51
|
Tanaka K, Besson V, Rivagorda M, Oury F, Marazzi G, Sassoon DA. Paternally expressed gene 3 (Pw1/Peg3) promotes sexual dimorphism in metabolism and behavior. PLoS Genet 2022; 18:e1010003. [PMID: 35025875 PMCID: PMC8791484 DOI: 10.1371/journal.pgen.1010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 01/26/2022] [Accepted: 12/20/2021] [Indexed: 01/06/2023] Open
Abstract
The paternally expressed gene 3 (Pw1/Peg3) is a mammalian-specific parentally imprinted gene expressed in stem/progenitor cells of the brain and endocrine tissues. Here, we compared phenotypic characteristics in Pw1/Peg3 deficient male and female mice. Our findings indicate that Pw1/Peg3 is a key player for the determination of sexual dimorphism in metabolism and behavior. Mice carrying a paternally inherited Pw1/Peg3 mutant allele manifested postnatal deficits in GH/IGF dependent growth before weaning, sex steroid dependent masculinization during puberty, and insulin dependent fat accumulation in adulthood. As a result, Pw1/Peg3 deficient mice develop a sex-dependent global shift of body metabolism towards accelerated adiposity, diabetic-like insulin resistance, and fatty liver. Furthermore, Pw1/Peg3 deficient males displayed reduced social dominance and competitiveness concomitant with alterations in the vasopressinergic architecture in the brain. This study demonstrates that Pw1/Peg3 provides an epigenetic context that promotes male-specific characteristics through sex steroid pathways during postnatal development. Pw1/Peg3 is under parental specific epigenetic regulation. We propose that Pw1/Peg3 confers a selective advantage in mammals by regulating sexual dimorphism. To address this question, we examined the consequences of Pw1/Peg3 loss of function in mice in an age- and sex-dependent context and found that Pw1/Peg3 mutants display reduced sexual dimorphism in growth, metabolism and behaviors. Our findings support the intralocus sexual conflict model of genomic imprinting where it contributes in sexual differentiation. Furthermore, our observations provide a unifying role of sex steroid signaling as a common property of Pw1/Peg3 expressing stem/progenitor cells and differentiated endocrine cells, both of which remain proliferative in response to gonadal hormones in adult life.
Collapse
Affiliation(s)
- Karo Tanaka
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Vanessa Besson
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Manon Rivagorda
- Hormonal Regulation of Brain Development and Functions, INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Franck Oury
- Hormonal Regulation of Brain Development and Functions, INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David A. Sassoon
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
- * E-mail:
| |
Collapse
|
52
|
El-Dairi R, Rysä J, Storvik M, Pasanen M, Huuskonen P. Aflatoxin B1 targeted gene expression profiles in human placental primary trophoblast cells. Curr Res Toxicol 2022; 3:100082. [PMID: 35814288 PMCID: PMC9263407 DOI: 10.1016/j.crtox.2022.100082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/25/2022] Open
Abstract
Gene expression profiles were studied in human primary trophoblast cells. 170 genes were significantly dysregulated in aflatoxin B1-exposed trophoblasts. AhR-mediated estrogen receptor signalling was dysregulated in response to AFB1. Transcripts involved in endocrine signalling and energy homeostasis were disrupted. Cellular growth and development, cell cycle and DNA repair processes were affected.
Aflatoxin B1 (AFB1) is a mycotoxin produced by Aspergillus flavus and A. parasiticus. A high exposure (40 nM and 1 µM AFB1 for 72 h) was used to study mechanistic effects of AFB1 on gene expression patterns in human primary trophoblast cells, isolated from full term placentae after delivery. Gene expression profiling was conducted, and Ingenuity pathway analysis (IPA) software was used to identify AFB1-regulated gene networks and regulatory pathways. In response to 40 nM AFB1, only 7 genes were differentially expressed whereas 1 µM AFB1 significantly dysregulated 170 genes (124 down- and 46 upregulated, ±1.5-fold, p < 0.05) in AFB1-exposed trophoblasts when compared to controls. The top downregulated genes were involved in endocrine signalling and biosynthesis of hormones, and lipid and carbohydrate metabolism. The top upregulated genes were involved in protein synthesis and regulation of cell cycle. The main canonical pathways identified by IPA were associated with endocrine signalling including growth hormone signalling, and corticotropin releasing hormone signalling. Furthermore, genes involved in aryl hydrocarbon receptor (AhR)-mediated estrogen receptor signalling were dysregulated in response to AFB1. Our findings indicate that a high concentration 72 h AFB1 exposure caused relatively moderate number of changes on transcript level to human placental primary trophoblast cells. However, these preliminary results need to be confirmed with human-relevant concentrations of AFB1.
Collapse
|
53
|
van der Velden LM, Maas P, van Amersfoort M, Timmermans-Sprang EPM, Mensinga A, van der Vaart E, Malergue F, Viëtor H, Derksen PWB, Klumperman J, van Agthoven A, Egan DA, Mol JA, Strous GJ. Small molecules to regulate the GH/IGF1 axis by inhibiting the growth hormone receptor synthesis. Front Endocrinol (Lausanne) 2022; 13:926210. [PMID: 35966052 PMCID: PMC9365994 DOI: 10.3389/fendo.2022.926210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Growth hormone (GH) and insulin-like growth factor-1 (IGF1) play an important role in mammalian development, cell proliferation and lifespan. Especially in cases of tumor growth there is an urgent need to control the GH/IGF1 axis. In this study we screened a 38,480-compound library, and in two consecutive rounds of analogues selection, we identified active lead compounds based on the following criteria: inhibition the GH receptor (GHR) activity and its downstream effectors Jak2 and STAT5, and inhibition of growth of breast and colon cancer cells. The most active small molecule (BM001) inhibited both the GH/IGF1 axis and cell proliferation with an IC50 of 10-30 nM of human cancer cells. BM001 depleted GHR in human lymphoblasts. In preclinical xenografted experiments, BM001 showed a strong decrease in tumor volume in mice transplanted with MDA-MB-231 breast cancer cells. Mechanistically, the drug acts on the synthesis of the GHR. Our findings open the possibility to inhibit the GH/IGF1 axis with a small molecule.
Collapse
Affiliation(s)
- Lieke M. van der Velden
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Peter Maas
- Specs Compound Handling, Zoetermeer, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| | | | | | - Anneloes Mensinga
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Elisabeth van der Vaart
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Fabrice Malergue
- Department of Research and Development, Beckman Coulter Life Science, Immunotech Marseille, Marseille, France
| | - Henk Viëtor
- Drug Discovery Factory (DDF) Ventures, Breukelen, Netherlands
| | - Patrick W B. Derksen
- Department of Pathology, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Judith Klumperman
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
| | - Andreas van Agthoven
- Department of Research and Development, Beckman Coulter Life Science, Immunotech Marseille, Marseille, France
| | - David A. Egan
- Cell Screening Core, Department of Cell Biology, Center for Molecular Medicine, University Medical Center, Utrecht, Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| | - Ger J. Strous
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Center (UMC) Utrecht, Utrecht, Netherlands
- *Correspondence: Ger J. Strous, ; Jan A. Mol, ; Peter Maas,
| |
Collapse
|
54
|
Gurevich E, Segev Y, Landau D. Growth Hormone and IGF1 Actions in Kidney Development and Function. Cells 2021; 10:cells10123371. [PMID: 34943879 PMCID: PMC8699155 DOI: 10.3390/cells10123371] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 01/17/2023] Open
Abstract
Growth hormone (GH) exerts multiple effects on different organs including the kidneys, either directly or via its main mediator, insulin-like-growth factor-1 (IGF-1). The GH/IGF1 system plays a key role in normal kidney development, glomerular hemodynamic regulation, as well as tubular water, sodium, phosphate, and calcium handling. Transgenic animal models demonstrated that GH excess (and not IGF1) may lead to hyperfiltration, albuminuria, and glomerulosclerosis. GH and IGF-1 play a significant role in the early development of diabetic nephropathy, as well as in compensatory kidney hypertrophy after unilateral nephrectomy. Chronic kidney disease (CKD) and its complications in children are associated with alterations in the GH/IGF1 axis, including growth retardation, related to a GH-resistant state, attributed to impaired kidney postreceptor GH-signaling and chronic inflammation. This may explain the safety of prolonged rhGH-treatment of short stature in CKD.
Collapse
Affiliation(s)
- Evgenia Gurevich
- Department of Nephrology, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, Petach Tikva 4920235, Israel;
| | - Yael Segev
- Shraga Segal Department of Microbiology and Immunology, Ben Gurion University, Beer Sheva 8410501, Israel;
| | - Daniel Landau
- Department of Nephrology, Schneider Children’s Medical Center of Israel, 14 Kaplan Street, Petach Tikva 4920235, Israel;
- Sackler School of Medicine, Tel Aviv University, P.O. Box 39040, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-3925-3651
| |
Collapse
|
55
|
Cao C, Jia Z, Shao M, Li R, Sun Q, Liu D. Prenatal exposure to polycyclic aromatic hydrocarbons could increase the risk of low birth weight by affecting the DNA methylation states in a Chinese cohort. Reprod Biol 2021; 21:100574. [PMID: 34794034 DOI: 10.1016/j.repbio.2021.100574] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 11/18/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), as a kind of endocrine disruptors, can enter the fetus body cross the placental barrier from prenatal PAHs exposure to cause adverse birth outcomes. However, it is controversial association between prenatal PAHs exposure and low birth weight (LBW) of their infants. So the present study aimed to estimate the effects of prenatal PAHs exposure during the pregnancy on the risk of LBW in a Chinese cohort through modifying the DNA methylation states. A longitudinal prospective study with 407 pregnant women was established from May to October 2019. The prenatal PAHs exposure during the pregnancy was assessed using the internal dose such as the PAHs metabolites and PAH-DNA adducts in the umbilical cord blood. The methylation levels of genomic DNA and growth-related genes (IGF1 and IGF2) were assessed, while the expressions of these genes were both determined by RT-PCR and Elisa methods. The growth outcomes and relevant Z-scores were recorded at birth. The correlations between the DNA methylation status and concentrations of PAHs, expression levels of growth-related genes and body weight/WAZ were investigated as the measures. According to the PAH-DNA adducts, the subjects were divided into two groups: PAHs-exposed group (PAH-DNA adducts>0, n = 55) and non-exposed group (PAH-DNA adducts = 0, n = 352). Compared with the non-exposed group, it displayed marked decreased birth weight, and increased concentrations of PAHs and DNA methylation levels of the global genomic, IGF1 and IGF2 with their lower expressions in the PAHs-exposed group. These hypermethylation (global genomic, CpG14 and CpG15 of IGF1, and CpG14 of IGF2) were positively correlated with the contents of PAHs in the umbilical cord blood, and negatively correlated with the growth outcomes and their expressions. Totally, prenatal PAHs exposures may contribute to an increased risk of LBW of their infants by modulating the DNA methylation states of genomic DNA and growth-related genes (IGF1 and IGF2) in the umbilical cord blood, which could provide the prenatal prevention of PAHs exposure from possible environmental media except from the occupation and tobacco usage to ensure the health of their infants.
