1
|
Huang J, Liang C, Huang J, Liu L. Update on diabetic retinopathy during pregnancy. Eur J Ophthalmol 2024; 34:1695-1706. [PMID: 38710196 DOI: 10.1177/11206721241248868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Diabetes mellitus (DM) leads to several vascular and neurological complications, including diabetic retinopathy (DR). As the population ages, health problems in certain groups, including children and pregnant women, are drawing more and more attention. Pregnancy is one of the independent risk factors for the development and progression of DR. Pregnancy-induced changes may contribute to or worsen DR, which can cause a tremendous burden on public health. It is essential for pregnant women with DR and their offspring to minimize the risk of vision loss from DR in this population and adverse outcomes by understanding the development and processes behind this process. Thus, we have updated the recent situation of epidemiology, evolution characteristics, risk factors, pathophysiology, pregnancy outcomes for a better understanding of the latest status of DR, helping to improve maternal and neonatal pregnancy outcomes, and promoting health for women with DR.
Collapse
Affiliation(s)
- Jiping Huang
- Chengnan Community Health Service Center of Yangchun, Yangjiang, Guangdong Province, China
| | - Chunlan Liang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Jining Huang
- Bao'an Center Hosipital, Shenzhen, Guangdong Province, China
| | - Lian Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Donato J, Kopchick JJ. New findings on brain actions of growth hormone and potential clinical implications. Rev Endocr Metab Disord 2024; 25:541-553. [PMID: 38060062 PMCID: PMC11156798 DOI: 10.1007/s11154-023-09861-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
Growth hormone (GH) is secreted by somatotropic cells of the anterior pituitary gland. The classical effects of GH comprise the stimulation of cell proliferation, tissue and body growth, lipolysis, and insulin resistance. The GH receptor (GHR) is expressed in numerous brain regions. Notably, a growing body of evidence indicates that GH-induced GHR signaling in specific neuronal populations regulates multiple physiological functions, including energy balance, glucose homeostasis, stress response, behavior, and several neurological/cognitive aspects. The importance of central GHR signaling is particularly evident when the organism is under metabolic stress, such as pregnancy, chronic food deprivation, hypoglycemia, and prolonged exercise. These particular situations are associated with elevated GH secretion. Thus, central GH action represents an internal signal that coordinates metabolic, neurological, neuroendocrine, and behavioral adaptations that are evolutionarily advantageous to increase the chances of survival. This review summarizes and discusses recent findings indicating that the brain is an important target of GH, and GHR signaling in different neuronal populations regulates essential physiological functions.
Collapse
Affiliation(s)
- Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Lineu Prestes, 1524, Sao Paulo, SP, 05508-000, Brazil.
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
3
|
Vasconcelos S, Moustakas I, Branco MR, Guimarães S, Caniçais C, van der Helm T, Ramalho C, Marques CJ, de Sousa Lopes SMC, Dória S. Syncytiotrophoblast Markers Are Downregulated in Placentas from Idiopathic Stillbirths. Int J Mol Sci 2024; 25:5180. [PMID: 38791219 PMCID: PMC11121380 DOI: 10.3390/ijms25105180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The trophoblast cells are responsible for the transfer of nutrients between the mother and the foetus and play a major role in placental endocrine function by producing and releasing large amounts of hormones and growth factors. Syncytiotrophoblast cells (STB), formed by the fusion of mononuclear cytotrophoblasts (CTB), constitute the interface between the foetus and the mother and are essential for all of these functions. We performed transcriptome analysis of human placental samples from two control groups-live births (LB), and stillbirths (SB) with a clinically recognised cause-and from our study group, idiopathic stillbirths (iSB). We identified 1172 DEGs in iSB, when comparing with the LB group; however, when we compared iSB with the SB group, only 15 and 12 genes were down- and upregulated in iSB, respectively. An assessment of these DEGs identified 15 commonly downregulated genes in iSB. Among these, several syncytiotrophoblast markers, like genes from the PSG and CSH families, as well as ALPP, KISS1, and CRH, were significantly downregulated in placental samples from iSB. The transcriptome analysis revealed underlying differences at a molecular level involving the syncytiotrophoblast. This suggests that defects in the syncytial layer may underlie unexplained stillbirths, therefore offering insights to improve clinical obstetrics practice.
Collapse
Affiliation(s)
- Sara Vasconcelos
- Genetics Service, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal (C.J.M.)
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Ioannis Moustakas
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands (T.v.d.H.); (S.M.C.d.S.L.)
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Miguel R. Branco
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Susana Guimarães
- Department of Pathology, Faculty of Medicine and Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Carla Caniçais
- Genetics Service, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal (C.J.M.)
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Talia van der Helm
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands (T.v.d.H.); (S.M.C.d.S.L.)
| | - Carla Ramalho
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Cristina Joana Marques
- Genetics Service, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal (C.J.M.)
