51
|
Popovics P, Cai R, Sha W, Rick FG, Schally AV. Growth hormone-releasing hormone antagonists reduce prostatic enlargement and inflammation in carrageenan-induced chronic prostatitis. Prostate 2018; 78:970-980. [PMID: 29786867 DOI: 10.1002/pros.23655] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/07/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Inflammation plays a key role in the etiology of benign prostatic hyperplasia (BPH) through multiple pathways involving the stimulation of proliferation by cytokines and growth factors as well as the induction of the focal occurrence of epithelial-to-mesenchymal transition (EMT). We have previously reported that GHRH acts as a prostatic growth factor in experimental BPH and in autoimmune prostatitis models and its blockade with GHRH antagonists offer therapeutic approaches for these conditions. Our current study was aimed at the investigation of the beneficial effects of GHRH antagonists in λ-carrageenan-induced chronic prostatitis and at probing the downstream molecular pathways that are implicated in GHRH signaling. METHODS To demonstrate the complications triggered by recurrent/chronic prostatic inflammation in Sprague-Dawley rats, 50 μL 3% carrageenan was injected into both ventral prostate lobes two times, 3 weeks apart. GHRH antagonist, MIA-690, was administered 5 days after the second intraprostatic injection at 20 μg daily dose for 4 weeks. GHRH-induced signaling events were identified in BPH-1 and in primary prostate epithelial (PrEp) cells at 5, 15, 30, and 60 min with Western blot. RESULTS Inflammation induced prostatic enlargement and increased the area of the stromal compartment whereas treatment with the GHRH antagonist significantly reduced these effects. This beneficial activity was consistent with a decrease in prostatic GHRH, inflammatory marker COX-2, growth factor IGF-1 and inflammatory and EMT marker TGF-β1 protein levels and the expression of multiple genes related to EMT. In vitro, GHRH stimulated multiple pathways involved in inflammation and growth in both BPH-1 and PrEp cells including NFκB p65, AKT, ERK1/2, EGFR, STAT3 and increased the levels of TGF-β1 and Snail/Slug. Most interestingly, GHRH also stimulated the transactivation of the IGF receptor. CONCLUSIONS The study demonstrates that GHRH antagonists could be beneficial for the treatment of prostatic inflammation and BPH in part by inhibiting the growth-promoting and inflammatory effects of locally produced GHRH.
Collapse
Affiliation(s)
- Petra Popovics
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida
| | - Renzhi Cai
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida
| | - Wei Sha
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida
| | - Ferenc G Rick
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida
- Department of Urology, Herbert Wertheim College of Medicine, Florida International, University, Miami, Florida
| | - Andrew V Schally
- Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, Florida
- Sylvester Comprehensive Cancer Center, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
52
|
Akirov A, Greenman Y, Glaser B, S'chigol I, Mansiterski Y, Eizenberg Y, Shraga-Slutzky I, Shimon I. IGF-1 levels may increase paradoxically with dopamine agonist treatment for prolactinomas. Pituitary 2018; 21:406-413. [PMID: 29728863 DOI: 10.1007/s11102-018-0891-5] [Citation(s) in RCA: 5] [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] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Hyperprolactinemia is common in acromegaly and in these patients, insulin-like growth factor (IGF)-1 level may decrease with dopamine agonist. We report a series of patients with prolactinoma and a paradoxical increase of IGF-1 levels during cabergoline treatment. METHODS Clinical characteristics and response to treatment of patients with prolactinomas, in whom normal or slightly elevated baseline IGF-1 levels increased with cabergoline. RESULTS The cohort consisted of ten prolactinoma patients (nine males, mean age 48 ± 14 years). Mean adenoma size was 23.8 ± 16.2 mm, with cavernous sinus invasion in eight. In five patients baseline IGF-1 levels were normal and in four levels were 1.2-1.5-fold the upper limit of the normal (ULN). One patient had IGF-1 measured shortly after initiating cabergoline and it was 1.4 × ULN. During cabergoline treatment (dose range 0.5-2 mg/week) PRL normalization was achieved in all and tumor shrinkage occurred in seven patients. The mean IGF-1 increase on cabergoline was 1.7 ± 0.4 × ULN. Cabergoline dose reduction or interruption was attempted in five patients and resulted in decreased IGF-1 levels in all, including normalization in two patients. Three patients were eventually diagnosed with acromegaly, one was referred for pituitary surgery followed by complete remission, another patient was switched to somatostatin analogue, and the third was treated by combination of somatostatin analogues with pegvisomant, with reduction of IGF-1 in all these patients. CONCLUSION IGF-1 levels may increase to clinically significant levels during cabergoline treatment for PRL-adenoma. We suggest IGF-1 monitoring in all patients treated with dopamine agonists and not only in those presenting symptoms of acromegaly.
Collapse
Affiliation(s)
- Amit Akirov
- Institute of Endocrinology, Rabin Medical Center, Beilinson Hospital, 4941492, Petach Tikva, Israel.
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Yona Greenman
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | | | - Yossi Mansiterski
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Maccabi Health Care Services, Tel Aviv, Israel
| | - Yoav Eizenberg
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Tel Aviv-Jaffa District Clalit Health Services, Tel Aviv, Israel
| | - Ilana Shraga-Slutzky
- Institute of Endocrinology, Rabin Medical Center, Beilinson Hospital, 4941492, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Shimon
- Institute of Endocrinology, Rabin Medical Center, Beilinson Hospital, 4941492, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
53
|
Lindsey RC, Rundle CH, Mohan S. Role of IGF1 and EFN-EPH signaling in skeletal metabolism. J Mol Endocrinol 2018; 61:T87-T102. [PMID: 29581239 PMCID: PMC5966337 DOI: 10.1530/jme-17-0284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/26/2018] [Indexed: 01/11/2023]
Abstract
Insulin-like growth factor 1(IGF1) and ephrin ligand (EFN)-receptor (EPH) signaling are both crucial for bone cell function and skeletal development and maintenance. IGF1 signaling is the major mediator of growth hormone-induced bone growth, but a host of different signals and factors regulate IGF1 signaling at the systemic and local levels. Disruption of the Igf1 gene results in reduced peak bone mass in both experimental animal models and humans. Additionally, EFN-EPH signaling is a complex system which, particularly through cell-cell interactions, contributes to the development and differentiation of many bone cell types. Recent evidence has demonstrated several ways in which the IGF1 and EFN-EPH signaling pathways interact with and depend upon each other to regulate bone cell function. While much remains to be elucidated, the interaction between these two signaling pathways opens a vast array of new opportunities for investigation into the mechanisms of and potential therapies for skeletal conditions such as osteoporosis and fracture repair.
Collapse
Affiliation(s)
- Richard C Lindsey
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Division of BiochemistryDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Center for Health Disparities and Molecular MedicineDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Charles H Rundle
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of MedicineLoma Linda University, Loma Linda, California, USA
| | - Subburaman Mohan
- Musculoskeletal Disease CenterVA Loma Linda Healthcare System, Loma Linda, California, USA
- Division of BiochemistryDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Center for Health Disparities and Molecular MedicineDepartment of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Department of MedicineLoma Linda University, Loma Linda, California, USA
| |
Collapse
|
54
|
Growth Hormone Secretion Patterns in German Landrace (DL) Fetuses and Piglets Compared to DL Piglets with Inherited 1,25-Dihydroxyvitamin D3 Deficiency. Nutrients 2018; 10:nu10050617. [PMID: 29762475 PMCID: PMC5986497 DOI: 10.3390/nu10050617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 11/24/2022] Open
Abstract
The regulation of growth hormone (GH) release during prenatal development and during early postnatal life is not entirely clarified. In this study plasma GH concentrations in pigs with inherited pseudo vitamin D deficiency type I (PDDR-I), which regularly show growth retardation, were compared during ontogeny with unaffected pigs of the same breed (German Landrace, DL) as control. Plasma GH concentrations were measured in plasma of chronically catheterized fetuses (beginning on day 101 after mating or after artificial insemination) and in piglets (day 37 postpartum (p.p.)—day 42 p.p.) of both lines. A growth curve beginning at day 7 p.p. was recorded for both lines. The relative amount of GH receptor (GHR) mRNA in liver was quantified by competitive reverse transcription polymerase chain reaction in piglets at day 42 p.p. A trend for higher GH concentrations was observed in PDDR-I fetuses (p < 0.1). In PDDR-I piglets compared to DL piglets higher plasma GH values (p < 0.01), were observed despite lower body weight. The relative quantity of GHR mRNA in liver was not significantly different between the two lines. Piglets with an inherited defect of vitamin D synthesis showed higher GH concentrations. A hormonal imprinting by low 1,25(OH)2D3 could be one reason for our observations and should be analysed in detail in future.
Collapse
|
55
|
Trifunović S, Milošević V. The Morpho-Functional Parameters of Rat Pituitary Hormone Producing Cells After Genistein Treatment. MACEDONIAN VETERINARY REVIEW 2018. [DOI: 10.1515/macvetrev-2017-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Abstract
Phytoestrogens are a diverse group of steroid–like compounds that occur naturally in many plants. There are various types of phytoestrogens, including the best-researched isoflavones which are commonly found in soy. The consumption of soy products has many health benefits, including protection against breast cancer, prostate cancer, menopausal symptoms, heart disease and osteoporosis. In contrast, use of hormonally active compounds-isoflavones may unfortunately interfere with the endocrine system and can have far-reaching consequences. Genistein, the most abundant soy-bean derived isoflavone, possesses a ring system similar to estrogens and acts through an estrogen receptor (ER)-mediated mechanism, by increasing or decreasing the transcription of ER-dependent target genes. Also, genistein can act on cells through ER non-dependent mechanisms, such as tyrosine kinase inhibitor. The neuroendocrine systems are responsible for the control of homeostatic processes in the body, including reproduction, growth, metabolism and energy balance, and stress responsiveness. It is well known, that estrogen is important for development of the neuroendocrine system in both sexes. At the pituitary level, estrogen is known to affect the regulation of all hormone producing (HP) cells, by direct and/or indirect mechanisms. Due to structural and functional resemblance to estrogen, the question may arise of whether and how genistein affects the morphofunctional features of pituitary HP cells. This review deals with the consequences of genistein’s effects on morphological, stereological and hormonal features of HP cells within the anterior pituitary gland. Transparency on this issue is needed because isoflavones are presently highly consumed. Inter alia, genistein as well as other isoflavones, are present in various dietary supplements and generally promoted as an accepted alternative to estrogen replacement therapy. Potential isoflavone biomedical exploitation is not only limited to estrogen replacement therapy, so it should be treated in a wider context of different ageing symptoms remediation.
Collapse
Affiliation(s)
- Svetlana Trifunović
- Department of Cytology, Institute for Biological Research “Siniša Stanković” , University of Belgrade , Bul Despot Stefan 142, 11060 Belgrade , Serbia
| | - Verica Milošević
- Department of Cytology, Institute for Biological Research “Siniša Stanković” , University of Belgrade , Bul Despot Stefan 142, 11060 Belgrade , Serbia
| |
Collapse
|
56
|
Vázquez-Borrego MC, Gahete MD, Martínez-Fuentes AJ, Fuentes-Fayos AC, Castaño JP, Kineman RD, Luque RM. Multiple signaling pathways convey central and peripheral signals to regulate pituitary function: Lessons from human and non-human primate models. Mol Cell Endocrinol 2018; 463:4-22. [PMID: 29253530 DOI: 10.1016/j.mce.2017.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/14/2017] [Accepted: 12/14/2017] [Indexed: 12/12/2022]
Abstract
The anterior pituitary gland is a key organ involved in the control of multiple physiological functions including growth, reproduction, metabolism and stress. These functions are controlled by five distinct hormone-producing pituitary cell types that produce growth hormone (somatotropes), prolactin (lactotropes), adrenocorticotropin (corticotropes), thyrotropin (thyrotropes) and follicle stimulating hormone/luteinizing hormone (gonadotropes). Classically, the synthesis and release of pituitary hormones was thought to be primarily regulated by central (neuroendocrine) signals. However, it is now becoming apparent that factors produced by pituitary hormone targets (endocrine and non-endocrine organs) can feedback directly to the pituitary to adjust pituitary hormone synthesis and release. Therefore, pituitary cells serve as sensors to integrate central and peripheral signals in order to fine-tune whole-body homeostasis, although it is clear that pituitary cell regulation is species-, age- and sex-dependent. The purpose of this review is to provide a comprehensive, general overview of our current knowledge of both central and peripheral regulators of pituitary cell function and associated intracellular mechanisms, focusing on human and non-human primates.
