Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Sci Rep ; 13(1): 18685, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37907720

ABSTRACT

The Developmental Origins of Health and Disease (DOHaD) concept has provided the framework to assess how early life experiences can shape health and disease throughout the life course. While maternal malnutrition has been proposed as a risk factor for the developmental programming of prostate cancer (PCa), the molecular mechanisms remain poorly understood. Using RNA-seq data, we demonstrated deregulation of miR-206-Plasminogen (PLG) network in the ventral prostate (VP) of young maternally malnourished offspring. RT-qPCR confirmed the deregulation of the miR-206-PLG network in the VP of young and old offspring rats. Considering the key role of estrogenic signaling pathways in prostate carcinogenesis, in vitro miRNA mimic studies also revealed a negative correlation between miR-206 and estrogen receptor α (ESR1) expression in PNT2 cells. Together, we demonstrate that early life estrogenization associated with the deregulation of miR-206 networks can contribute to the developmental origins of PCa in maternally malnourished offspring. Understanding the molecular mechanisms by which early life malnutrition affects offspring health can encourage the adoption of a governmental policy for the prevention of non-communicable chronic diseases related to the DOHaD concept.


Subject(s)
Malnutrition , MicroRNAs , Prostatic Neoplasms , Animals , Male , Rats , Malnutrition/complications , Malnutrition/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Origin of Life , Prostate/metabolism , Prostatic Neoplasms/genetics
2.
Reprod Fertil Dev ; 30(7): 969-979, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29207253

ABSTRACT

Besides androgenic dependence, other hormones also influence the prostate biology. Prolactin has been described as an important hormone associated with maintenance of prostatic morphophysiology; however, there is a lack of information on the involvement of prolactin during prostate development and growth. This study aimed to evaluate whether perinatal prolactin modulation interferes with rat ventral prostate (VP) development and maturation. Therefore, prolactin or bromocriptine (an inhibitor of prolactin release from the pituitary) were administered to Sprague Dawley rats from postnatal Day (PND) 12 to PND 21 or 35. Animals were then killed and serum hormonal quantification, VP morphological-stereological and immunohistochemical analyses and western blotting reactions were employed. Our results demonstrate that prolactin blockage increased serum testosterone on PND 21, which reflected an increase in anogenital distance. Although prolactin modulation did not interfere with VP weight, it modified VP morphology by dilating the acinar lumen and reducing epithelial cell height. Prolactin activated the signal transducer and activator of transcription (STAT) downstream pathway, increased androgen receptor expression and epithelial proliferation. In addition, prolactin and bromocriptine also increased expression of cytokeratin 18, a marker of luminal-differentiated cells. In conclusion, the VP responds to prolactin modulation through a mechanism of increasing the epithelial proliferative response and dynamics of cell differentiation, especially in animals treated for a more prolonged period.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Prolactin/metabolism , Prostate/growth & development , Animals , Bromocriptine/pharmacology , Hormone Antagonists/pharmacology , Keratin-18/metabolism , Male , Prolactin/pharmacology , Prostate/drug effects , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Androgen/metabolism , Testosterone/blood
3.
J Mol Histol ; 48(5-6): 403-415, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28988314

ABSTRACT

Despite the androgenic dependence, other hormones, growth factors, and cytokines are necessary to support prostatic growth and maintain the glandular structure; among them, prolactin is a non-steroidal hormone secreted mainly by the pituitary gland. However, extra-pituitary expression of prolactin, such as in the prostate, has also been demonstrated, highlighting the paracrine and autocrine actions of prolactin within the prostate. Here, we investigated whether prolactin modulation alters ventral prostate (VP) morphophysiology in adult castrated rats. Sprague Dawley rats were castrated and after 21 days, divided into ten experimental groups (n = 6/group): castrated control: castrated animals that did not receive treatment; castrated+testosterone: castrated animals that received T (4 mg/kg/day); castrated+PRL (PRL): castrated animals receiving prolactin (0.3 mg/kg/day); castrated+T+PRL: castrated animals that received a combination of testosterone and prolactin; and castrated+bromocriptine (BR): castrated animals that received bromocriptine (0.4 mg/kg/day). The control group included intact animals. The animals were treated for 3 or 10 consecutive days. At the end of experimental period, the animals were euthanized, and the blood and VP lobes were collected and analyzed by different methods. The main findings were that the administration of prolactin to castrated rats did not exert anabolic effects on the VP. Although we observed activation of downstream prolactin signaling after prolactin administration, this was not enough to overcome the prostatic androgen deficiency. Likewise, there was no additional glandular involution in the castrated group treated with bromocriptine. We concluded that despite stimulating the downstream signaling pathway, exogenous prolactin does not act on VP in the absence or presence of high levels of testosterone.


