1
|
Effect of Omega-3 or Omega-6 Dietary Supplementation on Testicular Steroidogenesis, Adipokine Network, Cytokines, and Oxidative Stress in Adult Male Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5570331. [PMID: 34257810 PMCID: PMC8260291 DOI: 10.1155/2021/5570331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/15/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022]
Abstract
This study was undertaken to elucidate the effect of omega-3 and omega-6 supplementation on the levels of different adipokines and cytokines, as well as the antioxidant system, in relation to male reproductive hormones and testicular functions. Adult male Sprague-Dawley rats were daily gavaged with either physiological saline (control group), sunflower oil (omega 6 group; 1 mL/kg body weight), or fish oil (omega-3 group; 1000 mg/kg body weight) for 12 weeks. The administration of omega-3 or omega-6 resulted in decreased serum concentrations of kisspeptin 1, gonadotropin-releasing hormone, luteinizing hormone, follicle-stimulating hormone, and testosterone. In addition, it downregulated the mRNA expression levels of steroidogenic genes. The intratesticular levels of apelin, adiponectin, and irisin were elevated while chemerin, leptin, resistin, vaspin, and visfatin were declined following the administration of either omega-3 or omega-6. The testicular concentration of interleukin 10 was increased while interleukin 1 beta, interleukin 6, tumor necrosis factor α, and nuclear factor kappa B were decreased after consumption of omega-3 or omega-6. In the testes, the levels of superoxide dismutase, catalase, glutathione peroxidase 1, and the total antioxidant capacity were improved. In conclusion, the administration of omega-3 or omega-6 adversely affects the process of steroidogenesis but improves the antioxidant and anti-inflammatory status of the reproductive system via modulating the levels of testicular adipokines.
Collapse
|
2
|
L-López F, Sarmento-Cabral A, Herrero-Aguayo V, Gahete MD, Castaño JP, Luque RM. Obesity and metabolic dysfunction severely influence prostate cell function: role of insulin and IGF1. J Cell Mol Med 2017; 21:1893-1904. [PMID: 28244645 PMCID: PMC5571563 DOI: 10.1111/jcmm.13109] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/01/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a major health problem that courses with severe comorbidities and a drastic impairment of homeostasis and function of several organs, including the prostate gland (PG). The endocrine–metabolic regulatory axis comprising growth hormone (GH), insulin and IGF1, which is drastically altered under extreme metabolic conditions such as obesity, also plays relevant roles in the development, modulation and homeostasis of the PG. However, its implication in the pathophysiological interplay between obesity and prostate function is still to be elucidated. To explore this association, we used a high fat–diet obese mouse model, as well as in vitro primary cultures of normal‐mouse PG cells and human prostate cancer cell lines. This approach revealed that most of the components of the GH/insulin/IGF1 regulatory axis are present in PGs, where their expression pattern is altered under obesity conditions and after an acute insulin treatment (e.g. Igfbp3), which might have some pathophysiological implications. Moreover, our results demonstrate, for the first time, that the PG becomes severely insulin resistant under diet‐induced obesity in mice. Finally, use of in vitro approaches served to confirm and expand the conception that insulin and IGF1 play a direct, relevant role in the control of normal and pathological PG cell function. Altogether, these results uncover a fine, germane crosstalk between the endocrine–metabolic status and the development and homeostasis of the PG, wherein key components of the GH, insulin and IGF1 axes could play a relevant pathophysiological role.
Collapse
Affiliation(s)
- Fernando L-López
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| | - André Sarmento-Cabral
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| | - Manuel D Gahete
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| | - Justo P Castaño
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| | - Raúl M Luque
- Maimónides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofía University Hospital, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Cordoba, Spain.,International Campus of Excellence on Agrifood, CeiA3, Cordoba, Spain
| |
Collapse
|
3
|
Relationships between blood hormonal concentrations and secondary fibre shedding in young cashmere-bearing goats at their first moult. ACTA ACUST UNITED AC 2016. [DOI: 10.1017/s1357729800054321] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractThe aim of this study was to evaluate secondary fibre shedding in relation to seasonal hormonal changes in young cashmere-bearing goats reared in southern Italy. We used 14 cashmere-bearing kids, seven males and seven females, of a Scottish breed-population of goats specialized in cashmere production. Monthly, a hair patch of 4 cm2was clipped from both left and right mid sides alternately and cashmere length and its yield were determined. Blood samples were taken every 2 weeks and plasma was assayed for prolactin (PRL), growth hormone (GH), tri-iodothyronine (T3), thyroxine (T4), progesterone and testosterone. With the same frequency, animals were monitored for onset of moult. Our data indicate that in young cashmere goats at their first moult, secondary fibre shedding is a seasonal event that is strictly related to changes in photoperiod and that PRL has a pivotal rôle in regulating this phenomenon. This corresponds with findings in adults. The observation that both thyroid hormones and sex steroids varied seasonally suggests that they are involved in the regulation of the moult cycle, but their precise rôle remains to be elucidated. Our data also indicate that GH seems to be implicated in the control of the moult cycle. The finding that maximal fibre shedding occurred earlier in males than females could be ascribed to differences in GH levels and to the earlier increase in plasma concentration of PRL observed in males.
Collapse
|
4
|
Sideris S, Aoun F, Martinez CN, Latifyan S, Awada A, Costante G, Gil T. Role of corticosteroids in prostate cancer progression: implications for treatment strategy in metastatic castration-resistant patients. J Endocrinol Invest 2016; 39:729-38. [PMID: 26786788 DOI: 10.1007/s40618-016-0430-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/27/2022]
Abstract
Corticosteroid agents (CA) are widely used in the treatment of metastatic castration-resistant prostate cancer (mCRPC) either as concomitant treatment with active agents such as docetaxel, cabazitaxel and abiraterone or in a palliative setting, predominantly due to their anti-inflammatory activity. However, the chronic use of CA has numerous side effects, especially in case of steroid-induced adrenal insufficiency. Furthermore, the latest clinical and preclinical data demonstrate that CA themselves are likely to promote tumour progression in certain populations of patients with mCRPC. Therefore, the role of CA in advanced disease should be carefully weighed for each patient and their withdrawal should be considered in some patients. This is necessary, especially in clinical trials that need good performance status patients to evaluate the activity and the safety of emerging drugs in mCRPC that do not require the concurrent use of CA. In oncology, there is no consensus on an algorithm of gradual steroid tapering and frequently the approach to this procedure is empirical. An algorithm is presented in this article based on clinical observations. Prospective studies are necessary to evaluate the efficacy and safety of the above-proposed algorithm in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- S Sideris
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - F Aoun
- Urology Department, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - C N Martinez
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - S Latifyan
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - A Awada
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| | - G Costante
- Endocrinology Department, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium.
| | - T Gil
- Medical Oncology Clinic, Jules Bordet Institute, Boulevard de Waterloo 121, 1000, Brussels, Belgium
| |
Collapse
|
5
|
Abstract
PURPOSE Prostate cancer cells are responsive to multiple hormones and growth factors that can affect cell function. These effects may include modulating cell proliferation and apoptosis, but the ability to impinge on the metastatic potential of prostate cancer cells by affecting cell motility should also be considered, as prostate tumor metastasis correlates with limited therapeutic options and poor prognosis. Human growth hormone (hGH) can affect the growth and survival of prostate cancer cells, but the effect of hGH on prostate cancer cell motility is unknown. In the present study, the potential for exogenous and autocrine hGH to directly affect prostate cancer cell motility was addressed. MATERIALS AND METHODS The effects of exogenous and autocrine hGH on the chemokinesis and chemotaxis of LNCaP prostate cancer cells were tested using cell monolayer wound healing and Boyden chamber invasion assays. The signaling pathways underlying these effects were resolved with chemical inhibitors and the correlation with cytoskeletal actin reorganization evaluated microscopically by staining cells with fluor-conjugated phalloidin. RESULTS Both exogenous and autocrine hGH augmented the migration and invasion of LNCaP cells, and hGH itself acted as a chemoattractant. This activity was dependent upon the STAT5, MEK1/2 and PI3K signaling pathways, and was accompanied by an alteration in cellular actin organization. CONCLUSIONS hGH may enhance the metastatic potential of prostate cancer cells, both as a stimulant of cellular motility and invasiveness and as a chemoattractant.
Collapse
Affiliation(s)
- Alona O Nakonechnaya
- Department of Biochemistry and Molecular Biology, Brody School of Medicine at East Carolina University , Greenville, NC , USA
| | | |
Collapse
|
6
|
Hull KL, Harvey S. Growth hormone and reproduction: a review of endocrine and autocrine/paracrine interactions. Int J Endocrinol 2014; 2014:234014. [PMID: 25580121 PMCID: PMC4279787 DOI: 10.1155/2014/234014] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 11/26/2014] [Indexed: 01/01/2023] Open
Abstract
The somatotropic axis, consisting of growth hormone (GH), hepatic insulin-like growth factor I (IGF-I), and assorted releasing factors, regulates growth and body composition. Axiomatically, since optimal body composition enhances reproductive function, general somatic actions of GH modulate reproductive function. A growing body of evidence supports the hypothesis that GH also modulates reproduction directly, exerting both gonadotropin-dependent and gonadotropin-independent actions in both males and females. Moreover, recent studies indicate GH produced within reproductive tissues differs from pituitary GH in terms of secretion and action. Accordingly, GH is increasingly used as a fertility adjunct in males and females, both humans and nonhumans. This review reconsiders reproductive actions of GH in vertebrates in respect to these new conceptual developments.