Collapse
Affiliation(s)
- M C Vázquez-Borrego
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - M D Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - A J Martínez-Fuentes
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - A C Fuentes-Fayos
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - J P Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - R D Kineman
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center, Research and Development Division, Chicago, IL, USA
| | - R M Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain; Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain.
| |
Collapse
|
57
|
Wu Q, Shah NP. High γ-aminobutyric acid production from lactic acid bacteria: Emphasis on Lactobacillus brevis as a functional dairy starter. Crit Rev Food Sci Nutr 2018; 57:3661-3672. [PMID: 26980301 DOI: 10.1080/10408398.2016.1147418] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
γ-Aminobutyric acid (GABA) and GABA-rich foods have shown anti-hypertensive and anti-depressant activities as the major functions in humans and animals. Hence, high GABA-producing lactic acid bacteria (LAB) could be used as functional starters for manufacturing novel fermented dairy foods. Glutamic acid decarboxylases (GADs) from LAB are highly conserved at the species level based on the phylogenetic tree of GADs from LAB. Moreover, two functionally distinct GADs and one intact gad operon were observed in all the completely sequenced Lactobacillus brevis strains suggesting its common capability to synthesize GABA. Difficulties and strategies for the manufacture of GABA-rich fermented dairy foods have been discussed and proposed, respectively. In addition, a genetic survey on the sequenced LAB strains demonstrated the absence of cell envelope proteinases in the majority of LAB including Lb. brevis, which diminishes their cell viabilities in milk environments due to their non-proteolytic nature. Thus, several strategies have been proposed to overcome the non-proteolytic nature of Lb. brevis in order to produce GABA-rich dairy foods.
Collapse
Affiliation(s)
- Qinglong Wu
- a Food and Nutritional Science, School of Biological Sciences , The University of Hong Kong , Hong Kong , Hong Kong
| | - Nagendra P Shah
- a Food and Nutritional Science, School of Biological Sciences , The University of Hong Kong , Hong Kong , Hong Kong
| |
Collapse
|
58
|
γ-Aminobutyric Acid (GABA): Biosynthesis, Role, Commercial Production, and Applications. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2018. [DOI: 10.1016/b978-0-444-64057-4.00013-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
59
|
Yang YM, Sehgal PB. Smooth Muscle-Specific BCL6+/- Knockout Abrogates Sex Bias in Chronic Hypoxia-Induced Pulmonary Arterial Hypertension in Mice. Int J Endocrinol 2018; 2018:3473105. [PMID: 30140283 PMCID: PMC6081567 DOI: 10.1155/2018/3473105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/07/2018] [Accepted: 06/24/2018] [Indexed: 12/18/2022] Open
Abstract
The "estrogen paradox" in pulmonary arterial hypertension (PAH) refers to observations that while there is a higher incidence of idiopathic PAH in women, rodent models of PAH show male dominance and estrogens are protective. To explain these differences, we previously proposed the neuroendocrine-STAT5-BCL6 hypothesis anchored in the sex-biased and species-specific patterns of growth hormone (GH) secretion by the pituitary, the targeting of the hypothalamus by estrogens to feminize GH secretion patterns, and the role of the transcription factors STAT5a/b and BCL6 as downstream mediators of this patterned GH-driven sex bias. As a test of this hypothesis, we previously reported that vascular smooth muscle cell- (SMC-) specific deletion of the STAT5a/b locus abrogated the male-dominant sex bias in the chronic hypoxia model of PAH in mice. In the present study, we confirmed reduced BCL6 expression in pulmonary arterial (PA) segments in both male and female SMC:STAT5a/b-/- mice. In order to test the proposed contribution of BCL6 to sex bias in PAH, we developed mice with SMC-specific deletion of BCL6+/- by crossing SM22α-Cre mice with BCL6-floxed mice and investigated sex bias in these mutant mice in the chronic hypoxia model of PAH. We observed that the male-bias observed in wild-type- (wt-) SM22α-Cre-positive mice was abrogated in the SMC:BCL6+/- knockouts-both males and females showed equivalent enhancement of indices of PAH. The new data confirm BCL6 as a contributor to the sex-bias phenotype observed in hypoxic PAH in mice and support the neuroendocrine-STAT5-BCL6 hypothesis of sex bias in this experimental model of vascular disease.
Collapse
Affiliation(s)
- Yang-Ming Yang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | - Pravin B. Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
60
|
Leonte A, Colzato LS, Steenbergen L, Hommel B, Akyürek EG. Supplementation of gamma-aminobutyric acid (GABA) affects temporal, but not spatial visual attention. Brain Cogn 2017; 120:8-16. [PMID: 29222993 DOI: 10.1016/j.bandc.2017.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 01/30/2023]
Abstract
In a randomized, double-blind, and placebo-controlled experiment, the acute effects of gamma-aminobutyric acid (GABA) supplementation on temporal and spatial attention in young healthy adults were investigated. A hybrid two-target rapid serial visual presentation task was used to measure temporal attention and integration. Additionally, a visual search task was used to measure the speed and accuracy of spatial attention. While temporal attention depends primarily on the distribution of limited attentional resources across time, spatial attention represents the engagement and disengagement by relevant and irrelevant stimuli across the visual field. Although spatial attention was unaffected by GABA supplementation altogether, we found evidence supporting improved performance in the temporal attention task. The attentional blink was numerically, albeit not significantly, attenuated at Lag 3, and significantly fewer order errors were committed at Lag 1, compared to the placebo condition. No effect was found on temporal integration rates. Although there is controversy about whether oral GABA can cross the blood-brain barrier, our results offer preliminary evidence that GABA intake might help to distribute limited attentional resources more efficiently, and can specifically improve the identification and ordering of visual events that occur in close temporal succession.
Collapse
Affiliation(s)
- Anna Leonte
- Department of Psychology, Experimental Psychology, University of Groningen, The Netherlands
| | - Lorenza S Colzato
- Institute for Psychological Research, Leiden Institute for Brain and Cognition, Leiden University, The Netherlands
| | - Laura Steenbergen
- Institute for Psychological Research, Leiden Institute for Brain and Cognition, Leiden University, The Netherlands
| | - Bernhard Hommel
- Institute for Psychological Research, Leiden Institute for Brain and Cognition, Leiden University, The Netherlands
| | - Elkan G Akyürek
- Department of Psychology, Experimental Psychology, University of Groningen, The Netherlands.
| |
Collapse
|
61
|
Khan KU, Zuberi A, Fernandes JBK, Ullah I, Sarwar H. An overview of the ongoing insights in selenium research and its role in fish nutrition and fish health. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1689-1705. [PMID: 28712005 DOI: 10.1007/s10695-017-0402-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/26/2017] [Indexed: 06/07/2023]
Abstract
In the present review, the ongoing researches about selenium research in fish nutrition have been comprehensively discussed. Selenium research is getting popularity in fish nutrition as it is required for the normal growth and proper physiological and biochemical functions in fish. Its deficiency or surplus amounts create severe problems in fish. It is available as inorganic form, organic form, and nano form. In fish, most of the previous research is about the selenium requirements for fish by using only one selenium source mainly the inorganic one. Selenium shows maximum biological activity and bioavailability when it is supplied in proper form. However, to differentiate the more bioavailable and less toxic form of selenium, sufficient information is needed about the comparative bioavailability of different selenium forms in different fish species. In fish, important data about the new forms of selenoproteins is still scarce. Therefore, it is necessary to focus on the determination and elucidation of the new selenoproteins in fish through the utilization of recent approaches of molecular biology and proteomics. The adaptation of these new approaches will replace the old fashioned methodologies regarding the selenium research in fish nutrition. Moreover, the use of molecular biology and proteomics-based new approaches in combination with selenium research will help in optimizing the area of fish nutrition and will improve the feed intake, growth performance, and more importantly the flesh quality which has a promising importance in the consumer market.
Collapse
Affiliation(s)
- Kifayat Ullah Khan
- Center of Aquaculture, São Paulo State University, Jaboticabal, SP, Brazil.
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Amina Zuberi
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | | | - Imdad Ullah
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Huda Sarwar
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
62
|
The Hypothalamic-Pituitary Axis and Autoantibody Related Disorders. Int J Mol Sci 2017; 18:ijms18112322. [PMID: 29099758 PMCID: PMC5713291 DOI: 10.3390/ijms18112322] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/27/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
This review summarized different studies reporting the presence of autoantibodies reacting against cells of the pituitary (APAs) and/or hypothalamus (AHAs). Both APAs and AHAs have been revealed through immunofluorescence using different kinds of substrates. Autoantibodies against gonadotropic cells were mainly found in patients affected by cryptorchidism and hypogonadotropic hypogonadism while those against prolactin cells were found in different kinds of patients, the majority without pituitary abnormalities. APAs to growth hormone (GH) cells have been associated with GH deficiency while those against the adrenocorticotropic cells have distinguished central Cushing's disease patients at risk of incomplete cure after surgical adenoma removal. AHAs to vasopressin cells have identified patients at risk of developing diabetes insipidus. APAs have been also found together with AHAs in patients affected by idiopathic hypopituitarism, but both were also present in different kinds of patients without abnormalities of the hypothalamic-pituitary axis. Despite some data being promising, the clinical use of pituitary and hypothalamus autoantibodies is still limited by the low diagnostic sensitivity, irreproducibility of the results, and the absence of autoantigen/s able to discriminate the autoimmune reaction involving the pituitary or the hypothalamus from the other autoimmune states.
Collapse
|
63
|
Höhne A, Schrader L, Weigend S, Petow S. Ghrelin plasma concentration does not covary with energy demand in adult laying hens. Domest Anim Endocrinol 2017; 61:77-83. [PMID: 28734138 DOI: 10.1016/j.domaniend.2017.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 06/21/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
Abstract
The peptide hormone ghrelin is suggested to be involved in food intake regulation in young growing chicken. Whether ghrelin is involved in the regulation of energetic balance associated with laying performance in adult laying hens was studied by use of 4 chicken lines that differ in laying performance and phylogeny (4 lines; 16 hens per line). As housing conditions are also known to affect energy demand, half of the hens per line were housed in single cages and the other half of hens were maintained in a floor housing system. Plasma samples were collected at 17 to 19, 33 to 35, 49 to 51, and 72 wk of age and analyzed with a chicken ghrelin ELISA Kit. From caged hens, individual food consumption and laying performance additionally was recorded. Due to its function in growth and its relationship with ghrelin, also GH plasma concentrations were analyzed. Ghrelin concentrations did not differ between the 4 lines at any of the test periods (all P > 0.05). Ghrelin was negatively related to food consumption only in the growing period of the high-performing lines (both P < 0.0001). During this phase, floor-housed hens showed greater ghrelin concentrations compared with caged hens (P < 0.0001). Our results suggest that in adult layers ghrelin is not involved in regulating energy intake related to laying performance but rather seems to be related to body growth and housing condition before start of lay, the latter possibly due to differences in hens' behavioral activity.