Collapse
Affiliation(s)
- Chunxia Cao
- Department of Pediatrics, Zibo Central Hospital, Shandong Province, 255000, China
| | - Zhiyi Jia
- Department of Pediatrics, Zibo Central Hospital, Shandong Province, 255000, China
| | - Mingyu Shao
- Department of Pediatrics, Zibo Central Hospital, Shandong Province, 255000, China
| | - Rongmiao Li
- Department of Thoracic Surgery, Huantai Country People's Hospital, Shandong Province, 255000, China
| | - Qi Sun
- Scientific Education and Communication Cooperation Office, Zibo Central Hospital, Shandong Province, 255000, China
| | - Dong Liu
- Department of Pediatrics, Zibo Central Hospital, Shandong Province, 255000, China.
| |
Collapse
|
56
|
Wang Y, Shimizu H, Xiang YY, Sugihara J, Lu WY, Liao XH, Cho HR, Toba H, Bai XH, Asa SL, Arvan P, Refetoff S, Liu M. XB130 Deficiency Causes Congenital Hypothyroidism in Mice due to Disorganized Apical Membrane Structure and Function of Thyrocytes. Thyroid 2021; 31:1650-1661. [PMID: 34470464 PMCID: PMC8917886 DOI: 10.1089/thy.2021.0195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background: Congenital hypothyroidism is often caused by genetic mutations that impair thyroid hormone (TH) production, resulting in growth and development defects. XB130 (actin filament associated protein 1 like 2) is an adaptor/scaffold protein that plays important roles in cell proliferation, migration, intracellular signal transduction, and tumorigenesis. It is highly expressed in thyrocytes, however, its function in the thyroid remains largely unexplored. Methods:Xb130-/- mice and their littermates were studied. Postnatal growth and growth hormone levels were measured, and responses to low or high-iodine diet, and levothyroxine treatment were examined. TH and thyrotropin in the serum and TH in the thyroid glands were quantified. Structure and function of thyrocytes in embryos and postnatal life were studied with histology, immunohistochemistry, immunofluorescence staining, Western blotting, and quantitative reverse transcription polymerase chain reaction. Results:Xb130-/- mice exhibited transient growth retardation postnatally, due to congenital hypothyroidism with reduced TH synthesis and secretion, which could be rescued by exogenous thyroxine supplementation. The thyroid glands of Xb130-/- mice displayed diminished thyroglobulin iodination and release at both embryonic and early postnatal stages. XB130 was found mainly on the apical membrane of thyroid follicles. Thyroid glands of embryonic and postnatal Xb130-/- mice exhibited disorganized apical membrane structure, delayed folliculogenesis, and abnormal formation of thyroid follicle lumina. Conclusion: XB130 critically regulates folliculogenesis by maintaining apical membrane structure and function of thyrocytes, and its deficiency leads to congenital hypothyroidism.
Collapse
Affiliation(s)
- Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Hiroki Shimizu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Yun-Yan Xiang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada
| | - Junichi Sugihara
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Wei-Yang Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Canada
| | | | - Hae-Ra Cho
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Hiroaki Toba
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Xiao-Hui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Sylvia L. Asa
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
- University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peter Arvan
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Samuel Refetoff
- Department of Medicine and Chicago, Illinois, USA
- Department of Pediatrics; and Chicago, Illinois, USA
- Department of Committee on Genetics; The University of Chicago, Chicago, Illinois, USA
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, and University of Toronto, Toronto, Ontario, Canada
- Department of Medicine; and University of Toronto, Toronto, Ontario, Canada
- Department of Institute of Medical Science; Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Address correspondence to: Mingyao Liu, MD, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Room: PMCRT2-814, Toronto M5G 1L7, Canada
| |
Collapse
|
57
|
Ramírez-Contreras CY, Mehran AE, Salehzadeh M, Mussai EX, Miller JW, Smith A, Ranger M, Holsti L, Soma KK, Devlin AM. Sex-specific effects of neonatal oral sucrose treatment on growth and liver choline and glucocorticoid metabolism in adulthood. Am J Physiol Regul Integr Comp Physiol 2021; 321:R802-R811. [PMID: 34612088 PMCID: PMC11961110 DOI: 10.1152/ajpregu.00091.2021] [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: 03/29/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 11/22/2022]
Abstract
Hospitalized preterm infants experience painful medical procedures. Oral sucrose is the nonpharmacological standard of care for minor procedural pain relief. Infants are treated with numerous doses of sucrose, raising concerns about potential long-term effects. The objective of this study was to determine the long-term effects of neonatal oral sucrose treatment on growth and liver metabolism in a mouse model. Neonatal female and male mice were randomly assigned to one of two oral treatments (n = 7-10 mice/group/sex): sterile water or sucrose. Pups were treated 10 times/day for the first 6 days of life with 0.2 mg/g body wt of respective treatments (24% solution; 1-4 μL/dose) to mimic what is given to preterm infants. Mice were weaned at age 3 wk onto a control diet and fed until age 16 wk. Sucrose-treated female and male mice gained less weight during the treatment period and were smaller at weaning than water-treated mice (P ≤ 0.05); no effect of sucrose treatment on body weight was observed at adulthood. However, adult sucrose-treated female mice had smaller tibias and lower serum insulin-like growth factor-1 than adult water-treated female mice (P ≤ 0.05); these effects were not observed in males. Lower liver S-adenosylmethionine, phosphocholine, and glycerophosphocholine were observed in adult sucrose-treated compared with water-treated female and male mice (P ≤ 0.05). Sucrose-treated female, but not male, mice had lower liver free choline and higher liver betaine compared with water-treated female mice (P < 0.01). Our findings suggest that repeated neonatal sucrose treatment has long-term sex-specific effects on growth and liver methionine and choline metabolism.
Collapse
Affiliation(s)
- Cynthia Y Ramírez-Contreras
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Arya E Mehran
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Melody Salehzadeh
- Department of Psychology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ei-Xia Mussai
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua W Miller
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey
| | - Andre Smith
- Department of Nutritional Sciences, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey
| | - Manon Ranger
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Nursing, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa Holsti
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Occupational Science and Occupational Therapy, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiran K Soma
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Psychology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
58
|
Schernthaner-Reiter MH, Trivellin G, Roetzer T, Hainfellner JA, Starost MF, Stratakis CA. Prkar1a haploinsufficiency ameliorates the growth hormone excess phenotype in Aip-deficient mice. Hum Mol Genet 2021; 29:2951-2961. [PMID: 32821937 DOI: 10.1093/hmg/ddaa178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/24/2022] Open
Abstract
Mutations of the regulatory subunit (PRKAR1A) of the cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA), leading to activation of the PKA pathway, are the genetic cause of Carney complex which is frequently accompanied by somatotroph tumors. Aryl hydrocarbon receptor-interacting protein (AIP) mutations lead to somatotroph tumorigenesis in mice and humans. The mechanisms of AIP-dependent pituitary tumorigenesis are still under investigation and evidence points to a connection between the AIP and PKA pathways. In this study, we explore the combined effects of Aip and Prkar1a deficiency on mouse phenotype and, specifically, pituitary histopathology. Aip+/- mice were compared with double heterozygous Aip+/-, Prkar1a+/- mice. The phenotype (including histopathology and serological studies) was recorded at 3, 6, 9 and 12 months of age. Detailed pituitary histological and immunohistochemical studies were performed at 12 months. Twelve-month old Aip+/- mice demonstrated phenotypic and biochemical evidence of GH excess including significantly elevated insulin-like growth factor 1 levels, larger weight and body length, higher hemoglobin and cholesterol levels and a higher frequency of growth plate thickening in comparison to Aip+/, Prkar1a+/- mice. Pituitary histopathology did not uncover any pituitary adenomas or somatotroph hyperplasia in either group. These results demonstrate a slow progression from elevated GH release to the formation of overt somatotropinomas in Aip+/- mice; the acromegalic phenotype of these mice is surprisingly ameliorated in Aip+/-, Prkar1a+/- mice. This highlights the complexities of interaction between the AIP and PKA pathway. Specifically targeting GH secretion rather than somatotroph proliferation may be an advantage in the medical treatment of AIP-dependent human acromegaly.
Collapse
Affiliation(s)
- Marie Helene Schernthaner-Reiter
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.,Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.,Laboratory of Cellular and Molecular Endocrinology and Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center - IRCCS, 20089 Rozzano, Italy
| | - Thomas Roetzer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A Hainfellner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Matthew F Starost
- Office of Research Services (ORS), Division of Veterinary Resources (DVR), Office of the Director, National Institutes of Health, Bethesda, MD 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
59
|
SINE Insertion in the Intron of Pig GHR May Decrease Its Expression by Acting as a Repressor. Animals (Basel) 2021; 11:ani11071871. [PMID: 34201672 PMCID: PMC8300111 DOI: 10.3390/ani11071871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary GH/IGF axis genes play a central role in the regulation of skeletal accretion during development and growth, and thus represent candidate genes for growth traits. Retrotransposon insertion polymorphisms are major contributors to structural variations. They tend to generate large effect mutations resulting in variations in target gene activity and phenotype due to the fact that they carry functional elements, such as enhancers, insulators, or promoters. In the present study, RIPs in four GH/IGF axis genes (GH, GHR, IGF1, and IGF1R) were investigated by comparative genomics and PCR. Four RIPs in the GHR gene and one RIP in the IGF1 gene were identified. Further analysis revealed that one RIP in the first intron of GHR might play a role in the regulation of GHR expression by acting as a repressor. These findings contribute to the understanding of the role of RIPs in the genetic variation of GH/IGF axis genes and phenotypic variation in pigs. Abstract The genetic diversity of the GH/IGF axis genes and their association with the variation of gene expression and phenotypic traits, principally represented by SNPs, have been extensively reported. Nevertheless, the impact of retrotransposon insertion polymorphisms (RIPs) on the GH/IGF axis gene activity has not been reported. In the present study, bioinformatic prediction and PCR verification were performed to screen RIPs in four GH/IGF axis genes (GH, GHR, IGF1 and IGF1R). In total, five RIPs, including one SINE RIP in intron 3 of IGF1, one L1 RIP in intron 7 of GHR, and three SINE RIPs in intron 1, intron 5 and intron 9 of GHR, were confirmed by PCR, displaying polymorphisms in diverse breeds. Dual luciferase reporter assay revealed that the SINE insertion in intron 1 of GHR significantly repressed the GHR promoter activity in PK15, Hela, C2C12 and 3T3-L1 cells. Furthermore, qPCR results confirmed that this SINE insertion was associated with a decreased expression of GHR in the leg muscle and longissimus dorsi, indicating that it may act as a repressor involved in the regulation of GHR expression. In summary, our data revealed that RIPs contribute to the genetic variation of GH/IGF axis genes, whereby one SINE RIP in the intron 1 of GHR may decrease the expression of GHR by acting as a repressor.
Collapse
|
60
|
Keshvari S, Caruso M, Teakle N, Batoon L, Sehgal A, Patkar OL, Ferrari-Cestari M, Snell CE, Chen C, Stevenson A, Davis FM, Bush SJ, Pridans C, Summers KM, Pettit AR, Irvine KM, Hume DA. CSF1R-dependent macrophages control postnatal somatic growth and organ maturation. PLoS Genet 2021; 17:e1009605. [PMID: 34081701 PMCID: PMC8205168 DOI: 10.1371/journal.pgen.1009605] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/15/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Homozygous mutation of the Csf1r locus (Csf1rko) in mice, rats and humans leads to multiple postnatal developmental abnormalities. To enable analysis of the mechanisms underlying the phenotypic impacts of Csf1r mutation, we bred a rat Csf1rko allele to the inbred dark agouti (DA) genetic background and to a Csf1r-mApple reporter transgene. The Csf1rko led to almost complete loss of embryonic macrophages and ablation of most adult tissue macrophage populations. We extended previous analysis of the Csf1rko phenotype to early postnatal development to reveal impacts on musculoskeletal development and proliferation and morphogenesis in multiple organs. Expression profiling of 3-week old wild-type (WT) and Csf1rko livers identified 2760 differentially expressed genes associated with the loss of macrophages, severe hypoplasia, delayed hepatocyte maturation, disrupted lipid metabolism and the IGF1/IGF binding protein system. Older Csf1rko rats developed severe hepatic steatosis. Consistent with the developmental delay in the liver Csf1rko rats had greatly-reduced circulating IGF1. Transfer of WT bone marrow (BM) cells at weaning without conditioning repopulated resident macrophages in all organs, including microglia in the brain, and reversed the mutant phenotypes enabling long term survival and fertility. WT BM transfer restored osteoclasts, eliminated osteopetrosis, restored bone marrow cellularity and architecture and reversed granulocytosis and B cell deficiency. Csf1rko rats had an elevated circulating CSF1 concentration which was rapidly reduced to WT levels following BM transfer. However, CD43hi non-classical monocytes, absent in the Csf1rko, were not rescued and bone marrow progenitors remained unresponsive to CSF1. The results demonstrate that the Csf1rko phenotype is autonomous to BM-derived cells and indicate that BM contains a progenitor of tissue macrophages distinct from hematopoietic stem cells. The model provides a unique system in which to define the pathways of development of resident tissue macrophages and their local and systemic roles in growth and organ maturation.