Collapse
Affiliation(s)
- Kerry L Hull
- Department of Biology, Bishop's University, Sherbrooke, QC, Canada J1M 1Z7 ; Centre de Recherche Clinique Etienne-Le Bel, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4
| | - Steve Harvey
- Department of Physiology, University of Alberta, Edmonton, AB, Canada T6G 2R3
| |
Collapse
|
7
|
Hsieh DJY, Huang CY, Pai P, Wang SGP, Tsai YL, Li CN, Kuo WW, Huang CY. Prolactin protects cardiomyocytes against intermittent hypoxia-induced cell damage by the modulation of signaling pathways related to cardiac hypertrophy and proliferation. Int J Cardiol 2014; 181:255-66. [PMID: 25531577 DOI: 10.1016/j.ijcard.2014.11.154] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 11/18/2014] [Accepted: 11/22/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Prolactin (PRL) is a multifunctional hormone that influences multiple physiological processes. It has been shown to have a protective effect on the cardiovascular system; however, the mechanisms of this effect are poorly understood. The purpose of the study was to elucidate the role of PRL in intermittent hypoxia (IH)-induced apoptosis in the cardiovascular system. METHOD AND RESULTS We established a hyperprolactinemic rat model by implanting two anterior pituitary (AP) glands into the renal capsule of male Sprague-Dawley rats. The rats were kept under normoxic conditions for 4weeks after implantation in order to reach the expression plateau of PRL in the plasma, and then treated with IH for 7 or 14days. Their hearts were then removed for histological and protein expression analyses. Cerebral cortex (CX)-grafted control rats challenged with IH displayed unique phenotypes such as a thicker heart wall, an abnormal myocardial architecture and an increased interstitial space of the left ventricle. They exhibited reduced expressions of p-JAK2, p-STAT5, cell cycle-dependent proteins (cyclin D1, cyclin E and cyclin A), IGF-IRα, PI3Kα, p-AKT and p-ERK1/2 in cardiomyocytes at 7days. CONCLUSIONS Our comprehensive analysis suggested that high plasma PRL can protect rat cardiomyocytes against IH through (1) the p-JAK2 and p-STAT5 pathways for transient cell proliferation, (2) the PI3Kα/AKT and MAPK survival pathways through IGF-I, and (3) the downregulation of IGF-II and ERK5, which inhibit cell hypertrophy.
Collapse
Affiliation(s)
- Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Peiying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Shyi-Gang P Wang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Department of Physiology, National Yang-Ming University, Taipei, Taiwan
| | - Ying-Lan Tsai
- Athletic Training and Health Department, National Taiwan Sport University, Taoyuan 333, Taiwan
| | - Chia-Ning Li
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
8
|
Nakonechnaya AO, Jefferson HS, Chen X, Shewchuk BM. Differential effects of exogenous and autocrine growth hormone on LNCaP prostate cancer cell proliferation and survival. J Cell Biochem 2013; 114:1322-35. [DOI: 10.1002/jcb.24473] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/27/2012] [Indexed: 12/12/2022]
|
9
|
Transcriptomic analysis of the developing and adult mouse cochlear sensory epithelia. PLoS One 2012; 7:e42987. [PMID: 22900075 DOI: 10.1371/journal.pone.0042987] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 07/16/2012] [Indexed: 01/13/2023] Open
Abstract
The adult mammalian cochlea lacks regenerative ability and the irreversible degeneration of cochlear sensory hair cells leads to permanent hearing loss. Previous data show that early postnatal cochlea harbors stem/progenitor-like cells and shows a limited regenerative/repair capacity. These properties are progressively lost later during the postnatal development. Little is known about the genes and pathways that are potentially involved in this difference of the regenerative/repair potentialities between early postnatal and adult mammalian cochlear sensory epithelia (CSE). The goal of our study is to investigate the transcriptomic profiles of these two stages. We used Mouse Genome 430 2.0 microarray to perform an extensive analysis of the genes expressed in mouse postnatal day-3 (P3) and adult CSE. Statistical analysis of microarray data was performed using SAM (Significance Analysis of Microarrays) software. We identified 5644 statistically significant differentially expressed transcripts with a fold change (FC) >2 and a False Discovery Rate (FDR) ≤0.05. The P3 CSE signature included 3,102 transcripts, among which were known genes in the cochlea, but also new transcripts such as, Hmga2 (high mobility group AT-hook 2) and Nrarp (Notch-regulated ankyrin repeat protein). The adult CSE overexpressed 2,542 transcripts including new transcripts, such as Prl (Prolactin) and Ar (Androgen receptor), that previously were not known to be expressed in the adult cochlea. Our comparative study revealed important genes and pathways differentially expressed between the developing and adult CSE. The identification of new candidate genes would be useful as potential markers of the maintenance or the loss of stem cells and regenerative/repair ability during mammalian cochlear development.
Collapse
|
10
|
Abstract
Prolactin is best known for its actions on the mammary gland. However, circulating prolactin is also detected in males and its receptor (PRLR) is expressed in the prostate, suggesting that the prostate is a target of prolactin. Germline knockout of prolactin or its receptor has failed to reveal a key role for prolactin signaling in mouse prostate physiology. However, several studies involving rodent models and human prostate cell lines and specimens have supported the contribution of the canonical PRLR-Jak2-Stat5a/b pathway to prostate cancer tumorigenesis and progression. Increased expression of prolactin in the prostate itself (rather than changes in circulating prolactin levels) and crosstalk with androgen receptor (AR) signaling are potential mechanisms for increased Stat5a/b signaling in prostate cancer. In the mouse prostate, prolactin overexpression results in disorganized expansion of the basal/stem cell compartment, which has been proposed to house putative prostate tumor-initiating cells. These findings provide new insight into the molecular and cellular targets by which locally produced prolactin could contribute to prostate cancer initiation and progression. A number of pharmacological inhibitors targeting various levels of the PRLR-Jak2-Stat5a/b pathway have been developed and are entering clinical trials for advanced prostate cancer.
Collapse
|
11
|
Aruldhas MM, Ramalingam N, Jaganathan A, John Sashi AM, Stanley JA, Nagappan AS, Vasavan J, Kannan A, Seshadri VN. Gestational and neonatal-onset hypothyroidism alters androgen receptor status in rat prostate glands at adulthood. Prostate 2010; 70:689-700. [PMID: 20033886 DOI: 10.1002/pros.21101] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Infertility associated with congenital and early childhood hypothyroidism is an important reproductive health problem in men. Nevertheless, the exact mechanism underlying hypothyroidism-induced changes in the prostate gland, an androgen-dependent organ that contributes a significant portion of the seminal plasma remains obscure. The present study tested the hypothesis "transient gestational- or neonatal-onset hypothyroidism may have duration dependent and lobe specific effect on androgen receptor (AR) status in the prostate glands of adult rats." METHODS Hypothyroidism was induced in pregnant and lactating rats by feeding 0.05% methimazole (MMI) through drinking water during fetal and neonatal milestones of testicular and prostatic development. Pregnant dams had MMI exposure from 9th day post-coitum (dpc) to 14 dpc (group II) or 21 dpc (group III). Lactating mothers had MMI exposure from day 1 post-partum (dpp) to 14 dpp (group IV) or up to 29 dpp (group V). AR status in the dorsolateral and ventral prostate lobes (DLP and VP) of the pups was assessed by RT-PCR, western blot and radio receptor assay. RESULTS AR mRNA expression consistently decreased in the DLP of all groups, whereas it increased in VP of group III and V rats. AR protein consistently decreased in both DLP and VP of all experimental rats. AR nuclear ligand-binding activity diminished in groups II and IV, whereas it increased in groups III and V. CONCLUSION The results obtained support the proposed hypothesis and indicate that an optimum thyroid activity during pre- and neonatal period determines AR status in the prostate glands at adulthood.
Collapse
Affiliation(s)
- Michael Maria Aruldhas
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, Tamil Nadu, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Giuffrida D, Perdichizzi A, Giuffrida MC, La Vignera S, D'Agata R, Vicari E, Calogero AE. Does prolactin induce apoptosis? Evidences in a prostate cancer in vitro model. J Endocrinol Invest 2010; 33:313-7. [PMID: 20009491 DOI: 10.1007/bf03346592] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Prolactin (PRL) regulates prostate growth and differentiation. Some studies have suggested that PRL has a pro-apoptotic effect on a myeloma cell line and in newt spermatogonia. The proliferative effect of PRL on prostate cancer cell lines is, however, a controversial area. AIM On this account, we evaluated the effects of PRL on the prostate cancer cell lines LNCaP and PC3 apoptosis and proliferation. MATERIALS AND METHODS LNCaP and PC3 cells were exposed to increasing concentrations of PRL for 24, 48, 72 and 96 hours. Staining with propidium iodide (PI) and TUNEL assay followed by flow cytometry were used to detect apoptosis. LNCaP and PC3 proliferation was assessed by optical microscopy counting. RESULTS PRL induced a dose-dependent decrease of DNA content and an increase of DNA fragmentation in LNCaP after 96 hours of incubation. These effects were observed with physiological concentrations of PRL and were counteracted by a prolactin receptor antagonist. On the other hand, PRL did not have any effect on DNA content or fragmentation in PC3 cells. No effect of PRL on LNCaP and PC3 proliferation was found. CONCLUSIONS This study indicates that PRL induces apoptosis in the androgen-responsive cell line LNCaP, whereas no effect was observed in the androgen-insensitive PC3 cell line. These findings suggest that androgen responsiveness may be required for PRL to be effective on prostatic cells.