Collapse
Affiliation(s)
- A Höhne
- Friedrich-Loeffler-Institut, Institute of Animal Welfare and Animal Husbandry, Celle, Germany.
| | - L Schrader
- Friedrich-Loeffler-Institut, Institute of Animal Welfare and Animal Husbandry, Celle, Germany
| | - S Weigend
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Mariensee, Germany
| | - S Petow
- Friedrich-Loeffler-Institut, Institute of Animal Welfare and Animal Husbandry, Celle, Germany
| |
Collapse
|
64
|
Foradori CD, Whitlock BK, Daniel JA, Zimmerman AD, Jones MA, Read CC, Steele BP, Smith JT, Clarke IJ, Elsasser TH, Keisler DH, Sartin JL. Kisspeptin Stimulates Growth Hormone Release by Utilizing Neuropeptide Y Pathways and Is Dependent on the Presence of Ghrelin in the Ewe. Endocrinology 2017; 158:3526-3539. [PMID: 28977590 DOI: 10.1210/en.2017-00303] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/13/2017] [Indexed: 12/31/2022]
Abstract
Although kisspeptin is the primary stimulator of gonadotropin-releasing hormone secretion and therefore the hypothalamic-pituitary-gonadal axis, recent findings suggest kisspeptin can also regulate additional neuroendocrine processes including release of growth hormone (GH). Here we show that central delivery of kisspeptin causes a robust rise in plasma GH in fasted but not fed sheep. Kisspeptin-induced GH secretion was similar in animals fasted for 24 hours and those fasted for 72 hours, suggesting that the factors involved in kisspeptin-induced GH secretion are responsive to loss of food availability and not the result of severe negative energy balance. Pretreatment with the neuropeptide Y (NPY) Y1 receptor antagonist, BIBO 3304, blocked the effects of kisspeptin-induced GH release, implicating NPY as an intermediary. Kisspeptin treatment induced c-Fos in NPY and GH-releasing hormone (GHRH) cells of the arcuate nucleus. The same kisspeptin treatment resulted in a reduction in c-Fos in somatostatin (SS) cells in the periventricular nucleus. Finally, blockade of systemic ghrelin release or antagonism of the ghrelin receptor eliminated or reduced the ability of kisspeptin to induce GH release, suggesting the presence of ghrelin is required for kisspeptin-induced GH release in fasted animals. Our findings support the hypothesis that during short-term fasting, systemic ghrelin concentrations and NPY expression in the arcuate nucleus rise. This permits kisspeptin activation of NPY cells. In turn, NPY stimulates GHRH cells and inhibits SS cells, resulting in GH release. We propose a mechanism by which kisspeptin conveys reproductive and hormone status onto the somatotropic axis, resulting in alterations in GH release.
Collapse
Affiliation(s)
- Chad D Foradori
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| | - Brian K Whitlock
- Department of Large Animal Clinical Sciences, University of Tennessee, Knoxville, Tennessee 37996
| | - Jay A Daniel
- Department of Animal Science, Berry College, Mt. Berry, Georgia 30149
| | - Arthur D Zimmerman
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| | - Melaney A Jones
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| | - Casey C Read
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| | - Barbara P Steele
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| | - Jeremy T Smith
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Crawley, Washington 6009, Australia
| | - Iain J Clarke
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia
| | - Theodore H Elsasser
- Animal Genomics and Improvement Laboratory, US Department of Agriculture, Agricultural Research Service, Beltsville, Maryland 20705
| | - Duane H Keisler
- Division of Animal Sciences, University of Missouri, Columbia, Missouri 65211
| | - James L Sartin
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, Alabama 36849
| |
Collapse
|
65
|
Jesuyon OMA. Effects of strain, sex, and season on body weight development of cane rat (Thryonomys swinderianus) in the humid tropics. Trop Anim Health Prod 2017; 50:5-10. [PMID: 28933037 DOI: 10.1007/s11250-017-1393-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/05/2017] [Indexed: 10/18/2022]
Abstract
The effects of strain (G), sex (H), and season (S) on live weight development of cane rats (Thryonomys swinderianus) reared in captive system were investigated during the dry (35.95 °C) and wet seasons (25.81 °C) from February to August of a tropical environment. The field experiment took place at the University of Ibadan. Seventy-nine cane rats spreading among three genotypes (Bamidele, FRIN, and Lawole) of different age and body weight groups were randomly distributed into cages using randomized complete block design in factorial. Data on live weight collected at 30 days interval over the experimental period was submitted for statistical analysis using the factorial ANOVA procedures of SAS® (2012). Strain was fixed factor while sex and season were random factors. Mean separation showed that G, H, S, and G×H, significantly (P < 0.05) affected live weight development of cane rats. G×H revealed sexual dimorphism. G×S and G×H×S did not demonstrate significant (P > 0.05) effects on live weight development in model. G×S showed mean weight gain levels of 0.21, 0.15, and 0.07 kg between wet and dry season for Bamidele, FRIN, and Lawole. The superiority of growth rate among genotypes between seasons were 57.1, 14.3, and 7.14 g/month for Bamidele > FRIN > Lawole, respectively. Lawole recorded highest body weight of 2.50 and 3.78 kg for female and male. FRIN recorded highest mixed body weight of 3.06 kg, highest body weights of 2.99 and 3.14 kg for dry and wet seasons. Bamidele demonstrated least average live weights of 2.76 and 2.97 kg for dry and wet seasons, the least body weight fluctuations between months in seasons and between seasons. Knowledge on the performance of cane rat genotypes by sex and season in captivity will provide information on adaptability to season and management systems for cane rat.
Collapse
Affiliation(s)
- Oluwatosin M A Jesuyon
- Animal Breeding and Genetics Unit, Department of Animal Production and Health, Federal University, PMB 373, Oye-Ekiti, Ekiti State, Nigeria.
| |
Collapse
|
66
|
Feng S, Xing C, Shen T, Qiao Y, Wang R, Chen J, Liao J, Lu Z, Yang X, Abd-Allah SM, Li J, Jing N, Tang K. Abnormal Paraventricular Nucleus of Hypothalamus and Growth Retardation Associated with Loss of Nuclear Receptor Gene COUP-TFII. Sci Rep 2017; 7:5282. [PMID: 28706241 PMCID: PMC5509697 DOI: 10.1038/s41598-017-05682-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/01/2017] [Indexed: 11/15/2022] Open
Abstract
The paraventricular nucleus of hypothalamus plays important roles in the regulation of energy balance and fetal growth. However, the molecular mechanisms underlying its formation and function have not been clearly elucidated. Various mutations in the human COUP-TFII gene, which encodes a nuclear receptor, result in growth retardation, congenital diaphragmatic hernia and congenital heart defects. Here, we show that COUP-TFII gene is expressed in the developing hypothalamus in mouse. The ventral forebrain-specific RXCre/+; COUP-TFIIF/F mutant mice display growth retardation. The development of the paraventricular nucleus of hypothalamus is compromised in the COUP-TFII mutant mainly because of increased apoptosis and mis-migration of the Brn2+ neurons. Moreover, hypoplastic anterior pituitary with blood cell clusters and shrunken posterior pituitary lacking AVP/OT neuron innervations are observed in the mutant, indicating the failure of formation of the hypothalamic-pituitary axis. Mechanistic studies show that the expression of Bdnf and Nrp1 genes is reduced in the mutant embryo, and that Bdnf is a direct downstream target of the COUP-TFII protein. Thus, our findings provide a novel functional validation that COUP-TFII gene promotes the expression of Bdnf and Nrp1 genes to ensure the appropriate morphogenesis of the hypothalamic-pituitary axis, especially the paraventricular nucleus of hypothalamus, and to prevent growth retardation.
Collapse
Affiliation(s)
- Su Feng
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Can Xing
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Tingyu Shen
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Yunbo Qiao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.,Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Ran Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Jun Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Jiaoyang Liao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Zhuo Lu
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Xiong Yang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Saber Mohamed Abd-Allah
- Theriogenology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Jinsong Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China. .,School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, China.
| | - Ke Tang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi, 330031, China.
| |
Collapse
|
67
|
Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017. [PMID: 28640444 DOI: 10.1002/cphy.c160027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.
Collapse
Affiliation(s)
- Katie M Troike
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Brooke E Henry
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Elizabeth A Jensen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Jonathan A Young
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
68
|
Kasuya E, Sutoh M, Yayou KI. The effects of l-DOPA and sulpiride on growth hormone secretion at different injection times in Holstein steers. Anim Sci J 2017; 88:1842-1848. [PMID: 28585780 DOI: 10.1111/asj.12850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/27/2017] [Indexed: 11/29/2022]
Abstract
The effects of l-DOPA, a precursor of dopamine (DA), and sulpiride, a D2 -type DA receptor blocker, on growth hormone (GH) and prolactin (PRL) secretion were investigated in steers. Eight Holstein steers (212.8 ± 7.8 kg body weight) were used. Lighting conditions were 12:12 L:D (lights on: 06.00-18.00 hours). Blood samplings were performed during the daytime (11.00-15.00 hours) and nighttime (23.00-03.00 hours). Intravenous injections of drugs or saline were performed at 12.00 hour for the daytime and 00.00 hour for the nighttime, respectively. Plasma GH and PRL concentrations were determined by radioimmunoassay. l-DOPA did not alter the GH secretion when it was injected at 12.00 hour (spontaneous GH level at its peak). On the other hand, l-DOPA increased GH secretion at 00.00 hour (GH level at its trough). Injection of sulpiride suppressed GH secretion at 12.00 hour but did not affect GH levels at 00.00 hour. l-DOPA inhibited and sulpiride stimulated PRL release during both periods. These results suggest that dopaminergic neurons have stimulatory action on GH secretion and inhibitory action on PRL secretion in cattle. In addition, injection time should be considered to evaluate the exact effects on GH secretion due to its ultradian rhythm of GH secretion in cattle.
Collapse
Affiliation(s)
- Etsuko Kasuya
- Animal Environment and Health Unit, Division of Animal Environment and Waste Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Madoka Sutoh
- Animal Environment and Health Unit, Division of Animal Environment and Waste Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Ken-Ichi Yayou
- Animal Environment and Health Unit, Division of Animal Environment and Waste Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| |
Collapse
|
69
|
Sarmento-Cabral A, Peinado JR, Halliday LC, Malagon MM, Castaño JP, Kineman RD, Luque RM. Adipokines (Leptin, Adiponectin, Resistin) Differentially Regulate All Hormonal Cell Types in Primary Anterior Pituitary Cell Cultures from Two Primate Species. Sci Rep 2017; 7:43537. [PMID: 28349931 PMCID: PMC5640086 DOI: 10.1038/srep43537] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/25/2017] [Indexed: 12/27/2022] Open
Abstract
Adipose-tissue (AT) is an endocrine organ that dynamically secretes multiple hormones, the adipokines, which regulate key physiological processes. However, adipokines and their receptors are also expressed and regulated in other tissues, including the pituitary, suggesting that locally- and AT-produced adipokines might comprise a regulatory circuit that relevantly modulate pituitary cell-function. Here, we used primary pituitary cell-cultures from two normal nonhuman-primate species [Papio-anubis/Macaca-fascicularis] to determine the impact of different adipokines on the functioning of all anterior-pituitary cell-types. Leptin and resistin stimulated GH-release, a response that was blocked by somatostatin. Conversely, adiponectin decreased GH-release, and inhibited GHRH-, but not ghrelin-stimulated GH-secretion. Furthermore: 1) Leptin stimulated PRL/ACTH/FSH- but not LH/TSH-release; 2) adiponectin stimulated PRL-, inhibited ACTH- and did not alter LH/FSH/TSH-release; and 3) resistin increased ACTH-release and did not alter PRL/LH/FSH/TSH-secretion. These effects were mediated through the activation of common (AC/PKA) and distinct (PLC/PKC, intra-/extra-cellular calcium, PI3K/MAPK/mTOR) signaling-pathways, and by the gene-expression regulation of key receptors/transcriptional-factors involved in the functioning of these pituitary cell-types (e.g. GHRH/ghrelin/somatostatin/insulin/IGF-I-receptors/Pit-1). Finally, we found that primate pituitaries expressed leptin/adiponectin/resistin. Altogether, these and previous data suggest that local-production of adipokines/receptors, in conjunction with circulating adipokine-levels, might comprise a relevant regulatory circuit that contribute to the fine-regulation of pituitary functions.
Collapse
Affiliation(s)
- André Sarmento-Cabral
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía (HURS), Córdoba, Spain.,CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain
| | - Juan R Peinado
- Department of Medical Sciences, Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Spain
| | - Lisa C Halliday
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, Illinois, USA
| | - María M Malagon
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía (HURS), Córdoba, Spain.,CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía (HURS), Córdoba, Spain.,CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain
| | - Rhonda D Kineman
- Research and Development Division, Jesse Brown Veterans Affairs Medical Center, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía (HURS), Córdoba, Spain.,CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain
| |
Collapse
|
70
|
Krishnan A, Muthusami S. Hormonal alterations in PCOS and its influence on bone metabolism. J Endocrinol 2017; 232:R99-R113. [PMID: 27895088 DOI: 10.1530/joe-16-0405] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/28/2016] [Indexed: 11/08/2022]
Abstract
According to the World Health Organization (WHO) polycystic ovary syndrome (PCOS) occurs in 4-8% of women worldwide. The prevalence of PCOS in Indian adolescents is 12.2% according to the Indian Council of Medical Research (ICMR). The National Institute of Health has documented that it affects approximately 5 million women of reproductive age in the United States. Hormonal imbalance is the characteristic of many women with polycystic ovarian syndrome (PCOS). The influence of various endocrine changes in PCOS women and their relevance to bone remains to be documented. Hormones, which include gonadotrophin-releasing hormone (GnRH), insulin, the leutinizing/follicle-stimulating hormone (LH/FSH) ratio, androgens, estrogens, growth hormones (GH), cortisol, parathyroid hormone (PTH) and calcitonin are disturbed in PCOS women. These hormones influence bone metabolism in human subjects directly as well as indirectly. The imbalance in these hormones results in increased prevalence of osteoporosis in PCOS women. Limited evidence suggests that the drugs taken during the treatment of PCOS increase the risk of bone fracture in PCOS patients through endocrine disruption. This review is aimed at the identification of the relationship between bone mineral density and hormonal changes in PCOS subjects and identifies potential areas to study bone-related disorders in PCOS women.
