1
|
Zhang W, Dao JJ, Li Q, Liu C, Qiao CM, Cui C, Shen YQ, Zhao WJ. Neuregulin 1 mitigated prolactin deficiency through enhancing TRPM8 signaling under the influence of melatonin in senescent pituitary lactotrophs. Int J Biol Macromol 2024; 275:133659. [PMID: 38969045 DOI: 10.1016/j.ijbiomac.2024.133659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The age-related alterations in pituitary function, including changes in prolactin (PRL) production contributes to the systemic susceptibility to age-related diseases. Our previous research has shown the involvement of Nrg1 in regulating the expression and secretion of PRL. However, the precise role of Nrg1 in mitigating the senescence of pituitary lactotrophs and the underlying mechanisms are yet to be comprehended. Here, data from the GEPIA database was used to evaluate the association between transient receptor potential cation channel subfamily M member 8 (TRPM8) and PRL in normal human pituitary tissues, followed by immunofluorescence verification using a human pituitary tissue microarray. TRPM8 levels showed a significant positive association with PRL expression in normal human pituitary tissues, and both TRPM8 and PRL levels declined during aging, suggesting that TRPM8 may regulate pituitary aging by affecting PRL production. It was also found that treatment with exogenous neuregulin 1 (Nrg1) markedly delayed the senescence of GH3 cells (rat lactotroph cell line) generated by D-galactose (D-gal). In addition, melatonin reduced the levels of senescence-related markers in senescent pituitary cells by promoting Nrg1 / ErbB4 signaling, stimulating PRL expression and secretion. Further investigation showed that Nrg1 attenuated senescence in pituitary cells by increasing TRPM8 expression. Downregulation of TRPM8 activation eliminated Nrg1-mediated amelioration of pituitary cell senescence. These findings demonstrate the critical function of Nrg1 / ErbB signaling in delaying pituitary lactotroph cell senescence and enhancing PRL production via promoting TRPM8 expression under the modulation of melatonin.
Collapse
Affiliation(s)
- Wei Zhang
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China; Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang 550000, Guizhou, China
| | - Ji-Ji Dao
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Qian Li
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chong Liu
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chen-Meng Qiao
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Chun Cui
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Yan-Qin Shen
- Department of Neurodegeneration and Neuroinjury, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Wei-Jiang Zhao
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, Jiangsu, China.
| |
Collapse
|
2
|
Rohayem J, Alexander EC, Heger S, Nordenström A, Howard SR. Mini-Puberty, Physiological and Disordered: Consequences, and Potential for Therapeutic Replacement. Endocr Rev 2024; 45:460-492. [PMID: 38436980 PMCID: PMC11244267 DOI: 10.1210/endrev/bnae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Indexed: 03/05/2024]
Abstract
There are 3 physiological waves of central hypothalamic-pituitary-gonadal (HPG) axis activity over the lifetime. The first occurs during fetal life, the second-termed "mini-puberty"-in the first months after birth, and the third at puberty. After adolescence, the axis remains active all through adulthood. Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by a deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) secretion or action. In cases of severe CHH, all 3 waves of GnRH pulsatility are absent. The absence of fetal HPG axis activation manifests in around 50% of male newborns with micropenis and/or undescended testes (cryptorchidism). In these boys, the lack of the mini-puberty phase accentuates testicular immaturity. This is characterized by a low number of Sertoli cells, which are important for future reproductive capacity. Thus, absent mini-puberty will have detrimental effects on later fertility in these males. The diagnosis of CHH is often missed in infants, and even if recognized, there is no consensus on optimal therapeutic management. Here we review physiological mini-puberty and consequences of central HPG axis disorders; provide a diagnostic approach to allow for early identification of these conditions; and review current treatment options for replacement of mini-puberty in male infants with CHH. There is evidence from small case series that replacement with gonadotropins to mimic "mini-puberty" in males could have beneficial outcomes not only regarding testis descent, but also normalization of testis and penile sizes. Moreover, such therapeutic replacement regimens in disordered mini-puberty could address both reproductive and nonreproductive implications.
Collapse
Affiliation(s)
- Julia Rohayem
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital of Eastern Switzerland, 9006 St. Gallen, Switzerland
- University of Muenster, 48149 Muenster, Germany
| | - Emma C Alexander
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sabine Heger
- Department of Pediatric Endocrinology, Children's Hospital Auf der Bult, 30173 Hannover, Germany
| | - Anna Nordenström
- Pediatric Endocrinology, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- Department of Paediatric Endocrinology, Royal London Children's Hospital, Barts Health NHS Trust, London E1 1FR, UK
| |
Collapse
|
3
|
Szabó F, Köves K, Gál L. History of the Development of Knowledge about the Neuroendocrine Control of Ovulation-Recent Knowledge on the Molecular Background. Int J Mol Sci 2024; 25:6531. [PMID: 38928237 PMCID: PMC11203711 DOI: 10.3390/ijms25126531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The physiology of reproduction has been of interest to researchers for centuries. The purpose of this work is to review the development of our knowledge on the neuroendocrine background of the regulation of ovulation. We first describe the development of the pituitary gland, the structure of the median eminence (ME), the connection between the hypothalamus and the pituitary gland, the ovarian and pituitary hormones involved in ovulation, and the pituitary cell composition. We recall the pioneer physiological and morphological investigations that drove development forward. The description of the supraoptic-paraventricular magnocellular and tuberoinfundibular parvocellular systems and recognizing the role of the hypophysiotropic area were major milestones in understanding the anatomical and physiological basis of reproduction. The discovery of releasing and inhibiting hormones, the significance of pulse and surge generators, the pulsatile secretion of the gonadotropin-releasing hormone (GnRH), and the subsequent pulsatility of luteinizing (LH) and follicle-stimulating hormones (FSH) in the human reproductive physiology were truly transformative. The roles of three critical neuropeptides, kisspeptin (KP), neurokinin B (NKB), and dynorphin (Dy), were also identified. This review also touches on the endocrine background of human infertility and assisted fertilization.