Collapse
Affiliation(s)
- Sahar Keshvari
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Melanie Caruso
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Ngari Teakle
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Lena Batoon
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Anuj Sehgal
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Omkar L. Patkar
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Michelle Ferrari-Cestari
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Cameron E. Snell
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Qld, Australia
| | - Alex Stevenson
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Felicity M. Davis
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Stephen J. Bush
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Clare Pridans
- Centre for Inflammation Research and Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Kim M. Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Allison R. Pettit
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
| | - Katharine M. Irvine
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
- * E-mail: (KMI); (DAH)
| | - David A. Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia
- * E-mail: (KMI); (DAH)
| |
Collapse
|
61
|
Trade‐offs between male fertility reduction and selected growth factors or the klotho response in a lipopolysaccharide-dependent mouse model. Toxicol Res 2021; 38:175-186. [PMID: 35415080 PMCID: PMC8960506 DOI: 10.1007/s43188-021-00098-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/01/2021] [Accepted: 04/20/2021] [Indexed: 11/06/2022] Open
Abstract
The increasing number of depression cases leads to a greater need for new antidepressant treatment development. It is postulated that antidepressants may harm male fertility, but the cellular mechanism is still poorly understood. The role of growth factors and klotho protein in maintaining normal male reproductive function is well documented. Hence, the study aimed to investigate the effect of the antidepressant drug – imipramine (tricyclic AD), and other substances with antidepressant potential (ALS), administered in combination or in combination with LPS (an animal model of depression) on gene expression and protein synthesis of IGF-2 (insulin-like growth factor 2), TGF-β1 (transforming growth factor β1), NGF (nerve growth factor), KGF (keratinocyte growth factor) and protein synthesis of VEGF-A (vascular endothelial growth factor A), IGF-IR (insulin-like growth factor receptor 1), EGFR (epidermal growth factor receptor) and klotho in the testis of mice. Mice were injected intraperitoneally with selected ALS and LPS or 10% DMSO (controls) (n = 7/group) once a day for 14 days. Animals were decapitated and testes collected for RNA and protein purification. PCR and western blot methods were employed for the evaluation of growth factors and klotho expression. The results obtained indicated a decreased level of most of the analyzed genes and proteins, except KGF; its expression increased after treatment with MTEP and IMI administrated individually and after NS-398, and IMI in combination with LPS. Our results may suggest that the tested ALS and LPS can contribute to a reduction of male fertility, but NS-398, IMI, and IMI+NS-398 may also act as stimulants after LPS.
Collapse
|
62
|
Zuo J, Huo T, Du X, Yang Q, Wu Q, Shen J, Liu C, Hung TC, Yan W, Li G. The joint effect of parental exposure to microcystin-LR and polystyrene nanoplastics on the growth of zebrafish offspring. JOURNAL OF HAZARDOUS MATERIALS 2021; 410:124677. [PMID: 33277077 DOI: 10.1016/j.jhazmat.2020.124677] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
The coexistence of nanoplastics (NPs) and various pollutants in the environment has become a problem that cannot be ignored. In order to identify the microcystin-LR (MCLR) bioaccumulation and the potential impacts on the early growth of F1 zebrafish (Danio rerio) offspring in the presence of polystyrene nanoplastics (PSNPs), PSNPs and MCLR were used to expose adult zebrafish for 21days. The exposure groups divided into MCLR (0, 0.9, 4.5 and 22.5μgL-1) alone groups and PSNP (100μgL-1) and MCLR co-exposure groups. F1 embryos were collected and developed to 120 h post-fertilization (hpf) in clear water. Compared with the exposure to MCLR only, the combined exposure increased the parental transfer of MCLR to the offspring and subsequently exacerbated the growth inhibition of F1 larvae. Further research clarified that combined exposure of PSNPs and MCLR could reduce the levels of thyroxine (T4) and 3, 5, 3'-triiodothyronine (T3) by altering the expression of hypothalamus-pituitary-thyroid (HPT) axis-related genes, eventually leading to growth inhibition of F1 larvae. Our results also exhibited combined exposure of PSNPs and MCLR could change the transcription of key genes of the GH/IGF axis compared with MCLR single exposure, suggesting the GH/IGF axis was a potential target for the growth inhibition of F1 larvae in PSNPs and MCLR co-exposure groups. The present study highlights the potential risks of coexistence of MCLR and PSNPs on development of fish offspring, and the environmental risks to aquatic ecosystems.
Collapse
Affiliation(s)
- Junli Zuo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Tangbin Huo
- Heilongjiang River Fishery Research Institute, Chinese Academy of Fishery Sciences, Harbin 150010, China
| | - Xue Du
- Heilongjiang River Fishery Research Institute, Chinese Academy of Fishery Sciences, Harbin 150010, China
| | - Qing Yang
- Key Laboratory of Ecological Impacts of Hydraulic-Projects and Restoration of Aquatic Ecosystem of Ministry of Water Resources, Institute of Hydroecology, Ministry of Water Resources and Chinese Academy of Sciences, Wuhan 430079, China
| | - Qin Wu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianzhong Shen
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chunsheng Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Tien-Chieh Hung
- Department of Biological and Agricultural Engineering, University of California-Davis, Davis, CA 95616, USA
| | - Wei Yan
- Institute of Agricultural Quality Standards & Testing Technology, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China.
| |
Collapse
|
63
|
Zhang L, Duan C, Guo Y, Zhang Y, Liu Y. Inhibition of prolactin promotes secondary skin follicle activation in cashmere goats. J Anim Sci 2021; 99:6167825. [PMID: 33693756 DOI: 10.1093/jas/skab079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/09/2021] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to investigate the involvement of prolactin (PRL) on development of secondary skin follicles in cashmere goats. Goats were randomly assigned to either a bromocriptine treatment or control group. Samples of cashmere fiber, blood, and skin were collected from all goats after 1 mo. The results indicated that the length, growth rate, and diameter of fibers were not influenced (P > 0.05) by the inhibition of PRL resulting from the treatment with bromocriptine. There was a tendency for increases in total follicle number, primary and secondary follicle numbers, and in the ratio of secondary to primary follicles following treatment with bromocriptine, but these differences were not significant (P > 0.05). The percentage of active secondary follicles in anagen was increased (P < 0.05) in the bromocriptine-treated goats, but there was no effect of treatment on the percentage of active primary follicles. Bromocriptine decreased (P < 0.05) circulating concentrations of PRL and Insulin-like growth factor 1 (IGF1) and increased (P < 0.05) those of melatonin (MT), but there was no effect of this treatment on the serum concentrations of cortisol, growth hormone, tetraiodothyronine, and triiodothyronine. In bromocriptine-treated goats, mRNA expressions of PRL and MT membrane receptor 1a (MTNR1a) were decreased (P < 0.05) and mRNA expression of MT nuclear receptor (RORα) was increased (P < 0.05), but there was no effect of the treatment on expression of long PRL receptor, short PRL receptor, MT membrane receptor 1b and IGF1. It is concluded that inhibition of PRL promotes secondary hair follicle development in the anagen phase, possibly by downregulating MTNR1a and up-regulating RORα gene expression in the skin.
Collapse
Affiliation(s)
- Lechao Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, PR China
| | - Chunhui Duan
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, PR China
| | - Yunxia Guo
- College of Life Science, Hebei Agricultural University, Baoding 071000, PR China
| | - Yingjie Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, PR China
| | - Yueqin Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, PR China
| |
Collapse
|
64
|
IGF-1 as selected growth factor multi-response to antidepressant-like substances activity in C57BL/6J mouse testis model. Acta Histochem 2021; 123:151685. [PMID: 33556704 DOI: 10.1016/j.acthis.2021.151685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/14/2021] [Accepted: 01/26/2021] [Indexed: 11/21/2022]
Abstract
Insulin-like growth factor (IGF-1) affects almost all cells in the body. Extremely important functions of this growth factor have been demonstrated in the brain and the reproductive system of both, females and males. Also, it is considered as a pro-inflammatory cytokine adjusting tissue homeostasis. However, it seems to play a special role in the male reproductive system and it may be disturbed by the application of antidepressants with different mechanisms of drug action during therapy. To date, the effect of antidepressant-like substances (ALS) on the course of physiological processes in male testicular cells is poorly understood. Therefore, the purpose of the research was to determine the presence, localizationof IGF-1R (insulin-like growth factor 1 β receptor) and mRNA gene expression of IGF-1R and IGF-1 after administration of 3-[(2-methyl-1,3-tiazol-4-yl)ethynyl]-pyridine (MTEP) and N-[2-(Cyclohexyloxy)-4-nitrophenyl]-methanesulfonamide (NS-398) in the different scheme in the testis of mice. Imipramine was used as a reference drug having a documented interaction with the mGluR5 receptors. The immunohistochemical analyses showed the localization of IGF-1R in Sertoli, Leydig, and germinal cells after all used substances. Differences in receptor localization were observed depending on the drugs applied and the type of analyzed cells. In contrast, there was a significant increase in IGF-1 gene expression after IMI + NS-398 and in IGF-1R after MTEP + NS-398 and IMI + NS-398 administration. It can, therefore, be assumed that the use of a combination of NS-398 with some ALS may run different mechanisms of action and affect the regulation of reproductive function in mouse testis through maintaining homeostasis at the molecular and immunological levels related to IGF.
Collapse
|
65
|
Bartke A, Hascup E, Hascup K, Masternak MM. Growth Hormone and Aging: New Findings. World J Mens Health 2021; 39:454-465. [PMID: 33663025 PMCID: PMC8255405 DOI: 10.5534/wjmh.200201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 12/21/2020] [Accepted: 01/02/2021] [Indexed: 01/04/2023] Open
Abstract
Complex relationships between growth hormone (GH) signaling and mammalian aging continue to attract attention of many investigators. Recent results include evidence that the impact of GH on genome maintenance (DNA damage and repair) is drastically different in normal as compared to cancer cells, consistent with GH promoting aging and cancer progression. Impact of GH on DNA methylation was studied as a possible mechanism linking actions of GH during early life to the trajectory of aging. Animals with reduced or enhanced GH signaling and novel animals with adipocyte-specific deletion of GH receptors were used to elucidate the effects of GH on white and brown adipose tissue, including the impact of this hormone on lipolysis, fibrosis, and thermogenesis. Effects of GH on adipose tissue related to lipid and energy metabolism emerge as mechanistic links between GH, healthspan, and lifespan. Treatment of healthy men with a combination of GH, dehydroepiandrosterone, and metformin was reported to restore thymus function and reduce epigenetic age. Studies of human subjects with deficiency of GH or GH receptors and studies of mice with the same endocrine syndromes identified several phenotypic changes related (positively or negatively) to the previously reported predisposition to healthy aging. Results of these and other recent studies advance present understanding of the mechanisms by which GH influences aging and longevity and of the trade-offs involved.
Collapse
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Erin Hascup
- Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin Hascup
- Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
66
|
Lasigliè D. Sirtuins and the prevention of immunosenescence. VITAMINS AND HORMONES 2021; 115:221-264. [PMID: 33706950 DOI: 10.1016/bs.vh.2020.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging of hematopoietic stem cells (HSCs) has been largely described as one underlying cause of senescence of the immune-hematopoietic system (immunosenescence). A set of well-defined hallmarks characterizes aged HSCs contributing to unbalanced hematopoiesis and aging-associated functional alterations of both branches of the immune system. In this chapter, the contribution of sirtuins, a family of conserved NAD+ dependent deacetylases with key roles in metabolism, genome integrity, aging and lifespan, to immunosenescence, will be addressed. In particular, the role of SIRT6 will be deeply analyzed highlighting a multifaceted part of this deacetylase in HSCs aging as well as in the immunosenescence of dendritic cells (DCs). These and other emerging data are currently paving the way for future design and development of rejuvenation means aiming at rescuing age-related changes in immune function in the elderly and combating age-associated hematopoietic diseases.
