Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 255(3): 103-116, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36069766

ABSTRACT

Recent reports indicate an increase in Leydig cell tumor (LCT) incidence. Radical orchiectomy is the standard therapy in children and adults, although it entails physical and psychosocial side effects. Testis-sparing surgery can be a consideration for benign LCT of 2.5 cm or less in size. Malignant LCTs respond poorly to conventional chemotherapy, so new treatment modalities are needed. In this study, we observed increased histidine decarboxylase expression and pro-angiogenic potential in LCT surgically resected from pediatric patients (fetal to pubertal) vs control samples from patients without endocrine or metabolic disorders which were collected at necropsy. We, therefore, evaluated for the first time the antitumor efficacy of two histidine decarboxylase inhibitors (α-methyl-dl-histidine dihydrochloride (α-MHD) and epigallocatechin gallate (EGCG)), alone and combined with carboplatin, in two preclinical models of LCT. MA-10 and R2C Leydig tumor cells, representing two different LCT subtypes, were used to generate syngeneic and xenograft mouse LCT models, respectively. In the syngeneic model, monotherapy with α-MHD effectively reduced tumor growth and angiogenesis. In the xenografts, which showed co-expression of histidine decarboxylase and CYP19, the combination of EGCG plus carboplatin was the most effective therapy, leading to LCT growth arrest and undetectable levels of plasmatic estradiol. Testicular and body weights remained unaltered. On the basis of this study, histidine decarboxylase may emerge as a novel pharmacological target for LCT treatment.


Subject(s)
Leydig Cell Tumor , Testicular Neoplasms , Animals , Aromatase , Carboplatin , Estradiol , Histidine , Histidine Decarboxylase/genetics , Humans , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/pathology , Leydig Cell Tumor/surgery , Male , Mice , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Testicular Neoplasms/surgery
2.
J Mol Endocrinol ; 63(3): 187-197, 2019 10.
Article in English | MEDLINE | ID: mdl-31416050

ABSTRACT

Testicular Leydig cells (LC) are modulated by several pathways, one of them being the histaminergic system. Heme oxygenase-1 (HO-1), whose upregulation comprises the primary response to oxidative noxae, has a central homeostatic role and might dysregulate LC functions when induced. In this report, we aimed to determine how hemin, an HO-1 inducer, affects LC proliferative capacity and whether HO-1 effects on LC functions are reversible. It was also evaluated if HO-1 interacts in any way with histamine, affecting its regulatory action over LC. MA-10 and R2C cell lines and immature rat LC were used as models. Firstly, we show that after a 24-h incubation with 25 µmol/L hemin, LC proliferation is reversibly impaired by cell cycle arrest in G2/M phase, with no evidence of apoptosis induction. Even though steroid production is abrogated after a 48-h exposure to 25 µmol/L hemin, steroidogenesis can be restored to control levels in a time-dependent manner if the inducer is removed from the medium. Regarding HO-1 and histamine interaction, it is shown that hemin abrogates histamine biphasic effect on steroidogenesis and proliferation. Working with histamine receptors agonists, we elucidated that HO-1 induction affects the regulation mediated by receptor types 1, 2 and 4. In summary, HO-1 induction arrests LC functions, inhibiting steroid production and cell cycle progression. Despite their reversibility, HO-1 actions might negatively influence critical phases of LC development and differentiation affecting their function as well as other androgen-dependent organs. What's more, we have described a hitherto unknown interaction between HO-1 induction and histamine effects.


Subject(s)
Heme Oxygenase-1/metabolism , Histamine/pharmacology , Leydig Cells/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Enzyme Induction/drug effects , G2 Phase/drug effects , Hemin/pharmacology , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mice , Mitosis/drug effects , Rats, Sprague-Dawley , Steroids/biosynthesis
3.
J Endocrinol ; 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30620715