Collapse
Affiliation(s)
- Flóra Szabó
- Division of Gastroenterology and Nutrition, Children’s Hospital of Richmond, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Katalin Köves
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
| | - Levente Gál
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA;
| |
Collapse
|
4
|
Martinez-Mayer J, Brinkmeier ML, O'Connell SP, Ukagwu A, Marti MA, Miras M, Forclaz MV, Benzrihen MG, Cheung LYM, Camper SA, Ellsworth BS, Raetzman LT, Pérez-Millán MI, Davis SW. Knockout mice with pituitary malformations help identify human cases of hypopituitarism. Genome Med 2024; 16:75. [PMID: 38822427 PMCID: PMC11140907 DOI: 10.1186/s13073-024-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.
Collapse
Affiliation(s)
- Julian Martinez-Mayer
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Sean P O'Connell
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA
| | - Arnold Ukagwu
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Marcelo A Marti
- Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mirta Miras
- Hospital De Niños de La Santísima Trinidad, Córdoba, Argentina
| | - Maria V Forclaz
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Maria G Benzrihen
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
- Department of Physiology and Biophyscis, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois, Champaign-Urbana, Urbana, IL, 61801, USA
| | - Maria I Pérez-Millán
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina.
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA.
| |
Collapse
|
5
|
Banik J, Moreira ARS, Lim J, Tomlinson S, Hardy LL, Lagasse A, Haney A, Crimmins MR, Boehm U, Odle AK, MacNicol MC, Childs GV, MacNicol AM. The Musashi RNA binding proteins direct the translational activation of key pituitary mRNAs. Sci Rep 2024; 14:5918. [PMID: 38467682 PMCID: PMC10928108 DOI: 10.1038/s41598-024-56002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
The pituitary functions as a master endocrine gland that secretes hormones critical for regulation of a wide variety of physiological processes including reproduction, growth, metabolism and stress responses. The distinct hormone-producing cell lineages within the pituitary display remarkable levels of cell plasticity that allow remodeling of the relative proportions of each hormone-producing cell population to meet organismal demands. The molecular mechanisms governing pituitary cell plasticity have not been fully elucidated. Our recent studies have implicated a role for the Musashi family of sequence-specific mRNA binding proteins in the control of pituitary hormone production, pituitary responses to hypothalamic stimulation and modulation of pituitary transcription factor expression in response to leptin signaling. To date, these actions of Musashi in the pituitary appear to be mediated through translational repression of the target mRNAs. Here, we report Musashi1 directs the translational activation, rather than repression, of the Prop1, Gata2 and Nr5a1 mRNAs which encode key pituitary lineage specification factors. We observe that Musashi1 further directs the translational activation of the mRNA encoding the glycolipid Neuronatin (Nnat) as determined both in mRNA reporter assays as well as in vivo. Our findings suggest a complex bifunctional role for Musashi1 in the control of pituitary cell function.
Collapse
Affiliation(s)
- Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Sophia Tomlinson
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Meghan R Crimmins
- Arkansas Children's Nutrition Center, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA.
| |
Collapse
|
6
|
Furtado AP, DeCourcey M, Miller MA, Chen AV, Martin LG, Guess SC, Wardrop KJ, de Souza C, Owen TJ. Cytologic features of canine melanotroph and corticotroph pituitary adenomas. Vet Clin Pathol 2024; 53:99-103. [PMID: 38238980 DOI: 10.1111/vcp.13311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 03/14/2024]
Abstract
BACKGROUND The introduction of intraoperative cytology revolutionized neurosurgical procedures in human medicine, providing real-time diagnostic guidance to surgeons and contributing to improved patient outcomes. In the realm of veterinary medicine, the understanding of pituitary tumors in dogs and cats remains limited due to challenges in obtaining antemortem samples of central nervous system lesions. OBJECTIVES The aim of this study was to describe the cytologic features of pituitary adenomas in 12 dogs that underwent hypophysectomy. METHODS The series included nine melanotroph adenomas and three corticotroph adenomas. Definitive diagnosis was based on histopathology and immunohistochemistry. RESULTS Cytologically, the adenomas had high numbers of bare nuclei and intact cells that were round to polygonal and situated individually or in small clusters. The intact cells had round to oval, eccentric nuclei with finely stippled chromatin and one to three prominent nucleoli and ample to abundant lightly basophilic to amphophilic, grainy cytoplasm with distinct borders, and variable numbers of discrete vacuoles. Mild-to-moderate anisocytosis and anisokaryosis, occasional binucleation, rare and atypical mitotic figures, and nuclear molding were also observed. CONCLUSIONS The results suggest that intraoperative cytology of canine pituitary adenomas holds promise as a valuable diagnostic tool, aiding swift differentiation from other sellar masses before histologic confirmation. Cytologic characterization of pituitary adenomas in dogs is exceptionally rare in the scientific literature, making this study one of the first to offer a comprehensive description of these cytologic features.