Collapse
Affiliation(s)
- Denise Lasigliè
- Istituto Comprensivo "Franco Marro", Ministero dell'Istruzione Ministero dell'Università e della Ricerca (M.I.U.R), Villar Perosa, TO, Italy.
| |
Collapse
|
67
|
Lepa C, Hoppe S, Stöber A, Skryabin BV, Sievers LK, Heitplatz B, Ciarimboli G, Neugebauer U, Lindenmeyer MT, Cohen CD, Drexler HC, Boor P, Weide T, Pavenstädt H, George B. TrkC Is Essential for Nephron Function and Trans-Activates Igf1R Signaling. J Am Soc Nephrol 2021; 32:357-374. [PMID: 33380522 PMCID: PMC8054883 DOI: 10.1681/asn.2020040424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/03/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Injury to kidney podocytes often results in chronic glomerular disease and consecutive nephron malfunction. For most glomerular diseases, targeted therapies are lacking. Thus, it is important to identify novel signaling pathways contributing to glomerular disease. Neurotrophic tyrosine kinase receptor 3 (TrkC) is expressed in podocytes and the protein transmits signals to the podocyte actin cytoskeleton. METHODS Nephron-specific TrkC knockout (TrkC-KO) and nephron-specific TrkC-overexpressing (TrkC-OE) mice were generated to dissect the role of TrkC in nephron development and maintenance. RESULTS Both TrkC-KO and TrkC-OE mice exhibited enlarged glomeruli, mesangial proliferation, basement membrane thickening, albuminuria, podocyte loss, and aspects of FSGS during aging. Igf1 receptor (Igf1R)-associated gene expression was dysregulated in TrkC-KO mouse glomeruli. Phosphoproteins associated with insulin, erb-b2 receptor tyrosine kinase (Erbb), and Toll-like receptor signaling were enriched in lysates of podocytes treated with the TrkC ligand neurotrophin-3 (Nt-3). Activation of TrkC by Nt-3 resulted in phosphorylation of the Igf1R on activating tyrosine residues in podocytes. Igf1R phosphorylation was increased in TrkC-OE mouse kidneys while it was decreased in TrkC-KO kidneys. Furthermore, TrkC expression was elevated in glomerular tissue of patients with diabetic kidney disease compared with control glomerular tissue. CONCLUSIONS Our results show that TrkC is essential for maintaining glomerular integrity. Furthermore, TrkC modulates Igf-related signaling in podocytes.
Collapse
Affiliation(s)
- Carolin Lepa
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Sascha Hoppe
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Antje Stöber
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Boris V. Skryabin
- Medical Faculty, Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische-Wilhelms-University, Münster, Germany
| | | | - Barbara Heitplatz
- Gerhard-Domagk Institute for Pathology, University Hospital Münster, Münster, Germany
| | | | - Ute Neugebauer
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Maja T. Lindenmeyer
- III. Medizinische Klinik und Poliklinik, University Hospital Hamburg-Eppendorf, Germany
| | - Clemens D. Cohen
- Klinik für Nieren-, Hochdruck- und Rheumaerkrankungen, München Klinik Harlaching, Germany
| | - Hannes C.A. Drexler
- Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Thomas Weide
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | | | - Britta George
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| |
Collapse
|
68
|
Luo Y, Zheng Z, Yang Y, Bai X, Yang H, Zhu H, Pan H, Chen S. Effects of growth hormone on cognitive, motor, and behavioral development in Prader-Willi syndrome children: a meta-analysis of randomized controlled trials. Endocrine 2021; 71:321-330. [PMID: 33222122 DOI: 10.1007/s12020-020-02547-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/03/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE The benefits of growth hormone (GH) therapy in Prader-Willi syndrome (PWS) children are well established, but there is still considerable controversy regarding whether GH treatment can improve cognitive, motor, and behavioral development in PWS children. The objectives of this meta-analysis were to quantitatively evaluate the effects of GH on cognitive, motor function, and behavioral development in PWS children. METHODS Randomized controlled trials (RCTs) examining the effects of GH on cognitive, motor, and behavioral development in PWS children were identified by searching the MEDLINE, EMBASE, and Cochrane Library databases. Intervention effects were represented by Hedges'g and pooled to calculate effect sizes using a random-effects model. RESULTS Ten relevant studies comprising data from 302 participants were finally included. We observed no significant difference in cognitive performance between the GH treatment group and the control group (p = 0.197). GH treatment was shown to remarkably improve motor development in PWS children compared with the control treatment (p < 0.001), with moderate positive treatment effects (Hedges'g [95% CI] = 0.71 [0.38, 1.03]). There were no significant differences between the GH group and the control group based on objective assessments of behavioral development (p = 0.53). CONCLUSIONS The meta-analysis suggested that GH treatment had a significantly positive effect on motor development, with moderate treatment effects in PWS children; however, there was no evidence of effects on cognitive or behavioral development.
Collapse
Affiliation(s)
- Yunyun Luo
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China
| | - Zhoude Zheng
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingying Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China
| | - Xi Bai
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China.
| | - Shi Chen
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Research Center for Behavior Medicine in Growth and Development, Beijing, China.
| |
Collapse
|
69
|
Affiliation(s)
- Horacio M Domené
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
| | - Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands.
| | - Stuart J Frank
- Department of Medicine, University of Alabama at Birmingham, Birmingham VAMC Medical Service, Birmingham, AL, USA.
| |
Collapse
|
70
|
Lekvijittada K, Hosomichi J, Maeda H, Hong H, Changsiripun C, Kuma YI, Oishi S, Suzuki JI, Yoshida KI, Ono T. Intermittent hypoxia inhibits mandibular cartilage growth with reduced TGF-β and SOX9 expressions in neonatal rats. Sci Rep 2021; 11:1140. [PMID: 33441835 PMCID: PMC7806651 DOI: 10.1038/s41598-020-80303-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/17/2020] [Indexed: 01/15/2023] Open
Abstract
Intermittent hypoxia (IH) has been associated with skeletal growth. However, the influence of IH on cartilage growth and metabolism is unknown. We compared the effects of IH on chondrocyte proliferation and maturation in the mandibular condyle fibrocartilage and tibial hyaline cartilage of 1-week-old male Sprague-Dawley rats. The rats were exposed to normoxic air (n = 9) or IH at 20 cycles/h (nadir, 4% O2; peak, 21% O2; 0% CO2) (n = 9) for 8 h each day. IH impeded body weight gain, but not tibial elongation. IH also increased cancellous bone mineral and volumetric bone mineral densities in the mandibular condylar head. The mandibular condylar became thinner, but the tibial cartilage did not. IH reduced maturative and increased hypertrophic chondrocytic layers of the middle and posterior mandibular cartilage. PCR showed that IH shifted proliferation and maturation in mandibular condyle fibrocartilage toward hypertrophic differentiation and ossification by downregulating TGF-β and SOX9, and upregulating collagen X. These effects were absent in the tibial growth plate hyaline cartilage. Our results showed that neonatal rats exposed to IH displayed underdeveloped mandibular ramus/condyles, while suppression of chondrogenesis marker expression was detected in the growth-restricted condylar cartilage.
Collapse
Affiliation(s)
- Kochakorn Lekvijittada
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.,Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Forensic Medicine, Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan. .,Department of Forensic Medicine, Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan.
| | - Hideyuki Maeda
- Department of Forensic Medicine, Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan
| | - Haixin Hong
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.,Department of Forensic Medicine, Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan
| | - Chidsanu Changsiripun
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Yo-Ichiro Kuma
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Shuji Oishi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Jun-Ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken-Ichi Yoshida
- Department of Forensic Medicine, Graduate School of Medicine, Tokyo Medical University, Tokyo, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| |
Collapse
|
71
|
Nishimura T, Suchy FP, Bhadury J, Igarashi KJ, Charlesworth CT, Nakauchi H. Generation of Functional Organs Using a Cell-Competitive Niche in Intra- and Inter-species Rodent Chimeras. Cell Stem Cell 2021; 28:141-149.e3. [PMID: 33373620 PMCID: PMC8025673 DOI: 10.1016/j.stem.2020.11.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/12/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
Interspecies organ generation via blastocyst complementation has succeeded in rodents, but not yet in evolutionally more distant species. Early developmental arrest hinders the formation of highly chimeric fetuses. We demonstrate that the deletion of insulin-like growth factor 1 receptor (Igf1r) in mouse embryos creates a permissive "cell-competitive niche" in several organs, significantly augmenting both mouse intraspecies and mouse/rat interspecies donor chimerism that continuously increases from embryonic day 11 onward, sometimes even taking over entire organs within intraspecies chimeras. Since Igf1r deletion allows the evasion of early developmental arrest, interspecies fetuses with high levels of organ chimerism can be generated via blastocyst complementation. This observation should facilitate donor cell contribution to host tissues, resulting in whole-organ generation via blastocyst complementation across wide evolutionary distances.
Collapse
Affiliation(s)
- Toshiya Nishimura
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fabian P Suchy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joydeep Bhadury
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Dept of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE 413 45, Gothenburg, Sweden
| | - Kyomi J Igarashi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carsten T Charlesworth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
72
|
Growth Hormone Upregulates Mediators of Melanoma Drug Efflux and Epithelial-to-Mesenchymal Transition In Vitro and In Vivo. Cancers (Basel) 2020; 12:cancers12123640. [PMID: 33291663 PMCID: PMC7761932 DOI: 10.3390/cancers12123640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Growth hormone (GH) action is strongly implicated in the progression and therapy resistance in several types of solid tumors which overexpress the GH receptor (GHR). The aim of our study was to characterize the effects of GH and its downstream effector insulin-like growth factor 1 (IGF-1) on melanoma using in vitro and in vivo models. We confirmed an IGF-1-independent role of elevated circulating GH in upregulating key mechanisms of therapy resistance and malignancy with analyses conducted at the molecular and cellular level. We identified that GH upregulates key mechanisms of therapy resistance and metastases in melanoma tumors in an IGF-1 dependent and independent manner by upregulating multidrug efflux pumps and EMT transcription factors. Our study reveals that GH action renders an intrinsic drug resistance phenotype to the melanoma tumors—a clinically crucial property of GH verifiable in other human cancers with GHR expression. Abstract Growth hormone (GH) and the GH receptor (GHR) are expressed in a wide range of malignant tumors including melanoma. However, the effect of GH/insulin-like growth factor (IGF) on melanoma in vivo has not yet been elucidated. Here we assessed the physical and molecular effects of GH on mouse melanoma B16-F10 and human melanoma SK-MEL-30 cells in vitro. We then corroborated these observations with syngeneic B16-F10 tumors in two mouse lines with different levels of GH/IGF: bovine GH transgenic mice (bGH; high GH, high IGF-1) and GHR gene-disrupted or knockout mice (GHRKO; high GH, low IGF-1). In vitro, GH treatment enhanced mouse and human melanoma cell growth, drug retention and cell invasion. While the in vivo tumor size was unaffected in both bGH and GHRKO mouse lines, multiple drug-efflux pumps were up regulated. This intrinsic capacity of therapy resistance appears to be GH dependent. Additionally, epithelial-to-mesenchymal transition (EMT) gene transcription markers were significantly upregulated in vivo supporting our current and recent in vitro observations. These syngeneic mouse melanoma models of differential GH/IGF action can be valuable tools in screening for therapeutic options where lowering GH/IGF-1 action is important.
Collapse
|
73
|
Domené S, Domené HM. The role of acid-labile subunit (ALS) in the modulation of GH-IGF-I action. Mol Cell Endocrinol 2020; 518:111006. [PMID: 32861700 DOI: 10.1016/j.mce.2020.111006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
Abstract
Acid-labile subunit (ALS) deficiency (ACLSD) constitutes the first monogenic defect involving a member of the Insulin-like Growth Factor (IGF) binding protein system. The lack of ALS completely disrupts the circulating IGF system. Autocrine/paracrine action of local produced IGF-I could explain the mild effect on growth. In the present work we have revised the more relevant clinical and biochemical consequences of complete ACLSD in 61 reported subjects from 31 families. Low birth weight and/or length, reduced head circumference, height between -2 and -3 SD, pubertal delay and insulin resistance are commonly observed. Partial ACLSD could be present in children initially labeled as idiopathic short stature, presenting low IGF-I levels, suggesting that one functional IGFALS allele is insufficient to stabilize ternary complexes. Dysfunction of the GH-IGF axis observed in ACLSD may eventually result in increased risk for type-2 diabetes and tumor progression. Consequently, long term surveillance is recommended in these patients.
Collapse
Affiliation(s)
- Sabina Domené
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá', (CEDIE) CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Horacio M Domené
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá', (CEDIE) CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
| |
Collapse
|
74
|
Frank SJ. Classical and novel GH receptor signaling pathways. Mol Cell Endocrinol 2020; 518:110999. [PMID: 32835785 PMCID: PMC7799394 DOI: 10.1016/j.mce.2020.110999] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
In this review, I summarize historical and recent features of the classical pathways activated by growth hormone (GH) through the cell surface GH receptor (GHR). GHR is a cytokine receptor superfamily member that signals by activating the non-receptor tyrosine kinase, JAK2, and members of the Src family kinases. Activation of the GHR engages STATs, PI3K, and ERK pathways, among others, and details of these now-classical pathways are presented. Modulating elements, including the SOCS proteins, phosphatases, and regulated GHR metalloproteolysis, are discussed. In addition, a novel physical and functional interaction of GHR with IGF-1R is summarized and discussed in terms of its mechanisms, consequences, and physiological and therapeutic implications.