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Susana M. Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands (T.v.d.H.); (S.M.C.d.S.L.)
| | - Sofia Dória
- Genetics Service, Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal (C.J.M.)
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
4
|
Sleumer B, van Faassen M, Vos MJ, den Besten G, Kema IP, van de Merbel NC. Simultaneous quantification of the 22-kDa isoforms of human growth hormone 1 and 2 in human plasma by multiplexed immunocapture and LC-MS/MS. Clin Chim Acta 2024; 554:117736. [PMID: 38142804 DOI: 10.1016/j.cca.2023.117736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
An LC-MS/MS method is presented for the simultaneous quantification of two structurally closely related protein biomarker isoforms, the 22-kDa isoforms of human growth hormone 1 and human growth hormone 2, in human plasma. It is based on multiplexed immunocapture using two monoclonal antibodies immobilized on magnetic beads, tryptic digestion and quantification of two specific signature peptides plus an additional peptide for estimation of total growth hormone related concentrations. A full validation according to international guidelines was performed across the clinically relevant concentration ranges of 0.5 to 50 ng/mL for growth hormone 1, and 2 to 50 ng/mL for growth hormone 2 and demonstrated satisfactory method performance in terms of accuracy, precision, stability and absence of interference. The method's applicability for routine analysis and its ability to effectively distinguish between GH1 and GH2 was demonstrated by the analysis of plasma samples from pregnant individuals to study the changes in growth hormone levels during pregnancy.
Collapse
Affiliation(s)
- Bas Sleumer
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, the Netherlands; Department of Analytical Biochemistry University of Groningen, A. Deusinglaan 1, 9700 AV Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Michel J Vos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Gijs den Besten
- Department of Clinical Chemistry, Isala, Dr. Van Heesweg 2, 8025 AB Zwolle, the Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, the Netherlands; Department of Analytical Biochemistry University of Groningen, A. Deusinglaan 1, 9700 AV Groningen, the Netherlands.
| |
Collapse
|
5
|
Abstract
Nutrient intake is obligatory for animal growth and development, but nutrients alone are not sufficient. Indeed, insulin and homologous hormones are required for normal growth even in the presence of nutrients. These hormones communicate nutrient status between organs, allowing animals to coordinate growth and metabolism with nutrient supply. Insulin and related hormones, such as insulin-like growth factors and insulin-like peptides, play important roles in development and metabolism, with defects in insulin production and signaling leading to hyperglycemia and diabetes. Here, we describe the insulin hormone family and the signal transduction pathways activated by these hormones. We highlight the roles of insulin signaling in coordinating maternal and fetal metabolism and growth during pregnancy, and we describe how secretion of insulin is regulated at different life stages. Additionally, we discuss the roles of insulin signaling in cell growth, stem cell proliferation and cell differentiation. We provide examples of the role of insulin in development across multiple model organisms: Caenorhabditis elegans, Drosophila, zebrafish, mouse and human.
Collapse
Affiliation(s)
- Miyuki Suzawa
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Michelle L. Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
6
|
Page L, Younge N, Freemark M. Hormonal Determinants of Growth and Weight Gain in the Human Fetus and Preterm Infant. Nutrients 2023; 15:4041. [PMID: 37764824 PMCID: PMC10537367 DOI: 10.3390/nu15184041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The factors controlling linear growth and weight gain in the human fetus and newborn infant are poorly understood. We review here the changes in linear growth, weight gain, lean body mass, and fat mass during mid- and late gestation and the early postnatal period in the context of changes in the secretion and action of maternal, placental, fetal, and neonatal hormones, growth factors, and adipocytokines. We assess the effects of hormonal determinants on placental nutrient delivery and the impact of preterm delivery on hormone expression and postnatal growth and metabolic function. We then discuss the effects of various maternal disorders and nutritional and pharmacologic interventions on fetal and perinatal hormone and growth factor production, growth, and fat deposition and consider important unresolved questions in the field.