Collapse
Affiliation(s)
- Adriana P Furtado
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| | - Michelle DeCourcey
- College of Veterinary Medicine, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Margaret A Miller
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Annie V Chen
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| | - Linda G Martin
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| | - Sarah C Guess
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| | - K Jane Wardrop
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| | - Cleverson de Souza
- Department of Comparative, Diagnostic and Population Medicine, University of Florida, Gainesville, Florida, USA
| | - Tina J Owen
- Veterinary Teaching Hospital, Washington State University, Pullman, Washington, USA
| |
Collapse
|
7
|
Wang S, Qin Q, Jiang D, Xiao Y, Ye L, Jiang X, Guo Q. Re-analysis of gene mutations found in pituitary stalk interruption syndrome and a new hypothesis on the etiology. Front Endocrinol (Lausanne) 2024; 15:1338781. [PMID: 38464967 PMCID: PMC10920343 DOI: 10.3389/fendo.2024.1338781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/09/2024] [Indexed: 03/12/2024] Open
Abstract
Background Pituitary stalk interruption syndrome (PSIS) is a complex clinical syndrome characterized by varied pituitary hormone deficiencies, leading to severe manifestations across multiple systems. These include lifelong infertility, short stature, mental retardation, and potentially life-threatening pituitary crises if not promptly diagnosed and treated. Despite extensive research, the precise pathogenesis of PSIS remains unclear. Currently, there are two proposed theories regarding the pathogenic mechanisms: the genetic defect theory and the perinatal injury theory. Methods We systematically searched English databases (PubMed, Web of Science, Embase) and Chinese databases (CNKI, WanFang Med Online, Sinomed) up to February 24, 2023, to summarize studies on gene sequencing in PSIS patients. Enrichment analyses of reported mutated genes were subsequently performed using the Metascape platform. Results Our study included 37 articles. KEGG enrichment analysis revealed mutated genes were enriched in the Notch signaling pathway, Wnt signaling pathway, and Hedgehog signaling pathway. GO enrichment analysis demonstrated mutated genes were enriched in biological processes such as embryonic development, brain development, axon development and guidance, and development of other organs. Conclusion Based on our summary and analyses, we propose a new hypothesis: disruptions in normal embryonic development, partially stemming from the genetic background and/or specific gene mutations in individuals, may increase the likelihood of abnormal fetal deliveries, where different degrees of traction during delivery may lead to different levels of pituitary stalk interruption and posterior lobe ectopia. The clinical diversity observed in PSIS patients may result from a combination of genetic background, specific mutations, and variable degrees of traction during delivery.
Collapse
Affiliation(s)
- Shengjie Wang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Deyue Jiang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Xiao
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lingtong Ye
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Qinghua Guo
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Cho YW, Fu Y, Huang CCJ, Wu X, Ng L, Kelley KA, Vella KR, Berg AH, Hollenberg AN, Liu H, Forrest D. Thyroid hormone-regulated chromatin landscape and transcriptional sensitivity of the pituitary gland. Commun Biol 2023; 6:1253. [PMID: 38081939 PMCID: PMC10713718 DOI: 10.1038/s42003-023-05546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor β (TRβ) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRβ binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRβ-deficiency blunts T3-regulated gene expression, indicating that TRβ confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.
Collapse
Affiliation(s)
- Young-Wook Cho
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yulong Fu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chen-Che Jeff Huang
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuefeng Wu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lily Ng
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kevin A Kelley
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen R Vella
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Anders H Berg
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, California, 90048, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Weill Department of Medicine Weill Cornell Medicine, New York, New York, 10065, USA
| | - Hong Liu
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Douglas Forrest
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
9
|
Chen Q, Leshkowitz D, Li H, van Impel A, Schulte-Merker S, Amit I, Rizzoti K, Levkowitz G. Neural plate progenitors give rise to both anterior and posterior pituitary cells. Dev Cell 2023; 58:2652-2665.e6. [PMID: 37683631 DOI: 10.1016/j.devcel.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1-positive astroglial pituicytes of the neurohypophysis and prop1-positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3+) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells.
Collapse
Affiliation(s)
- Qiyu Chen
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Hanjie Li
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Present address: CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, UK
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
10
|
Cavaleri D, Capogrosso CA, Guzzi P, Bernasconi G, Re M, Misiak B, Crocamo C, Bartoli F, Carrà G. Blood concentrations of anterior pituitary hormones in drug-naïve people with first-episode psychosis: A systematic review and meta-analysis. Psychoneuroendocrinology 2023; 158:106392. [PMID: 37778198 DOI: 10.1016/j.psyneuen.2023.106392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023]
Abstract
INTRODUCTION The role of anterior pituitary hormones - i.e., adrenocorticotropic hormone (ACTH), luteinizing and follicle stimulating hormones (LH and FSH), growth hormone (GH), prolactin (PRL), and thyroid-stimulating hormone (TSH) - in early schizophrenia and psychoses unclear. We thus performed a systematic review and meta-analysis on the blood concentrations of ACTH, LH and FSH, GH, PRL, and TSH in drug-naïve people with first-episode psychosis (FEP) as compared with healthy controls. METHODS We searched Embase, MEDLINE, and PsycInfo for articles indexed until September 2022. Data quality was appraised. Random-effects meta-analyses were carried out, generating pooled standardized mean differences (SMDs). Between-study heterogeneity was estimated using the I2 statistic. Sensitivity and meta-regression analyses were performed. RESULTS Twenty-six studies were included. Drug-naïve people with FEP, compared to healthy subjects, had higher blood concentrations of ACTH (k = 7; N = 548; SMD = 0.62; 95%CI: 0.29 to 0.94; p < 0.001; I2 = 60.9%) and PRL (k = 17; N = 1757; SMD = 0.85; 95%CI: 0.56 to 1.14; p < 0.001; I2 = 85.5%) as well as lower levels of TSH (k = 6; N = 677; SMD = -0.34; 95%CI: -0.54 to -0.14; p = 0.001; I2 = 29.1%). Meta-regressions did not show any moderating effect of age (p = 0.78), sex (p = 0.21), or symptom severity (p = 0.87) on PRL concentrations in drug-naïve FEP. Available data were not sufficient to perform meta-analyses on FSH, LH, and GH. CONCLUSIONS Drug-naïve people with FEP have altered ACTH, PRL, and TSH blood concentrations, supporting the hypothesis that an abnormal anterior pituitary hormone secretion may be involved in the onset of schizophrenia and psychoses. Further research is needed to elucidate the role of pituitary hormones in FEP.