ABSTRACT

Insulin resistance is the decreased ability of insulin to mediate metabolic actions. In the ovary, insulin controls ovulation and oocyte quality. Alterations in ovarian insulin signaling pathway could compromise ovarian physiology. Here, we aimed to investigate the effects of fetal programming on ovarian insulin signaling and evaluate the effect of metformin treatment. Pregnant rats were hyperandrogenized with testosterone and female offspring born to those dams were employed; at adulthood, prenatally hyperandrogenized (PH) offspring presented two phenotypes: irregular ovulatory (PHiov) and anovulatory (PHanov). Half of each group was orally treated with metformin. Metformin treatment improved the estrous cyclicity in both PH groups. Both PH groups showed low mRNA levels of IR, IRS1 and Glut4. IRS2 was decreased only in PHanov. Metformin upregulated the mRNA levels of some of the mediators studied. Protein expression of IR, IRS1/2 and GLUT4 was decreased in both PH groups. In PHiov, metformin restored the expression of all the mediators, whereas, in PHanov, metformin restored only that of IR and IRS1/2. IRS1 phosphorylation was measured in tyrosine residues, which activates the pathway, and in serine residues, which impairs insulin action. PHiov presented high IRS1 phosphorylation on tyrosine and serine residues, whereas PHanov showed high serine phosphorylation and low tyrosine phosphorylation. Metformin treatment lowered serine phosphorylation only in PHanov rats. Our results suggest that PHanov rats have a defective insulin action, partially restored with metformin. PHiov rats had less severe alterations, and metformin treatment was more effective in this phenotype.

4.
Open Vet J ; 8(1): 77-85, 2018.
Article in English | MEDLINE | ID: mdl-29721436

ABSTRACT

11ß-Hydroxysteroid dehydrogenase 1 (11ß-HSD1) is an enzyme that activates cortisone into cortisol in tissues. Alterations in this enzyme are related to the development of metabolic syndrome, obesity and hyperadrenocorticism (HAC). Endothelial nitric oxide synthase (eNOS) produces nitric oxide and is related to the regulation of adrenal steroidogenesis. The aim of the study was to evaluate 11ß-HSD1 and eNOS expression in dogs with HAC. Visceral adipose tissue samples were taken to evaluate 11ß-HSD1 expression by immunohistochemistry and western blotting. In parallel, adrenal gland samples were collected to evaluate eNOS expression by immunohistochemistry. 11ß-HSD1 expression was significantly higher in the adipocytes of dogs with HAC than in those of the control dogs. eNOS expression in the adrenal cortex (zona fasciculata) was significantly lower in the dogs with HAC than in the control dogs. 11ß-HSD1 overexpression and eNOS underexpression could play a role in the maintenance of hypercortisolism in dogs with HAC.

5.
Eur J Cancer ; 91: 125-135, 2018 03.
Article in English | MEDLINE | ID: mdl-29367056

ABSTRACT

Leydig-cell tumours (LCTs) are rare endocrine tumours of the testicular interstitium, with recent increased incidence. Symptoms include precocious puberty in children; and erectile dysfunction, infertility and/or gynaecomastia, in adults. So far, scientific evidence points to aromatase (CYP19) overexpression and excessive oestrogen and insulin-like growth factor (IGF) -1 production as responsible for Leydig-cell tumourigenesis. LCTs are usually benign; however, malignant LCTs respond poorly to chemo/radiotherapy, highlighting the need to identify novel targets for treatment. Herein, we investigated the potential role of the histamine receptor H4 (HRH4) as a therapeutic target for LCTs using R2C rat Leydig tumour cells, a well-documented in vitro model for Leydigioma. Also, we studied for the first time the expression of CYP19, IGF-1R, oestrogen receptor (ER) α, ERß, androgen receptor (AR) and HRH4 in human prepubertal LCTs versus normal prepubertal testes (NPTs). HRH4 agonist treatment inhibited steroidogenesis and proliferation in R2C cells and also negatively affected their pro-angiogenic capacity in vitro and in vivo, as assessed by evaluating the proliferative activity of human umbilical vein endothelial cells and by means of the quail chorioallantoic membrane assay, respectively. Moreover, E2 and IGF-1 inhibited HRH4 mRNA and protein levels. In human prepubertal LCTs, CYP19, IGF-1R, ERα and ERß were overexpressed compared with NPTs. In contrast, HRH4 staining was weak in LCTs, but moderate/strong and confined to the interstitium in NPTs. Importantly, HRH4 was absent or barely detectable in seminiferous tubules or germ cells. Overall, our results point to HRH4 as a novel therapeutic target in LCTs.