Collapse
Affiliation(s)
- Abhaya Krishnan
- Department of BiochemistryKarpagam University, Coimbatore, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of BiochemistryKarpagam University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
71
|
Birzniece V, Ho KKY. Sex steroids and the GH axis: Implications for the management of hypopituitarism. Best Pract Res Clin Endocrinol Metab 2017; 31:59-69. [PMID: 28477733 DOI: 10.1016/j.beem.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Growth hormone (GH) regulates somatic growth, substrate metabolism and body composition. Sex hormones exert profound effect on the secretion and action of GH. Estrogens stimulate the secretion of GH, but inhibit the action of GH on the liver, an effect that occurs when administered orally. Estrogens suppress GH receptor signaling by stimulating the expression proteins that inhibit cytokine receptor signaling. This effect of estrogens is avoided when physiological doses of estrogens are administered via a non-oral route. Estrogen-like compounds, such as selective estrogen receptor modulators, possess dual properties of inhibiting the secretion as well as the action of GH. In contrast, androgens stimulate GH secretion, driving IGF-1 production. In the periphery, androgens enhance the action of GH. The differential effects of estrogens and androgens influence the dose of GH replacement in patients with hypopituitarism on concomitant treatment with sex steroids. Where possible, a non-oral route of estrogen replacement is recommended for optimizing cost-benefit of GH replacement in women with GH deficiency. Adequate androgen replacement in conjunction with GH replacement is required to achieve the full anabolic effect in men with hypopituitarism.
Collapse
Affiliation(s)
- Vita Birzniece
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; School of Medicine, University of New South Wales, NSW 2052, Australia.
| | - Ken K Y Ho
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; Centres for Health Research, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
72
|
Nishikawa K, Furube E, Morita S, Horii-Hayashi N, Nishi M, Miyata S. Structural Reconstruction of the Perivascular Space in the Adult Mouse Neurohypophysis During an Osmotic Stimulation. J Neuroendocrinol 2017; 29. [PMID: 28072496 DOI: 10.1111/jne.12456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/19/2016] [Accepted: 01/08/2017] [Indexed: 12/11/2022]
Abstract
Oxytocin (OXT) and arginine vasopressin (AVP) neuropeptides in the neurohypophysis (NH) control lactation and body fluid homeostasis, respectively. Hypothalamic neurosecretory neurones project their axons from the supraoptic and paraventricular nuclei to the NH to make contact with the vascular surface and release OXT and AVP. The neurohypophysial vascular structure is unique because it has a wide perivascular space between the inner and outer basement membranes. However, the significance of this unique vascular structure remains unclear; therefore, we aimed to determine the functional significance of the perivascular space and its activity-dependent changes during salt loading in adult mice. The results obtained revealed that pericytes were the main resident cells and defined the profile of the perivascular space. Moreover, pericytes sometimes extended their cellular processes or 'perivascular protrusions' into neurohypophysial parenchyma between axonal terminals. The vascular permeability of low-molecular-weight (LMW) molecules was higher at perivascular protrusions than at the smooth vascular surface. Axonal terminals containing OXT and AVP were more likely to localise at perivascular protrusions than at the smooth vascular surface. Chronic salt loading with 2% NaCl significantly induced prominent changes in the shape of pericytes and also increased the number of perivascular protrusions and the surface area of the perivascular space together with elevations in the vascular permeability of LMW molecules. Collectively, these results indicate that the perivascular space of the NH acts as the main diffusion route for OXT and AVP and, in addition, changes in the shape of pericytes and perivascular reconstruction occur in response to an increased demand for neuropeptide release.
Collapse
Affiliation(s)
- K Nishikawa
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - E Furube
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
| | - S Morita
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
- Department of Anatomy and Neuroscience, Nara Medical University, Kashihara, Nara, Japan
| | - N Horii-Hayashi
- Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - M Nishi
- Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - S Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Kyoto, Japan
- The Center for Advanced Insect Research Promotion (CAIRP), Kyoto Institute of Technology, Kyoto, Japan
| |
Collapse
|
73
|
Rigamonti AE, Grugni G, Arreghini M, Capodaglio P, De Col A, Agosti F, Sartorio A. GH Responsiveness to Combined GH-Releasing Hormone and Arginine Administration in Obese Patients with Fibromyalgia Syndrome. Int J Endocrinol 2017; 2017:3106041. [PMID: 28744309 PMCID: PMC5506478 DOI: 10.1155/2017/3106041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 02/04/2023] Open
Abstract
Reportedly, fibromyalgia (FM) is frequently associated with reduced IGF-1 levels and GH hyporesponsiveness to different GH stimulation tests. Since there is a high prevalence of obesity in FM, and obesity itself is characterized by hyposomatotropism, the aim of this study was to assess IGF-1 levels and GH responsiveness in sixteen severely obese women suffering from FM, who, subdivided into two subgroups on the basis of their age-dependent IGF-1 values (> or <-2 SDS), underwent the combined GHRH plus arginine test. Four out of 16 obese women with FM (25%) had low IGF-1 SDS values, 2 cases of this subgroup (12.5%) failing also to normally respond to the test. Among patients with normal GH responses, 4 showed a delayed GH peak. The subgroup with low IGF-1 SDS values had higher BMI than that with normal IGF-1 SDS. GH peak and area under the curve were not correlated with CRP, ESR, or tender point score, while significant correlations were found with fat-free mass and fat mass. In conclusion, this study shows the existence of a high prevalence of GH-IGF-1 dysfunction in patients with both FM and obesity, presumably as a consequence of the obese rather than fibromyalgic condition.
Collapse
Affiliation(s)
- Antonello E. Rigamonti
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- *Antonello E. Rigamonti:
| | - Graziano Grugni
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Verbania, Italy
- Division of Auxology, Istituto Auxologico Italiano, IRCCS, Verbania, Italy
| | - Marco Arreghini
- Orthopedic Rehabilitation Unit, IRCCS, Istituto Auxologico Italiano, IRCCS, Verbania, Italy
| | - Paolo Capodaglio
- Orthopedic Rehabilitation Unit, IRCCS, Istituto Auxologico Italiano, IRCCS, Verbania, Italy
| | - Alessandra De Col
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Verbania, Italy
| | - Fiorenza Agosti
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Verbania, Italy
| | - Alessandro Sartorio
- Experimental Laboratory for Auxo-Endocrinological Research, Istituto Auxologico Italiano, IRCCS, Milan and Verbania, Italy
- Division of Auxology, Istituto Auxologico Italiano, IRCCS, Verbania, Italy
| |
Collapse
|
74
|
Couvelard A, Pélaprat D, Dokmak S, Sauvanet A, Voisin T, Couvineau A, Ruszniewski P. Antisecretory Effects of Chimeric Somatostatin/Dopamine Receptor Ligands on Gastroenteropancreatic Neuroendocrine Tumors. Pancreas 2017; 46:631-638. [PMID: 28375946 DOI: 10.1097/mpa.0000000000000813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The recent finding that gastroenteropancreatic neuroendocrine tumors expressed the dopaminergic D2 receptor in addition to somatostatin (sst) receptors suggested that multiple targeting approaches might decrease hormone hypersecretion more effectively than sst agonists alone. METHODS To test this hypothesis, (i) we measured the expression of sst receptor type 2 (sst2 receptor) and D2 receptor in 11 gastroenteropancreatic neuroendocrine tumors and (ii) we compared the ability of lanreotide, cabergoline, their combination, and sst/D2 chimeric ligands to decrease chromogranin A (CgA), gastrin, or serotonin release in primary cultures derived from these tumors. RESULTS Moderate to high positivity was observed for sst2 receptor and D2 receptor, the latter being more expressed in pancreatic tumors. Lanreotide decreased CgA secretion in all cultures, but only 3 tumors responded to cabergoline. No additivity was observed in lanreotide. BIM 23A781 decreased CgA release to the same extent as lanreotide, whereas the other chimeric ligands were less efficient. However, BIM 23A781 was 50 times less potent than lanreotide. Similar patterns were found for gastrin or serotonin. CONCLUSION No improvement was brought by the sst/D2 combination or chimeric ligands. Factors that underlie these tissue-specific differences remain to be elucidated.
Collapse
Affiliation(s)
- Anne Couvelard
- From the *Faculté de Médecine Xavier Bichat, INSERM U1149, Centre de Recherche sur l'Inflammation (CRI), DHU Unity, Université Paris Diderot; †Université Paris Diderot; ‡Département de Pathologie Beaujon-Bichat, Hôpital Bichat, DHU UNITY, AP-HP, Paris; and §Département de Chirurgie Pancréatico-Biliaire and ∥Département de Gastroentérologie-Pancréatologie, Hôpital Beaujon, DHU UNITY, AP-HP, Clichy, France
| | | | | | | | | | | | | |
Collapse
|
75
|
Miyata S. Advances in Understanding of Structural Reorganization in the Hypothalamic Neurosecretory System. Front Endocrinol (Lausanne) 2017; 8:275. [PMID: 29089925 PMCID: PMC5650978 DOI: 10.3389/fendo.2017.00275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic neurosecretory system synthesizes neuropeptides in hypothalamic nuclei and releases them from axonal terminals into the circulation in the neurohypophysis (NH) and median eminence (ME). This system plays a crucial role in regulating body fluid homeostasis and social behaviors as well as reproduction, growth, metabolism, and stress responses, and activity-dependent structural reorganization has been reported. Current knowledge on dynamic structural reorganization in the NH and ME, in which the axonal terminals of neurosecretory neurons directly contact the basement membrane (BM) of a fenestrated vasculature, is discussed herein. Glial cells, pituicytes in the NH and tanycytes in the ME, engulf axonal terminals and interpose their cellular processes between axonal terminals and the BM when hormonal demands are low. Increasing demands for neurosecretion result in the retraction of the cellular processes of glial cells from axonal terminals and the BM, permitting increased neurovascular contact. The shape conversion of pituicytes and tanycytes is mediated by neurotransmitters and sex steroid hormones, respectively. The NH and ME have a rough vascular BM profile of wide perivascular spaces and specialized extension structures called "perivascular protrusions." Perivascular protrusions, the insides of which are occupied by the cellular processes of vascular mural cells pericytes, contribute to increasing neurovascular contact and, thus, the efficient diffusion of hypothalamic neuropeptides. A chronic physiological stimulation has been shown to increase perivascular protrusions via the shape conversion of pericytes and the profile of the vascular surface. Continuous angiogenesis occurs in the NH and ME of healthy normal adult rodents depending on the signaling of vascular endothelial growth factor (VEGF). The inhibition of VEGF signaling suppresses the proliferation of endothelial cells (ECs) and promotes their apoptosis, which results in decreases in the population of ECs and axonal terminals. Pituicytes and tanycytes are continuously replaced by the proliferation and differentiation of stem/progenitor cells, which may be regulated by matching those of ECs and axonal terminals. In conclusion, structural reorganization in the NH and ME is caused by the activity-dependent shape conversion of glial cells and vascular mural cells as well as the proliferation of endothelial and glial cells by angiogenesis and gliogenesis, respectively.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, The Center for Advanced Insect Research Promotion (CAIRP), Kyoto Institute of Technology, Kyoto, Japan
- *Correspondence: Seiji Miyata,
| |
Collapse
|
76
|
|
77
|
Najafi P, Moghadasi M. The effect of yoga training on enhancement of Adrenocorticotropic hormone (ACTH) and cortisol levels in female patients with multiple sclerosis. Complement Ther Clin Pract 2016; 26:21-25. [PMID: 28107844 DOI: 10.1016/j.ctcp.2016.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 11/16/2022]
Abstract
The effect of 8 weeks yoga training on cortisol and Adrenocorticotropic hormone (ACTH) levels in female patients with Multiple Sclerosis (MS) is examined. Twenty four MS female patients with Expanded Disability Status Scale (EDSS) 1 to 5.5 participated in this study as the subject. The participants were divided into control (n = 10) or training group (n = 14) randomly. Training group performed 90 min yoga training per session, 3 days a week for 8 weeks. Assessments include body composition measurement and blood sampling 48 h before first session and 48 h after the intervention. The results demonstrated that ACTH increased and cortisol decreased compared to the control group (P < 0.05); In conclusion, it seems that yoga training modulates ACTH level in concomitant with reduction in cortisol level in female patients with MS.