Collapse
Affiliation(s)
- D Giuffrida
- Department of Biomedical Sciences, University of Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
13
|
Anbalagan J, Sashi AM, Vengatesh G, Stanley JA, Neelamohan R, Aruldhas MM. Mechanism underlying transient gestational-onset hypothyroidism-induced impairment of posttesticular sperm maturation in adult rats. Fertil Steril 2010; 93:2491-7. [PMID: 20303481 DOI: 10.1016/j.fertnstert.2010.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 01/28/2010] [Accepted: 02/01/2010] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To understand the mechanism underlying gestational-onset hypothyroidism-induced male infertility. DESIGN Controlled laboratory study. SETTING Research laboratory in a university department of endocrinology. ANIMAL(S) Wistar rat. INTERVENTION(S) Pregnant rats were exposed to methimazole from embryonic days 9 to 14, 18, and 21, covering specific fetal periods of differentiation and development of male reproductive tract organs. MAIN OUTCOME MEASURE(S) Fertility of male rats was assessed by testing sperm count, forward motility, and in vivo fertilizing ability. Secretory activity of the epididymis was evaluated by quantifying sialic acid, carnitine, and glycerylphosphorylcholine. Bioavailability of androgens was assessed by quantifying testosterone in serum and testicular interstitial fluid and epididymal 5alpha-reductase activity/mRNA expression. Androgen receptor (AR) status in the epididymis was tested by detecting the expression levels of its mRNA and protein, as well as ligand binding activity. Data were analyzed statistically by one-way analysis of variance. RESULT(S) Gestational exposure to methimazole decreased sperm forward motility, in vivo fertilizing ability, bioavailability of androgens, AR status, and secretory activity of the epididymis in adult rats. CONCLUSION(S) Transient gestational-onset hypothyroidism affects male fertility by impairing posttesticular sperm maturation process in the epididymis, owing to subnormal androgen(s) bioavailability, AR expression, and AR functional activity.
Collapse
Affiliation(s)
- Jaganathan Anbalagan
- Department of Endocrinology, Dr. A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | | | | | | | | | | |
Collapse
|
14
|
Hoffman JR, Kraemer WJ, Bhasin S, Storer T, Ratamess NA, Haff GG, Willoughby DS, Rogol AD. Position stand on androgen and human growth hormone use. J Strength Cond Res 2009; 23:S1-S59. [PMID: 19620932 DOI: 10.1519/jsc.0b013e31819df2e6] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hoffman, JR, Kraemer, WJ, Bhasin, S, Storer, T, Ratamess, NA, Haff, GG, Willoughby, DS, and Rogol, AD. Position stand on Androgen and human growth hormone use. J Strength Cond Res 23(5): S1-S59, 2009-Perceived yet often misunderstood demands of a sport, overt benefits of anabolic drugs, and the inability to be offered any effective alternatives has fueled anabolic drug abuse despite any consequences. Motivational interactions with many situational demands including the desire for improved body image, sport performance, physical function, and body size influence and fuel such negative decisions. Positive countermeasures to deter the abuse of anabolic drugs are complex and yet unclear. Furthermore, anabolic drugs work and the optimized training and nutritional programs needed to cut into the magnitude of improvement mediated by drug abuse require more work, dedication, and preparation on the part of both athletes and coaches alike. Few shortcuts are available to the athlete who desires to train naturally. Historically, the NSCA has placed an emphasis on education to help athletes, coaches, and strength and conditioning professionals become more knowledgeable, highly skilled, and technically trained in their approach to exercise program design and implementation. Optimizing nutritional strategies are a vital interface to help cope with exercise and sport demands (). In addition, research-based supplements will also have to be acknowledged as a strategic set of tools (e.g., protein supplements before and after resistance exercise workout) that can be used in conjunction with optimized nutrition to allow more effective adaptation and recovery from exercise. Resistance exercise is the most effective anabolic form of exercise, and over the past 20 years, the research base for resistance exercise has just started to develop to a significant volume of work to help in the decision-making process in program design (). The interface with nutritional strategies has been less studied, yet may yield even greater benefits to the individual athlete in their attempt to train naturally. Nevertheless, these are the 2 domains that require the most attention when trying to optimize the physical adaptations to exercise training without drug use.Recent surveys indicate that the prevalence of androgen use among adolescents has decreased over the past 10-15 years (). The decrease in androgen use among these students may be attributed to several factors related to education and viable alternatives (i.e., sport supplements) to substitute for illegal drug use. Although success has been achieved in using peer pressure to educate high school athletes on behaviors designed to reduce the intent to use androgens (), it has not had the far-reaching effect desired. It would appear that using the people who have the greatest influence on adolescents (coaches and teachers) be the primary focus of the educational program. It becomes imperative that coaches provide realistic training goals for their athletes and understand the difference between normal physiological adaptation to training or that is pharmaceutically enhanced. Only through a stringent coaching certification program will academic institutions be ensured that coaches that they hire will have the minimal knowledge to provide support to their athletes in helping them make the correct choices regarding sport supplements and performance-enhancing drugs.The NSCA rejects the use of androgens and hGH or any performance-enhancing drugs on the basis of ethics, the ideals of fair play in competition, and concerns for the athlete's health. The NSCA has based this position stand on a critical analysis of the scientific literature evaluating the effects of androgens and human growth hormone on human physiology and performance. The use of anabolic drugs to enhance athletic performance has become a major concern for professional sport organizations, sport governing bodies, and the federal government. It is the belief of the NSCA that through education and research we can mitigate the abuse of androgens and hGH by athletes. Due to the diversity of testosterone-related drugs and molecules, the term androgens is believed to be a more appropriate term for anabolic steroids.
Collapse
Affiliation(s)
- Jay R Hoffman
- Department of Health and Exercise Science, The College of New Jersey, Ewing, 08628, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Ohlsson C, Mohan S, Sjögren K, Tivesten A, Isgaard J, Isaksson O, Jansson JO, Svensson J. The role of liver-derived insulin-like growth factor-I. Endocr Rev 2009; 30:494-535. [PMID: 19589948 PMCID: PMC2759708 DOI: 10.1210/er.2009-0010] [Citation(s) in RCA: 300] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IGF-I is expressed in virtually every tissue of the body, but with much higher expression in the liver than in any other tissue. Studies using mice with liver-specific IGF-I knockout have demonstrated that liver-derived IGF-I, constituting a major part of circulating IGF-I, is an important endocrine factor involved in a variety of physiological and pathological processes. Detailed studies comparing the impact of liver-derived IGF-I and local bone-derived IGF-I demonstrate that both sources of IGF-I can stimulate longitudinal bone growth. We propose here that liver-derived circulating IGF-I and local bone-derived IGF-I to some extent have overlapping growth-promoting effects and might have the capacity to replace each other (= redundancy) in the maintenance of normal longitudinal bone growth. Importantly, and in contrast to the regulation of longitudinal bone growth, locally derived IGF-I cannot replace (= lack of redundancy) liver-derived IGF-I for the regulation of a large number of other parameters including GH secretion, cortical bone mass, kidney size, prostate size, peripheral vascular resistance, spatial memory, sodium retention, insulin sensitivity, liver size, sexually dimorphic liver functions, and progression of some tumors. It is clear that a major role of liver-derived IGF-I is to regulate GH secretion and that some, but not all, of the phenotypes in the liver-specific IGF-I knockout mice are indirect, mediated via the elevated GH levels. All of the described multiple endocrine effects of liver-derived IGF-I should be considered in the development of possible novel treatment strategies aimed at increasing or reducing endocrine IGF-I activity.
Collapse
Affiliation(s)
- Claes Ohlsson
- Division of Endocrinology, Institute of Medicine, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Chen Y, Sood S, Krishnamurthy VMR, Rotwein P, Rabkin R. Endotoxin-induced growth hormone resistance in skeletal muscle. Endocrinology 2009; 150:3620-6. [PMID: 19443577 PMCID: PMC2717874 DOI: 10.1210/en.2008-1703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammation-induced skeletal muscle wasting is a serious clinical problem and arises in part because of resistance to GH-stimulated IGF-I expression. Although it is established that in the liver, resistance develops because of impaired signaling through the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5 (STAT5) transduction pathway, together with a more distal defect in STAT5 DNA-binding activity, the situation in skeletal muscle is unclear. Accordingly, we set out to characterize the mechanisms behind the skeletal muscle resistance to GH in rats with acute inflammation induced by endotoxin. Endotoxin caused significant declines in GH-stimulated STAT5a/b phosphorylation and IGF-I gene expression, and this occurred despite a lack of change in signaling protein levels or phosphorylation of JAK2. In whole muscle, GH-stimulated phospho-STAT5a/b levels were reduced by half, and in the nucleus, phospho-STAT5b levels were similarly reduced. Furthermore, the binding of phosphorylated STAT5b to DNA was reduced and to a similar extent to the reduction in nuclear phosphorylated STAT5b. Interestingly, GH-induced androgen receptor gene expression was also suppressed. Thus, it appears that skeletal muscle resistance to GH-stimulated IGF-I expression in acute endotoxemia arises from a defect in STAT5b signaling, with a proportionate reduction in STAT5b DNA binding. Finally, it appears that resistance to GH-induced androgen receptor expression also develops and, together with the attenuated GH-induced IGF-I expression, likely plays an important role in the muscle wasting that arises in endotoxin-induced inflammation.