Collapse
Affiliation(s)
- Laura Page
- Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Noelle Younge
- Neonatology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Michael Freemark
- Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC 27710, USA;
- The Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
7
|
Wu G, Dong Y, Hu Q, Ma H, Xu Q, Xu K, Chen H, Yang Z, He M. HGH1 and the immune landscape: a novel prognostic marker for immune-desert tumor microenvironment identification and immunotherapy outcome prediction in human cancers. Cell Cycle 2023; 22:1969-1985. [PMID: 37811868 PMCID: PMC10761050 DOI: 10.1080/15384101.2023.2260163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/21/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
HGH1 homolog, a protein-coding gene, plays a crucial role in human growth and development. However, its role in human cancer remains unclear. For the first time, this study comprehensively evaluated the potential involvement of HGH1 in cancer prognosis and immunological function. To achieve this, data from various databases, including The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, cBioPortal, Tumor Immune Estimation Resource and Immune Cell Abundance Identifier, were collated, as well as from one large clinical study, three immunotherapy cohorts and in vitro experiments. This study aims to elucidate the role of HGH1 expression in cancer prognosis and immune response. Our findings revealed a significant association between increased HGH1 expression and a worse prognosis across various cancer types. Predominantly, copy number variations were identified as the most common genetic mutations. Additionally, HGH1 was observed to not only regulate cell cycle-related functions to promote cell proliferation but also influence autoimmunity-related functions within both the innate and adaptive immune systems, along with other relevant immune-related signaling pathways. Gene set enrichment analysis and gene set variation analysis were used to substantiate these findings. HGH1 overexpression contributed to an immune-deficient (immune-desert) tumor microenvironment, which was characterized by a significant expression of immune-related features such as immune-related gene and pathway expression and the number of immune-infiltrating cells. Furthermore, the correlation between HGH1 expression and tumor mutational burden in four cancers and microsatellite instability in eight cancers was observed. This suggests that HGH1 has potential as an immunotherapeutic target. Immunotherapy data analysis supports this notion, demonstrating that patients with low HGH1 expression treated with immune checkpoint inhibitors exhibit improved survival rates and a higher likelihood of responding to immunotherapy than patients with high HGH1 expression. Collectively, these findings highlight the significant role of HGH1 in human cancers, illuminating its involvement in tumorigenesis and cancer immunity. Elevated HGH1 expression was identified to be indicative of an immune-desert tumor microenvironment. Consequently, the targeting of HGH1, particularly in combination with immune checkpoint inhibitor therapy, holds promise for enhancing therapeutic outcomes in patients with cancer.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Yipeng Dong
- School of Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qin Hu
- Shanghai Medical College, Fudan University, Shanghai, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Qun Xu
- School of Medicine, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kun Xu
- Department of Chemotherapy, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Hongyu Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Shanghai Medical College, Fudan University, Shanghai, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
8
|
Paganoni AJJ, Cannarella R, Oleari R, Amoruso F, Antal R, Ruzza M, Olivieri C, Condorelli RA, La Vignera S, Tolaj F, Cariboni A, Calogero AE, Magni P. Insulin-like Growth Factor 1, Growth Hormone, and Anti-Müllerian Hormone Receptors Are Differentially Expressed during GnRH Neuron Development. Int J Mol Sci 2023; 24:13073. [PMID: 37685880 PMCID: PMC10487694 DOI: 10.3390/ijms241713073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are key neuroendocrine cells in the brain as they control reproduction by regulating hypothalamic-pituitary-gonadal axis function. In this context, anti-Müllerian hormone (AMH), growth hormone (GH), and insulin-like growth factor 1 (IGF1) were shown to improve GnRH neuron migration and function in vitro. Whether AMH, GH, and IGF1 signaling pathways participate in the development and function of GnRH neurons in vivo is, however, currently still unknown. To assess the role of AMH, GH, and IGF1 systems in the development of GnRH neuron, we evaluated the expression of AMH receptors (AMHR2), GH (GHR), and IGF1 (IGF1R) on sections of ex vivo mice at different development stages. The expression of AMHR2, GHR, and IGF1R was assessed by immunofluorescence using established protocols and commercial antibodies. The head sections of mice were analyzed at E12.5, E14.5, and E18.5. In particular, at E12.5, we focused on the neurogenic epithelium of the vomeronasal organ (VNO), where GnRH neurons, migratory mass cells, and the pioneering vomeronasal axon give rise. At E14.5, we focused on the VNO and nasal forebrain junction (NFJ), the two regions where GnRH neurons originate and migrate to the hypothalamus, respectively. At E18.5, the median eminence, which is the hypothalamic area where GnRH is released, was analyzed. At E12.5, double staining for the neuronal marker ß-tubulin III and AMHR2, GHR, or IGF1R revealed a signal in the neurogenic niches of the olfactory and VNO during early embryo development. Furthermore, IGF1R and GHR were expressed by VNO-emerging GnRH neurons. At E14.5, a similar expression pattern was found for the neuronal marker ß-tubulin III, while the expression of IGF1R and GHR began to decline, as also observed at E18.5. Of note, hypothalamic GnRH neurons labeled for PLXND1 tested positive for AMHR2 expression. Ex vivo experiments on mouse sections revealed differential protein expression patterns for AMHR2, GHR, and IGF1R at any time point in development between neurogenic areas and hypothalamic compartments. These findings suggest a differential functional role of related systems in the development of GnRH neurons.
Collapse
Affiliation(s)
- Alyssa J. J. Paganoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (S.L.V.); (A.E.C.)