Subject(s)
Antineoplastic Agents/pharmacology , Guanidines/pharmacology , Histamine Agonists/pharmacology , Imidazoles/pharmacology , Leydig Cell Tumor/drug therapy , Receptors, Histamine H4/agonists , Testicular Neoplasms/drug therapy , Thiourea/analogs & derivatives , Age Factors , Angiogenesis Inhibitors/pharmacology , Animals , Aromatase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Coturnix/embryology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infant , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/pathology , Male , Molecular Targeted Therapy , Neovascularization, Pathologic , Rats , Receptor, IGF Type 1 , Receptors, Androgen/metabolism , Receptors, Histamine H4/metabolism , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Steroid Synthesis Inhibitors/pharmacology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Thiourea/pharmacology
6.
J Endocrinol ; 230(1): 67-79, 2016 07.
Article in English | MEDLINE | ID: mdl-27179108

ABSTRACT

Prenatal hyperandrogenism is hypothesized as one of the main factors contributing to the development of polycystic ovary syndrome (PCOS). PCOS patients have high risk of developing fatty liver and steatosis. This study aimed to evaluate the role of prenatal hyperandrogenism in liver lipid metabolism and fatty liver development. Pregnant rats were hyperandrogenized with testosterone. At pubertal age, the prenatally hyperandrogenized (PH) female offspring displayed both ovulatory (PHov) and anovulatory (PHanov) phenotypes that mimic human PCOS features. We evaluated hepatic transferases, liver lipid content, the balance between lipogenesis and fatty acid oxidation pathway, oxidant/antioxidant balance and proinflammatory status. We also evaluated the general metabolic status through growth rate curve, basal glucose and insulin levels, glucose tolerance test, HOMA-IR index and serum lipid profile. Although neither PH group showed signs of liver lipid content, the lipogenesis and fatty oxidation pathways were altered. The PH groups also showed impaired oxidant/antioxidant balance, a decrease in the proinflammatory pathway (measured by prostaglandin E2 and cyclooxygenase-2 levels), decreased glucose tolerance, imbalance of circulating lipids and increased risk of metabolic syndrome. We conclude that prenatal hyperandrogenism generates both PHov and PHanov phenotypes with signs of liver alterations, imbalance in lipid metabolism and increased risk of developing metabolic syndrome. The anovulatory phenotype showed more alterations in liver lipogenesis and a more impaired balance of insulin and glucose metabolism, being more susceptible to the development of steatosis.


Subject(s)
Hyperandrogenism/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Cyclooxygenase 2/metabolism , Female , Glucose Tolerance Test , Inflammation/metabolism , Insulin/blood , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Lipids/blood , Liver/drug effects , Metabolic Syndrome/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Testosterone/pharmacology
7.
J Endocrinol ; 223(3): 241-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253872

ABSTRACT

The histamine H4 receptor (HRH4), discovered only 13 years ago, is considered a promising drug target for allergy, inflammation, autoimmune disorders and cancer, as reflected by a steadily growing number of scientific publications and patent applications. Although the presence of HRH4 has been evidenced in the testis, its specific localization or its role has not been established. Herein, we sought to identify the possible involvement of HRH4 in the regulation of Leydig cell function. We first evaluated its expression in MA-10 Leydig tumor cells and then assessed the effects of two HRH4 agonists on steroidogenesis and proliferation. We found that HRH4 is functionally expressed in MA-10 cells, and that its activation leads to the inhibition of LH/human chorionic gonadotropin-induced cAMP production and StAR protein expression. Furthermore, we observed decreased cell proliferation after a 24-h HRH4 agonist treatment. We then detected for the sites of HRH4 expression in the normal rat testis, and detected HRH4 immunostaining in the Leydig cells of rats aged 7-240 days, while 21-day-old rats also presented HRH4 expression in male gametes. Finally, we evaluated the effect of HRH4 activation on the proliferation of normal progenitor and immature rat Leydig cell culture, and both proved to be susceptible to the anti-proliferative effect of HRH4 agonists. Given the importance of histamine (2-(1H-imidazol-4-yl)ethanamine) in human (patho)physiology, continued efforts are directed at elucidating the emerging properties of HRH4 and its ligands. This study reveals new sites of HRH4 expression, and should be considered in the design of selective HRH4 agonists for therapeutic purposes.