Collapse
Affiliation(s)
- Yu Chen
- Department of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
17
|
Action, localization and structure-function relationship of growth factors and their receptors in the prostate. ACTA ACUST UNITED AC 2009. [DOI: 10.1017/s0962279900001265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Whereas the direct action of sex steroids, namely of androgens, on prostate cell division was questioned as early as in the 1970s, and remains so, the interest in prostatic growth factors (GFs) is rather recent but has expanded tremendously in the last five years. This lag period can be partly explained by the fact that, at the time, androgen receptors had just been discovered, and newly developed hormonal regimens or strategies to treat patients with prostate carcinoma (PCa) or epithelioma had generated great enthusiasm and hopes in the medical and scientific community. Another point to consider was the difficulty in maintaining prostate tissues in organ cultures and the relative novelty of culturing prostate epithelial cells in monolayers. Failures of sex steroids to elicit a direct positive response on prostate cell divisionin vitro, as seenin vivo, were interpreted as resulting from inappropriate models or culture conditions. However, the increasing number of reports confirming the lack of mitogenic activity of sex steroidsin vitro, coupled with the powerful mitogenic activity of GFs displayed in other systems, the discovery of GF receptors (GF-Rs), and the elucidation of their signalling pathways showing sex steroid receptors as potential substrates of GF-activated protein kinases gradually led to an increased interest in the putative role of GFs in prostate physiopathology. Of utmost importance was the recognition that hormone refractiveness was responsible for PCa progression, and for the poor outcome of patients with advanced disease under endocrine therapies. This problem remains a major issue and it raises several key questions that need to be solved at the fundamental and clinical levels.
Collapse
|
18
|
Abstract
Stromal-epithelial interactions mediated by paracrine signaling mechanisms dictate prostate development and progression of prostate cancer. The regulatory role of androgens in both the prostate stromal and epithelial compartments set the prostate apart from many other organs and tissues with regard to gene targeting. The identification of androgen-dependent prostate epithelial promoters has allowed successful gene targeting to the prostate epithelial compartment. Currently, there are no transgenic mouse models available to specifically alter gene expression within the prostate stromal compartment. As a primary metastatic site for prostate cancer is bone, the functional dissection of the bone stromal compartment is important for understanding stromal-epithelial interactions associated with metastatic tumor growth. Use of currently available methodologies for the expression or deletion of gene expression in recent research studies has advanced our understanding of the stroma. However, the complexity of stromal heterogeneity within the prostate remains a challenge to obtaining compartment or cell-lineage-specific in vivo models necessary for furthering our understanding of prostatic developmental, benign, tumorigenic, and metastatic growth.
Collapse
Affiliation(s)
- Roger S Jackson
- Department of Urologic Surgery, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232-2765, USA
| | | | | |
Collapse
|
19
|
Sullivan DA. Tearful relationships? Sex, hormones, the lacrimal gland, and aqueous-deficient dry eye. Ocul Surf 2007; 2:92-123. [PMID: 17216082 DOI: 10.1016/s1542-0124(12)70147-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sex and the endocrine system exert a significant influence on the physiology and pathophysiology of the lacrimal gland. The purpose of this article is to briefly review the nature and magnitude of these interactions between sex, hormones and lacrimal tissue, and to address how they may relate to the pathogenesis of aqueous-deficient dry eye. Towards this end, this article has a 3-fold approach: first, to summarize the influence of androgens, estrogens, glucocorticoids, mineralocorticoids, retinoic acid, prolactin, alpha-melanocyte stimulating hormone, adrenocorticotropic hormone, luteinizing hormone, follicle-stimulating hormone, growth hormone, thyroid-stimulating hormone, arginine vasopressin, oxytocin, thyroxine, parathyroid hormone, insulin, glucagon, melatonin, human chorionic gonadotropin and cholecystokinin on the structure and function of the lacrimal gland; second, to discuss the mechanism of action of each hormone on lacrimal tissue; and third, to discuss the clinical relevance of the endocrine-lacrimal gland interrelationship, with a particular focus on each hormone's role (i.e. if relevant) in the development of aqueous-tear deficiency.
Collapse
Affiliation(s)
- David A Sullivan
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
20
|
Meinbach DS, Lokeshwar BL. Insulin-like growth factors and their binding proteins in prostate cancer: Cause or consequence?☆. Urol Oncol 2006; 24:294-306. [PMID: 16818181 DOI: 10.1016/j.urolonc.2005.12.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2005] [Revised: 12/27/2005] [Accepted: 12/28/2005] [Indexed: 12/29/2022]
Abstract
Insulin-like growth factors (IGFs) promote growth and survival of many types of tumor cells. Epidemiologic studies have implicated carcinogenesis with high levels of IGFs in circulation or in tissues. The levels of IGF binding proteins (IGFBPs) have been associated with reduced risk for prostate and other cancers. Experimental studies have implicated high levels of IGF-I directly and IGFBP-3 inversely in prostate cancer growth, survival, and progression. However, recent evidence suggests a much weaker association of IGF-I with prostate cancer development and a stronger antagonistic association of IGFBP-3 with prostate cancer progression. Considering the clonal heterogeneity and unpredictable progression pattern of prostate cancer, the role of any single growth factor or its regulator (IGFBP) as a single determining factor is limited. This review is a critical appraisal of the role of IGFs, IGFBP, and IGF-I receptor (the IGF axis) in both experimental and clinical prostate cancer genesis and progression.
Collapse
Affiliation(s)
- David S Meinbach
- Department of Urology, Leonard Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | | |
Collapse
|
21
|
Fuhrman B, Barba M, Schünemann HJ, Hurd T, Quattrin T, Cartagena R, Carruba G, Muti P. Basal growth hormone concentrations in blood and the risk for prostate cancer: a case-control study. Prostate 2005; 64:109-15. [PMID: 15666390 DOI: 10.1002/pros.20203] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To assess the relationship between basal serum growth hormone (GH) levels and prostate cancer risk. METHODS We conducted a population-based case-control study; cases included 68 men, aged 45-85 years, diagnosed with incident, primary, histologically confirmed, and clinically apparent (stage B and higher) prostate cancer. Controls included 240 men, frequency matched on age and residential area. Age, race, BMI, waist circumference, history of enlarged prostate, education, and current smoking status, were all considered as possible confounders. RESULTS We found a statistically significant trend of decreasing prostate cancer risk across increasing GH quintiles, in both crude (OR: 0.31, 95% CI: 0.12-0.83, P for trend 0.01) and adjusted models (OR: 0.35, 95% CI: 0.12-1.05, P for trend 0.03), in the highest compared to the lowest quintile, respectively. CONCLUSIONS Lower basal levels of GH in serum are associated with increased prostate cancer risk. The inverse association may be explained by the negative feedback loop generated by IGF-1 produced by the tumor on GH secretion.
Collapse
Affiliation(s)
- Barbara Fuhrman
- Department of Social and Preventive Medicine, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Weiss-Messer E, Merom O, Adi A, Karry R, Bidosee M, Ber R, Kaploun A, Stein A, Barkey RJ. Growth hormone (GH) receptors in prostate cancer: gene expression in human tissues and cell lines and characterization, GH signaling and androgen receptor regulation in LNCaP cells. Mol Cell Endocrinol 2004; 220:109-23. [PMID: 15196705 DOI: 10.1016/j.mce.2004.03.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2004] [Revised: 03/03/2004] [Accepted: 03/12/2004] [Indexed: 11/18/2022]
Abstract
Various hormones and growth factors have been implicated in progression of prostate cancer, but their role and the underlying molecular mechanism(s) involved remain poorly understood. In this study, we investigated the role of human growth hormone (GH) and its receptor (GHR) in human prostate cancer. We first demonstrated mRNA expression of GHR and of its exon 9-truncated isoform (GHR(tr)) in benign prostate hyperplasia (BPH) and prostate adenocarcinoma patient tissues, as well as in LNCaP, PC3 and DU145 human prostate cancer cell lines. GHR mRNA levels were 80% higher and GHR(tr) only 25% higher, in the carcinoma tissues than in BPH. Both isoforms were also expressed in LNCaP and PC3 cell lines and somewhat less so in DU145 cells. The LNCaP cell GHR protein was further characterized, on the basis of its M(r) of 120kDa, its binding to two different GHR monoclonal antibodies, its high affinity and purely somatogenic binding to (125)I-hGH and its ability to secrete GH binding protein, all characteristic of a functional GHR. Furthermore, GH induced rapid, time- and dose-dependent signaling events in LNCaP cells, including phosphorylation of JAK2 tyrosine kinase, of GHR itself and of STAT5A (JAK2-STAT5A pathway), of p42/p44 MAPK and of Akt/PKB. No effect of GH (72h) could be shown on basal or androgen-induced LNCaP cell proliferation nor on PSA secretion. Interestingly, however, GH caused a rapid (2-12h) though transient striking increase in immunoreactive androgen receptor (AR) levels (< or =5-fold), followed by a slower (24-48h) reduction (< or = 80%), with only modest parallel changes in serine-phosphorylated AR. In conclusion, the GH-induced activation of signaling pathways, its effects on AR protein in LNCaP cells and the isoform-specific regulation of GHR in prostate cancer patient tissues, suggest that GH, most likely in concert with other hormones and growth factors, may play an important role in progression of human prostate cancer.