Collapse
Affiliation(s)
- Parisa Najafi
- Department of Exercise Physiology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Mehrzad Moghadasi
- Department of Exercise Physiology, Shiraz Branch, Islamic Azad University, Shiraz, Iran.
| |
Collapse
|
78
|
Veldhuis JD, Erickson D, Yang R, Takahashi P, Bowers C. Endogenous Estrogen Regulates Somatostatin-Induced Rebound GH Secretion in Postmenopausal Women. J Clin Endocrinol Metab 2016; 101:4298-4304. [PMID: 27459535 PMCID: PMC5095244 DOI: 10.1210/jc.2016-2080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Systemic concentrations of T, estradiol (E2), GH, IGF-1, and IGF binding protein-3 decline in healthy aging individuals. Conversely, T and E2 stimulate GH and IGF-1 production in hypogonadal patients. HYPOTHESIS Because E2 stimulates GH secretion, putatively via the nuclear estrogen receptor-α and E2 and GH fall with menopause, we postulated that diminished endogenous E2 contributes to low GH output in older women. LOCATION The study was conducted at the Mayo Center for Clinical and Translational Science. STUDY DESIGN This was a randomized, double-blind, controlled study in 60 healthy postmenopausal women treated with the following: 1) double placebo; 2) anastrozole, a potent inhibitor of aromatase-enzyme activity, which mediates E2 synthesis from T; and/or 3) fulvestrant, a selective estrogen receptor-α antagonist. METHODS GH pulse generation was quantified by frequent GH sampling before and after short-term iv somatostatin infusion, thought to induce hypothalamic GHRH-mediated rebound-like GH secretion. RESULTS On anastrozole, E2 fell from 3.1 ± 0.35 pg/mL to 0.36 ± 0.04 pg/mL, and estrone from 13 ± 1.4 pg/mL to 1.9 ± 0.01 pg/mL (P < .001) by mass spectrometry. Estrogen values were unchanged by fulvestrant. T concentrations did not change. One-hour peak GH rebound after somatostatin infusion declined markedly during both estrogen-deprivation schedules (P < .001). Mean (150 min) maximal GH rebound decreased comparably (P < .001). Measures of GH rebound correlated negatively with computed tomography-estimated abdominal visceral fat (all P < .05). CONCLUSION These data suggest a previously unrecognized dependence of hypothalamo-pituitary GH regulation on low levels of endogenous estrogen after menopause.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit (J.D.V., D.E., R.Y.), Mayo Clinic College of Medicine, Center for Translational Science Activities, Mayo Clinic, and Department of Primary Care Internal Medicine (P.T.), Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| | - Dana Erickson
- Endocrine Research Unit (J.D.V., D.E., R.Y.), Mayo Clinic College of Medicine, Center for Translational Science Activities, Mayo Clinic, and Department of Primary Care Internal Medicine (P.T.), Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| | - Rebecca Yang
- Endocrine Research Unit (J.D.V., D.E., R.Y.), Mayo Clinic College of Medicine, Center for Translational Science Activities, Mayo Clinic, and Department of Primary Care Internal Medicine (P.T.), Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| | - Paul Takahashi
- Endocrine Research Unit (J.D.V., D.E., R.Y.), Mayo Clinic College of Medicine, Center for Translational Science Activities, Mayo Clinic, and Department of Primary Care Internal Medicine (P.T.), Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| | - Cyril Bowers
- Endocrine Research Unit (J.D.V., D.E., R.Y.), Mayo Clinic College of Medicine, Center for Translational Science Activities, Mayo Clinic, and Department of Primary Care Internal Medicine (P.T.), Mayo Clinic, Rochester, Minnesota 55905; and Tulane University Health Sciences Center (C.B.), Endocrinology and Metabolism Section, Peptide Research Section, New Orleans, Louisiana 70112
| |
Collapse
|
79
|
Growth Hormone-Releasing Hormone and Its Analogues: Significance for MSCs-Mediated Angiogenesis. Stem Cells Int 2016; 2016:8737589. [PMID: 27774107 PMCID: PMC5059609 DOI: 10.1155/2016/8737589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/19/2016] [Accepted: 07/03/2016] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising candidates for regenerative medicine because of their multipotency, immune-privilege, and paracrine properties including the potential to promote angiogenesis. Accumulating evidence suggests that the inherent properties of cytoprotection and tissue repair by native MSCs can be enhanced by various preconditioning stimuli implemented prior to cell transplantation. Growth hormone-releasing hormone (GHRH), a stimulator in extrahypothalamus systems including tumors, has attracted great attentions in recent years because GHRH and its agonists could promote angiogenesis in various tissues. GHRH and its agonists are proangiogenic in responsive tissues including tumors, and GHRH antagonists have been tested as antitumor agents through their ability to suppress angiogenesis and cell growth. GHRH-R is expressed by MSCs and evolving work from our laboratory indicates that treatment of MSCs with GHRH agonists prior to cell transplantation markedly enhanced the angiogenic potential and tissue reparative properties of MSCs through a STAT3 signaling pathway. In this review we summarized the possible effects of GHRH analogues on cell growth and development, as well as on the proangiogenic properties of MSCs. We also discussed the relationship between GHRH analogues and MSC-mediated angiogenesis. The analyses provide new insights into molecular pathways of MSCs-based therapies and their augmentation by GHRH analogues.
Collapse
|
80
|
Lindsey RC, Mohan S. Skeletal effects of growth hormone and insulin-like growth factor-I therapy. Mol Cell Endocrinol 2016; 432:44-55. [PMID: 26408965 PMCID: PMC4808510 DOI: 10.1016/j.mce.2015.09.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 10/23/2022]
Abstract
The growth hormone/insulin-like growth factor (GH/IGF) axis is critically important for the regulation of bone formation, and deficiencies in this system have been shown to contribute to the development of osteoporosis and other diseases of low bone mass. The GH/IGF axis is regulated by a complex set of hormonal and local factors which can act to regulate this system at the level of the ligands, receptors, IGF binding proteins (IGFBPs), or IGFBP proteases. A combination of in vitro studies, transgenic animal models, and clinical human investigations has provided ample evidence of the importance of the endocrine and local actions of both GH and IGF-I, the two major components of the GH/IGF axis, in skeletal growth and maintenance. GH- and IGF-based therapies provide a useful avenue of approach for the prevention and treatment of diseases such as osteoporosis.
Collapse
Affiliation(s)
- Richard C Lindsey
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA Healthcare System, Loma Linda, CA 92357, USA; Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
81
|
Zhang Y, D'Argenio DZ. Feedback control indirect response models. J Pharmacokinet Pharmacodyn 2016; 43:343-58. [PMID: 27394724 DOI: 10.1007/s10928-016-9479-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 06/13/2016] [Indexed: 11/29/2022]
Abstract
A general framework is introduced for modeling pharmacodynamic processes that are subject to autoregulation, which combines the indirect response (IDR) model approach with methods from classical feedback control of engineered systems. The canonical IDR models are modified to incorporate linear combinations of feedback control terms related to the time course of the difference (the error signal) between the pharmacodynamic response and its basal value. Following the well-established approach of traditional engineering control theory, the proposed feedback control indirect response models incorporate terms proportional to the error signal itself, the integral of the error signal, the derivative of the error signal or combinations thereof. Simulations are presented to illustrate the types of responses produced by the proposed feedback control indirect response model framework, and to illustrate comparisons with other PK/PD modeling approaches incorporating feedback. In addition, four examples from literature are used to illustrate the implementation and applicability of the proposed feedback control framework. The examples reflect each of the four mechanisms of drug action as modeled by each of the four canonical IDR models and include: selective serotonin reuptake inhibitors and extracellular serotonin; histamine H2-receptor antagonists and gastric acid; growth hormone secretagogues and circulating growth hormone; β2-selective adrenergic agonists and potassium. The proposed feedback control indirect response approach may serve as an exploratory modeling tool and may provide a bridge for development of more mechanistic systems pharmacology models.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - David Z D'Argenio
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
82
|
Maliza R, Fujiwara K, Tsukada T, Azuma M, Kikuchi M, Yashiro T. Effects of retinoic acid on growth hormone-releasing hormone receptor, growth hormone secretagogue receptor gene expression and growth hormone secretion in rat anterior pituitary cells. Endocr J 2016; 63:555-61. [PMID: 27052215 DOI: 10.1507/endocrj.ej16-0086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Retinoic acid (RA) is an important signaling molecule in embryonic development and adult tissue. The actions of RA are mediated by the nuclear receptors retinoic acid receptor (RAR) and retinoid X receptor (RXR), which regulate gene expression. RAR and RXR are widely expressed in the anterior pituitary gland. RA was reported to stimulate growth hormone (GH) gene expression in the anterior pituitary cells. However, current evidence is unclear on the role of RA in gene expression of growth hormone-releasing hormone receptor (Ghrh-r), growth hormone secretagogue receptor (Ghs-r) and somatostatin receptors (Sst-rs). Using isolated anterior pituitary cells of rats, we examined the effects of RA on gene expression of these receptors and GH release. Quantitative real-time PCR revealed that treatment with all-trans retinoic acid (ATRA; 10(-6) M) for 24 h increased gene expression levels of Ghrh-r and Ghs-r; however, expressions of Sst-r2 and Sst-r5 were unchanged. Combination treatment with the RAR-agonist Am80 and RXR-agonist PA024 mimicked the effects of ATRA on Ghrh-r and Ghs-r gene expressions. Exposure of isolated pituitary cells to ATRA had no effect on basal GH release. In contrast, ATRA increased growth hormone-releasing hormone (GHRH)- and ghrelin-stimulated GH release from cultured anterior pituitary cells. Our results suggest that expressions of Ghrh-r and Ghs-r are regulated by RA through the RAR-RXR receptor complex and that RA enhances the effects of GHRH and ghrelin on GH release from the anterior pituitary gland.
Collapse
Affiliation(s)
- Rita Maliza
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | | | | | | | | | | |
Collapse
|
83
|
Lee RJ, Fanning JR, Coccaro EF. GH response to intravenous clonidine challenge correlates with history of childhood trauma in personality disorder. J Psychiatr Res 2016; 76:38-43. [PMID: 26874268 DOI: 10.1016/j.jpsychires.2015.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 11/16/2015] [Accepted: 11/19/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Childhood trauma is a risk factor for personality disorder. We have previously shown that childhood trauma is associated with increased central corticotrophin-releasing hormone concentration in adults with personality disorder. In the brain, the release of corticotrophin-releasing hormone can be stimulated by noradrenergic neuronal activity, raising the possibility that childhood trauma may affect the hypothalamic-pituitary adrenal (HPA) axis by altering brain noradrenergic function. In this study, we sought to test the hypothesis that childhood trauma is associated with blunted growth hormone response to the α-2 adrenergic autoreceptor agonist clonidine. METHODS All subjects provided written informed consent. Twenty personality disordered and twenty healthy controls (without personality disorder or Axis I psychopathology) underwent challenge with clonidine, while plasma Growth Hormone (GH) concentration was monitored by intravenous catheter. On a different study session, subjects completed the Childhood Trauma Questionnaire and underwent diagnostic interviews. RESULTS Contrary to our a priori hypothesis, childhood trauma was associated with enhanced GH response to clonidine. This positive relationship was present in the group of 40 subjects and in the subgroup 20 personality disordered subjects, but was not detected in the healthy control subjects when analyzed separately. The presence of personality disorder was unrelated to the magnitude of GH response. DISCUSSION Childhood trauma is positively correlated with GH response to clonidine challenge in adults with personality disorder. Enhanced rather that blunted GH response differentiates childhood trauma from previously identified negative predictors of GH response, such as anxiety or mood disorder.