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 10681, USA
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Federica Amoruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Renata Antal
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Marco Ruzza
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Chiara Olivieri
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (S.L.V.); (A.E.C.)
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (S.L.V.); (A.E.C.)
| | - Fationa Tolaj
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (S.L.V.); (A.E.C.)
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (A.J.J.P.); (R.O.); (F.A.); (R.A.); (M.R.); (C.O.); (F.T.); (P.M.)
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| |
Collapse
|
9
|
Tavares MR, Frazao R, Donato J. Understanding the role of growth hormone in situations of metabolic stress. J Endocrinol 2023; 256:JOE-22-0159. [PMID: 36327147 DOI: 10.1530/joe-22-0159] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Growth hormone (GH) is secreted by the anterior pituitary gland and plays a key role in controlling tissue and body growth. While basal GH secretion is considerably reduced along adulthood and aging, several situations of metabolic stress can lead to robust increases in circulating GH levels. The objective of the present review is to summarize and discuss the importance of GH regulating different physiological functions in situations of metabolic stress, including prolonged food restriction, hypoglycemia, exercise, pregnancy, and obesity. The presented data indicate that GH increases hunger perception/food intake, fat mobilization, blood glucose levels, and insulin resistance and produces changes in energy expenditure and neuroendocrine responses during metabolic challenges. When all these effects are considered in the context of situations of metabolic stress, they contribute to restore homeostasis by (1) helping the organism to use appropriate energy substrates, (2) preventing hypoglycemia or increasing the availability of glucose, (3) stimulating feeding to provide nutrients in response to energy-demanding activities or to accelerate the recovery of energy stores, and (4) affecting the activity of neuronal populations involved in the control of metabolism and stress response. Thus, the central and peripheral effects of GH coordinate multiple adaptations during situations of metabolic stress that ultimately help the organism restore homeostasis, increasing the chances of survival.
Collapse
Affiliation(s)
- Mariana Rosolen Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
10
|
Selective quantification of the 22-kDa isoform of human growth hormone 1 in serum and plasma by immunocapture and LC-MS/MS. Anal Bioanal Chem 2022; 414:6187-6200. [PMID: 35838770 PMCID: PMC9314277 DOI: 10.1007/s00216-022-04188-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/16/2022] [Accepted: 06/20/2022] [Indexed: 11/01/2022]
Abstract
The human growth hormone GH1 (22 kDa) is a commonly measured biomarker for diagnosis and during treatment of growth disorders, but its quantification by ligand binding assays may be compromised by the occurrence of a number of isoforms. These can interfere in the assays and lead to differences in results between laboratories and potentially even in the treatment of patients. We present an LC-MS/MS method that is able to distinguish the major growth hormone isoform (GH1, 22 kDa) from other isoforms and quantify it without any interference across the clinically relevant concentration range of 0.5 to 50 ng/mL. Analysis involves purification of a 100-µL serum sample by immunocapture using an anti-GH-directed antibody, tryptic digestion, and LC-MS/MS quantification of an isoform-specific signature peptide for GH1 (22 kDa). A tryptic peptide occurring in all GH isoforms is monitored in the same 16-min analytical run as a read-out for total GH. Stable-isotope-labeled forms of these two peptides are included as internal standards. Full validation of the method according to recent guidelines, against a recombinant form of the analyte in rat plasma calibrators, demonstrated intra-assay and inter-assay imprecision below 6% across the calibration range for both signature peptides and recoveries between 94 and 102%. An excellent correlation was found between nominal and measured concentrations of the WHO reference standard for GH1 (22 kDa). Addition of up to 1000 ng/mL biotin or the presence of a 100-fold excess of GH binding protein did not affect the measurement. Equivalent method performance was found for analysis of GH in serum, EDTA, and heparin plasma. Analyte stability was demonstrated during all normal sample storage conditions. Comparison with the IDS-iSYS GH immunoassay showed a good correlation with the LC-MS/MS method for the isoform-specific signature peptide, but a significant positive bias was observed for the LC-MS/MS results of the peptide representing total GH. This seems to confirm the actual occurrence of other GH isoforms in serum. Finally, in serum from pregnant individuals, no quantifiable GH1 (22 kDa) was found, but relatively high concentrations of total GH.