Collapse
Affiliation(s)
- Daniele Cavaleri
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.
| | | | - Pierluca Guzzi
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Gianna Bernasconi
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Martina Re
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Błażej Misiak
- Department of Psychiatry, Wroclaw Medical University, Pasteura 10 Street, 50-367 Wroclaw, Poland
| | - Cristina Crocamo
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Francesco Bartoli
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Carrà
- Department of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy; Division of Psychiatry, University College London, Maple House 149, London W1T 7BN, United Kingdom
| |
Collapse
|
11
|
Constantin S, Sokanovic SJ, Mochimaru Y, Smiljanic K, Sivcev S, Prévide RM, Wray S, Balla T, Stojilkovic SS. Postnatal Development and Maintenance of Functional Pituitary Gonadotrophs Is Dependent on PI4-Kinase A. Endocrinology 2023; 164:bqad168. [PMID: 37935042 PMCID: PMC10652335 DOI: 10.1210/endocr/bqad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Postnatal development of functional pituitary gonadotrophs is necessary for maturation of the hypothalamic-pituitary-gonadal axis, puberty, and reproduction. Here we examined the role of PI4-kinase A, which catalyzes the biosynthesis of PI4P in mouse reproduction by knocking out this enzyme in cells expressing the gonadotropin-releasing hormone (GnRH) receptor. Knockout (KO) mice were infertile, reflecting underdeveloped gonads and reproductive tracts and lack of puberty. The number and distribution of hypothalamic GnRH neurons and Gnrh1 expression in postnatal KOs were not affected, whereas Kiss1/kisspeptin expression was increased. KO of PI4-kinase A also did not alter embryonic establishment and neonatal development and function of the gonadotroph population. However, during the postnatal period, there was a progressive loss of expression of gonadotroph-specific genes, including Fshb, Lhb, and Gnrhr, accompanied by low gonadotropin synthesis. The postnatal gonadotroph population also progressively declined, reaching approximately one-third of that observed in controls at 3 months of age. In these residual gonadotrophs, GnRH-dependent calcium signaling and calcium-dependent membrane potential changes were lost, but intracellular administration of inositol-14,5-trisphosphate rescued this signaling. These results indicate a key role for PI4-kinase A in the postnatal development and maintenance of a functional gonadotroph population.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Srdjan J Sokanovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuta Mochimaru
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sonja Sivcev
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rafael M Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Li Y, Wang J, Wang R, Chang Y, Wang X. Gut bacteria induce IgA expression in pituitary hormone-secreting cells during aging. iScience 2023; 26:107747. [PMID: 37692284 PMCID: PMC10492204 DOI: 10.1016/j.isci.2023.107747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/17/2023] [Accepted: 08/24/2023] [Indexed: 09/12/2023] Open
Abstract
Pituitary hormone decline is a hallmark of aging. However, the precise gene regulation mechanism during pituitary aging is unclear. Here, we characterized the cell population alteration and global transcriptional change during pituitary aging through single-cell RNA sequencing (scRNA-seq). We found that mRNA-encoding components of protein translational machinery declined the most in the pituitary during aging. Remarkably, Immunoglobulin A (IgA) was found to be expressed in hormone-secreting cells, and the IgA expression level increased dramatically in aged pituitary. Moreover, the pituitary IgA expression was regulated by gut microbiota. The non-hematopoietic origin of the IgA+ cells in the pituitary was further confirmed through bone marrow transplantation. Somatotropes were identified as the most prominent IgA-producing cells through lineage tracing. Thus, pituitary hormone-secreting cells can generate IgA in an age-dependent manner, and such a process is influenced by gut bacteria.
Collapse
Affiliation(s)
- Yehua Li
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Jiawen Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Rui Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Ying Chang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
13
|
Laporte E, Vankelecom H. Organoid models of the pituitary gland in health and disease. Front Endocrinol (Lausanne) 2023; 14:1233714. [PMID: 37614709 PMCID: PMC10442803 DOI: 10.3389/fendo.2023.1233714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The pituitary gland represents the hub of our endocrine system. Its cells produce specific hormones that direct multiple vital physiological processes such as body growth, fertility, and stress. The gland also contains a population of stem cells which are still enigmatic in phenotype and function. Appropriate research models are needed to advance our knowledge on pituitary (stem cell) biology. Over the last decade, 3D organoid models have been established, either derived from the pituitary stem cells or from pluripotent stem cells, covering both healthy and diseased conditions. Here, we summarize the state-of-the-art of pituitary-allied organoid models and discuss applications of these powerful in vitro research and translational tools to study pituitary development, biology, and disease.
Collapse
Affiliation(s)
- Emma Laporte
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Taga S, Suga H, Nakano T, Kuwahara A, Inoshita N, Kodani Y, Nagasaki H, Sato Y, Tsumura Y, Sakakibara M, Soen M, Miwata T, Ozaki H, Kano M, Watari K, Ikeda A, Yamanaka M, Takahashi Y, Kitamoto S, Kawaguchi Y, Miyata T, Kobayashi T, Sugiyama M, Onoue T, Yasuda Y, Hagiwara D, Iwama S, Tomigahara Y, Kimura T, Arima H. Generation and purification of ACTH-secreting hPSC-derived pituitary cells for effective transplantation. Stem Cell Reports 2023; 18:1657-1671. [PMID: 37295423 PMCID: PMC10444568 DOI: 10.1016/j.stemcr.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023] Open
Abstract
Pituitary organoids are promising graft sources for transplantation in treatment of hypopituitarism. Building on development of self-organizing culture to generate pituitary-hypothalamic organoids (PHOs) using human pluripotent stem cells (hPSCs), we established techniques to generate PHOs using feeder-free hPSCs and to purify pituitary cells. The PHOs were uniformly and reliably generated through preconditioning of undifferentiated hPSCs and modulation of Wnt and TGF-β signaling after differentiation. Cell sorting using EpCAM, a pituitary cell-surface marker, successfully purified pituitary cells, reducing off-target cell numbers. EpCAM-expressing purified pituitary cells reaggregated to form three-dimensional pituitary spheres (3D-pituitaries). These exhibited high adrenocorticotropic hormone (ACTH) secretory capacity and responded to both positive and negative regulators. When transplanted into hypopituitary mice, the 3D-pituitaries engrafted, improved ACTH levels, and responded to in vivo stimuli. This method of generating purified pituitary tissue opens new avenues of research for pituitary regenerative medicine.