Collapse
Affiliation(s)
- Esther Weiss-Messer
- Department of Pharmacology, Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Technion, P.O.B. 9649, Haifa 31096, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Binart N, Melaine N, Pineau C, Kercret H, Touzalin AM, Imbert-Bolloré P, Kelly PA, Jégou B. Male reproductive function is not affected in prolactin receptor-deficient mice. Endocrinology 2003; 144:3779-82. [PMID: 12933648 DOI: 10.1210/en.2003-0409] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mice with a targeted disruption of the prolactin (PRL) receptor gene were used to study the physiological role of PRL in the control of the male reproductive function. Fertility parameters as well as body and reproductive organ weights (epididymis and testes) were unaffected in PRL receptor knockout mice. Testicular histology and sperm reserves were also normal. Compared with wild-type animals, knockout mice had no significant difference in basal plasma LH, FSH, and testosterone levels, and the weight of seminal vesicles and prostate was unaffected. Moreover, no alteration was detected in human chorionic gonadotropin-induced testosterone levels. It is concluded that the absence of PRL signaling is not detrimental to male testicular function and to fertility in the mouse.
Collapse
Affiliation(s)
- Nadine Binart
- Hormone Targets, Institut National de la Santé et de la Recherche Médicale Unité 584, Faculté de Médecine Necker-Enfants Malades, 75015 Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Joakim Larsson DG, Sperry TS, Thomas P. Regulation of androgen receptors in Atlantic croaker brains by testosterone and estradiol. Gen Comp Endocrinol 2002; 128:224-30. [PMID: 12392696 DOI: 10.1016/s0016-6480(02)00503-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A nuclear androgen receptor (AR1), distinctly different from the mammalian AR, has previously been identified in the brain of the Atlantic croaker, Micropogonias undulatus. Interestingly, brain AR1 levels were higher in gonadally recrudesced than in regressed fish. Therefore, the possible involvement of gonadal steroids in the regulation of brain AR1 levels was investigated in the present study. Saturation analysis of [3H]testosterone binding showed that brain AR1 levels were significantly reduced (2-3-fold) in either the nuclear or cytosolic fractions of both males and females three weeks after gonadectomy. Implantation of gonadectomized females with testosterone or estradiol-17beta one week prior to sampling resulted in physiological plasma steroid concentrations and restored brain AR1 levels to 60-105% of those in intact or sham-operated fish. These results suggest that gonadal factors, including both androgens and estrogens, are involved in the physiological regulation of brain androgen receptors in a teleost species during the reproductive cycle.
Collapse
Affiliation(s)
- D G Joakim Larsson
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, USA. joakim.larson.@fysiologi.gu.se
| | | | | |
Collapse
|
25
|
Kindblom J, Dillner K, Ling C, Törnell J, Wennbo H. Progressive prostate hyperplasia in adult prolactin transgenic mice is not dependent on elevated serum androgen levels. Prostate 2002; 53:24-33. [PMID: 12210477 DOI: 10.1002/pros.10113] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Transgenic mice overexpressing the rat prolactin (PRL) gene under control of the metallothionein-1 promoter (Mt-1) develop a dramatic prostatic enlargement. These animals also display significantly elevated testosterone serum levels. In this study, we aim to clarify the role of circulating androgen levels in the promotion of abnormal prostate growth in the adult PRL transgenic mouse prostate. METHODS Prostate morphology and androgen-receptor distribution patterns were analyzed in castrated and testosterone substituted adult PRL transgenic and in wild-type males. RESULTS Progressive prostatic hyperplasia in adult PRL transgenic males was not affected by substitution to serum testosterone levels corresponding to wild-type. Furthermore, prolonged testosterone treatment in adult wild-type males did not produce any significant changes in prostate growth or morphology compared with wild-type controls. Immunohistochemical studies revealed a significantly increased proportion of androgen receptor positive epithelial cells in all lobes of the PRL transgenic prostate versus wild-type. CONCLUSION The present study demonstrates that progressive prostate hyperplasia in adult PRL transgenic mice is not dependent on elevated serum androgen levels. Furthermore, prolonged androgen treatment in adult wild-type male mice appears to have no significant effect on prostate growth. In addition, our results suggest that prolonged hyperprolactinemia results in changes in prostate epithelial and stromal cell androgen receptor distribution.
Collapse
Affiliation(s)
- Jon Kindblom
- Department of Pharmacology and Physiology, Göteborg University, Sweden
| | | | | | | | | |
Collapse
|
26
|
Dillner K, Kindblom J, Flores-Morales A, Pang ST, Törnell J, Wennbo H, Norstedt G. Molecular characterization of prostate hyperplasia in prolactin-transgenic mice by using cDNA representational difference analysis. Prostate 2002; 52:139-49. [PMID: 12111705 DOI: 10.1002/pros.10102] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Transgenic mice overexpressing the rat prolactin (PRL) gene develop a dramatic enlargement of the prostate gland. The objective of this study was to characterize the molecular mechanisms in the prostate of importance for the prostate hyperplasia seen in these transgenic mice. METHODS cDNA representational difference analysis (cDNA RDA) was used to isolate differentially expressed transcripts in the prostate hyperplasia of the transgenic mice compared with wildtype littermates. Furthermore, cDNA microarray analysis was used to verify the RDA output. RESULTS Here we report 10 transcripts, some of them described to be involved in proliferation and apoptosis, which are differentially expressed in the enlarged transgenic prostates compared with controls. CONCLUSION The identified differentially expressed transcripts presented herein supports molecular similarities between the prostate hyperplasia of PRL-transgenic mice and human BPH that may contribute to explain the molecular basis of prostate hyperplasia.
Collapse
Affiliation(s)
- Karin Dillner
- Department of Pharmacology and Physiology, Göteborg University, Göteborg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
27
|
Chopin LK, Veveris-Lowe TL, Philipps AF, Herington AC. Co-expression of GH and GHR isoforms in prostate cancer cell lines. Growth Horm IGF Res 2002; 12:126-136. [PMID: 12175650 DOI: 10.1054/ghir.2002.0271] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Prostate cancer is a significant cause of morbidity and mortality in Western males. While it is known that androgens play a central role in prostate cancer development and progression, other hormones and growth factors are also involved in prostate growth. Insulin-like growth factor-I (IGF-I) plasma levels have been associated with prostate cancer risk, and growth hormone (GH), a major factor regulating IGF levels, also appears to have a role in prostate cancer cell growth. Most significantly, GH has been shown to increase the rate of cell proliferation in prostate cancer cell lines. We have now demonstrated the co-expression of GH and GH receptor (GHR) mRNA isoforms in the ALVA41, PC3, DU145, LNCaP prostate cancer cells by reverse transcription polymerase chain reaction. Sequence analysis has confirmed that these cell lines express the pituitary form of GH mRNA and also the placental mRNA isoform. These prostate cancer cell lines also express the full-length mRNA for the GHR and the exon 3 deleted isoform. We have also demonstrated the presence of GH and GHR proteins in these cell lines by immunohistochemistry. GH expression has not been described previously in human prostate cancer cells. The co-expression of GH and its receptor would enable an autocrine-paracrine pathway to exist in the prostate that would be capable of stimulating prostate growth, either directly via the GHR or indirectly via IGF production. The GH axis in the prostate could therefore be an important additional target for the future development of prostate cancer therapies.
Collapse
Affiliation(s)
- L K Chopin
- Centre for Molecular Biotechnology, Queensland University of Technology, GPO Box 2434, Brisbane, Q 4001, Australia.
| | | | | | | |
Collapse
|
28
|
Hull KL, Harvey S. Growth hormone: roles in male reproduction. Endocrine 2000; 13:243-50. [PMID: 11216634 DOI: 10.1385/endo:13:3:243] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2000] [Revised: 05/22/2000] [Accepted: 05/22/2000] [Indexed: 11/11/2022]
Abstract
Growth hormone (GH), as its name suggests, is obligatory for growth and development. It is, however, also required for sexual differentiation and pubertal maturation and participates in gonadal steroidogenesis and gametogenesis. These roles are likely to reflect the endocrine actions of pituitary GH, directly at gonadal sites and indirectly via hepatic insulin-like growth factor-1. However, because GH is also produced in gonadal tissues, it may act in paracrine or autocrine ways to regulate local processes that are strategically regulated by pituitary GH. The concept that GH is a major regulator of male reproduction is the focus of this review.