Collapse
|
11
|
Paragliola RM, Carrozza C, Corsello SM, Salvatori R. The biochemical diagnosis of acromegaly: revising the role of measurement of IGF-I and GH after glucose load in 5 questions. Expert Rev Endocrinol Metab 2022; 17:205-224. [PMID: 35485763 DOI: 10.1080/17446651.2022.2069558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Acromegaly is a rare disorder characterized by the excessive secretion of growth hormone (GH), mostly caused by pituitary adenomas. While in full-blown cases the diagnosis is easy to establish, milder cases are more challenging. Additionally, establishing whether full cure after surgery is reached may be difficult. AREAS COVERED In this article, we will review the challenges posed by the variability in measurements of GH and its main effector insulin-like growth factor I (IGF-I) due to both biological changes, co-morbidities, and assays variability. EXPERT OPINION Interpretation of GH and IGF-I assays is important in establishing an early diagnosis of acromegaly, in avoiding misdiagnosis, and in establishing if cure is achieved by surgery. Physicians should be familiar with the variables that affect measurements of these 2 hormones, and with the performance of the assays available in their practice.
Collapse
Affiliation(s)
- Rosa Maria Paragliola
- Unit of Endocrinology, Department of Translational Medicine and Surgery - Universita' Cattolica del Sacro Cuore, Fondazione Policlinico "Gemelli", IRCCS, Rome, Italy
| | - Cinzia Carrozza
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology - Università Cattolica Del Sacro Cuore, Fondazione Policlinico "Gemelli," IRCCS, Rome, Italy
| | - Salvatore M Corsello
- Unit of Endocrinology, Department of Translational Medicine and Surgery - Universita' Cattolica del Sacro Cuore, Fondazione Policlinico "Gemelli", IRCCS, Rome, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Roberto Salvatori
- Division of Endocrinology Diabetes and Metabolism and Pituitary Center, Johns Hopkins University, Baltimore MD, USA
| |
Collapse
|
12
|
|
13
|
Gantenbein KV, Kanaka-Gantenbein C. Highlighting the trajectory from intrauterine growth restriction to future obesity. Front Endocrinol (Lausanne) 2022; 13:1041718. [PMID: 36440208 PMCID: PMC9691665 DOI: 10.3389/fendo.2022.1041718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
During the last decades several lines of evidence reported the association of an adverse intrauterine environment, leading to intrauterine restriction, with future disease, such as obesity and metabolic syndrome, both leading to increased cardiovascular and cancer risk. The underlying explanation for this association has firstly been expressed by the Barker's hypothesis, the "thrifty phenotype hypothesis". According to this hypothesis, a fetus facing an adverse intrauterine environment adapts to this environment through a reprogramming of its endocrine-metabolic status, during the crucial window of developmental plasticity to save energy for survival, providing less energy and nutrients to the organs that are not essential for survival. This theory evolved to the concept of the developmental origin of health and disease (DOHaD). Thus, in the setting of an adverse, f. ex. protein restricted intrauterine environment, while the energy is mainly directed to the brain, the peripheral organs, f.ex. the muscles and the liver undergo an adaptation that is expressed through insulin resistance. The adaptation at the hepatic level predisposes to future dyslipidemia, the modifications at the vascular level to endothelial damage and future hypertension and, overall, through the insulin resistance to the development of metabolic syndrome. All these adaptations are suggested to take place through epigenetic modifications of the expression of genes without change of their amino-acid sequence. The epigenetic modifications leading to future obesity and cardiovascular risk are thought to induce appetite dysregulation, promoting food intake and adipogenesis, facilitating obesity development. The epigenetic modifications may even persist into the next generation even though the subsequent generation has not been exposed to an adverse intrauterine environment, a notion defined as the "transgenerational transfer of environmental information". As a consequence, if the increased public health burden and costs of non-communicable chronic diseases such as obesity, hypertension, metabolic syndrome and type 2 diabetes have to be minimized, special attention should be laid to the healthy lifestyle habits of women of reproductive age, including healthy diet and physical activity to be established long before any pregnancy takes place in order to provide the best conditions for both somatic and mental health of future generations.
Collapse
Affiliation(s)
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital, Athens, Greece
- *Correspondence: Christina Kanaka-Gantenbein, ,
| |
Collapse
|
14
|
Metabolic-endocrine disruption due to preterm birth impacts growth, body composition, and neonatal outcome. Pediatr Res 2022; 91:1350-1360. [PMID: 34040160 PMCID: PMC9197767 DOI: 10.1038/s41390-021-01566-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Despite optimized nutrition, preterm-born infants grow slowly and tend to over-accrete body fat. We hypothesize that the premature dissociation of the maternal-placental-fetal unit disrupts the maintenance of physiological endocrine function in the fetus, which has severe consequences for postnatal development. This review highlights the endocrine interactions of the maternal-placental-fetal unit and the early perinatal period in both preterm and term infants. We report on hormonal levels (including tissue, thyroid, adrenal, pancreatic, pituitary, and placental hormones) and nutritional supply and their impact on infant body composition. The data suggest that the premature dissociation of the maternal-placental-fetal unit leads to a clinical picture similar to panhypopituitarism. Further, we describe how the premature withdrawal of the maternal-placental unit, neonatal morbidities, and perinatal stress can cause differences in the levels of growth-promoting hormones, particularly insulin-like growth factors (IGF). In combination with the endocrine disruption that occurs following dissociation of the maternal-placental-fetal unit, the premature adaptation to the extrauterine environment leads to early and fast accretion of fat mass in an immature body. In addition, we report on interventional studies that have aimed to compensate for hormonal deficiencies in infants born preterm through IGF therapy, resulting in improved neonatal morbidity and growth. IMPACT: Preterm birth prematurely dissociates the maternal-placental-fetal unit and disrupts the metabolic-endocrine maintenance of the immature fetus with serious consequences for growth, body composition, and neonatal outcomes. The preterm metabolic-endocrine disruption induces symptoms resembling anterior pituitary failure (panhypopituitarism) with low levels of IGF-1, excessive postnatal fat mass accretion, poor longitudinal growth, and failure to thrive. Appropriate gestational age-adapted nutrition alone seems insufficient for the achievement of optimal growth of preterm infants. Preliminary results from interventional studies show promising effects of early IGF-1 supplementation on postnatal development and neonatal outcomes.
