , Lung/pathology , Male , Mice , Time Factors , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
19.
J Steroid Biochem Mol Biol ; 56(1-6 Spec No): 113-7, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8603031

ABSTRACT

Androgens are involved in many regulatory processes in mammary and endometrial epithelium, but their role in the development and progression of breast and endometrial carcinoma is poorly understood. Androgen receptors (AR) are found in normal epithelium as well as in more than 50% of specimen from both tumor types. The occurrence of AR is correlated with estrogen and progesterone receptors. Androgen receptor positive cell lines were established during the last few years in our laboratory from malignant mammary (MFM-223) and endometrial (MFE-296) tumors supplementing the small number of androgen-responsive cell lines published so far. In this paper some aspects of the role of androgens in these two types of hormone responsive female cancers are presented. The proliferation of ZR-75-1, MFM-223 and MFE-296 cells is inhibited by androgens. The progestin medroxyprogesterone acetate inhibits the proliferation of estrogen- and progesterone receptor negative MFM-223 cells via the androgen receptor. Some steroid metabolites with distinct estrogenic properties like androst-5-ene-3 beta,17 beta-diol possess androgenic properties in this model system. Androgens stimulate the in vitro secretion of gross cystic disease fluid proteins by human mammary cancer cells. These proteins are normally found in benign breast cysts in vivo. The occurrence of gross cystic disease is correlated with an increased risk of breast cancer. The AR is autoregulated in MFM-223 mammary cancer cells on the protein and mRNA level. In MFE-296 cells with endometrial origin AR protein was increased after incubation with androgens.


Subject(s)
Androgen Antagonists/therapeutic use , Androgens/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Apolipoproteins , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Endometrial Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Glycoproteins , Membrane Transport Proteins , Neoplasm Proteins/drug effects , Receptors, Androgen/drug effects , Androgen Antagonists/pharmacology , Androstane-3,17-diol/pharmacology , Androstane-3,17-diol/therapeutic use , Androstenediol/pharmacology , Androstenediol/therapeutic use , Antineoplastic Agents, Hormonal/pharmacology , Apolipoproteins D , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Carcinoma/pathology , Carcinoma/physiopathology , Carrier Proteins/metabolism , Endometrial Neoplasms/physiopathology , Female , Fibrocystic Breast Disease/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medroxyprogesterone Acetate/pharmacology , Medroxyprogesterone Acetate/therapeutic use , Neoplasm Proteins/physiology , Pleural Effusion/pathology , Receptors, Androgen/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...