Collapse
Affiliation(s)
- Stuart J Frank
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, 1720 2nd Avenue South, BDB 485, AL, 35294-0012, USA; Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA; Endocrinology Section, Medical Service, Veterans Affairs Medical Center, Birmingham, AL, 35233, USA.
| |
Collapse
|
75
|
Landi E, Karabatas L, Scaglia P, Pisciottano F, Gutiérrez M, Ramírez L, Bergadá I, Rey RA, Jasper HG, Domené HM, Plazas PV, Domené S. Expression of acid-labile subunit (ALS) in developing and adult zebrafish and its role in dorso-ventral patterning during development. Gen Comp Endocrinol 2020; 299:113591. [PMID: 32828812 DOI: 10.1016/j.ygcen.2020.113591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 11/24/2022]
Abstract
Mammalian acid-labile subunit (ALS) is a serum protein that binds binary complexes between Insulin-like growth factors (IGFs) and Insulin-like growth factor-binding proteins (IGFBPs) extending their half-life and keeping them in the vasculature. Human ALS deficiency (ACLSD), due to homozygous or compound heterozygous mutations in IGFALS, leads to moderate short stature with reduced levels of IGF-I and IGFBP-3. There is only one corresponding zebrafish ortholog gene and it has not yet been studied. In this study we elucidate the role of igfals during zebrafish development. In zebrafish embryos igfals mRNA is expressed throughout development, mainly in the brain and subsequently also in the gut and swimbladder. To determine its role during development, we knocked down igfals gene product using morpholinos (MOs). Igfals morphant embryos displayed dorsalization in different degrees of severity, including a shortened trunk and loss of tail. Furthermore, co-injection of human IGFALS (hIGFALS) mRNA was able to rescue the MO-induced phenotype. Finally, overexpression of either hIGFALS or zebrafish igfals (zigfals) mRNA leads to ventralization of embryos including a reduced head and enlarged tail. These findings suggest that als plays an important role in dorso-ventral patterning during zebrafish development.
Collapse
Affiliation(s)
- Estefanía Landi
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Liliana Karabatas
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Paula Scaglia
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Francisco Pisciottano
- Instituto de Biología y Medicina Experimental (IBYME), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina.
| | - Mariana Gutiérrez
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Laura Ramírez
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Héctor Guillermo Jasper
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Horacio Mario Domené
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| | - Paola Viviana Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Paraguay 2155, C1121ABG Buenos Aires, Argentina.
| | - Sabina Domené
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.
| |
Collapse
|
76
|
Lodjak J, Verhulst S. Insulin-like growth factor 1 of wild vertebrates in a life-history context. Mol Cell Endocrinol 2020; 518:110978. [PMID: 32798584 DOI: 10.1016/j.mce.2020.110978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Broad variation in intra- and interspecific life-history traits is largely shaped by resource limitation and the ensuing allocation trade-offs that animals are forced to make. Insulin-like growth factor 1 (IGF-1), a growth-hormone-dependent peptide, may be a key player in the regulation of allocation processes. In laboratory animals, the effects of IGF-1 on growth- and development (positive), reproduction (positive), and longevity (negative) are well established. We here review the evidence on these effects in wild vertebrates, where animals are more likely to face resource limitation and other challenges. We point out the similarities and dissimilarities in patterns of IGF-1 functions obtained in these two different study settings and discuss the knowledge we need to develop a comprehensive picture of the role of IGF-1 in mediating life-history variation of wild vertebrates.
Collapse
Affiliation(s)
- Jaanis Lodjak
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu, 51014, Estonia; Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands
| |
Collapse
|
77
|
Jiang Z, Derrick-Roberts AL, Byers S. Altered IHH signaling contributes to reduced chondrocyte proliferation in the growth plate of MPS VII mice. Mol Genet Metab Rep 2020; 25:100668. [PMID: 33117654 PMCID: PMC7582094 DOI: 10.1016/j.ymgmr.2020.100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 10/29/2022] Open
Abstract
Bone elongation is driven by chondrocyte proliferation and hypertrophy in the growth plate. Both processes are modulated by multiple signaling pathways including the Indian Hedgehog (IHH) signaling pathway. Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders characterized by accumulation of glycosaminoglycans (GAGs) in multiple tissues and organs, leading to a range of clinical symptoms including bone shortening through mechanisms that are not fully understood. Using MPS VII mice, we previously observed a reduction in the number of proliferating and hypertrophic chondrocytes and a reduced gene expression of Ihh in the tibial growth plate. We further demonstrate here that IHH secretion by MPS VII chondrocytes was reduced both in vitro and in vivo. While normal chondrocytes showed no response to exogenous IHH, proliferation of MPS VII chondrocytes was stimulated in response to exogenous IHH in vitro. This was accompanied by an elevated gene expression of patched receptor (Ptch1). The results from this study suggested that reduced proliferation in MPS VII growth plate may be partially due to dysfunction of the IHH signaling pathway.
Collapse
Affiliation(s)
- Zhirui Jiang
- Genetics and Evolution, The University of Adelaide, Adelaide, SA, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Ainslie L.K. Derrick-Roberts
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Paediatrics, The University of Adelaide, Adelaide, SA, Australia
| | - Sharon Byers
- Genetics and Evolution, The University of Adelaide, Adelaide, SA, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Paediatrics, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
78
|
Forbes BE, Blyth AJ, Wit JM. Disorders of IGFs and IGF-1R signaling pathways. Mol Cell Endocrinol 2020; 518:111035. [PMID: 32941924 DOI: 10.1016/j.mce.2020.111035] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
The insulin-like growth factor (IGF) system comprises two ligands, IGF-I and IGF-II, that regulate multiple physiological processes, including mammalian development, metabolism and growth, through the type 1 IGF receptor (IGF-1R). The growth hormone (GH)-IGF-I axis is the major regulator of longitudinal growth. IGF-II is expressed in many tissues, notably the placenta, to regulate human pre- and post-natal growth and development. This review provides a brief introduction to the IGF system and summarizes findings from reports arising from recent larger genomic sequencing studies of human genetic mutations in IGF1 and IGF2 and genes of proteins regulating IGF action, namely the IGF-1R, IGF-1R signaling pathway components and the IGF binding proteins (IGFBPs). A perspective on the effect of homozygous mutations on structure and function of the IGFs and IGF-1R is also given and this is related to the effects on growth.
Collapse
Affiliation(s)
- Briony E Forbes
- Discipline of Medical Biochemistry, Flinders Health and Medical Research Institute, Flinders University, Australia.
| | - Andrew J Blyth
- Discipline of Medical Biochemistry, Flinders Health and Medical Research Institute, Flinders University, Australia
| | - Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
79
|
Negron SG, Ercan-Sencicek AG, Freed J, Walters M, Lin Z. Both proliferation and lipogenesis of brown adipocytes contribute to postnatal brown adipose tissue growth in mice. Sci Rep 2020; 10:20335. [PMID: 33230135 PMCID: PMC7683731 DOI: 10.1038/s41598-020-77362-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/26/2020] [Indexed: 02/03/2023] Open
Abstract
Brown adipose tissue (BAT) is the primary non-shivering thermogenesis organ in mammals, which plays essential roles in maintaining the body temperature of infants. Although the development of BAT during embryogenesis has been well addressed in rodents, how BAT grows after birth remains unknown. Using mouse interscapular BAT (iBAT) as an example, we studied the cellular and molecular mechanisms that regulate postnatal BAT growth. By analyzing the developmental dynamics of brown adipocytes (BAs), we found that BAs size enlargement partially accounts for iBAT growth. By investigating the BAs cell cycle activities, we confirmed the presence of proliferative BAs in the neonatal mice. Two weeks after birth, most of the BAs exit cell cycle, and the further expansion of the BAT was mainly due to lipogenesis-mediated BAs volume increase. Microscopy and fluorescence-activated cell sorting analyses suggest that most BAs are mononuclear and diploid. Based on the developmental dynamics of brown adipocytes, we propose that the murine iBAT has two different growth phases between birth and weaning: increase of BAs size and number in the first two weeks, and BAs size enlargement thereafter. In summary, our data demonstrate that both lipogenesis and proliferation of BAs contribute to postnatal iBAT growth in mice.
Collapse
Affiliation(s)
- Steven G Negron
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY, 13501, USA
| | - A Gulhan Ercan-Sencicek
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY, 13501, USA
- Department of Neurosurgery, Program On Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jessica Freed
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY, 13501, USA
| | - Madeline Walters
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY, 13501, USA
| | - Zhiqiang Lin
- Masonic Medical Research Institute, 2150 Bleecker Street, Utica, NY, 13501, USA.
| |
Collapse
|
80
|
Strous GJ, Almeida ADS, Putters J, Schantl J, Sedek M, Slotman JA, Nespital T, Hassink GC, Mol JA. Growth Hormone Receptor Regulation in Cancer and Chronic Diseases. Front Endocrinol (Lausanne) 2020; 11:597573. [PMID: 33312162 PMCID: PMC7708378 DOI: 10.3389/fendo.2020.597573] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
The GHR signaling pathway plays important roles in growth, metabolism, cell cycle control, immunity, homeostatic processes, and chemoresistance via both the JAK/STAT and the SRC pathways. Dysregulation of GHR signaling is associated with various diseases and chronic conditions such as acromegaly, cancer, aging, metabolic disease, fibroses, inflammation and autoimmunity. Numerous studies entailing the GHR signaling pathway have been conducted for various cancers. Diverse factors mediate the up- or down-regulation of GHR signaling through post-translational modifications. Of the numerous modifications, ubiquitination and deubiquitination are prominent events. Ubiquitination by E3 ligase attaches ubiquitins to target proteins and induces proteasomal degradation or starts the sequence of events that leads to endocytosis and lysosomal degradation. In this review, we discuss the role of first line effectors that act directly on the GHR at the cell surface including ADAM17, JAK2, SRC family member Lyn, Ubc13/CHIP, proteasome, βTrCP, CK2, STAT5b, and SOCS2. Activity of all, except JAK2, Lyn and STAT5b, counteract GHR signaling. Loss of their function increases the GH-induced signaling in favor of aging and certain chronic diseases, exemplified by increased lung cancer risk in case of a mutation in the SOCS2-GHR interaction site. Insight in their roles in GHR signaling can be applied for cancer and other therapeutic strategies.
Collapse
Affiliation(s)
- Ger J. Strous
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
- BIMINI Biotech B.V., Leiden, Netherlands
| | - Ana Da Silva Almeida
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Joyce Putters
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Julia Schantl
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Magdalena Sedek
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Johan A. Slotman
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tobias Nespital
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Gerco C. Hassink
- Department of Cell Biology, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jan A. Mol
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
81
|
Werner H, Sarfstein R, Nagaraj K, Laron Z. Laron Syndrome Research Paves the Way for New Insights in Oncological Investigation. Cells 2020; 9:cells9112446. [PMID: 33182502 PMCID: PMC7696416 DOI: 10.3390/cells9112446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
Laron syndrome (LS) is a rare genetic endocrinopathy that results from mutation of the growth hormone receptor (GH-R) gene and is typically associated with dwarfism and obesity. LS is the best characterized entity under the spectrum of the congenital insulin-like growth factor-1 (IGF1) deficiencies. Epidemiological analyses have shown that LS patients do not develop cancer, whereas heterozygous family members have a cancer prevalence similar to the general population. To identify genes and signaling pathways differentially represented in LS that may help delineate a biochemical and molecular basis for cancer protection, we have recently conducted a genome-wide profiling of LS patients. Studies were based on our collection of Epstein–Barr virus (EBV)-immortalized lymphoblastoid cell lines derived from LS patients, relatives and healthy controls. Bioinformatic analyses identified differences in gene expression in several pathways, including apoptosis, metabolic control, cytokine biology, Jak-STAT and PI3K-AKT signaling, etc. Genes involved in the control of cell cycle, motility, growth and oncogenic transformation are, in general, down-regulated in LS. These genetic events seem to have a major impact on the biological properties of LS cells, including proliferation, apoptosis, response to oxidative stress, etc. Furthermore, genomic analyses allowed us to identify novel IGF1 downstream target genes that have not been previously linked to the IGF1 signaling pathway. In summary, by ‘mining’ genomic data from LS patients, we were able to generate clinically-relevant information in oncology and, potentially, related disciplines.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (R.S.); (K.N.)