Collapse
|
15
|
Wasinski F, Teixeira PDS, List EO, Kopchick JJ, Donato J. Growth hormone receptor contributes to the activation of STAT5 in the hypothalamus of pregnant mice. Neurosci Lett 2021; 770:136402. [PMID: 34929316 DOI: 10.1016/j.neulet.2021.136402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 11/19/2022]
Abstract
Growth hormone (GH) receptor (GHR) signaling induces the phosphorylation of the signal transducer and activator of transcription 5 (pSTAT5) in the cells of several tissues including in the hypothalamus. During pregnancy, several STAT5-recruiting hormones (e.g., prolactin, GH and placental lactogens) are highly secreted. However, the precise contribution of GHR signaling to the surge of pSTAT5 immunoreactive neurons that occurs in the hypothalamus of pregnant mice is currently unknown. Thus, the objective of the present study was to determine whether GHR expression in neurons is required for inducing pSTAT5 expression in several hypothalamic nuclei during pregnancy. Initially, we demonstrated that late pregnant C57BL/6 mice (gestational day 14 to 18) exhibited increased pulsatile GH secretion compared to virgin females. Next, we confirmed that neuron-specific GHR ablation robustly reduces hypothalamic Ghr mRNA levels and prevents GH-induced pSTAT5 in the arcuate, paraventricular and ventromedial hypothalamic nuclei. Subsequently, the number of pSTAT5 immunoreactive cells was determined in the hypothalamus of late pregnant mice. Although neuron-specific GHR ablation did not affect the number of pSTAT5 immunoreactive cells in the paraventricular nucleus of the hypothalamus, reduced pSTAT5 expression was observed in the arcuate and ventromedial nuclei of pregnant neuron-specific GHR knockouts, compared to control pregnant mice. In summary, a subset of hypothalamic neurons requires GHR signaling to express pSTAT5 during pregnancy. These findings contribute to the understanding of the endocrine factors that affect the activation of transcription factors in the brain during pregnancy.
Collapse
Affiliation(s)
- Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo 05508000, Brazil
| | - Pryscila D S Teixeira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo 05508000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, Sao Paulo 05508000, Brazil.
| |
Collapse
|
16
|
Nomura Y, Rompala G, Pritchett L, Aushev V, Chen J, Hurd YL. Natural disaster stress during pregnancy is linked to reprogramming of the placenta transcriptome in relation to anxiety and stress hormones in young offspring. Mol Psychiatry 2021; 26:6520-6530. [PMID: 33981007 PMCID: PMC8586067 DOI: 10.1038/s41380-021-01123-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/29/2022]
Abstract
Prenatal stress can lead to long-term adverse effects that increase the risk of anxiety and other emotional disorders in offspring. The in utero underpinnings contributing to such phenotypes remain unknown. We profiled the transcriptome of placental specimens from women who lived through Hurricane Sandy during pregnancy compared to those pregnant during non-Sandy conditions. Following birth, longitudinal assessments were conducted in their offspring during childhood (~3-4 years old) to measure steroid hormones (in hair) and behavioral and emotional problems. This revealed a significant link between prenatal Sandy stress (PNSS) and child HPA dysfunction, evident by altered cortisol, dehydroepiandrosterone (DHEA), and cortisol:DHEA levels. In addition, PNSS was associated with significantly increased anxiety and aggression. These findings coincided with significant reorganization of the placental transcriptome via vascular, immune, and endocrine gene pathways. Interestingly, many of the most prominently altered genes were known to be uniquely expressed in syncytiotrophoblast (STB)-subtype of placental cells and harbored glucocorticoid response elements in promoter regions. Finally, several vascular development- and immune-related placental gene sets were found to mediate the relationship between PNSS and childhood phenotypes. Overall, these findings suggest that natural disaster-related stress during pregnancy reprograms the placental molecular signature, potentially driving long-lasting changes in stress regulation and emotional health. Further examination of placental mechanisms may elucidate the environment's contribution to subsequent risk for anxiety disorders later in life.