Collapse
Affiliation(s)
- Shiori Taga
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, Hyogo 650-0047, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan.
| | - Tokushige Nakano
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Atsushi Kuwahara
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, Hyogo 650-0047, Japan
| | - Naoko Inoshita
- Department of Pathology, Moriyama Memorial Hospital, 4-3-1 Kitakasai, Edogawa-ku, Tokyo 134-0081, Japan
| | - Yu Kodani
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hiroshi Nagasaki
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Yoshitaka Sato
- Department of Virology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Yusuke Tsumura
- Department of Pediatrics, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Mayu Sakakibara
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Mika Soen
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Tsutomu Miwata
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Hajime Ozaki
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Mayuko Kano
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa 216-8511, Japan
| | - Kenji Watari
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, Hyogo 650-0047, Japan
| | - Atsushi Ikeda
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, Hyogo 650-0047, Japan
| | - Mitsugu Yamanaka
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka 554-0022, Japan
| | - Yasuhiko Takahashi
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Sachiko Kitamoto
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan
| | - Yohei Kawaguchi
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Takashi Miyata
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Yoshitaka Tomigahara
- Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Osaka 554-8558, Japan; Nihon Medi-Physics Co., Ltd., Koto-ku, Tokyo 136-0075, Japan
| | - Toru Kimura
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, Hyogo 650-0047, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Graduate School of Medicine, Nagoya University, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
15
|
Moreira ARS, Lim J, Urbaniak A, Banik J, Bronson K, Lagasse A, Hardy L, Haney A, Allensworth M, Miles TK, Gies A, Byrum SD, Wilczynska A, Boehm U, Kharas M, Lengner C, MacNicol MC, Childs GV, MacNicol AM, Odle AK. Musashi Exerts Control of Gonadotrope Target mRNA Translation During the Mouse Estrous Cycle. Endocrinology 2023; 164:bqad113. [PMID: 37477898 PMCID: PMC10402870 DOI: 10.1210/endocr/bqad113] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
The anterior pituitary controls key biological processes, including growth, metabolism, reproduction, and stress responses through distinct cell types that each secrete specific hormones. The anterior pituitary cells show a remarkable level of cell type plasticity that mediates the shifts in hormone-producing cell populations that are required to meet organismal needs. The molecular mechanisms underlying pituitary cell plasticity are not well understood. Recent work has implicated the pituitary stem cell populations and specifically, the mRNA binding proteins of the Musashi family in control of pituitary cell type identity. In this study we have identified the target mRNAs that mediate Musashi function in the adult mouse pituitary and demonstrate the requirement for Musashi function in vivo. Using Musashi RNA immunoprecipitation, we identify a cohort of 1184 mRNAs that show specific Musashi binding. Identified Musashi targets include the Gnrhr mRNA, which encodes the gonadotropin-releasing hormone receptor (GnRHR), and the Fshb mRNA, encoding follicle-stimulating hormone (FSH). Reporter assays reveal that Musashi functions to exert repression of translation of the Fshb mRNA, in addition to the previously observed repression of the Gnrhr mRNA. Importantly, mice engineered to lack Musashi in gonadotropes demonstrate a failure to repress translation of the endogenous Gnrhr and Fshb mRNAs during the estrous cycle and display a significant heterogeneity in litter sizes. The range of identified target mRNAs suggests that, in addition to these key gonadotrope proteins, Musashi may exert broad regulatory control over the pituitary proteome in a cell type-specific manner.
Collapse
Affiliation(s)
- Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Katherine Bronson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Linda Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melody Allensworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children's Research Institute, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - Ania Wilczynska
- Bit.bio, The Dorothy Hodgkin Building, Babraham Research Campus, Cambridge CB22 3FH, UK
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Michael Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
16
|
Ferreira NGBP, Madeira JLO, Gergics P, Kertsz R, Marques JM, Trigueiro NSS, Benedetti AFF, Azevedo BV, Fernandes BHV, Bissegatto DD, Biscotto IP, Fang Q, Ma Q, Ozel AB, Li J, Camper SA, Jorge AAL, Mendonça BB, Arnhold IJP, Carvalho LR. Homozygous CDH2 variant may be associated with hypopituitarism without neurological disorders. Endocr Connect 2023; 12:e220473. [PMID: 37166408 PMCID: PMC10388658 DOI: 10.1530/ec-22-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/11/2023] [Indexed: 05/12/2023]
Abstract
Context Congenital hypopituitarism is a genetically heterogeneous condition. Whole exome sequencing (WES) is a promising approach for molecular diagnosis of patients with this condition. Objectives The aim of this study is to conduct WES in a patient with congenital hypopituitarism born to consanguineous parents, CDH2 screening in a cohort of patients with congenital hypopituitarism, and functional testing of a novel CDH2 variant. Design Genomic DNA from a proband and her consanguineous parents was analyzed by WES. Copy number variants were evaluated. The genetic variants were filtered for population frequency (ExAC, 1000 genomes, gnomAD, and ABraOM), in silico prediction of pathogenicity, and gene expression in the pituitary and/or hypothalamus. Genomic DNA from 145 patients was screened for CDH2 by Sanger sequencing. Results One female patient with deficiencies in growth hormone, thyroid-stimulating hormone, adrenocorticotropic hormone, luteinizing hormone, and follicle-stimulating hormone and ectopic posterior pituitary gland contained a rare homozygous c.865G>A (p.Val289Ile) variant in CDH2. To determine whether the p.Val289Ile variant in CDH2 affects cell adhesion properties, we stably transfected L1 fibroblast lines, labeled the cells with lipophilic dyes, and quantified aggregation. Large aggregates formed in cells expressing wildtype CDH2, but aggregation was impaired in cells transfected with variant CDH2 or non-transfected. Conclusion A homozygous CDH2 allelic variant was found in one hypopituitarism patient, and the variant impaired cell aggregation function in vitro. No disease-causing variants were found in 145 other patients screened for CDH2 variants. Thus, CDH2 is a candidate gene for hypopituitarism that needs to be tested in different populations. Significance statement A female patient with hypopituitarism was born from consanguineous parents and had a homozygous, likely pathogenic, CDH2 variant that impairs cell aggregation in vitro. No other likely pathogenic variants in CDH2 were identified in 145 hypopituitarism patients.