Collapse
Affiliation(s)
- K L Hull
- Bishop's University, Lennoxville, Quebec, Canada
| | | |
Collapse
|
29
|
Untergasser G, Rumpold H, Hermann M, Dirnhofer S, Jilg G, Berger P. Proliferative disorders of the aging human prostate: involvement of protein hormones and their receptors. Exp Gerontol 1999; 34:275-87. [PMID: 10363793 DOI: 10.1016/s0531-5565(98)00063-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The majority of elderly men is affected by benign and malignant diseases of the prostate. Both proliferative disorders, i.e., benign hyperplasia of the prostate (BPH) and prostate cancer (PCa)-which has recently emerged as the most common male malignancy in industrialized countries-seem to be governed by endocrine factors such as sex steroid hormones, but auto/paracrine factors are involved as well. Age-related changes in levels and ratios of endocrine factors as androgens, estrogens, gonadotropins, and prolactin (PRL) and changes in the balance between auto/paracrine growth-stimulatory and growth-inhibitory factors such as insulin-like growth factors (IGFs), epidermal growth factor (EGF), nerve growth factor (NGF), IGF-binding proteins (IGFBPs), and transforming growth factor beta (TGFbeta) are meant to be responsible for abnormal prostatic growth. We investigated the existence of putative local regulatory circuits involving the protein hormones, human growth hormone (hGH), human placental lactogen (hPL), and hPRL, and their corresponding receptors in prostatic tissue specimens (transurethral resections of the prostate, TURP; n = 11), in the prostatic cancer cell lines PC3, Du145, LnCap, a virus-transformed BPH cell line (BPH-1), and in a normal healthy prostate by RT-PCRs and highly specific and sensitive immunofluorometric assays (IFMA). Neither hPRL nor hGH was detected at the mRNA or protein levels in prostatic tissue and cell lines, with the exception of 2 of 11 prostatic TURP-samples, which showed weak expression of the PL-A/B genes. PRL- and GH-receptors were expressed in all normal and pathological prostatic specimens. Surprisingly, PRL-receptor expression was not detectable in prostatic cancer cell lines. The trophic effects of exogenous hGH, hPL, and hPRL were investigated by cell proliferation assays (WST-I) in prostatic primary cell cultures and PCa cell lines. hGH significantly (p < 0.005) increased cell proliferation up to 138+/-3.2% (1 nM hGH), while hPL and hPRL revealed only moderate effects. Our data suggest that local auto/paracrine networks of protein hormone actions are not involved in the pathology of BPH or prostatic cancer. On the other hand, systemic pituitary-derived hGH can increase the proliferative response of BPH and PCa, acting directly on the target organ prostate, via the hGH-R. In this case, envisaged GH substitution in elderly people must be viewed at with caution because age-related declines in GH/IGF-I could act as a protective mechanism against abnormal cell growth.
Collapse
Affiliation(s)
- G Untergasser
- Institute for Biomedical Aging Research of the Austrian Academy of Sciences, Innsbruck
| | | | | | | | | | | |
Collapse
|
30
|
Leav I, Merk FB, Lee KF, Loda M, Mandoki M, McNeal JE, Ho SM. Prolactin receptor expression in the developing human prostate and in hyperplastic, dysplastic, and neoplastic lesions. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:863-70. [PMID: 10079264 PMCID: PMC1866401 DOI: 10.1016/s0002-9440(10)65333-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/02/1998] [Indexed: 10/18/2022]
Abstract
In situ hybridization and immunohistochemistry were used to localize and compare the expression of the long form of the human prolactin receptor in fetal, prepubertal, and adult prostate. Results were then compared with hyperplastic, dysplastic, and neoplastic lesions. Both receptor message and protein were predominately localized in epithelial cells of the fetal, neonatal, prepubertal, and normal adult prostate. In hyperplastic lesions the expression of the receptor was unchanged with respect to normal epithelial cells. Irrespective of grade, markedly enhanced expression of the receptor was evident in dysplastic lesions. In lower Gleason grade carcinomas the intensity of receptor signal at the message and protein levels approximated that found in normal prostatic epithelium. However, in foci within higher grade cancers, receptor expression appeared diminished. Results from our study suggest that prolactin action plays a role in the development and maintenance of the human prostate and may also participate in early neoplastic transformation of the gland. Diminution of receptor expression in high grade neoplasms could reflect the emergence of a population of cells that are no longer responsive to the peptide hormone.
Collapse
Affiliation(s)
- I Leav
- Department of Pathology, Tufts University, School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
BACKGROUND Although essential, androgens alone are not sufficient to induce normal growth and functionality of the prostate. Nonandrogenic hormones must also be involved in the proliferation of the prostate cancer cells which do not respond to antiandrogenic therapy and which thus become androgen-independent. Prolactin, but also growth hormone and luteinizing hormone, are potentially able to act on both normal and abnormal prostatic cells. METHODS In this review we summarize data from the literature concerning the physiological and pathological implications of prolactin, growth hormone, and luteinizing hormone on the prostate. RESULTS In rodent prostates, prolactin and growth hormone can induce a variety of effects independently of androgens (e.g., transactivation of certain genes, or synthesis of the major secretion products). Moreover, hyperprolactinemia is responsible for inflammation and dysplasia of the gland, while growth hormone promotes the development of prostate tumors in vivo in the mouse and rat. Growth hormone acts on the gland directly, through prostatic growth hormone receptors, and/or indirectly via the stimulation of insulin-like growth factor-I (IGF-I) synthesis in the liver. Luteinizing hormone receptor is expressed in rat and human prostates. Luteinizing hormone increases the amount of various transcripts in the rat prostate through an androgen-independent pathway. CONCLUSIONS Prolactin, growth hormone, and luteinizing hormone, alone or synergistically with androgens, play physiologically significant roles in the normal prostate. The involvement of these hormones in the development of benign prostatic hyperplasia and prostatic carcinoma is an issue that needs to be addressed.
Collapse
Affiliation(s)
- E Reiter
- Station de Physiologie de la Reproduction des Mammifères Domestiques, Institut National de la Recherche Agronomique, URA CNRS 1291, Nouzilly, France.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Sullivan DA, Wickham LA, Rocha EM, Kelleher RS, da Silveira LA, Toda I. Influence of gender, sex steroid hormones, and the hypothalamic-pituitary axis on the structure and function of the lacrimal gland. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998; 438:11-42. [PMID: 9634860 DOI: 10.1007/978-1-4615-5359-5_2] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- D A Sullivan
- Schepens Eye Research Institute, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
33
|
Bole-Feysot C, Goffin V, Edery M, Binart N, Kelly PA. Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocr Rev 1998; 19:225-68. [PMID: 9626554 DOI: 10.1210/edrv.19.3.0334] [Citation(s) in RCA: 1047] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PRL is an anterior pituitary hormone that, along with GH and PLs, forms a family of hormones that probably resulted from the duplication of an ancestral gene. The PRLR is also a member of a larger family, known as the cytokine class-1 receptor superfamily, which currently has more than 20 different members. PRLRs or binding sites are widely distributed throughout the body. In fact, it is difficult to find a tissue that does not express any PRLR mRNA or protein. In agreement with this wide distribution of receptors is the fact that now more than 300 separate actions of PRL have been reported in various vertebrates, including effects on water and salt balance, growth and development, endocrinology and metabolism, brain and behavior, reproduction, and immune regulation and protection. Clearly, a large proportion of these actions are directly or indirectly associated with the process of reproduction, including many behavioral effects. PRL is also becoming well known as an important regulator of immune function. A number of disease states, including the growth of different forms of cancer as well as various autoimmune diseases, appear to be related to an overproduction of PRL, which may act in an endocrine, autocrine, or paracrine manner, or via an increased sensitivity to the hormone. The first step in the mechanism of action of PRL is the binding to a cell surface receptor. The ligand binds in a two-step process in which site 1 on PRL binds to one receptor molecule, after which a second receptor molecule binds to site 2 on the hormone, forming a homodimer consisting of one molecule of PRL and two molecules of receptor. The PRLR contains no intrinsic tyrosine kinase cytoplasmic domain but associates with a cytoplasmic tyrosine kinase, JAK2. Dimerization of the receptor induces tyrosine phosphorylation and activation of the JAK kinase followed by phosphorylation of the receptor. Other receptor-associated kinases of the Src family have also been shown to be activated by PRL. One major pathway of signaling involves phosphorylation of cytoplasmic State proteins, which themselves dimerize and translocate to nucleus and bind to specific promoter elements on PRL-responsive genes. In addition, the Ras/Raf/MAP kinase pathway is also activated by PRL and may be involved in the proliferative effects of the hormone. Finally, a number of other potential mediators have been identified, including IRS-1, PI-3 kinase, SHP-2, PLC gamma, PKC, and intracellular Ca2+. The technique of gene targeting in mice has been used to develop the first experimental model in which the effect of the complete absence of any lactogen or PRL-mediated effects can be studied. Heterozygous (+/-) females show almost complete failure to lactate after the first, but not subsequent, pregnancies. Homozygous (-/-) females are infertile due to multiple reproductive abnormalities, including ovulation of premeiotic oocytes, reduced fertilization of oocytes, reduced preimplantation oocyte development, lack of embryo implantation, and the absence of pseudopregnancy. Twenty per cent of the homozygous males showed delayed fertility. Other phenotypes, including effects on the immune system and bone, are currently being examined. It is clear that there are multiple actions associated with PRL. It will be important to correlate known effects with local production of PRL to differentiate classic endocrine from autocrine/paracrine effects. The fact that extrapituitary PRL can, under some circumstances, compensate for pituitary PRL raises the interesting possibility that there may be effects of PRL other than those originally observed in hypophysectomized rats. The PRLR knockout mouse model should be an interesting system by which to look for effects activated only by PRL or other lactogenic hormones. On the other hand, many of the effects reported in this review may be shared with other hormones, cytokines, or growth factors and thus will be more difficult to study. (ABSTRACT TRUNCATED)