Collapse
Affiliation(s)
- Yoko Nomura
- Queens College, CUNY, Psychology, New York, NY, United States,CUNY, The Graduate Center, Psychology, Graduate School of Public Health, New York, NY, United States,Icahn School of Medicine at Mount Sinai, Psychiatry and Neuroscience, Addiction Institute of Mount Sinai, New York, NY, United States
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, Psychiatry and Neuroscience, Addiction Institute of Mount Sinai, New York, NY, United States
| | - Lexi Pritchett
- Queens College, CUNY, Psychology, New York, NY, United States,CUNY, The Graduate Center, Psychology, Graduate School of Public Health, New York, NY, United States
| | - Vasily Aushev
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai
| | - Yasmin L. Hurd
- Icahn School of Medicine at Mount Sinai, Psychiatry and Neuroscience, Addiction Institute of Mount Sinai, New York, NY, United States
| |
Collapse
|
17
|
Wallis M. Do some viruses use growth hormone, prolactin and their receptors to facilitate entry into cells?: Episodic evolution of hormones and receptors suggests host-virus arms races; related placental lactogens may provide protective viral decoys. Bioessays 2021; 43:e2000268. [PMID: 33521987 DOI: 10.1002/bies.202000268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/30/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022]
Abstract
The molecular evolution of pituitary growth hormone and prolactin in mammals shows two unusual features: episodes of markedly accelerated evolution and, in some species, complex families of related proteins expressed in placenta and resulting from multiple gene duplications. Explanations of these phenomena in terms of physiological adaptations seem unconvincing. Here, I propose an alternative explanation, namely that these evolutionary features reflect the use of the hormones (and their receptors) as viral receptors. Episodes of rapid evolution can then be explained as due to "arms races" in which changes in the hormone lead to reduced interaction with the virus, and subsequent changes in the virus counteract this. Placental paralogues of the hormones could provide decoys that bind viruses, and protect the foetus against infection. The hypothesis implies that the extensive changes introduced into growth hormone, prolactin and their receptors during the course of mammalian evolution reflect viral interactions, not endocrine adaptations.
Collapse
Affiliation(s)
- Michael Wallis
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
18
|
Jensen RB, Boas M, Nielsen JE, Maroun LL, Jørgensen A, Larsen T, Main KM, Juul A. A common deletion in the growth hormone receptor gene (d3-GHR) in the offspring is related to maternal placental GH levels during pregnancy. Growth Horm IGF Res 2020; 55:101360. [PMID: 33096343 DOI: 10.1016/j.ghir.2020.101360] [Citation(s) in RCA: 1] [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: 03/15/2020] [Revised: 09/02/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND A common growth hormone receptor polymorphism with deletion of exon 3 (d3-GHR) has previously been linked to increased postnatal growth on the one hand and decreased fetal growth on the other. Regulation of fetal growth is positively dependent on secretion of placental GH (hGH-V). OBJECTIVE We explored the effect of the fetal d3-GHR genotype on maternal serum levels of hGH-V and fetal growth. The cellular localization of hGH-V synthesis and the GH receptors were determined in normal placentas. METHODS 43 healthy mother-child pairs were examined during pregnancy with measurements of hGH-V during third trimester, and serial ultrasound measurements determined fetal growth rate. Birth anthropometrics were obtained. The GHR genotype of the child was analysed postnatally. Immunohistochemical (IHC) analysis was conducted on four placentas. RESULTS The presence of the d3-GHR genotype was associated with a markedly reduced concentration of hGH-V in maternal serum (β -0.52, SE 0.24, p = 0.04) compared to those who had a fl/fl genotype. Accordingly, a tendency towards reduced fetal growth rate during third trimester (β -25.8, SE 12.7, p = 0.05) and a lower birth weight were found among carriers of the d3-GHR allele, but these associations did not reach statistical significance (p = 0.08). IHC analysis showed expression of placental GH and GHR in the villous syncytiotrophoblast, the extravillous trophoblast, and the decidual cells and smooth muscle cells in chorionic vessels. CONCLUSIONS The presence of the d3-GHR polymorphism in the fetus was associated with lower maternal serum levels of hGH-V, decreased fetal growth rate in third trimester and lower birth weight compared to the wildtype.