Collapse
Affiliation(s)
- Nathalia G B P Ferreira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Joao L O Madeira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Peter Gergics
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Renata Kertsz
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Juliana M Marques
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Nicholas S S Trigueiro
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | | | - Bruna V Azevedo
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Bianca H V Fernandes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Universidade de São Paulo, Zebrafish Facility, São Paulo, São Paulo, Brazil
| | - Debora D Bissegatto
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Isabela P Biscotto
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Qing Fang
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Qianyi Ma
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Asye B Ozel
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Jun Li
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Sally A Camper
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice B Mendonça
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Ivo J P Arnhold
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Luciani R Carvalho
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
17
|
Costanza D, Coluccia P, Auletta L, Castiello E, Navas L, Greco A, Meomartino L. Computed Tomographic Assessment of Pituitary Gland Dimensions in Domestic Short-Haired Cats. Animals (Basel) 2023; 13:1935. [PMID: 37370445 DOI: 10.3390/ani13121935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/27/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The detection of subtle changes in the pituitary dimensions has relevant clinical implications. In cats, a few studies have established the cut-off values of the pituitary gland's dimensions using small and inhomogeneous samples. The aims of this study were: to determine by computed tomography (CT) the pituitary linear dimensions and the pituitary-to-brain (P:B) ratio in a sample of domestic short-haired (DSH) cats; to assess the effects of sex, age, and weight on pituitary dimensions; and to evaluate the inter- and intra-observer agreement for such measurements. All skull CTs of DSH cats performed over four years using a multidetector CT and a standardized protocol were retrospectively reviewed. The exclusion criteria were: clinical, laboratory, or CT alterations of the pituitary gland, brain diseases, fractures of the neurocranium, and diabetes. The pituitary dimensions and brain area were assessed by two different observers using multiplanar reconstructions and automated segmentation tools. Fifty-one cats were included in the final sample. The intraclass correlation coefficients for intra- and inter-observer reliability were good/excellent, and moderate/good, respectively. No differences between sexes were detected, and negligible correlations were found between age and weight. According to this study, a pituitary gland with a height > 4 mm or a P:B ratio > 0.49 mm should be considered enlarged.
Collapse
Affiliation(s)
- Dario Costanza
- Interdepartmental Center of Veterinary Radiology, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| | - Pierpaolo Coluccia
- Interdepartmental Center of Veterinary Radiology, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| | - Luigi Auletta
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milano, Via dell'Università 6, 26900 Lodi, Italy
| | - Erica Castiello
- Interdepartmental Center of Veterinary Radiology, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| | - Luigi Navas
- Department of Veterinary Medicine and Animal Production, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| | - Adelaide Greco
- Interdepartmental Center of Veterinary Radiology, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| | - Leonardo Meomartino
- Interdepartmental Center of Veterinary Radiology, University of Napoli "Federico II", Via Federico Delpino 1, 80137 Napoli, Italy
| |
Collapse
|
18
|
Mniai EM, Bourial A, Salam S, Mahi M, Rami A. Short Stature: Think About the Pituitary Stalk Interruption Syndrome. Cureus 2023; 15:e35700. [PMID: 37012935 PMCID: PMC10066793 DOI: 10.7759/cureus.35700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 03/06/2023] Open
Abstract
Pituitary stalk interruption syndrome (PSIS) is an uncommon congenital defect of the pituitary gland. It is considered one of the rare endocrinal causes of abnormally short stature. Herein, we present a case of a four-year-old girl who consulted for short stature and delayed growth. The patient's history did not include any past medical or surgical pathology. Birth history revealed a full-term delivery with a breech presentation. Clinically, the patient had a small stature, beneath the third percentile. Magnetic resonance imaging findings, through a typical triad, were consistent with PSIS. We describe through this report, what we believe is a rare typical case of PSIS. This case was discovered in a young patient with pituitary dwarfism. We hope that the concise and synthesized structure of this case report will help physicians acquire the necessary reflexes to notice and diagnose the already underdiagnosed PSIS.
Collapse
|
19
|
Altmann R, Scharnreitner I, Auer C, Hirtler L, Springer C, Falschlehner S, Arzt W. Visualization of the Third Ventricle, the Future Cavum Septi Pellucidi, and the Cavum Veli Interpositi at 11+3 to 13+6 Gestational Weeks on 3D Transvaginal Ultrasound Including Normative Data. ULTRASCHALL IN DER MEDIZIN (STUTTGART, GERMANY : 1980) 2023; 44:e72-e82. [PMID: 35213924 DOI: 10.1055/a-1683-6141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVES To show the development of the third ventricle, commissural plate, future cavum septi pellucidi, and cavum veli interpositi in weeks 12-14 by transvaginal 3D ultrasound. METHODS This is a prospective transvaginal 3D study carried out to define the third ventricle and the diencephalic midline structures surrounding it. 93 of 387 fetuses in which the commissural plate with the future cavum septi pellucidi, cavum veli interpositi, and the roof of the third ventricle could be well visualized, were selected with the choroid plexus of the third ventricle and the pituitary gland serving as leading structures. In a small number of fetuses, the optic chiasm could also be displayed. In addition, the following measurements were performed: third ventricle craniocaudal and anteroposterior, roof of the third ventricle/cavum veli interpositi, and fcsp. RESULTS The sonomorphologic characteristics of the commissural plate, the future cavum septi pellucidi, and the cavum veli interpositi are described IN 9% OF THE FETUSES examined. Measurements of the third ventricle, cavum veli interpositi, and the roof of the third ventricle show the following results: 3rd V cc = 3.895 + 0.091*CRL mm; 3rd V ap = 4.175 + 0.036*CRL mm; CVI ap = 2.223 + 0.029*CRL mm; CVI cc = 0.139 + 0.02*CRL mm. CONCLUSION Transvaginal neurosonography enables visualization and measurement of the normal fetal third ventricle at 12-14 weeks of gestation including visualization of the future cavum septi pellucidi and the cavum veli interpositi. BEFORE USE IN PATIENTS CAN BE CONSIDERED, FURTHER SCIENTIFIC WORK IS REQUIRED.