- Shalom and Varda Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| | - Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (R.S.); (K.N.)
| | - Karthik Nagaraj
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (R.S.); (K.N.)
| | - Zvi Laron
- Endocrine and Diabetes Research Unit, Schneider Children’s Medical Center, Petah Tikva 49292, Israel;
| |
Collapse
|
82
|
Lmod3 promotes myoblast differentiation and proliferation via the AKT and ERK pathways. Exp Cell Res 2020; 396:112297. [DOI: 10.1016/j.yexcr.2020.112297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
|
83
|
McRae HM, Eccles S, Whitehead L, Alexander WS, Gécz J, Thomas T, Voss AK. Downregulation of the GHRH/GH/IGF1 axis in a mouse model of Börjeson-Forssman-Lehman syndrome. Development 2020; 147:dev.187021. [PMID: 32994169 DOI: 10.1242/dev.187021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 09/09/2020] [Indexed: 12/28/2022]
Abstract
Börjeson-Forssman-Lehmann syndrome (BFLS) is an intellectual disability and endocrine disorder caused by plant homeodomain finger 6 (PHF6) mutations. Individuals with BFLS present with short stature. We report a mouse model of BFLS, in which deletion of Phf6 causes a proportional reduction in body size compared with control mice. Growth hormone (GH) levels were reduced in the absence of PHF6. Phf6 - /Y animals displayed a reduction in the expression of the genes encoding GH-releasing hormone (GHRH) in the brain, GH in the pituitary gland and insulin-like growth factor 1 (IGF1) in the liver. Phf6 deletion specifically in the nervous system caused a proportional growth defect, indicating a neuroendocrine contribution to the phenotype. Loss of suppressor of cytokine signaling 2 (SOCS2), a negative regulator of growth hormone signaling partially rescued body size, supporting a reversible deficiency in GH signaling. These results demonstrate that PHF6 regulates the GHRH/GH/IGF1 axis.
Collapse
Affiliation(s)
- Helen M McRae
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Victoria 3052, Australia
| | - Samantha Eccles
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Lachlan Whitehead
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Victoria 3052, Australia
| | - Warren S Alexander
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Victoria 3052, Australia
| | - Jozef Gécz
- Adelaide Medical School and the Robinson Research Institute, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Tim Thomas
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia .,Department of Medical Biology, The University of Melbourne, Victoria 3052, Australia
| | - Anne K Voss
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia .,Department of Medical Biology, The University of Melbourne, Victoria 3052, Australia
| |
Collapse
|
84
|
Magill L, Laemmer C, Woelfle J, Fimmers R, Gohlke B. Early start of growth hormone is associated with positive effects on auxology and metabolism in Prader-Willi-syndrome. Orphanet J Rare Dis 2020; 15:283. [PMID: 33046090 PMCID: PMC7552493 DOI: 10.1186/s13023-020-01527-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prader-Willi-Syndrome (PWS) is characterized by hypothalamic-pituitary dysfunction. Recent research suggests starting growth hormone-treatment (GHT) as soon as possible. The aim of this study is to analyze possible differences in auxological parameters, carbohydrate and lipid metabolism between two groups of children with PWS that started GHT either during or after their first year of life. STUDY DESIGN Retrospective longitudinal study of 62 children (31 males) with genetically confirmed PWS. Upon diagnosis all children were offered GHT, some started immediately, others commenced later. Cohort A (n = 21; 11 males) started GHT at 0.3-0.99 yrs. (mean 0.72 yrs) and Cohort B (n = 41; 20 males) commenced GHT at 1.02-2.54 yrs. (mean 1.42 yrs) of age. Fasting morning blood samples and auxological parameters were obtained before the start of therapy and semi-annually thereafter. Differences between the two cohorts were estimated with a linear mixed-effect model. RESULTS Mean length/height-SDSPWS differed significantly between the groups [1 yr: A: 0.37 (±0.83) vs B: 0.05 (±0.56); 5 yrs.: A: 0.81 (±0.67) vs B: 0.54 (±0.64); p = 0.012]. No significant differences were found in BMI, lean body mass or body fat. Low-density cholesterol was significantly lower in A than in B [LDL: 1 yr: A: 79 (±20) mg/dl vs B: 90 (±19) mg/dl; 5 yrs.: A: 91(±18) mg/dl vs 104 (±26) mg/dl; p = 0.024]. We found significant differences in the glucose homeostasis between the groups [fasting insulin: p = 0.012; HOMA-IR: p = 0.006; HbA1c: p < 0.001; blood glucose: p = 0.022]. CONCLUSIONS An early start of GHT during the first year of life seems to have a favorable effect on height-SDS and metabolic parameters.
Collapse
Affiliation(s)
- Lucy Magill
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital, University of Bonn, Venusberg-Campus, Building 30, 53127, Bonn, Germany
| | - Constanze Laemmer
- Pediatric Endocrinology and Diabetology, St. Bernward Hospital, Treibestraße 9, 31134, Hildesheim, Germany
| | - Joachim Woelfle
- Children's University Hospital Erlangen, Loschgestrasse 15, 91054, Erlangen, Germany
| | - Rolf Fimmers
- University Hospital of Bonn, Institute for Medical Biometry, University of Bonn, Venusberg-Campus, 53127, Bonn, Germany
| | - Bettina Gohlke
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital, University of Bonn, Venusberg-Campus, Building 30, 53127, Bonn, Germany.
| |
Collapse
|
85
|
Fermented Oyster Extract Promotes Insulin-Like Growth Factor-1-Mediated Osteogenesis and Growth Rate. Mar Drugs 2020; 18:md18090472. [PMID: 32962034 PMCID: PMC7551862 DOI: 10.3390/md18090472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Fermented oyster (Crassostrea gigas) extract (FO) prevents ovariectomy-induced osteoporosis by inhibiting osteoclastogenesis and activating osteogenesis. However, the molecular mechanisms underlying FO-mediated bone formation and growth rate are unclear. In the current study, we found that FO significantly upregulated the expression of growth-promoting genes in zebrafish larvae including insulin-like growth factor 1 (zigf-1), insulin-like growth factor binding protein 3 (zigfbp-3), growth hormone-1 (zgh-1), growth hormone receptor-1 (zghr-1), growth hormone receptor alpha (zghra), glucokinase (zgck), and cholecystokinin (zccka). In addition, zebrafish larvae treated with 100 μg/mL FO increased in total body length (3.89 ± 0.13 mm) at 12 days post fertilization (dpf) compared to untreated larvae (3.69 ± 0.02 mm); this effect was comparable to that of the β-glycerophosphate-treated zebrafish larvae (4.00 ± 0.02 mm). Furthermore, FO time- and dose-dependently increased the extracellular release of IGF-1 from preosteoblast MC3T3-E1 cells, which was accompanied by high expression of IGF-1. Pharmacological inhibition of IGF-1 receptor (IGF-1R) using picropodophyllin (PPP) significantly reduced FO-mediated vertebrae formation (from 9.19 ± 0.31 to 5.53 ± 0.35) and growth performance (from 3.91 ± 0.02 to 3.69 ± 0.01 mm) in zebrafish larvae at 9 dpf. Similarly, PPP significantly decreased FO-induced calcium deposition in MC3T3-E1 cells by inhibiting GSK-3β phosphorylation at Ser9. Additionally, DOI hydrochloride, a potent stabilizer of GSK-3β, reduced FO-induced nuclear translocation of RUNX2. Transient knockdown of IGF-1Rα/β using specific silencing RNA also resulted in a significant decrease in calcium deposition and reduction in GSK-3β phosphorylation at Ser9 in MC3T3-E1 cells. Altogether, these results indicate that FO increased phosphorylated GSK-3β at Ser9 by activating the autocrine IGF-1-mediated IGF-1R signaling pathway, thereby promoting osteogenesis and growth performance. Therefore, FO is a potential nutritional supplement for bone formation and growth.
Collapse
|
86
|
Cappa M, Maghnie M, Carbone V, Chioma L, Errichiello C, Giavoli C, Giordano M, Guazzarotti L, Klain A, Montini G, Murer L, Parpagnoli M, Pecoraro C, Pesce S, Verrina E. Summary of Expert Opinion on the Management of Children With Chronic Kidney Disease and Growth Failure With Human Growth Hormone. Front Endocrinol (Lausanne) 2020; 11:587. [PMID: 33013690 PMCID: PMC7493742 DOI: 10.3389/fendo.2020.00587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Background: The management of children and adolescents with chronic kidney disease (CKD) and growth failure candidate for recombinant human growth hormone therapy (rhGH) is based on an appraisal of the literature established on a 2006 consensus statement and 2019 Clinical practice recommendations. The performance of these guidelines has never been tested. Aims: The objective of this study was to establish the level of adherence to international guidelines based on the 2006 consensus and the 2019 criteria that lead to the initiation of growth hormone treatment by both pediatric endocrinologists and pediatric nephrologists. Methods: A multidisciplinary team of pediatric endocrinologists and pediatric nephrologists, members of the Italian Society of Pediatric Endocrinology or of the Italian Society of Pediatric Nephrology, discussed and reviewed the main issues related to the management of pediatric patients with CKD who need treatment with rhGH. Experts developed 11 questions focusing on risk assessment and decision makings in October 2019 and a survey was sent to forty pediatric endocrinologists (n = 20) and nephrologists (n = 20) covering the whole national territory. The results were then analyzed and discussed in light of current clinical practice guidelines and recent recommendations. Results: Responses were received from 32 of the 40 invited specialists, 17 of whom were pediatric endocrinologists (42.5%) and 15 pediatric nephrologists (37.5%). Although all the centers that participated in the survey agreed to follow the clinical and biochemical diagnostic work-up and the criteria for the treatment of patients with CKD, among the Italian centers there was a wide variety of decision-making processes. Conclusions: Despite current guidelines for the management of children with CKD and growth failure, its use varies widely between centers and rhGH is prescribed in a relatively small number of patients and rarely after kidney transplantation. Several raised issues are not taken into account by international guidelines and a multidisciplinary approach with mutual collaboration between specialists will improve patient care based on their unmet needs.
Collapse
Affiliation(s)
- Marco Cappa
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Mohamad Maghnie
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Vincenza Carbone
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital “Giovanni XXIII”, Bari, Italy
| | - Laura Chioma
- Unit of Endocrinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Claudia Giavoli
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Mario Giordano
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital “Giovanni XXIII”, Bari, Italy
| | - Laura Guazzarotti
- Pediatric Endocrinology and Auxology, Adolescence Unit - Department of Women's and Children's Health, Pediatric Department - Padua University Hospital, Padua, Italy
| | - Antonella Klain
- Pediatric Endocrinology Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis, and Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Giuliana and Bernardo Caprotti Chair of Pediatrics, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Luisa Murer
- Pediatric Nephrology, Dialysis and Transplant Unit, Department of Woman's and Child's Health, Azienda Ospedaliera –University of Padova, Padua, Italy
| | - Maria Parpagnoli
- Auxo-Endocrinology and Gynecology Meyer Children's University Hospital, Florence, Italy
| | - Carmine Pecoraro
- Pediatric Nephrology and Dialysis Unit, Santobono-Pausilipon Hospital, Naples, Italy
| | - Sabino Pesce
- Pediatric Endocrinology and Metabolic Diseases, Pediatric Hospital “Giovanni XXIII”, Bari, Italy
| | - Enrico Verrina
- Unit of Dialysis, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
87
|
Kong L, Liu G, Deng M, Lian Z, Han Y, Sun B, Guo Y, Liu D, Li Y. Growth retardation-responsive analysis of mRNAs and long noncoding RNAs in the liver tissue of Leiqiong cattle. Sci Rep 2020; 10:14254. [PMID: 32868811 PMCID: PMC7459292 DOI: 10.1038/s41598-020-71206-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
As an important type of non-coding RNA molecule, long non-coding RNAs (lncRNAs) have varied roles in many biological processes, and have been studied extensively over the past few years. However, little is known about lncRNA-mediated regulation during cattle growth and development. Therefore, in the present study, RNA sequencing was used to determine the expression level of mRNAs and lncRNAs in the liver of adult Leiqiong cattle under the condition of growth retardation and normal growth. We totally detected 1,124 and 24 differentially expressed mRNAs and lncRNAs, respectively. The differentially expressed mRNAs were mainly associated with growth factor binding, protein K63-linked ubiquitination and cellular protein metabolic process; additionally, they were significantly enriched in the growth and development related pathways, including PPAR signaling pathway, vitamin B6 metabolism, glyoxylate and dicarboxylate metabolism. Combined analysis showed that the co-located differentially expressed lncRNA Lnc_002583 might positively influence the expression of the corresponding genes IFI44 and IFI44L, exerting co-regulative effects on Leiqiong cattle growth and development. Thus, we made the hypothesis that Lnc_002583, IFI44 and IFI44L might function synergistically to regulate the growth of Leiqiong cattle. This study provides a catalog of Leiqiong cattle liver mRNAs and lncRNAs, and will contribute to a better understanding of the molecular mechanism underlying growth regulataion.