Collapse
Affiliation(s)
- Rikke Beck Jensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark.
| | - Malene Boas
- Department of Pediatrics, Herlev Hospital, University of Copenhagen, Denmark
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
| | - Lisa Leth Maroun
- Department of Pathology, Rigshospitalet, University of Copenhagen, Denmark
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
| | - Torben Larsen
- Department of Obstetrics and Gynecology, Holbæk Hospital, Holbæk, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Denmark
| |
Collapse
|
19
|
Vickers MH, Perry JK. The 20-kDa Placental GH Variant: A New and Improved Growth Hormone? Endocrinology 2020; 161:5902562. [PMID: 32894774 DOI: 10.1210/endocr/bqaa147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Mark H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Teixeira PDS, Couto GC, Furigo IC, List EO, Kopchick JJ, Donato J. Central growth hormone action regulates metabolism during pregnancy. Am J Physiol Endocrinol Metab 2019; 317:E925-E940. [PMID: 31479305 PMCID: PMC7132326 DOI: 10.1152/ajpendo.00229.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The maternal organism undergoes numerous metabolic adaptations to become prepared for the demands associated with the coming offspring. These metabolic adaptations involve changes induced by several hormones that act at multiple levels, ultimately influencing energy and glucose homeostasis during pregnancy and lactation. Previous studies have shown that central growth hormone (GH) action modulates glucose and energy homeostasis. However, whether central GH action regulates metabolism during pregnancy and lactation is still unknown. In the present study, we generated mice carrying ablation of GH receptor (GHR) in agouti-related protein (AgRP)-expressing neurons, in leptin receptor (LepR)-expressing cells or in the entire brain to investigate the role played by central GH action during pregnancy and lactation. AgRP-specific GHR ablation led to minor metabolic changes during pregnancy and lactation. However, while brain-specific GHR ablation reduced food intake and body adiposity during gestation, LepR GHR knockout (KO) mice exhibited increased leptin responsiveness in the ventromedial nucleus of the hypothalamus during late pregnancy, although their offspring showed reduced growth rate. Additionally, both Brain GHR KO and LepR GHR KO mice had lower glucose tolerance and glucose-stimulated insulin secretion during pregnancy, despite presenting increased insulin sensitivity, compared with control pregnant animals. Our findings revealed that during pregnancy central GH action regulates food intake, fat retention, as well as the sensitivity to insulin and leptin in a cell-specific manner. Together, the results suggest that GH acts in concert with other "gestational hormones" to prepare the maternal organism for the metabolic demands of the offspring.
Collapse
Affiliation(s)
- Pryscila D S Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gisele C Couto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Chanson P, Vialon M, Caron P. An update on clinical care for pregnant women with acromegaly. Expert Rev Endocrinol Metab 2019; 14:85-96. [PMID: 30696300 DOI: 10.1080/17446651.2019.1571909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/16/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION As pregnancy is rare in women with acromegaly, only case reports and few series have been published. AREAS COVERED All case reports and publications dealing with pregnancy in patients with acromegaly were collated. Information concerning the effects of acromegaly on pregnancy outcomes, the impact of pregnancy on GH/IGF-I measurements, acromegaly comorbidity and pituitary adenoma size, the effects of treatment of acromegaly on fetus outcomes were retrieved and analyzed. EXPERT COMMENTARY Based on the small number of reported cases, pregnancy is generally uneventful, except for a potential increased incidence of gestational hypertension and diabetes mellitus. Medical therapy of acromegaly (dopamine agonists, somatostatin analogs, growth hormone-receptor antagonists) is generally interrupted before or at diagnosis of pregnancy. In very rare patients with a pituitary adenoma, particularly a macroadenoma that has not been surgically treated before pregnancy, or if a surgical remnant persists, or when acromegaly is revealed during pregnancy, tumor volume may increase and cause symptoms through a mass effect. Close monitoring of clinical manifestations and imaging are necessary during pregnancy in these cases. In the rare cases of symptomatic tumor enlargement during pregnancy, medical treatment with dopamine agonists or eventually somatostatin analogs may be attempted before resorting to transsphenoidal surgery.
Collapse
Affiliation(s)
- Philippe Chanson
- a Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Mladies Rares de l'Hypophyse , Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre , Le Kremlin Bicêtre , France
- b Unité Mixte de Recherche S1185 Facultéde Médecine Paris-Sud , University Paris-Sud , Le Kremlin Bicêtre , France
- c Unit 1185, Institut National de la Santé et de laRecherche Médicale (INSERM) , Le Kremlin Bicêtre , France
| | - Magaly Vialon
- d Service d'Endocrinologie et des Maladies Métaboliques , Centre Hospitalier Universitaire de Toulouse, Hôpital Larrey , Toulouse , France
| | - Philippe Caron
- d Service d'Endocrinologie et des Maladies Métaboliques , Centre Hospitalier Universitaire de Toulouse, Hôpital Larrey , Toulouse , France
| |
Collapse
|