Subject(s)
Aging/metabolism , Castration , Hormone Replacement Therapy , Prolactin/metabolism , Prostate/metabolism , Testosterone/therapeutic use , Animals , Blotting, Western , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Rats, Sprague-Dawley , Receptors, Androgen/metabolism
4.
Gen Comp Endocrinol ; 246: 258-269, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28041790

ABSTRACT

Experimental data demonstrated the negative impact of maternal protein malnutrition (MPM) on rat prostate development, but the mechanism behind the impairment of prostate growth has not been well understood. Male Sprague Dawley rats, borned to dams fed a normal protein diet (CTR group, 17% protein diet), were compared with those borned from dams fed a low protein diet (6% protein diet) during gestation (GLP group) or gestation and lactation (GLLP). The ventral prostate lobes (VP) were removed at post-natal day (PND) 10 and 21, and analyzed via different methods. The main findings were low birth weight, a reduction in ano-genital distance (AGD, a testosterone-dependent parameter), and an impairment of prostate development. A delay in prostate morphogenesis was associated with a reduced testosterone levels and angiogenic process through downregulation of aquaporin-1 (AQP-1), insulin/IGF-1 axis and VEGF signaling pathway. Depletion of the microvascular network, which occurs in parallel to the impairment of proliferation and differentiation of the epithelial cells, affects the bidirectional flux between blood vessels impacting prostatic development. In conclusion, our data support the hypothesis that a reduction in microvascular angiogenesis, especially in the subepithelial compartment, is associated to the impairment of prostate morphogenesis in the offspring of MPM dams.


Subject(s)
Fetal Development , Fetal Nutrition Disorders/pathology , Microvessels/embryology , Neovascularization, Pathologic/pathology , Prostate/pathology , Protein-Energy Malnutrition/physiopathology , Animals , Animals, Newborn , Blotting, Western , Female , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Lactation/physiology , Male , Pregnancy , Prostate/blood supply , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/blood , Vascular Endothelial Growth Factor A/metabolism
5.
Anat Rec (Hoboken) ; 300(2): 291-299, 2017 02.
Article in English | MEDLINE | ID: mdl-27788294

ABSTRACT

Gestational diabetes mellitus (GDM) has increased in recent years. Although the cellular and molecular mechanisms involved in GDM-increased risk factors to offspring remained poorly understood, some studies suggested an association between an increase in oxidative stress induced by maternal hyperglycemia and complications for both mothers and newborns. Here, we investigated the impact of maternal hyperglycemia followed by maternal insulin replacement during lactation on the expression of antioxidant enzymes and mast cell number in offspring ventral prostate (VP) at puberty. Pregnant rats were divided into three groups: control (CT); streptozotocin-induced maternal hyperglycemia (MH); and MH plus maternal insulin replacement during lactation (MHI). Male offspring were euthanized at postnatal day (PND) 60 and the VP was removed and processed for histology and Western blotting analyses. Maternal hyperglycemia delayed prostate maturation, and increased mast cell number catalase (CAT), superoxide dismutase (SOD), glutatione-s-transferase (GST-pi), and cyclooxygenase-2 (Cox-2) expression in the offspring of hyperglycemic dams. Maternal insulin replacement restored VP structure, mast cell number and antioxidant protein expression, except for Cox-2, which remained higher in the MHI group. Thus, an increase in oxidative stress induced by intrauterine hyperglycemia impacts prostate development and maturation, which persists until puberty. The overall improvement of maternal metabolism after insulin administration contributes to the restoration of prostate antioxidant enzymes and secretory function. Taken together, our results highlighted that imbalanced physiological maternal-fetal interaction contributes to the impairment of reproductive performance of the offspring from diabetic mothers. Anat Rec, 300:291-299, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes, Gestational/metabolism , Mast Cells/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prostate/metabolism , Animals , Blood Glucose/metabolism , Cell Count , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/drug therapy , Diabetes, Gestational/enzymology , Diabetes, Gestational/pathology , Female , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/pharmacology , Insulin/therapeutic use , Male , Mast Cells/drug effects , Mast Cells/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pregnancy , Prenatal Exposure Delayed Effects/enzymology , Prenatal Exposure Delayed Effects/pathology , Prostate/drug effects , Prostate/enzymology , Prostate/pathology , Rats , Rats, Wistar
6.
Gen Comp Endocrinol ; 206: 60-71, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24983773

ABSTRACT

Clinical and experimental studies have shown that exposure to adverse conditions during the critical stages of embryonic, fetal or neonatal development lead to a significantly increased risk of later disease. Diabetes during pregnancy has been linked to increased risk of obesity and diabetes in offspring. Here, we investigated whether mild gestational diabetes mellitus (GDM) followed or not by maternal insulin replacement affects the ventral prostate (VP) structure and function in male offspring at puberty and adulthood. Pregnant rats were divided into the following 3 groups: control (CT); streptozotocin (STZ)-induced diabetes (D); and D plus insulin replacement during lactation (GDI). The male offspring from different groups were euthanized at postnatal day (PND) 60 and 120. Biometrical parameters, hormonal levels and prostates were evaluated. Mild-GDM promoted reduction in the glandular parenchyma and increased collagen deposition. Insulin replacement during lactation restored the VP morphology. Most importantly, mild-GDM decreased the androgen-induced secretory function as determined by prostatein expression, and insulin replacement reversed this effect. Our results demonstrated that mild GDM impairs VP parenchyma maturation, which is associated with an increase in the fibromuscular stroma compartment. Functionally, the reduction in the VP parenchyma decreases the glandular secretory activity as demonstrated by low expression of prostatein, a potent immunosuppressor factor that protects sperm from immunologic damage into the feminine reproductive tract. This change could lead to impairment of reproductive function in male offspring from diabetic mothers. Maternal insulin replacement during the weaning period apparently restores the prostate function in male offspring.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetes, Gestational/physiopathology , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Lactation/drug effects , Prostate/metabolism , Sexual Maturation/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Female , Humans , Hypoglycemic Agents/pharmacology , Immunoenzyme Techniques , Insulin/pharmacology , Male , Pregnancy , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...