Collapse
Affiliation(s)
- Lingxuan Kong
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Guangbin Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Ming Deng
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Zhiquan Lian
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Yinru Han
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Baoli Sun
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Yongqing Guo
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China
| | - Dewu Liu
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China.
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China.
| | - Yaokun Li
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, GD, China.
- National Local Joint Engineering Research Center of Livestock and Poutry, South China Agricultural University, Guangzhou, 510642, GD, China.
| |
Collapse
|
88
|
Bøllehuus Hansen L, Kaludjerovic J, Nielsen JE, Rehfeld A, Poulsen NN, Ide N, Skakkebaek NE, Frederiksen H, Juul A, Lanske B, Blomberg Jensen M. Influence of FGF23 and Klotho on male reproduction: Systemic vs direct effects. FASEB J 2020; 34:12436-12449. [PMID: 32729975 DOI: 10.1096/fj.202000061rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 01/05/2023]
Abstract
Currently, no treatment exists to improve semen quality in most infertile men. Here, we demonstrate systemic and direct effects of Fibroblast growth factor 23 (FGF23) and Klotho, which normally regulate vitamin D and mineral homeostasis, on testicular function. Direct effects are plausible because KLOTHO is expressed in both germ cells and spermatozoa and forms with FGFR1 a specific receptor for the bone-derived hormone FGF23. Treatment with FGF23 increased testicular weight in wild-type mice, while mice with global loss of either FGF23 or Klotho had low testicular weight, reduced sperm count, and sperm motility. Mice with germ cell-specific Klotho (gcKL) deficiency neither had a change in sperm count nor sperm motility. However, a tendency toward fewer pregnancies was detected, and significantly fewer Klotho heterozygous pups originated from gcKL knockdown mice than would be expected by mendelian inheritance. Moreover, gcKL mice had a molecular phenotype with higher testicular expression of Slc34a2 and Trpv5 than wild-type littermates, which suggests a regulatory role for testicular phosphate and calcium homeostasis. KLOTHO and FGFR1 were also expressed in human germ cells and spermatozoa, and FGF23 treatment augmented the calcium response to progesterone in human spermatozoa. Moreover, cross-sectional data revealed that infertile men with the highest serum Klotho levels had significantly higher serum Inhibin B and total sperm count than men with the lowest serum Klotho concentrations. In conclusion, this translational study suggests that FGF23 and Klotho influence gonadal function and testicular mineral ion homeostasis both directly and indirectly through systemic changes in vitamin D and mineral homeostasis.
Collapse
Affiliation(s)
- Lasse Bøllehuus Hansen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard Medical School, Boston, MA, USA
| | - Jovana Kaludjerovic
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard Medical School, Boston, MA, USA
| | - John Erik Nielsen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Rehfeld
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nadia Nicholine Poulsen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Noriko Ide
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard Medical School, Boston, MA, USA
| | - Niels Erik Skakkebaek
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Beate Lanske
- Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard Medical School, Boston, MA, USA
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Division of Bone and Mineral Research, Harvard School of Dental Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
89
|
Gawne R, McKenna KZ, Levin M. Competitive and Coordinative Interactions between Body Parts Produce Adaptive Developmental Outcomes. Bioessays 2020; 42:e1900245. [DOI: 10.1002/bies.201900245] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Richard Gawne
- Allen Discovery Center at Tufts University Medford MA 02155
| | - Kenneth Z. McKenna
- Division of Biological SciencesSection of Cellular and Developmental BiologyUniversity of California San Diego La Jolla CA 92093
| | - Michael Levin
- Allen Discovery Center at Tufts University Medford MA 02155
| |
Collapse
|
90
|
Fernández-Iglesias Á, Fuente R, Gil-Peña H, Alonso-Durán L, García-Bengoa M, Santos F, López JM. Innovative Three-Dimensional Microscopic Analysis of Uremic Growth Plate Discloses Alterations in the Process of Chondrocyte Hypertrophy: Effects of Growth Hormone Treatment. Int J Mol Sci 2020; 21:ijms21124519. [PMID: 32630463 PMCID: PMC7350242 DOI: 10.3390/ijms21124519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic kidney disease (CKD) alters the morphology and function of the growth plate (GP) of long bones by disturbing chondrocyte maturation. GP chondrocytes were analyzed in growth-retarded young rats with CKD induced by adenine intake (AD), control rats fed ad libitum (C) or pair-fed with the AD group (PF), and CKD rats treated with growth hormone (ADGH). In order to study the alterations in the process of GP maturation, we applied a procedure recently described by our group to obtain high-quality three-dimensional images of whole chondrocytes that can be used to analyze quantitative parameters like cytoplasm density, cell volume, and shape. The final chondrocyte volume was found to be decreased in AD rats, but GH treatment was able to normalize it. The pattern of variation in the cell cytoplasm density suggests that uremia could be causing a delay to the beginning of the chondrocyte hypertrophy process. Growth hormone treatment appears to be able to compensate for this disturbance by triggering an early chondrocyte enlargement that may be mediated by Nkcc1 action, an important membrane cotransporter in the GP chondrocyte enlargement.
Collapse
Affiliation(s)
- Ángela Fernández-Iglesias
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
- Instituto de Investigación sanitaria del Principado de Asturias (ISPA), 33012 Oviedo, Spain
| | - Rocío Fuente
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
| | - Helena Gil-Peña
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
- Instituto de Investigación sanitaria del Principado de Asturias (ISPA), 33012 Oviedo, Spain
- Department of Pediatrics, Hospital Universitario Central de Asturias (HUCA), 33013 Oviedo, Asturias, Spain
| | - Laura Alonso-Durán
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
- Instituto de Investigación sanitaria del Principado de Asturias (ISPA), 33012 Oviedo, Spain
| | - María García-Bengoa
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
| | - Fernando Santos
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
- Instituto de Investigación sanitaria del Principado de Asturias (ISPA), 33012 Oviedo, Spain
- Department of Pediatrics, Hospital Universitario Central de Asturias (HUCA), 33013 Oviedo, Asturias, Spain
- Correspondence: ; Tel.: +34-985102728
| | - José Manuel López
- Division of Pediatrics, Department of Medicine, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain; (A.F.-I.); (R.F.); (H.G.-P.); (L.A.-D.); (M.G.-B.); (J.M.L.)
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, CP 33006 Oviedo, Asturias, Spain
| |
Collapse
|
91
|
Walz M, Chau L, Walz C, Sawitzky M, Ohde D, Brenmoehl J, Tuchscherer A, Langhammer M, Metzger F, Höflich C, Hoeflich A. Overlap of Peak Growth Activity and Peak IGF-1 to IGFBP Ratio: Delayed Increase of IGFBPs versus IGF-1 in Serum as a Mechanism to Speed up and down Postnatal Weight Gain in Mice. Cells 2020; 9:cells9061516. [PMID: 32580353 PMCID: PMC7348928 DOI: 10.3390/cells9061516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/02/2020] [Accepted: 06/17/2020] [Indexed: 11/16/2022] Open
Abstract
Forced expression of insulin-like growth factor binding proteins (IGFBPs) in transgenic mice has clearly revealed inhibitory effects on somatic growth. However, by this approach, it cannot be solved if or how IGFBPs rule insulin-like growth factor (IGF)-dependent growth under normal conditions. In order to address this question, we have used growth-selected mouse models (obese and lean) and studied IGF-1 and IGFBPs in serum with respect to longitudinal growth activity in males and females compared with unselected controls. In mice of both genders, body weights were recorded and daily weight gains were calculated. Between 2 and 54 weeks of age, serum IGF-1 was determined by ELISA and intact IGFBP-2, -3 and -4 were quantified by Western ligand blotting. The molar ratio of IGF-1 to the sum of IGFBP-2 to -4 was calculated for all groups and plotted against the daily weight gain curve. Growth-selected mice are characterized by higher daily weight gains and extended periods of elevated growth activity if compared to matched unselected controls. Therefore, adult mice from the obese and lean groups can achieve more than twofold increased body weight in both genders (p < 0.001). Between 2 and 11 weeks of age, in obese and lean mice of both genders, serum IGF-1 concentrations are increased more prominently if compared to unselected controls (p < 0.001). Instead, substantial decreases of IGFBPs, particularly of IGFBP-2, are observed in males and females of all groups at the age of 2 to 4 weeks (p < 0.001). Due to the strong increase of IGF-1 but not of IGFBPs between two and four weeks of age, the ratio of IGF-1 to IGFBP-2 to -4 in serum significantly increased in all groups and genders (p < 0.05). Notably, the IGF-1 to IGFBP ratio was higher in male and female obese mice if compared to unselected controls (p < 0.05).
Collapse
Affiliation(s)
- Michael Walz
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Luong Chau
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Christina Walz
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Mandy Sawitzky
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Daniela Ohde
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Julia Brenmoehl
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Armin Tuchscherer
- Institute of Genetics and Biometry, Leibniz-Institute for Farm Animal Biology (FBN), 18197 Dummerstorf, Germany; (A.T.); (M.L.)
| | - Martina Langhammer
- Institute of Genetics and Biometry, Leibniz-Institute for Farm Animal Biology (FBN), 18197 Dummerstorf, Germany; (A.T.); (M.L.)
| | | | | | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
- Correspondence: ; Tel.: +49-(0)38208-68744; Fax: +49-(0)38208-68-702
| |
Collapse
|
92
|
Bright GM, Fierro-Renoy JF. A rationale for the treatment of short stature in children with the combination of recombinant human growth hormone (rhGH) and recombinant human insulin-like growth factor-I (rhIGF-I). Growth Horm IGF Res 2020; 52:101318. [PMID: 32252003 DOI: 10.1016/j.ghir.2020.101318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Both rhGH and rhIGF-I are signaling molecules with the capacity to restore the rate of growth in certain subsets of slowly growing children. In some instances, heights attained at or near the time of cessation of linear growth are indistinguishable from the height distribution of the community as a whole or from the height distribution expected based on the heights of biological parents. The GH: IGF-I signaling system is sequential, forming a continuous loop wherein GH will stimulate production of IGF-I and IGF-I will inhibit production of GH. This feature suggests that a deficiency of GH will be accompanied by a deficiency of IGF-I and that treatment of GH deficiency with rhGH will restore IGF-I and the subnormal growth of combined GH: IGF-I deficiency. Although logical, this proposition is not always true. rhGH and rhIGF-I are distinct polypeptides, with distinct cell surface receptors and distinct intracellular signaling pathways both capable of amplifying distinct, yet overlapping, patterns of gene replication, protein synthesis and metabolic activities. These features suggest that neither treatment with rhGH nor rhIGF-I alone will invariably recapitulate the combined activities of the GH: IGF-I system, At the present time, this proposition appears both logical and true. The possibility that combined rhGH and rhIGF-I treatment can accomplish that which neither monotherapy can has been examined in gene knock-out experiments in animals and direct comparisons of GH, IGF-I and combined GH: IGF- treatments in animals and in children with short stature, normal GH and low IGF-I (primary IGF-I deficiency). In these experimental models, the growth rates with combined rhGH and rhIGF-I treatment exceed those of either monotherapy. The extent to which this proposition can be generalized to various short stature populations remains to be determined.