Subject(s)
Cell Proliferation , Leydig Cells/metabolism , Progesterone/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Animals , Blotting, Western , Bucladesine/pharmacology , Cell Line, Tumor , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Guanidines/pharmacology , Histamine Agonists/pharmacology , Immunohistochemistry , Indoles/pharmacology , Leydig Cells/drug effects , Male , Microscopy, Confocal , Oximes/pharmacology , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, Histamine H4 , Testis/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology
8.
J Endocrinol ; 221(1): 15-28, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424290

ABSTRACT

Histamine (HA) is a neurotransmitter synthesized in most mammalian tissues exclusively by histidine decarboxylase enzyme. Among the plethora of actions mediated by HA, the modulatory effects on steroidogenesis and proliferation in Leydig cells (LCs) have been described recently. To determine whether the effects on LCs reported could be extrapolated to all steroidogenic systems, in this study, we assessed the effect of this amine on adrenal proliferation and steroidogenesis, using two adrenocortical cell lines as experimental models, murine Y1 cells and human NCI-H295R cells. Even when steroidogenesis was not modified by HA in adrenocortical cells, the biogenic amine inhibited the proliferation of H295R cells. This action was mediated by the activation of HRH1 subtype and an increase in the production of inositol phosphates as second messengers, causing cell-cycle arrest in the G2/M phase. These results indicate a new role for HA in the proliferation of human adrenocortical cells that could contribute to a better understanding of tumor pathology as well as to the development of new therapeutic agents.


Subject(s)
Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Cell Proliferation , Histamine/metabolism , Steroids/metabolism , Animals , Cell Line , G2 Phase Cell Cycle Checkpoints , Humans , M Phase Cell Cycle Checkpoints , Mice
9.
Biol Reprod ; 87(6): 150, 2012 Jun.
Article in English | MEDLINE | ID: mdl-23077168

ABSTRACT

Mast cells (MC) occur normally in the testis with a species-specific distribution, yet their precise role remains unclear. Testicular MC express histidine decarboxylase (HDC), the unique enzyme responsible for histamine (HA) generation. Evidence to date supports a role for HA as a local regulator of steroidogenesis via functional H1 and H2 receptor subtypes (HRH1 and HRH2, respectively) present in Leydig cells. Given that HA is a well-known modulator of physiological and pathological proliferation in many different cell types, we aimed in the present study to evaluate whether HA might contribute to the regulation of Leydig cell number as well as to the control of androgen production. Herein, we demonstrate, to our knowledge for the first time, that MA-10 Leydig tumor cells, but not normal immature Leydig cells (ILC), exhibit a proliferative response upon stimulation with HA that involves HRH2 activation, transient elevation of cAMP levels, and increased extracellular signal-regulated kinase (ERK) phosphorylation. Our results also reveal that MA-10 cells show significantly heightened HDC expression compared to normal ILC or whole-testicular lysate and that inhibition of HDC activity decreases MA-10 cell proliferation, suggesting a possible correlation between autocrine overproduction of HA and abnormally increased proliferation in Leydig cells. The facts that germ cells are also both source and target of HA and that multiple testicular cells are susceptible to HA action underline the importance of the present study, which we hope will serve as a first step for further research into regulation of non-MC-related HDC expression within the testis and its significance for testicular function.


Subject(s)
Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine/metabolism , Leydig Cell Tumor/metabolism , Leydig Cells/metabolism , Receptors, Histamine H2/metabolism , Second Messenger Systems , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Histamine Agonists/metabolism , Histamine Agonists/pharmacology , Histidine Decarboxylase/antagonists & inhibitors , Histidine Decarboxylase/biosynthesis , Histidine Decarboxylase/metabolism , Leydig Cell Tumor/drug therapy , Leydig Cell Tumor/enzymology , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/enzymology , Male , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Histamine H2/chemistry , Second Messenger Systems/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
10.
Biol Reprod ; 80(1): 144-52, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18768916

ABSTRACT

This study was conducted to shed light on the so far unexplored intracellular mechanisms underlying negative modulation of Leydig cell steroidogenesis by histamine (HA). Using the MA-10 cell line and highly purified rat Leydig cells as experimental models, we examined the effect of the amine on biochemical steps known to be modulated by HA or involved in LH/hCG action. In agreement with previous findings, HA at 10 microM showed a potent inhibitory effect on hCG-stimulated steroid synthesis, regardless of the gonadotropin concentration used. Moreover, HA decreased not only LH/hCG-induced cAMP production but also steroid synthesis stimulated by the permeable cAMP analog dibutyryl cAMP (db-cAMP). Considering the post-cAMP sites of HA action, it is shown herein that HA markedly inhibited db-cAMP-stimulated steroidogenic acute regulatory (STAR) protein expression, as well as steps catalyzed by P450-dependent enzymes, mainly the conversion of cholesterol to pregnenolone by cholesterol side-chain cleavage enzyme (CYP11A). The antisteroidogenic action of HA was blocked by addition of the phospholipase C (PLC) inhibitor U73122, and HA significantly augmented inositol triphosphate (IP3) production, suggesting a major role for the PLC/IP3 pathway in HA-induced inhibition of Leydig cell function. Finally, HA increased nitric oxide synthase (NOS) activity, and the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) markedly attenuated the effect of the amine on steroid synthesis. On the basis of our findings, HA antagonizes the gonadotropin action in Leydig cells at steps before and after cAMP formation. NOS activation is the main intracellular mechanism by which HA exerts its antisteroidogenic effects.