Collapse
Affiliation(s)
- Reinhard Altmann
- Prenatal Medicine, Kepler University Hospital Med Campus IV, Linz, Austria
| | - Iris Scharnreitner
- Prenatal Medicine, Kepler University Hospital Med Campus IV, Linz, Austria
| | - Christian Auer
- Department of Neurosurgery, Kepler University Hospital, Linz, Austria
| | - Lena Hirtler
- Center for Anatomy and Cell Biology, Medical University of Vienna, Wien, Austria
| | - Claudia Springer
- Prenatal Medicine, Kepler University Hospital Med Campus IV, Linz, Austria
| | - Stephanie Falschlehner
- Department of Gynecology and Obstetrics, Kepler University Hospital Med Campus IV, Linz, Austria
| | - Wolfgang Arzt
- Prenatal Medicine, Kepler University Hospital Med Campus IV, Linz, Austria
| |
Collapse
|
20
|
Pituitary-Gland-Based Genes Participates in Intrauterine Growth Restriction in Piglets. Genes (Basel) 2022; 13:genes13112141. [DOI: 10.3390/genes13112141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a major problem associated with piglet growth performance. The incidence of IUGR is widespread in Rongchang pigs. The pituitary gland is important for regulating growth and metabolism, and research has identified genes associated with growth and development. The pituitary gland of newborn piglets with normal birth weight (NBW group, n = 3) and (IUGR group, n = 3) was collected for transcriptome analysis. A total of 323 differentially expression genes (DEGs) were identified (|log2(fold-change)| > 1 and q value < 0.05), of which 223 were upregulated and 100 were downregulated. Gene Ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses showed that the DEGs were mainly related to the extracellular matrix, regulation of the multicellular organismal process, tissue development and angiogenesis, which participate in the growth and immune response in IUGR piglets. Moreover, 7 DEGs including IGF2, THBS1, ITGA1, ITGA8, EPSTI1, FOSB, and UCP2 were associated with growth and immune response. Furthermore, based on the interaction network analysis of the DEGs, two genes, IGF2 and THBS1, participated in cell proliferation, embryonic development and angiogenesis. IGF2 and THBS1 were also the main genes participating in the IUGR. This study identified the core genes involved in IUGR in piglets and provided a reference for exploring the effect of the pituitary gland on piglet growth.
Collapse
|
21
|
Novel Variant in Exon 3 of the BMP4 Gene Resulted in Ectopic Posterior Pituitary, Craniocervical Junction Dysmorphism and Limb Anomaly. Case Rep Pediatr 2022; 2022:8059409. [PMID: 35633847 PMCID: PMC9135578 DOI: 10.1155/2022/8059409] [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: 02/11/2022] [Revised: 04/07/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction. Pituitary differentiation involves a large number of transcription factors. In particular, BMP4 expression is fundamental for pituitary gland commitment from the ventral diencephalon, suppressing Shh expression in Rathke's pouch. Pathogenic variants in BMP4 are reported in the literature with a broad phenotypic spectrum, including pituitary and brain malformations. Case Presentation. A five-year-old girl came to medical attention following a mild cervical trauma with onset of cervical pain. On clinical examination at birth, postaxial polydactyly type B of the left hand was observed and removed at 10 months of age. A cervical radiography was performed, and a suspicion of craniocervical junction malformation was made. A magnetic resonance imaging of the cervical spine was made, showing an ectopic posterior pituitary, associated with dysmorphism of the craniocervical junction. The anthropometric parameters were pubertal Tanner stage 1, weight 16 kg (z-score: −1.09), height 107 cm (z-score: −0.76), and BMI 14 kg/m2 (z-score: −0.92). Normal hormonal assessment was detected. Genetic analysis via next generation sequencing showed a novel de novo heterozygous variant (c.277 G > T, p.Glu93∗) in exon 3 of BMP4. Discussion. We described a novel mutation in BMP4, resulting in ectopic posterior pituitary with normal hormonal assessment, associated to craniocervical junction dysmorphism and limb anomaly. It is important to monitor patient's growth and puberty and to screen the onset of symptoms related to the deficiency of one or more anterior as well as posterior pituitary hormones.
Collapse
|
22
|
Abstract
Congenital hypopituitarism is the deficiency in 1 or more hormones produced by the anterior pituitary or released by the posterior pituitary and has an estimated incidence of 1 in 4,000 to 10,000. Due to the critical role the pituitary plays in growth, metabolic, and reproductive processes, early diagnosis is essential to prevent devastating and often preventable outcomes. However, in neonates with congenital hypopituitarism, symptoms are often nonspecific and tend to overlap with other disease processes, making diagnosis extremely challenging in the neonatal period. This review highlights the embryology and organogenesis of the pituitary gland, genetic causes of hypopituitarism, clinical presentations in the neonatal period, and methods to diagnose and treat select deficiencies with a focus on anterior pituitary hormones.