Collapse
Affiliation(s)
- Royce J Lee
- Clinical Neuroscience & Psychopharmacology Research Unit, Department of Psychiatry and Behavioral Neuroscience, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Jennifer R Fanning
- Clinical Neuroscience & Psychopharmacology Research Unit, Department of Psychiatry and Behavioral Neuroscience, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Emil F Coccaro
- Clinical Neuroscience & Psychopharmacology Research Unit, Department of Psychiatry and Behavioral Neuroscience, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
84
|
Do A, Menon V, Zhi X, Gesing A, Wiesenborn DS, Spong A, Sun L, Bartke A, Masternak MM. Thyroxine modifies the effects of growth hormone in Ames dwarf mice. Aging (Albany NY) 2016; 7:241-55. [PMID: 25935838 PMCID: PMC4429089 DOI: 10.18632/aging.100739] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ames dwarf (df/df) mice lack growth hormone (GH), thyroid stimulating hormone and prolactin. Treatment of juvenile df/df mice with GH alone stimulates somatic growth, reduces insulin sensitivity and shortens lifespan. Early‐life treatment with thyroxine (T4) alone produces modest growth stimulation but does not affect longevity. In this study, we examined the effects of treatment of juvenile Ames dwarf mice with a combination of GH + T4 and compared them to the effects of GH alone. Treatment of female and male dwarfs with GH + T4 between the ages of 2 and 8 weeks rescued somatic growth yet did not reduce lifespan to match normal controls, thus contrasting with the previously reported effects of GH alone. While the male dwarf GH + T4 treatment group had no significant effect on lifespan, the female dwarfs undergoing treatment showed a decrease in maximal longevity. Expression of genes related to GH and insulin signaling in the skeletal muscle and white adipose tissue (WAT) of female dwarfs was differentially affected by treatment with GH + T4 vs. GH alone. Differences in the effects of GH + T4 vs. GH alone on insulin target tissues may contribute to the differential effects of these treatments on longevity.
Collapse
Affiliation(s)
- Andrew Do
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Vinal Menon
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.,Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC 29209, USA
| | - Xu Zhi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.,Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Adam Gesing
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Denise S Wiesenborn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.,Department of Medical Biochemistry and Molecular Biology, University of Saarland, 66421 Homburg, Germany.,Department of Biotechnology, University of Applied Sciences Kaiserslautern, 66482 Zweibrücken, Germany
| | - Adam Spong
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Liou Sun
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.,Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
85
|
Fridlyand LE, Tamarina NA, Schally AV, Philipson LH. Growth Hormone-Releasing Hormone in Diabetes. Front Endocrinol (Lausanne) 2016; 7:129. [PMID: 27777568 PMCID: PMC5056186 DOI: 10.3389/fendo.2016.00129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 12/13/2022] Open
Abstract
Growth hormone-releasing hormone (GHRH) is produced by the hypothalamus and stimulates growth hormone synthesis and release in the anterior pituitary gland. In addition, GHRH is an important regulator of cellular functions in many cells and organs. Expression of GHRH G-Protein Coupled Receptor (GHRHR) has been demonstrated in different peripheral tissues and cell types, including pancreatic islets. Among the peripheral activities, recent studies demonstrate a novel ability of GHRH analogs to increase and preserve insulin secretion by beta-cells in isolated pancreatic islets, which makes them potentially useful for diabetes treatment. This review considers the role of GHRHR in the beta-cell and addresses the unique engineered GHRH agonists and antagonists for treatment of type 2 diabetes mellitus. We discuss the similarity of signaling pathways activated by GHRHR in pituitary somatotrophs and in pancreatic beta-cells and possible ways as to how the GHRHR pathway can interact with glucose and other secretagogues to stimulate insulin secretion. We also consider the hypothesis that novel GHRHR agonists can improve glucose metabolism in Type 2 diabetes by preserving the function and survival of pancreatic beta-cells. Wound healing and cardioprotective action with new GHRH agonists suggest that they may prove useful in ameliorating certain diabetic complications. These findings highlight the future potential therapeutic effectiveness of modulators of GHRHR activity for the development of new therapeutic approaches in diabetes and its complications.
Collapse
Affiliation(s)
- Leonid E. Fridlyand
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- *Correspondence: Leonid E. Fridlyand,
| | - Natalia A. Tamarina
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Andrew V. Schally
- VA Medical Center, Miami, FL, USA
- Department of Pathology and Medicine, Division of Endocrinology and Hematology-Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Louis H. Philipson
- Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
86
|
Abstract
The regulation of organ size is essential to human health and has fascinated biologists for centuries. Key to the growth process is the ability of most organs to integrate organ-extrinsic cues (eg, nutritional status, inflammatory processes) with organ-intrinsic information (eg, genetic programs, local signals) into a growth response that adapts to changing environmental conditions and ensures that the size of an organ is coordinated with the rest of the body. Paired organs such as the vertebrate limbs and the long bones within them are excellent models for studying this type of regulation because it is possible to manipulate one member of the pair and leave the other as an internal control. During development, growth plates at the end of each long bone produce a transient cartilage model that is progressively replaced by bone. Here, we review how proliferation and differentiation of cells within each growth plate are tightly controlled mainly by growth plate-intrinsic mechanisms that are additionally modulated by extrinsic signals. We also discuss the involvement of several signaling hubs in the integration and modulation of growth-related signals and how they could confer remarkable plasticity to the growth plate. Indeed, long bones have a significant ability for "catch-up growth" to attain normal size after a transient growth delay. We propose that the characterization of catch-up growth, in light of recent advances in physiology and cell biology, will provide long sought clues into the molecular mechanisms that underlie organ growth regulation. Importantly, catch-up growth early in life is commonly associated with metabolic disorders in adulthood, and this association is not completely understood. Further elucidation of the molecules and cellular interactions that influence organ size coordination should allow development of novel therapies for human growth disorders that are noninvasive and have minimal side effects.
Collapse
Affiliation(s)
- Alberto Roselló-Díez
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| |
Collapse
|
87
|
Sadagurski M, Landeryou T, Cady G, Kopchick JJ, List EO, Berryman DE, Bartke A, Miller RA. Growth hormone modulates hypothalamic inflammation in long-lived pituitary dwarf mice. Aging Cell 2015; 14:1045-54. [PMID: 26268661 PMCID: PMC4693470 DOI: 10.1111/acel.12382] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 12/11/2022] Open
Abstract
Mice in which the genes for growth hormone (GH) or GH receptor (GHR(-/-) ) are disrupted from conception are dwarfs, possess low levels of IGF-1 and insulin, have low rates of cancer and diabetes, and are extremely long-lived. Median longevity is also increased in mice with deletion of hypothalamic GH-releasing hormone (GHRH), which leads to isolated GH deficiency. The remarkable extension of longevity in hypopituitary Ames dwarf mice can be reversed by a 6-week course of GH injections started at the age of 2 weeks. Here, we demonstrate that mutations that interfere with GH production or response, in the Snell dwarf, Ames dwarf, or GHR(-/-) mice lead to reduced formation of both orexigenic agouti-related peptide (AgRP) and anorexigenic proopiomelanocortin (POMC) projections to the main hypothalamic projection areas: the arcuate nucleus (ARH), paraventricular nucleus (PVH), and dorsomedial nucleus (DMH). These mutations also reduce hypothalamic inflammation in 18-month-old mice. GH injections, between 2 and 8 weeks of age, reversed both effects in Ames dwarf mice. Disruption of GHR specifically in liver (LiGHRKO), a mutation that reduces circulating IGF-1 but does not lead to lifespan extension, had no effect on hypothalamic projections or inflammation, suggesting an effect of GH, rather than peripheral IGF-1, on hypothalamic development. Hypothalamic leptin signaling, as monitored by induction of pStat3, is not impaired by GHR deficiency. Together, these results suggest that early-life disruption of GH signaling produces long-term hypothalamic changes that may contribute to the longevity of GH-deficient and GH-resistant mice.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Department of Internal Medicine Division of Geriatric and Palliative Medicine University of Michigan Ann Arbor MI USA
| | - Taylor Landeryou
- Department of Pathology and Geriatrics Center University of Michigan Ann Arbor MI USA
| | - Gillian Cady
- Department of Pathology and Geriatrics Center University of Michigan Ann Arbor MI USA
| | | | - Edward O. List
- Edison Biotechnology Institute Ohio University Athens OH USA
| | | | - Andrzej Bartke
- Department of Internal Medicine–Geriatrics Research Southern Illinois University School of Medicine Springfield IL USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center University of Michigan Ann Arbor MI USA
| |
Collapse
|
88
|
Miyata S. New aspects in fenestrated capillary and tissue dynamics in the sensory circumventricular organs of adult brains. Front Neurosci 2015; 9:390. [PMID: 26578857 PMCID: PMC4621430 DOI: 10.3389/fnins.2015.00390] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/05/2015] [Indexed: 12/20/2022] Open
Abstract
The blood-brain barrier (BBB) generally consists of endothelial tight junction barriers that prevent the free entry of blood-derived substances, thereby maintaining the extracellular environment of the brain. However, the circumventricular organs (CVOs), which are located along the midlines of the brain ventricles, lack these endothelial barriers and have fenestrated capillaries; therefore, they have a number of essential functions, including the transduction of information between the blood circulation and brain. Previous studies have demonstrated the extensive contribution of the CVOs to body fluid and thermal homeostasis, energy balance, the chemoreception of blood-derived substances, and neuroinflammation. In this review, recent advances have been discussed in fenestrated capillary characterization and dynamic tissue reconstruction accompanied by angiogenesis and neurogliogenesis in the sensory CVOs of adult brains. The sensory CVOs, including the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO), and area postrema (AP), have size-selective and heterogeneous vascular permeabilities. Astrocyte-/tanycyte-like neural stem cells (NSCs) sense blood- and cerebrospinal fluid-derived information through the transient receptor potential vanilloid 1, a mechanical/osmotic receptor, Toll-like receptor 4, a lipopolysaccharide receptor, and Nax, a Na-sensing Na channel. They also express tight junction proteins and densely and tightly surround mature neurons to protect them from blood-derived neurotoxic substances, indicating that the NSCs of the CVOs perform BBB functions while maintaining the capacity to differentiate into new neurons and glial cells. In addition to neurogliogenesis, the density of fenestrated capillaries is regulated by angiogenesis, which is accompanied by the active proliferation and sprouting of endothelial cells. Vascular endothelial growth factor (VEGF) signaling may be involved in angiogenesis and neurogliogenesis, both of which affect vascular permeability. Thus, recent findings advocate novel concepts for the CVOs, which have the dynamic features of vascular and parenchymal tissues.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, Kyoto Institute of TechnologyKyoto, Japan
| |
Collapse
|
89
|
Sehgal PB, Yang YM, Miller EJ. Hypothesis: Neuroendocrine Mechanisms (Hypothalamus-Growth Hormone-STAT5 Axis) Contribute to Sex Bias in Pulmonary Hypertension. Mol Med 2015; 21:688-701. [PMID: 26252185 PMCID: PMC4749490 DOI: 10.2119/molmed.2015.00122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/30/2015] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension (PH) is a disease with high morbidity and mortality. The prevalence of idiopathic pulmonary arterial hypertension (IPAH) and hereditary pulmonary arterial hypertension (HPAH) is approximately two- to four-fold higher in women than in men. Paradoxically, there is an opposite male bias in typical rodent models of PH (chronic hypoxia or monocrotaline); in these models, administration of estrogenic compounds (for example, estradiol-17β [E2]) is protective. Further complexities are observed in humans ingesting anorexigens (female bias) and in rodent models, such as after hypoxia plus SU5416/Sugen (little sex bias) or involving serotonin transporter overexpression or dexfenfluramine administration (female bias). These complexities in sex bias in PH remain incompletely understood. We recently discovered that conditional deletion of signal transducer and activator of transcription 5a/b (STAT5a/b) in vascular smooth muscle cells abrogated the male bias in PH in hypoxic mice and that late-stage obliterative lesions in patients of both sexes with IPAH and HPAH showed reduced STAT5a/b, reduced Tyr-P-STAT5 and reduced B-cell lymphoma 6 protein (BCL6). In trying to understand the significance of these observations, we realized that there existed a well-characterized E2-sensitive central neuroendocrine mechanism of sex bias, studied over the last 40 years, that, at its peripheral end, culminated in species-specific male ("pulsatile") versus female ("more continuous") temporal patterns of circulating growth hormone (GH) levels leading to male versus female patterned activation of STAT5a/b in peripheral tissues and thus sex-biased expression of hundreds of genes. In this report, we consider the contribution of this neuroendocrine mechanism (hypothalamus-GH-STAT5) in the generation of sex bias in different PH situations.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, United States of America
- Department of Medicine, New York Medical College, Valhalla, New York, United States of America
| | - Yang-Ming Yang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Edmund J Miller
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| |
Collapse
|
90
|
Sehgal PB, Yang YM, Yuan H, Miller EJ. STAT5a/b contribute to sex bias in vascular disease: A neuroendocrine perspective. JAKSTAT 2015; 4:1-20. [PMID: 27141328 DOI: 10.1080/21623996.2015.1090658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
Previous studies have elucidated a neuroendocrine mechanism consisting of the hypothalamus (growth hormone releasing hormone, GHRH) - pituitary (growth hormone, GH) - STAT5a/b axis that underlies sex-biased gene expression in the liver. It is now established that male vs female patterned secretion of GHRH, and thus of circulating GH levels ("pulsatile" vs "more continuous" respectively), leading to differently patterned activation of PY-STAT5a/b in hepatocytes results in sex-biased gene expression of cohorts of hundreds of downstream genes. This review outlines new data in support of a STAT5a/b-based mechanism of sex bias in the vascular disease pulmonary hypertension (PH). Puzzling observations in PH include its 2-4-fold higher prevalence in women but a male-dominance in many rodent models, and, paradoxically, inhibition of PH development by estrogens in such models. We observed that conditional deletion of STAT5a/b in vascular smooth muscle cells (SMC) in mice converted the male-dominant model of chronic hypoxia-induced PH into a female-dominant phenotype. In human idiopathic PH, there was reduced STAT5a/b and PY-STAT5 in cells in late-stage obliterative pulmonary arterial lesions in both men and women. A juxtaposition of the prior liver data with the newer PH-related data drew attention to the hypothalamus-GH-STAT5 axis, which is the major target of estrogens at the level of the hypothalamus. This hypothesis explains many of the puzzling aspects of sex bias in PH in humans and rodent models. The extension of STAT5-anchored mechanisms of sex bias to vascular disease emphasizes the contribution of central neuroendocrine processes in generating sexual dimorphism in different tissues and cell types.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Departments of Cell Biology & Anatomy; New York Medical College; Valhalla, NY USA; Department of Medicine; New York Medical College; Valhalla, NY USA
| | - Yang-Ming Yang
- Departments of Cell Biology & Anatomy; New York Medical College ; Valhalla, NY USA
| | - Huijuan Yuan
- Departments of Cell Biology & Anatomy; New York Medical College ; Valhalla, NY USA
| | - Edmund J Miller
- Center for Heart and Lung Research; The Feinstein Institute for Medical Research ; Manhasset, NY USA
| |
Collapse
|
91
|
Kasuya E. Secretory pattern and regulatory mechanism of growth hormone in cattle. Anim Sci J 2015; 87:178-82. [PMID: 26260675 PMCID: PMC5042056 DOI: 10.1111/asj.12418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 11/30/2022]
Abstract
The ultradian rhythm of growth hormone (GH) secretion has been known in several animal species for years and has recently been observed in cattle. Although the physiological significance of the rhythm is not yet fully understood, it appears essential for normal growth. In this review, previous studies concerning the GH secretory pattern in cattle, including its ultradian rhythm, are introduced and the regulatory mechanism is discussed on the basis of recent findings.