Subject(s)
Histamine/pharmacology , Leydig Cells/metabolism , Nitric Oxide Synthase Type II/metabolism , Progesterone/biosynthesis , Receptors, Histamine/metabolism , Testosterone/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Chorionic Gonadotropin/pharmacology , Cyclic AMP/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Inositol Phosphates/metabolism , Leydig Cells/drug effects , Leydig Cells/enzymology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/biosynthesis , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
11.
Buenos Aires; Ministerio de Salud de la Nación; 2004. (120412).
Monography in Spanish | ARGMSAL | ID: biblio-993374
12.
Buenos Aires; Ministerio de Salud de la Nación; 2003. (120411).
Monography in Spanish | ARGMSAL | ID: biblio-993375

ABSTRACT

This study shows that NO inhibits basal, ACTH and Angll-induced aldosterone synthesis in bovine adrenal glomerulosa cells. This inhibition is independent of soluble guanylyl cyclases cGMP levels, protein kinase G activity, cAMP production or phospholipase C activity. NO inhibits aldosterogenesis through, at least, a direct inhibition of cytochrome P450 steroidogenic enzymes, including P450scc enzyme activity. We present evidence that NO can be synthesized endogenously, since we have partially characterized NO synthase activity in glomeralosa cells extracts. In conclusion, our results support the hypothesis that NO can act as autocrine and/or paracrine modulator of aldosterone secretion from adrenal gland, thus contributing to the regulation of physiological aldosterone levels induced by hormones such as angiotensin II or ACTH.


Subject(s)
Testicular Diseases , Fellowships and Scholarships
13.
Buenos Aires; Ministerio de Salud de la Nación; 2003.
Monography in Spanish | BINACIS | ID: biblio-1217742

ABSTRACT

This study shows that NO inhibits basal, ACTH and Angll-induced aldosterone synthesis in bovine adrenal glomerulosa cells. This inhibition is independent of soluble guanylyl cyclases cGMP levels, protein kinase G activity, cAMP production or phospholipase C activity. NO inhibits aldosterogenesis through, at least, a direct inhibition of cytochrome P450 steroidogenic enzymes, including P450scc enzyme activity. We present evidence that NO can be synthesized endogenously, since we have partially characterized NO synthase activity in glomeralosa cells extracts. In conclusion, our results support the hypothesis that NO can act as autocrine and/or paracrine modulator of aldosterone secretion from adrenal gland, thus contributing to the regulation of physiological aldosterone levels induced by hormones such as angiotensin II or ACTH.


Subject(s)
Testicular Diseases , Fellowships and Scholarships
14.
Buenos Aires; Ministerio de Salud de la Nación; 2004.
Monography in Spanish | LILACS-Express | BINACIS | ID: biblio-1217743
15.
Buenos Aires; Ministerio de Salud de la Nación; 2004. (120412).
Monography in Spanish | BINACIS | ID: bin-120412
16.
Buenos Aires; Ministerio de Salud de la Nación; 2003. (120411).
Monography in Spanish | BINACIS | ID: bin-120411

ABSTRACT

This study shows that NO inhibits basal, ACTH and Angll-induced aldosterone synthesis in bovine adrenal glomerulosa cells. This inhibition is independent of soluble guanylyl cyclases cGMP levels, protein kinase G activity, cAMP production or phospholipase C activity. NO inhibits aldosterogenesis through, at least, a direct inhibition of cytochrome P450 steroidogenic enzymes, including P450scc enzyme activity. We present evidence that NO can be synthesized endogenously, since we have partially characterized NO synthase activity in glomeralosa cells extracts. In conclusion, our results support the hypothesis that NO can act as autocrine and/or paracrine modulator of aldosterone secretion from adrenal gland, thus contributing to the regulation of physiological aldosterone levels induced by hormones such as angiotensin II or ACTH.


Subject(s)
Testicular Diseases , Fellowships and Scholarships
SELECTION OF CITATIONS
SEARCH DETAIL
...