Collapse
Affiliation(s)
- C Bole-Feysot
- INSERM Unité 344-Endocrinologie Moléculaire, Faculté de Médecine Necker, Paris, France
| | | | | | | | | |
Collapse
|
34
|
Nixon AJ, Ford CA, Oldham JM, Pearson AJ. Localisation of insulin-like growth factor receptors in skin follicles of sheep (Ovis aries) and changes during an induced growth cycle. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART A, PHYSIOLOGY 1997; 118:1247-57. [PMID: 9505430 DOI: 10.1016/s0300-9629(97)00048-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pelage growth cycles are regulated by circulating prolactin in many mammals, but the intercellular mediators of this signaling are unknown. Binding sites for insulin-like growth factors (IGFs) were examined in sheep skin to show changes in distribution and abundance of IGF receptors associated with a prolactin stimulus and the subsequent hair follicle growth cycle. Follicle cycles were induced in New Zealand Wiltshire ewes by a surge in plasma prolactin following a 4-month period of prolactin suppression with bromocriptine. Eight treated and three control sheep were slaughtered at intervals over 43 days during the follicle growth cycle. At 12-20 days after the elevation of prolactin, wool follicles passed through brief catagen and telogen phases, followed by a return to anagen. IGF binding sites were localized in skin sections by incubation with 125I-IGF-I or 125I-IGF-II. Displacement with competitive binding inhibitors (unlabeled IGF-I, IGF-II, des(1-3)IGF-I, des(1-6)IGF-II, or insulin) and affinity cross-linking showed that these binding sites were predominantly IGF type 1 and type 2 (mannose-6-phosphate) receptors. The radioligands bound especially to follicle germinal cells and prekeratinocytes. Increases in specific binding of both radioligands were observed after the rise in prolactin, but prior to anatomical changes in follicles associated with cessation of growth. For IGF-I, highest binding density was observed during catagen in the germinal matrix and dermal papilla cells. For IGF-II, peak density occurred during late anagen/early catagen in the germinal matrix and during telogen in the dermal papilla. These cycle associated changes in receptor availability suggest that IGF receptors are involved in control of the wool growth.
Collapse
Affiliation(s)
- A J Nixon
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand.
| | | | | | | |
Collapse
|
35
|
Abstract
A number of tissues, including the brain, pituitary, immune system, placenta, mammary gland, and testis, may be self-contained units of GH regulation, production, and action. The production of GH and GH-releasing factors outside the hypothalamo-pituitary axis complements, rather than replaces, the traditional endocrine interactions between GH-releasing factors, GH, and its target tissues.
Collapse
Affiliation(s)
- S Harvey
- Department of Physiology, University of Alberta, Edmonton, Canada.
| | | |
Collapse
|
36
|
Tørring N, Vinter-Jensen L, Pedersen SB, Sørensen FB, Flyvbjerg A, Nexø E. Systemic administration of insulin-like growth factor I (IGF-I) causes growth of the rat prostate. J Urol 1997; 158:222-7. [PMID: 9186364 DOI: 10.1097/00005392-199707000-00075] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE To investigate the effects of insulin-like growth factor I (IGF-I) and epidermal growth factor (EGF) on the rat prostate. In addition, we investigated the effect of ornithine decarboxylase (ODC) inhibition with alpha-diflouromethylornitine (DFMO) on the expected growth of the prostate. MATERIALS AND METHODS Eight week old Wistar rats were allocated into groups of eight, receiving systemic treatment for 3 and 7 days with either IGF-I (400 micrograms/rat/day) or EGF (30 micrograms/rat/day) alone or in combination with DFMO. RESULTS Systemic treatment with IGF-I for 7 days resulted in a 29% (p < 0.01) increase in the mean wet weight of the ventral prostate. The mean weight of the dorsolateral lobe of the prostate increased by 39% (p < 0.05), and the seminal vesicle and coagulating gland increased 69% (p < 0.05) compared to controls. The ODC-activity in the prostate was significantly increased by IGF-I after 3 days of treatment, and administration of IGF-I concomitantly with DFMO significantly inhibited ODC activity and the weight increase of the prostate. Stereological examination of the prostate in the IGF-I-treated animals showed growth of the epithelial component of the gland. Systemic treatment with EGF did not affect the mean weight of the prostate or the seminal vesicle compared to controls. CONCLUSION The results demonstrate that treatment with IGF-I but not EGF for 7 days induces profound growth of the rat prostate and the seminal vesicle, and that the growth is dependent on an intact ODC-activity.
Collapse
Affiliation(s)
- N Tørring
- Department of Clinical Biochemistry-KH, Aarhus University, Denmark
| | | | | | | | | | | |
Collapse
|
37
|
Kumar VL, Majumder PK, Kumar V. Androgen deprivation causes up-regulation of androgen receptor transcript in the rat prostate. Mol Cell Biochem 1997; 171:133-8. [PMID: 9201706 DOI: 10.1023/a:1006805503284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have studied the effect of androgenic deprivation on the level of androgen receptor transcript in the rat ventral prostate. The rats were treated with estradiol benzoate, flutamide and [D Trp6, des Gly10]gonadotropin releasing hormone (GnRH) for different time periods. These treatments produced a significant decrease in the weight of prostate. Total RNA isolated from the ventral prostates was hybridized with the cDNA probe for androgen receptor. Densitometric analysis of the autoradiographic signal revealed a rise in the level of androgen receptor RNA following treatment of rats with estradiol benzoate and flutamide. Treatment of rats with [D Trp6, des Gly10] GnRH brought about a transient rise in the level of androgen receptor RNA. Thus, our results indicate that androgenic deprivation up-regulates the level of androgen receptor transcript.
Collapse
Affiliation(s)
- V L Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | |
Collapse
|
38
|
|
39
|
Reiter E, Poncin J, Hennuy B, Bruyninx M, Klug M, Cornet A, Closset J, Hennen G. Luteinizing hormone increases the abundance of various transcripts, independently of the androgens, in the rat prostate. Biochem Biophys Res Commun 1997; 233:108-12. [PMID: 9144405 DOI: 10.1006/bbrc.1997.6412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Differential display analysis was carried out to find, in the rat prostate, genes that could be regulated by Luteinizing Hormone (LH), independently of the androgens. Hypophysectomized and castrated adult rats were treated with either LH, testosterone or saline. Regulated discrete bands have been eluted and reamplified. After Northern blotting, the levels of mRNA corresponding to 8 PCR fragments were significantly increased by LH treatment. None of these inserts were found to be induced by testosterone. One insert was subcloned, sequenced and identified as the ribosomial protein S 23. A competitive RT-PCR assay was carried out on the full length S 23 cDNA and confirmed that its mRNA levels were stimulated by LH but not by testosterone. These results strongly suggest that the LH membrane receptor, previously shown to be expressed in the rat prostate, has a physiological significance in this organ. Moreover, it appears that the effect of LH on the rat prostate are independent of the androgens.
Collapse
Affiliation(s)
- E Reiter
- Biochemistry and Laboratory of Endocrinology, University of Liège, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Franklin RB, Zou J, Gorski E, Yang YH, Costello LC. Prolactin regulation of mitochondrial aspartate aminotransferase and protein kinase C in human prostate cancer cells. Mol Cell Endocrinol 1997; 127:19-25. [PMID: 9099897 DOI: 10.1016/s0303-7207(96)03972-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Citrate production is a major physiological function of the prostate that is regulated by testosterone and prolactin. Mitochondrial aspartate aminotransferase (mAAT) is a key enzyme in the metabolic pathway of prostate citrate production. In addition, prolactin stimulates expression of mAAT in the rat lateral prostate. In this report we establish the role of prolactin in the regulation of mAAT in two prostate cancer cell lines, LNCaP and PC-3. LNCaP cells respond to hormonal stimulation with increased secretion of prostate specific products. PC-3 cells, on the other hand, are testosterone independent and apparently do not respond to other growth factors either. Results showed that both LNCaP and PC-3 cells responded to prolactin with increased mAAT activity and an increased steady state level of mAAT mRNA. Prolactin also increased protein kinase C (PKC) activity in both these cell lines. Treatment of LNCaP and PC-3 cells with the phorbol ester 12-O-tetradecanoylphorbol (TPA) caused the same effect on mAAT activity and mRNA level as prolactin. The results suggest that the diacylglycerol-PKC signal transduction system mediates the prolactin effect on mAAT. In addition, these results also show that the prolactin effect on mAAT is independent of androgens since PC-3 cells reportedly lack androgen receptor expression. Thus, these results provide evidence that prolactin is a physiological regulator of prostate function in human as well as rat prostate. In addition, the results also show that though prostate cancer cells are androgen independent, they remain responsive to prolactin. This could have important implications for the treatment and management of prostate cancer.