Collapse
Affiliation(s)
- Etsuko Kasuya
- Animal Physiology Research Unit, Division of Animal Sciences, National Institute of Agrobiological Sciences, Tsukuba, Japan
| |
Collapse
|
92
|
Rigamonti AE, Grugni G, Marazzi N, Bini S, Bidlingmaier M, Sartorio A. Unaltered ratio of circulating levels of growth hormone/GH isoforms in adults with Prader-Willi syndrome after GHRH plus arginine administration. Growth Horm IGF Res 2015; 25:168-173. [PMID: 26059749 DOI: 10.1016/j.ghir.2015.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/28/2022]
Abstract
Human growth hormone (GH) is a heterogeneous protein hormone consisting of several isoforms, the most abundant being 22 kDa- and 20 kDa-GH. The availability of analytical methods to measure these GH isoforms might represent a valuable diagnostic tool to investigate GH hyposecretory states, including Prader-Willi syndrome (PWS), one of the most common causes of syndromic obesity. The aim of the present study was to measure circulating levels of 22 kDa- and 20 kDa-GH in PWS adults (n=14; M/F: 5/9; genotype DEL15/UPD15: 12/2; age: 19.0±3.7 years; BMI: 29.9±8.7 kg/m2) after combined GH releasing hormone (GHRH) plus arginine (ARG) administration. The results were analysed subdividing the study population in obese vs. nonobese (6/8) and GH deficient vs. nonGH deficient (GHD) (6/8) subjects, according to appropriate BMI-related diagnostic cut-off limits of GH peak response to the provocative test. Circulating levels of 22 kDa-GH were measured by a chemiluminescent method based on a detection monoclonal antibody targeting an epitope in the loop connecting helix 1 and 2 of GH, which is missing in 20 kDa-GH; the 20 kDa-GH was measured using a time resolved fluorescence assay based on two monoclonal antibodies with no cross-reactivity to 22-kDa GH. GHRH plus ARG significantly stimulated the secretions of 22 kDa- and 20 kDa-GH in nonobese (at 30, 45, 60 and 90 min and at 45, 60, 90 and 120 min vs. 0 min, p<0.05, with GH peaks of 15.8±10.3 ng/ml and 2.7±1.2 ng/ml, respectively) and in nonGHD PWS (at 30, 45 and 60 min and at 45, 60 and 90 min vs 0 min, p<0.05, with GH peaks of 12.5±9.0 ng/ml and 2.0±1.8 ng/ml, respectively). No significant GHRH plus ARG-induced changes in 22 kDa- and 20 kDa-GH were observed in obese or GHD PWS patients, the only exception being the increase of 22 kDa-GH (p<0.05) 60 min after the stimulus administration in GHD group (with GH peaks of 6.9±4.7 ng/ml and 0.8±0.6 ng/ml in obese subjects and 8.5±6.0 ng/ml and 1.2±1.0 ng/ml in GHD subjects for 22 kDa- and 20 kDa-GH, respectively). The GH responses for both isoforms were significantly higher in nonobese than in obese PWS patients (at 45 and 60 min for 22 kDa-GH and at 45, 60, 90 and 120 min for 20 kDa-GH, p<0.05), while no differences were detected between GHD vs. nonGHD groups. As previously reported in healthy subjects, the ratios of circulating levels of 22 kDa- to 20 kDa-GH remained constant after GHRH plus ARG both in obese/non-obese and GHD/non-GHD groups, thus suggesting the preservation of a normal balance in GH isoforms in PWS.
Collapse
Affiliation(s)
- A E Rigamonti
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy.
| | - G Grugni
- Istituto Auxologico Italiano, IRCCS, Experimental Laboratory for Auxo-endocrinological Research, Milan and Verbania, Italy; Istituto Auxologico Italiano, IRCCS, Division of Auxology, Verbania, Italy
| | - N Marazzi
- Istituto Auxologico Italiano, IRCCS, Experimental Laboratory for Auxo-endocrinological Research, Milan and Verbania, Italy
| | - S Bini
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - M Bidlingmaier
- Endocrine Research Laboratories, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - A Sartorio
- Istituto Auxologico Italiano, IRCCS, Experimental Laboratory for Auxo-endocrinological Research, Milan and Verbania, Italy; Istituto Auxologico Italiano, IRCCS, Division of Auxology, Verbania, Italy
| |
Collapse
|
93
|
Ye RS, Li M, Qi QE, Cheng X, Chen T, Li CY, Wang SB, Shu G, Wang LN, Zhu XT, Jiang QY, Xi QY, Zhang YL. Comparative Anterior Pituitary miRNA and mRNA Expression Profiles of Bama Minipigs and Landrace Pigs Reveal Potential Molecular Network Involved in Animal Postnatal Growth. PLoS One 2015; 10:e0131987. [PMID: 26134288 PMCID: PMC4489742 DOI: 10.1371/journal.pone.0131987] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/09/2015] [Indexed: 12/29/2022] Open
Abstract
The anterior pituitary is the most important endocrine organ modulating animal postnatal growth, mainly by controlling growth hormone (GH) gene transcription, synthesis, and secretion. As an ideal model for animal postnatal growth studies, the Bama minipig is characterized as having a lower growth performance and fewer individual differences compared with larger pig breeds. In this study, anterior pituitaries from Bama minipig and Landrace pig were used for miRNA and mRNA expression profile analysis using miRNA microarrays and mRNA-seq. Consequently, a total of 222 miRNAs and 12,909 transcripts were detected, and both miRNAs and mRNAs in the two breeds showed high correlation (r > 0.97). Additionally, 41 differentially expressed miRNAs and 2,254 transcripts were identified. Pathways analysis indicated that 32 pathways significantly differed in the two breeds. Importantly, two GH-regulation-signalling pathways, cAMP and inositol 1, 4, 5-triphosphate (IP3), and multiple GH-secretion-related transcripts were significantly down-regulated in Bama minipigs. Moreover, TargetScan and RNAHybrid algorithms were used for predicting differentially expressed miRNAs (DE miRNAs) and differentially expressed mRNAs (DE mRNAs) interaction. By examining their fold-changes, interestingly, most DE miRNA-DE mRNA target pairs (63.68-71.33%) presented negatively correlated expression pattern. A possible network among miRNAs, mRNAs, and GH-regulation pathways was also proposed. Among them, two miRNA-mRNA interactions (Y-47 targets FSHB; ssc-miR-133a-3p targets GNAI3) were validated by dual-luciferase assay. These data will be helpful in understanding the possible molecular mechanisms involved in animal postnatal growth.
Collapse
Affiliation(s)
- Rui-Song Ye
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Meng Li
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qi-En Qi
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiao Cheng
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ting Chen
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Chao-Yun Li
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Song-Bo Wang
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Gang Shu
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Li-Na Wang
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiao-Tong Zhu
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qing-Yan Jiang
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qian-Yun Xi
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- * E-mail: (YLZ); (QYX)
| | - Yong-Liang Zhang
- Chinese National Engineering Research Center for Breeding Swine Industry, SCAU-Alltech Research Joint Alliance, Guandong Provincial Key Lab of Agro-Animal Genomics And Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- * E-mail: (YLZ); (QYX)
| |
Collapse
|
94
|
Proudan N, Peroski M, Grignol G, Merchenthaler I, Dudas B. Juxtapositions between the somatostatinergic and growth hormone-releasing hormone (GHRH) neurons in the human hypothalamus. Neuroscience 2015; 297:205-10. [DOI: 10.1016/j.neuroscience.2015.03.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/06/2015] [Accepted: 03/24/2015] [Indexed: 11/29/2022]
|
95
|
Ross AW, Russell L, Helfer G, Thomson LM, Dalby MJ, Morgan PJ. Photoperiod regulates lean mass accretion, but not adiposity, in growing F344 rats fed a high fat diet. PLoS One 2015; 10:e0119763. [PMID: 25789758 PMCID: PMC4366045 DOI: 10.1371/journal.pone.0119763] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/16/2015] [Indexed: 01/10/2023] Open
Abstract
In this study the effects of photoperiod and diet, and their interaction, were examined for their effects on growth and body composition in juvenile F344 rats over a 4-week period. On long (16L:8D), relative to short (8L:16D), photoperiod food intake and growth rate were increased, but percentage adiposity remained constant (ca 3-4%). On a high fat diet (HFD), containing 22.8% fat (45% energy as fat), food intake was reduced, but energy intake increased on both photoperiods. This led to a small increase in adiposity (up to 10%) without overt change in body weight. These changes were also reflected in plasma leptin and lipid levels. Importantly while both lean and adipose tissue were strongly regulated by photoperiod on a chow diet, this regulation was lost for adipose, but not lean tissue, on HFD. This implies that a primary effect of photoperiod is the regulation of growth and lean mass accretion. Consistent with this both hypothalamic GHRH gene expression and serum IGF-1 levels were photoperiod dependent. As for other animals and humans, there was evidence of central hyposomatotropism in response to obesity, as GHRH gene expression was suppressed by the HFD. Gene expression of hypothalamic AgRP and CRH, but not NPY nor POMC, accorded with the energy balance status on long and short photoperiod. However, there was a general dissociation between plasma leptin levels and expression of these hypothalamic energy balance genes. Similarly there was no interaction between the HFD and photoperiod at the level of the genes involved in thyroid hormone metabolism (Dio2, Dio3, TSHβ or NMU), which are important mediators of the photoperiodic response. These data suggest that photoperiod and HFD influence body weight and body composition through independent mechanisms but in each case the role of the hypothalamic energy balance genes is not predictable based on their known function.