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW Bone elongation is a complex process driven by multiple intrinsic (hormones, growth factors) and extrinsic (nutrition, environment) variables. Bones grow in length by endochondral ossification in cartilaginous growth plates at ends of developing long bones. This review provides an updated overview of the important factors that influence this process. RECENT FINDINGS Insulin-like growth factor-1 (IGF-1) is the major hormone required for growth and a drug for treating pediatric skeletal disorders. Temperature is an underrecognized environmental variable that also impacts linear growth. This paper reviews the current state of knowledge regarding the interaction of IGF-1 and environmental factors on bone elongation. Understanding how internal and external variables regulate bone lengthening is essential for developing and improving treatments for an array of bone elongation disorders. Future studies may benefit from understanding how these unique relationships could offer realistic new approaches for increasing bone length in different growth-limiting conditions.
Collapse
Affiliation(s)
- Holly L Racine
- Department of Natural Sciences and Mathematics, West Liberty University, West Liberty, WV, 26074, USA
| | - Maria A Serrat
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
- Department of Orthopaedics, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
94
|
Cheng Y, Chen T, Song J, Qi Q, Wang C, Xi Q, Liu S, Hao L, Zhang Y. miR-709 inhibits GHRP6 induced GH synthesis by targeting PRKCA in pituitary. Mol Cell Endocrinol 2020; 506:110763. [PMID: 32084499 DOI: 10.1016/j.mce.2020.110763] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Pituitary growth hormone (GH) plays an essential role in processes of organism growth and metabolism. MicroRNA (miRNA) could also participate in diverse biological processes. However, the role of miRNA in the regulation of pituitary GH during the growth process remains unclear. In this study, we firstly confirmed that the second highly expressed pituitary miRNA (miR-709) significantly inhibited the GH synthesis and suppressed the viability of GH3 cells. The bioinformatics analysis and dual luciferase report system were used to ascertain the PRKCA is the direct target gene of miR-709, which is the coding gene of PKCα. Then the transcription and translation levels of Prkca were obvious reduced by the over-expression of miR-709 in GH3 cells, followed by the inhibition of the transcription factor (CREB1) of Gh1 gene and the ERK1/2 signaling pathway or the possible cross-talk signaling pathway (cAMP/PKA signaling pathway) detected by western blot, suggesting that ERK1/2 maybe an important factor involved in the GH3 cell viability mediated by PKCα. At last, GHRP6 increased PKCα and GH expression but reduced miR-709 expression in vitro and vivo assays, and this conclusion was further confirmed by the result of GHRP6 attenuated the inhibition of miR-709 on GH expression. These findings will provide new molecular mechanism on the regulation of pituitary GH.
Collapse
Affiliation(s)
- Yunyun Cheng
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Jie Song
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University, Foshan, 528231, China
| | - Chunli Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China
| | - Songcai Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China
| | - Linlin Hao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xian Road, Changchun, 130062, China.
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, China.
| |
Collapse
|
95
|
Ok SM, Kim JH, Kim JS, Jeong EG, Park YM, Jeon HM, Heo JY, Ahn YW, Yu SN, Park HR, Kim KH, Ahn SC, Jeong SH. Local Injection of Growth Hormone for Temporomandibular Joint Osteoarthritis. Yonsei Med J 2020; 61:331-340. [PMID: 32233176 PMCID: PMC7105408 DOI: 10.3349/ymj.2020.61.4.331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/09/2020] [Accepted: 02/18/2020] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Osteoarthritis (OA) of the temporomandibular joint (TMJ) elicits cartilage and subchondral bone defects. Growth hormone (GH) promotes chondrocyte growth. The aim of this study was to evaluate the efficacy of intra-articular injections of GH to treat TMJ-OA. MATERIALS AND METHODS Monosodium iodoacetate (MIA) was used to induce OA in the TMJs of rats. After confirming the induction of OA, recombinant human GH was injected into the articular cavities of rats. Concentrations of GH and IGF-1 were measured in the blood and synovial fluid, and OA grades of cartilage and subchondral bone degradation were recorded by histological examination and micro-computed tomography. RESULTS MIA-induced OA in the rat TMJ upregulated insulin-like growth factor-1 (IGF-1) rather than GH levels. GH and IGF-1 concentrations were increased after local injection of GH, compared with controls. Locally injected GH lowered osteoarthritic scores in the cartilage and subchondral bone of the TMJ. CONCLUSION Intra-articular injection of GH improved OA scores in rat TMJs in both cartilage and subchondral bone of the condyles without affecting condylar bone growth. These results suggest that intra-articular injection of human GH could be a suitable treatment option for TMJ-OA patients in the future.
Collapse
Affiliation(s)
- Soo Min Ok
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, Korea
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Jin Hwa Kim
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Ji Su Kim
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Eun Gyo Jeong
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Yang Mi Park
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Hye Mi Jeon
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Jun Young Heo
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, Korea
| | - Yong Woo Ahn
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, Korea
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea
| | - Sun Nyoung Yu
- Department of Microbiology & Immunology, Pusan National University School of Medicine, Yangsan, Korea
| | - Hae Ryoun Park
- Department of Oral Pathology, Pusan National University School of Dentistry, Yangsan, Korea
| | - Kyung Hee Kim
- Deptartment of Oral Medicine, Busan Paik Hospital, Inje University, Busan, Korea
| | - Soon Cheol Ahn
- Department of Microbiology & Immunology, Pusan National University School of Medicine, Yangsan, Korea
| | - Sung Hee Jeong
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, Korea
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, Korea.
| |
Collapse
|
96
|
Kassem N, Kassem MM, Pedersen SF, Pedersen PA, Kragelund BB. Yeast recombinant production of intact human membrane proteins with long intrinsically disordered intracellular regions for structural studies. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183272. [PMID: 32169592 DOI: 10.1016/j.bbamem.2020.183272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/08/2020] [Accepted: 03/09/2020] [Indexed: 01/07/2023]
Abstract
Membrane proteins exist in lipid bilayers and mediate solute transport, signal transduction, cell-cell communication and energy conversion. Their activities are fundamental for life, which make them prominent subjects of study, but access to only a limited number of high-resolution structures complicates their mechanistic understanding. The absence of such structures relates mainly to difficulties in expressing and purifying high quality membrane protein samples in large quantities. An additional layer of complexity stems from the presence of intra- and/or extra-cellular domains constituted by unstructured intrinsically disordered regions (IDR), which can be hundreds of residues long. Although IDRs form key interaction hubs that facilitate biological processes, these are regularly removed to enable structural studies. To advance mechanistic insight into intact intrinsically disordered membrane proteins, we have developed a protocol for their purification. Using engineered yeast cells for optimized expression and purification, we have purified to homogeneity two very different human membrane proteins each with >300 residues long IDRs; the sodium proton exchanger 1 and the growth hormone receptor. Subsequent to their purification we have further explored their incorporation into membrane scaffolding protein nanodiscs, which will enable future structural studies.
Collapse
Affiliation(s)
- Noah Kassem
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark
| | - Maher M Kassem
- Machine Learning, Department of Computer Science, University of Copenhagen, Universitetsparken 5, DK-2100, Copenhagen Ø, Denmark
| | - Stine F Pedersen
- Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark
| | - Per Amstrup Pedersen
- Cell Biology and Physiology, Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark.
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
97
|
Heallen TR, Kadow ZA, Wang J, Martin JF. Determinants of Cardiac Growth and Size. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037150. [PMID: 31615785 DOI: 10.1101/cshperspect.a037150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Within the realm of zoological study, the question of how an organism reaches a specific size has been largely unexplored. Recently, studies performed to understand the regulation of organ size have revealed that both cellular signals and external cues contribute toward the determination of total cell mass within each organ. The establishment of final organ size requires the precise coordination of cell growth, proliferation, and survival throughout development and postnatal life. In the mammalian heart, the regulation of size is biphasic. During development, cardiomyocyte proliferation predominantly determines cardiac growth, whereas in the adult heart, total cell mass is governed by signals that regulate cardiac hypertrophy. Here, we review the current state of knowledge regarding the extrinsic factors and intrinsic mechanisms that control heart size during development. We also discuss the metabolic switch that occurs in the heart after birth and precedes homeostatic control of postnatal heart size.
Collapse
Affiliation(s)
- Todd R Heallen
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, Texas 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Zachary A Kadow
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - James F Martin
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, Texas 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
98
|
Kopchick JJ, Berryman DE, Puri V, Lee KY, Jorgensen JOL. The effects of growth hormone on adipose tissue: old observations, new mechanisms. Nat Rev Endocrinol 2020; 16:135-146. [PMID: 31780780 PMCID: PMC7180987 DOI: 10.1038/s41574-019-0280-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2019] [Indexed: 12/18/2022]
Abstract
The ability of growth hormone (GH) to induce adipose tissue lipolysis has been known for over five decades; however, the molecular mechanisms that mediate this effect and the ability of GH to inhibit insulin-stimulated glucose uptake have scarcely been documented. In this same time frame, our understanding of adipose tissue has evolved to reveal a complex structure with distinct types of adipocyte, depot-specific differences, a biologically significant extracellular matrix and important endocrine properties mediated by adipokines. All these aforementioned features, in turn, can influence lipolysis. In this Review, we provide a historical and current overview of the lipolytic effect of GH in humans, mice and cultured cells. More globally, we explain lipolysis in terms of GH-induced intracellular signalling and its effect on obesity, insulin resistance and lipotoxicity. In this regard, findings that define molecular mechanisms by which GH induces lipolysis are described. Finally, data are presented for the differential effect of GH on specific adipose tissue depots and on distinct classes of metabolically active adipocytes. Together, these cellular, animal and human studies reveal novel cellular phenotypes and molecular pathways regulating the metabolic effects of GH on adipose tissue.
Collapse
Affiliation(s)
- John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA.
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Vishwajeet Puri
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Kevin Y Lee
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Diabetes, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
99
|
Petersen M, Gandhi PS, Buchardt J, Alanentalo T, Fels JJ, Johansen NL, Helding-Kvist P, Vad K, Thygesen P. Tissue Distribution and Receptor Activation by Somapacitan, a Long Acting Growth Hormone Derivative. Int J Mol Sci 2020; 21:ijms21041181. [PMID: 32053994 PMCID: PMC7072805 DOI: 10.3390/ijms21041181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 11/16/2022] Open
Abstract
Somapacitan is a long-acting, once-weekly, albumin-binding growth hormone (GH) derivative. The reversible albumin-binding properties leads to prolonged circulation half-life. Here, we investigated and compared somapacitan with human GH on downstream receptor signaling in primary hepatocytes and hepatocellular models and using isothermal titration calorimetry to characterize receptor binding of somapacitan in the presence or absence of human serum albumin (HSA). With non-invasive fluorescence imaging we quantitatively visualize and compare the temporal distribution and examine the tissue-specific growth hormone receptor (GHR) activation at distribution sites. We found that signaling kinetics were slightly more rapid and intense for GH compared with somapacitan. Receptor binding isotherms were characterized by a high and a low affinity interaction site with or without HSA. Using in vivo optical imaging we found prolonged systemically biodistribution of somapacitan compared with GH, which correlated with plasma pharmacokinetics. Ex vivo mouse organ analysis revealed that the temporal fluorescent intensity in livers dosed with somapacitan was significantly increased compared with GH-dosed livers and correlated with the degree of downstream GHR activation. Finally, we show that fluorescent-labeled analogs distributed to the hypertrophic zone in the epiphysis of proximal tibia of hypophysectomized rats and that somapacitan and GH activate the GHR signaling in epiphyseal tissues.
Collapse
Affiliation(s)
- Maj Petersen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Jens Buchardt
- Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Tomas Alanentalo
- Umeå Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | | | | | | | - Knud Vad
- Global Development, Novo Nordisk A/S, 2860 Søborg, Denmark
| | - Peter Thygesen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
- Correspondence: ; Tel.: +45-3075-4617
| |
Collapse
|
100
|
Abstract
Post-natal bone development is characterized by substantial longitudinal bone growth and changes in skeletal size and shape. Bone is in a dynamic process of continuous remodeling which helps to regulate calcium homeostasis, repair micro-damage to bones from everyday stress, and to shape the skeleton during growth. Bone growth is regulated by systemic hormones and locally generated factors. Understanding their mechanisms of action enables us to obtain a better appreciation of the cellular and molecular basis of bone remodeling and could therefore be valuable in approaches to new therapies. This article will review molecular and cellular control of skeletal growth in the post-natal period, the physiology of each bone cell with their systemic and local regulators, as well as the physiology of bone remodeling.
Collapse
Affiliation(s)
- Rania Ali El-Farrash
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Radwa Hassan Ali
- Physiology Department, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Noha Mokhtar Barakat
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| |
Collapse
|