Collapse
Affiliation(s)
- R B Franklin
- Department of Oral and Craniofacial Biological Sciences, University of Maryland, Dental School, Baltimore 21201, USA
| | | | | | | | | |
Collapse
|
41
|
Yamashita A, Hayashi N, Sugimura Y, Cunha GR, Kawamura J. Influence of diethylstilbestrol, Leuprolelin (a luteinizing hormone-releasing hormone analog), Finasteride (a 5 alpha-reductase inhibitor), and castration on the lobar subdivisions of the rat prostate. Prostate 1996; 29:1-14. [PMID: 8685049 DOI: 10.1002/(sici)1097-0045(199607)29:1<1::aid-pros1>3.0.co;2-k] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The effects of various means of interfering with androgen action on rat coagulating gland, ventral prostate, lateral type 1 prostate, lateral type 2 prostate, and dorsal prostate were examined morphologically and quantitatively by assessing DNA content, wet weight, protein content, and zinc concentrations. Adult male Sprague-Dawley rats were subjected to 2 weeks of interfering with androgen action by treatment with Leuprolelin (a luteinizing hormone-releasing hormone analog), Finasteride (a 5 alpha-reductase inhibitor), or diethylstilbestrol (DES), or by physical castration. For all prostatic lobes, inhibition of 5 alpha-reductase elicited the smallest reduction in prostatic wet weight, DNA and protein contents, and zinc concentration. The most profound reductions in all parameters were elicited by castration. Treatments with DES and Leuprolelin gave intermediate effects with DES being the more effective in reducing all parameters in all prostatic lobes. Morphological changes elicited by all forms of androgen blockade were reduction of epithelial height, relative increase of connective tissue, reduction in ductal diameter, length, and number. The order of effectiveness of the various treatments on morphological features was as described above. While all forms of androgen blockade elicited similar effects throughout the prostate, differences in response to all forms of interference with androgen action were observed in different lobes of the prostate with regard to wet weight, DNA and protein contents, and zinc concentration as well as morphological effects. Regressive changes at the morphological level were particularly striking in the coagulating gland and ventral prostate, and indistinct in the lateral type 2 prostate. Prostatic zinc concentration in both normal and androgen-deprived rats was the highest in the lateral type 2 prostate and was reduced by interfering with androgen action to the greatest extent in the dorsolateral prostate (lateral type 1 and type 2, and dorsal prostate). The distribution of zinc correlated with the expression of metallothionein, which was detected by immunocytochemistry only in the lateral type 2 prostate of both normal and androgen deprived rats. Intraprostatic heterogeneity of zinc and metallothionein expression emphasizes interlobar differences in biological function within the rat prostate. The mechanism of development of regional heterogeneity within the prostate may shed light on the pathogenesis of prostatic proliferative diseases (prostatic hyperplasia and prostatic cancer) that initially owe their development to focal changes within large cell populations.
Collapse
Affiliation(s)
- A Yamashita
- Department of Urology, Mie University, School of Medicine, Mieken, Japan
| | | | | | | | | |
Collapse
|
42
|
Wang F, McKeehan WL. Insulin-like growth factor-1 messenger RNA isoforms in rat prostate and prostate tumors. In Vitro Cell Dev Biol Anim 1996; 32:265-8. [PMID: 8792155 DOI: 10.1007/bf02723058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
43
|
Reiter E, Lardinois S, Klug M, Sente B, Hennuy B, Bruyninx M, Closset J, Hennen G. Androgen-independent effects of prolactin on the different lobes of the immature rat prostate. Mol Cell Endocrinol 1995; 112:113-22. [PMID: 7589778 DOI: 10.1016/0303-7207(95)03596-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In this study, we have examined the respective roles of androgens and prolactin (Prl) on rat prostate development and function. Hypophysectomized immature rats, castrated or not after hypophysectomy and treated or not with a 5 alpha-reductase inhibitor, were used to study the different aspects of Prl action on the rat prostate and its synergy with androgens in vivo. Using Northern blot analysis and quantitation of prostatic mRNAs, we have shown that Prl significantly increases the steady-state levels of transcripts coding for several lobe-specific proteins: the C3 subunit of prostatein, probasin, and RWB. We have confirmed these observations in vitro, on explants of immature rat prostate treated with either saline, Prl, or testosterone. In addition, we have demonstrated by a nuclear run-on assay that Prl significantly enhances the transcription rate of the C3 gene in the rat prostate. We conclude that the effects of Prl concern all lobes of the organ and are, at least in part, androgen-independent. Moreover, Prl is able, via an androgen-independent pathway, to increase the rate of transcription of the C3 gene, one of the major products of the rat prostate.
Collapse
Affiliation(s)
- E Reiter
- Laboratory of Endocrinology, University of Liège, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Drivdahl RH, Loop SM, Andress DL, Ostenson RC. IGF-binding proteins in human prostate tumor cells: expression and regulation by 1,25-dihydroxyvitamin D3. Prostate 1995; 26:72-9. [PMID: 7531847 DOI: 10.1002/pros.2990260203] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The expression of the six known insulin-like growth factor binding proteins (IGFBPs) and their corresponding messenger RNAs has been examined in three cell lines established from surgical and biopsy specimens of human prostate carcinoma. All three cell lines produced both IGFBP-4 and IGFBP-6 and the respective mRNAs; expression of IGFBP-6 has not been previously demonstrated in human prostate tumor cells. No other binding proteins were detected. The levels of IGFBP mRNAs were not regulated by androgens or IGF-1, but the level of IGFBP-6 mRNA was sharply increased by 1,25-dihydroxyvitamin D3 (1,25(OH)D3). The stimulation was dose-dependent with a maximum effect at 10 nM 1,25(OH)D3 and a clearly discernible effect at 0.1 nM. The results support a role for vitamin D in the control of prostate tumor growth, mediated at least in part by interaction with IGFs and specific IGFBPs.
Collapse
Affiliation(s)
- R H Drivdahl
- Research Service, Department of Veterans Affairs Medical Center, Tacoma, Washington 98493
| | | | | | | |
Collapse
|
45
|
Abstract
The mechanism by which steroids influence cell proliferation is poorly understood although an understanding of this process might facilitate the development of strategies to modulate the tissue-specific activity of steroid hormones. In this article, the evidence that steroid hormones interact with the insulin-like growth factor (IGF) signal transduction pathway is reviewed for three different tissues. In osteoblasts, oestradiol stimulates the production of IGF-I which appears to act as an autocrine growth factor. In uterine tissue, oestradiol increases the synthesis of IGF-I in the stroma which then modulates the proliferation of epithelial cells although there is also evidence that oestradiol can modulate the sensitivity of uterine epithelial cells to IGFs. In breast cancer, oestrogens may increase IGF-II synthesis in epithelial cells, increase the sensitivity of breast cancer cells to IGFs (possibly by modulating type I IGF receptor levels) as well as resulting components of the IGF signal transduction pathway resulting in induction of immediate early genes. There therefore appears to be a variety of ways in which oestradiol interact with the IGF signal transduction pathway and these may be applicable to other malignant and normal tissues and other groups of steroid hormones.
Collapse
Affiliation(s)
- B R Westley
- Department of Pathology, Royal Victoria Infirmary, Newcastle upon Tyne, England
| | | |
Collapse
|
46
|
Kumar MV, Jones EA, Grossmann ME, Blexrud MD, Tindall DJ. Identification and characterization of a suppressor element in the 5'-flanking region of the mouse androgen receptor gene. Nucleic Acids Res 1994; 22:3693-8. [PMID: 7937079 PMCID: PMC308348 DOI: 10.1093/nar/22.18.3693] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Androgens play an important role in the development and maintenance of male reproductive organs through the androgen receptor (AR). In order to study the mechanism of regulation of AR at the molecular level, a 1571 bp fragment in the 5'-flanking region of the mouse androgen receptor (mAR) gene was isolated and sequenced. Transfection of 5'-deletion constructs cloned into vectors containing the chloramphenicol acetyl transferase (CAT) gene indicated the presence of a promoter in the sequence -146 to +131. These experiments also suggested the presence of a suppressor element. Further characterization indicated that the suppressor is present between -486 to -351. It is functional in the context of the natural AR promoter and the heterologous thymidine kinase promoter. Transfection of a -546/ + 131 construct in which the putative suppressor element (-421 to -448) had been deleted caused increased basal CAT activity suggesting that the suppressor is limited to this 28 bp element in the 5'-flanking region of the mouse AR gene.
Collapse
Affiliation(s)
- M V Kumar
- Department of Urology, Mayo Foundation, Rochester, MN 55905
| | | | | | | | | |
Collapse
|
47
|
Desgrandchamps F, Teillac P. The role of growth factors in the pathogenesis of benign prostatic hyperplasia. Biomed Pharmacother 1994. [DOI: 10.1016/s0753-3322(09)80004-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|