Collapse
Affiliation(s)
- Alexander W. Ross
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Laura Russell
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Gisela Helfer
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Lynn M. Thomson
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Matthew J. Dalby
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Peter J. Morgan
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
96
|
Yang YM, Yuan H, Edwards JG, Skayian Y, Ochani K, Miller EJ, Sehgal PB. Deletion of STAT5a/b in vascular smooth muscle abrogates the male bias in hypoxic pulmonary hypertension in mice: implications in the human disease. Mol Med 2015; 20:625-38. [PMID: 25470773 DOI: 10.2119/molmed.2014.00180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 11/20/2014] [Indexed: 12/27/2022] Open
Abstract
Chronic hypoxia typically elicits pulmonary hypertension (PH) in mice with a male-dominant phenotype. There is an opposite-sex bias in human PH, with a higher prevalence in women, but greater survival (the "estrogen paradox"). We investigated the involvement of the STAT5a/b species, previously established to mediate sexual dimorphism in other contexts, in the sex bias in PH. Mice with heterozygous or homozygous deletions of the STAT5a/b locus in vascular smooth muscle cells (SMCs) were generated in crosses between STAT5a/b(fl/fl) and transgelin (SM22α)-Cre(+/+) parents. Wild-type (wt) males subjected to chronic hypoxia showed significant PH and pulmonary arterial remodeling, with wt females showing minimal changes (a male-dominant phenotype). However, in conditional STAT5(+/-) or STAT5(-/-) mice, hypoxic females showed the severest manifestations of PH (a female-dominant phenotype). Immunofluorescence studies on human lung sections showed that obliterative pulmonary arterial lesions in patients with idiopathic pulmonary arterial hypertension (IPAH) or hereditary pulmonary arterial hypertension (HPAH), both male and female, overall had reduced STAT5a/b, reduced PY-STAT5 and reduced endoplasmic reticulum (ER) GTPase atlastin-3 (ATL3). Studies of SMCs and endothelial cell (EC) lines derived from vessels isolated from lungs of male and female IPAH patients and controls revealed instances of coordinate reductions in STAT5a, STAT5b and ATL3 in IPAH-derived cells, including SMCs and ECs from the same patient. Taken together, these data provide the first definitive evidence for a contribution of STAT5a/b to the sex bias in PH in the hypoxic mouse and implicate reduced STAT5 in the pathogenesis of the human disease.
Collapse
Affiliation(s)
- Yang-Ming Yang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Huijuan Yuan
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York, United States of America
| | - Yester Skayian
- Department of Physiology, New York Medical College, Valhalla, New York, United States of America
| | - Kanta Ochani
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Edmund J Miller
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Pravin B Sehgal
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America.,Department of Medicine, New York Medical College, Valhalla, New York, United States of America
| |
Collapse
|
97
|
Ibáñez-Costa A, Córdoba-Chacón J, Gahete MD, Kineman RD, Castaño JP, Luque RM. Melatonin regulates somatotrope and lactotrope function through common and distinct signaling pathways in cultured primary pituitary cells from female primates. Endocrinology 2015; 156:1100-10. [PMID: 25545385 PMCID: PMC4330310 DOI: 10.1210/en.2014-1819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Melatonin (MT) is secreted by the pineal gland and exhibits a striking circadian rhythm in its release. Depending on the species studied, some pituitary hormones also display marked circadian/seasonal patterns and rhythms of secretion. However, the precise relationship between MT and pituitary function remains controversial, and studies focusing on the direct role of MT in normal pituitary cells are limited to nonprimate species. Here, adult normal primate (baboons) primary pituitary cell cultures were used to determine the direct impact of MT on the functioning of all pituitary cell types from the pars distalis. MT increased GH and prolactin (PRL) expression/release in a dose- and time-dependent fashion, a response that was blocked by somatostatin. However, MT did not significantly affect ACTH, FSH, LH, or TSH expression/release. MT did not alter GHRH- or ghrelin-induced GH and/or PRL secretions, suggesting that MT may activate similar signaling pathways as ghrelin/GHRH. The effects of MT on GH/PRL release, which are likely mediated through MT1 receptor, involve both common (adenylyl cyclase/protein kinase A/extracellular calcium-channels) and distinct (phospholipase C/intracellular calcium-channels) signaling pathways. Actions of MT on pituitary cells also included regulation of the expression of other key components for the control of somatotrope/lactotrope function (GHRH, ghrelin, and somatostatin receptors). These results show, for the first time in a primate model, that MT directly regulates somatotrope/lactotrope function, thereby lending support to the notion that the actions of MT on these cells might substantially contribute to the define daily patterns of GH and PRL observed in primates and perhaps in humans.
Collapse
Affiliation(s)
- Alejandro Ibáñez-Costa
- Department of Cell Biology, Physiology, and Immunology (A.I.-C., J.C.-C., M.D.G., J.P.C., R.M.L.), University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofia; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición; and Campus de Excelencia Internacional Agroalimentario (ceiA3), E-14014 Córdoba, Spain; and Department of Medicine (J.C.-C., R.D.K.), Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago and Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | | | | | | | | | | |
Collapse
|
98
|
Sosial E, Nussinovitch I. Multiple Ca2+ channel-dependent components in growth hormone secretion from rat anterior pituitary somatotrophs. J Neuroendocrinol 2015; 27:166-76. [PMID: 25442738 DOI: 10.1111/jne.12240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022]
Abstract
The involvement of L-type Ca(2+) channels in both 'basal' and 'stimulated' growth hormone (GH) secretion is well established; however, knowledge regarding the involvement of non-L-type Ca(2+) channels is lacking. We investigated whether non-L-type Ca(2+) channels regulate GH secretion from anterior pituitary (AP) cells. To this end, GH secretion was monitored from dissociated AP cells, which were incubated for 15 min with 2 mm K(+) ('basal' secretion) or 60 mm K(+) ('stimulated' secretion). The role of non-L-type Ca(2+) influx was investigated using specific channel blockers, including ω-agatoxin-IVA, ω-conotoxin GVIA or SNX-482, to block P/Q-, N- or R-type Ca(2+) channels, respectively. Our results demonstrate that P/Q-, N- and R-type Ca(2+) channels contributed 21.2 ± 1.9%, 20.2 ± 7.6% and 11.4 ± 1.8%, respectively, to 'basal' GH secretion and 18.3 ± 1.0%, 24.4 ± 5.4% and 14.2 ± 4.8%, respectively, to 'stimulated' GH secretion. After treatment with a 'cocktail' that comprised the previously described non-L-type blockers, non-L-type Ca(2+) channels contributed 50.9 ± 0.4% and 45.5 ± 2.0% to 'basal' and 'stimulated' GH secretion, respectively. Similarly, based on the effects of nifedipine (10 μM), L-type Ca(2+) channels contributed 34.2 ± 3.7% and 54.7 ± 4.1% to 'basal' and 'stimulated' GH secretion, respectively. Interestingly, the relative contributions of L-type/non-L-type Ca(2+) channels to 'stimulated' GH secretion were well correlated with the relative contributions of L-type/non-L-type Ca(2+) channels to voltage-gated Ca(2+) influx in AP cells. Finally, we demonstrated that compartmentalisation of Ca(2+) channels is important for GH secretion. Lipid raft disruption (methyl-β-cyclodextrin, 10 mm) abrogated the compartmentalisation of Ca(2+) channels and substantially reduced 'basal' and 'stimulated' GH secretion by 43.2 ± 3.4% and 58.4 ± 4.0%, respectively. In summary, we have demonstrated that multiple Ca(2+) channel-dependent pathways regulate GH secretion. The proper function of these pathways depends on their compartmentalisation within AP cell membranes.
Collapse
Affiliation(s)
- E Sosial
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | | |
Collapse
|
99
|
Trifunović S, Manojlović-Stojanoski M, Ajdžanović V, Nestorović N, Ristić N, Medigović I, Milošević V. Effects of genistein on stereological and hormonal characteristics of the pituitary somatotrophs in rats. Endocrine 2014; 47:869-77. [PMID: 24752394 DOI: 10.1007/s12020-014-0265-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/01/2014] [Indexed: 01/08/2023]
Abstract
The hypothalamic-pituitary somatotropic system plays a pivotal role in the regulation of physiological processes and metabolism, which is modulated by gonadal steroids. Considering that genistein belongs to the phytoestrogen family and acts via similar mechanisms to estrogens, the present study was designed to demonstrate whether genistein modulates the morphofunctional characteristic of somatotrophs [growth hormone (GH) cells] in adult rats in comparison with the effects of estradiol. In the study, the orchidectomized adult rats were used as an appropriate model system for testing the effects of this hormone-like substance. Changes in the pituitary somatotrophs were evaluated histologically and stereologically, while GH level was determined biochemically. Using immunolabelling and stereological methods, we showed that orchidectomy (Orx) provoked the decrease of GH cell volume density. After estradiol treatment of Orx rats, the most prominent change concerned the pituitary relative intensity of GH fluorescence and circulating GH level, which were elevated 77 % and 4.7-fold, respectively. Clearly, in contrast to orchidectomy, estradiol treatment enhanced the GH cells activity. Genistein treatment increased pituitary weight and volume, GH cell volume density, the total number of GH cells, and GH blood concentration (1.3-fold) in comparison to the Orx group. Although identical tendencies followed estradiol and genistein administration, the changes observed after genistein treatment were milder compared to estradiol treatment.
Collapse
|
100
|
Huang G, He C, Meng F, Li J, Zhang J, Wang Y. Glucagon-like peptide (GCGL) is a novel potential TSH-releasing factor (TRF) in Chickens: I) Evidence for its potent and specific action on stimulating TSH mRNA expression and secretion in the pituitary. Endocrinology 2014; 155:4568-80. [PMID: 25076122 DOI: 10.1210/en.2014-1331] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our recent study proposed that the novel glucagon-like peptide (GCGL), encoded by a glucagon-like gene identified in chickens and other lower vertebrates, is likely a hypophysiotropic factor in nonmammalian vertebrates. To test this hypothesis, in this study, we investigated the GCGL action on chicken pituitaries. The results showed that: 1) GCGL, but not TRH, potently and specifically stimulates TSH secretion in intact pituitaries incubated in vitro or in cultured pituitary cells monitored by Western blotting or a cell-based luciferase reporter assay; 2) GCGL (0.1nM-10nM) dose dependently induces the mRNA expression of TSHβ but not 5 other hormone genes in cultured pituitary cells examined by quantitative real-time RT-PCR, an action likely mediated by intracellular adenylate cyclase/cAMP/protein kinase A and phospholipase C/inositol 1,4,5-trisphosphate/Ca(2+) signaling pathways coupled to GCGL receptor (GCGLR); 3) GCGLR mRNA is mainly localized in pituitary cephalic lobe demonstrated by in situ hybridization, where TSH-cells reside, further supporting a direct action of GCGL on thyrotrophs. The potent and specific action of GCGL on pituitary TSH expression and secretion, together with the partial accordance shown among the temporal expression profiles of GCGL in the hypothalamus and GCGLR and TSHβ in the pituitary, provides the first collective evidence that hypothalamic GCGL is most likely to be a novel TSH-releasing factor functioning in chickens. The discovery of this novel potential TSH-releasing factor (GCGL) in a nonmammalian vertebrate species, ie, chickens, would facilitate our comprehensive understanding of the hypothalamic control of pituitary-thyroid axis across vertebrates.
Collapse
Affiliation(s)
- Guian Huang
- Key Laboratory of Bioresources and Ecoenvironment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, People's Republic of China
| | | | | | | | | | | |
Collapse
|