Collapse
Affiliation(s)
- Geoanna Bautista
- Department of Pediatrics, Division of Neonatology, University of California, Davis Children's Hospital, Sacramento, CA
| |
Collapse
|
23
|
Castets S, Villanueva C, Vergier J, Brue T, Saveanu A, Reynaud R. Clinical, radiological, and molecular diagnosis of congenital pituitary diseases causing short stature. Arch Pediatr 2022; 28:8S33-8S38. [PMID: 37870532 DOI: 10.1016/s0929-693x(22)00041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Short stature in children can be caused by congenital pituitary disorders involving at least one form of growth hormone deficiency. Clinical and radiological evaluations of the index case and family history assessments are essential to guide genetic diagnostic testing and interpret results. The first-line approach is panel testing of genes involved in pituitary development with variants known to be pathogenic in this context. It identifies a genetic cause in less than 10% of cases, however. Whole-exome and whole-genome sequencing techniques may provide original information but also raise new questions regarding the pathophysiological role of identified variants. These new tools can make genetic counselling more complex. The role of clinicians in these interpretations is therefore important. © 2022 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- S Castets
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France.
| | - C Villanueva
- Hospices Civils de Lyon (HCL), Hôpital Femme Mère Enfant (HFME), Service d'Endocrinologie pédiatrique, Bron, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France
| | - J Vergier
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France
| | - T Brue
- Hospices Civils de Lyon (HCL), Hôpital Femme Mère Enfant (HFME), Service d'Endocrinologie pédiatrique, Bron, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Service d'Endocrinologie, Hôpital de la Conception, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - A Saveanu
- Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Marseille Medical Genetics (MMG), U 1251, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Laboratoire de Biologie Moléculaire, Hôpital de la Conception, Marseille, France
| | - R Reynaud
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Marseille Medical Genetics (MMG), U 1251, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| |
Collapse
|
24
|
Yan JL, Chen MY, Chen YL, Chuang CC, Hsu PW, Wei KC, Chang CN. Surgical Outcome and Evaluation of Strategies in the Management of Growth Hormone-Secreting Pituitary Adenomas After Initial Transsphenoidal Pituitary Adenectomy Failure. Front Endocrinol (Lausanne) 2022; 13:756855. [PMID: 35498411 PMCID: PMC9048041 DOI: 10.3389/fendo.2022.756855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 03/09/2022] [Indexed: 12/02/2022] Open
Abstract
Acromegaly is a systemic disease that requires multidisciplinary treatment to achieve the best clinical outcome. This study aimed to evaluate the outcomes of the endoscopic transsphenoidal approach (TSA) as the primary treatment for somatotroph adenomas and further investigate patients who had suboptimal surgical results. This retrospective study included 83 patients with somatotroph adenomas treated by TSA at our institution from 1999 to 2010. Biochemical remission was defined as hGH <1 and <2.5 ng/ml. Factors associated with failure of TSA and strategy of secondary treatments for refractory and recurrent disease were analyzed. The mean age of patients was 41.1 ± 11.3 years, and the mean follow-up time was 54.2 ± 44.3 months. Approximately 44.5% of patients had residual tumors after TSA. Larger tumor size, higher GH level before the operation, and the existence of residual tumors were associated with TSA failure. Forty-one patients had an inadequate response to TSA or a recurrent lesion, and of these patients, 37 had residual tumor after TSA. Octreotide results in good outcomes in the treatment of DGSA patients, and SRS/EXRT generates good results in treating patients who receive second treatments when remission cannot be reached 6 months after TSA operation.
Collapse
Affiliation(s)
- Jiun-Lin Yan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital of the Chang Gung Medical Foundation, Keelung, Taiwan
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- *Correspondence: Jiun-Lin Yan, ; Chen-Nen Chang,
| | - Mao-Yu Chen
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital of the Chang Gung Medical Foundation, Keelung, Taiwan
| | - Yao-Liang Chen
- Department of Radiology, Keelung Chang Gung Memorial Hospital of the Chang Gung Medical Foundation, Keelung, Taiwan
| | - Chi-Cheng Chuang
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Peng-Wei Hsu
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Radiology, Keelung Chang Gung Memorial Hospital of the Chang Gung Medical Foundation, Keelung, Taiwan
| | - Chen-Nen Chang
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital of the Chang Gung Medical Foundation, Keelung, Taiwan
- Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Department of Neurosurgery, Xiamen Chang Gung Hospital, Xiamen, Taiwan
- *Correspondence: Jiun-Lin Yan, ; Chen-Nen Chang,
| |
Collapse
|
25
|
Liu C, Nakano-Tateno T, Satou M, Chik C, Tateno T. Emerging role of signal transducer and activator of transcription 3 (STAT3) in pituitary adenomas. Endocr J 2021; 68:1143-1153. [PMID: 34248112 DOI: 10.1507/endocrj.ej21-0106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Pituitary adenomas are benign tumours that can cause an individual various clinical manifestations including tumour mass effects and/or the diverse effects of abnormal pituitary hormone secretion. Given the morbidity and limited treatment options for pituitary adenomas, there is a need for better biomarkers and treatment options. One molecule that is of specific interest is the signal transducer and activator of transcription 3 (STAT3), a transcription factor that plays a critical role in mediating cytokine-induced changes in gene expression. In addition, STAT3 controls cell proliferation by regulating mitochondrial activity. Not only does activation of STAT3 play a crucial role in tumorigenesis, including pituitary tumorigenesis, but a number of studies also demonstrate pharmacological STAT3 inhibition as a promising treatment approach for many types of tumours, including pituitary tumours. This review will focus on the role of STAT3 in different pituitary adenomas, in particular, growth hormone-producing adenomas and null cell adenomas. Furthermore, how STAT3 is involved in the cell proliferation and hormone regulation in pituitary adenomas and its potential role as a molecular therapeutic target in pituitary adenomas will be summarized.
Collapse
Affiliation(s)
- Cyndy Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tae Nakano-Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Motoyasu Satou
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
- Department of Biochemistry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Constance Chik
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Toru Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
26
|
Ranke MB. Short and Long-Term Effects of Growth Hormone in Children and Adolescents With GH Deficiency. Front Endocrinol (Lausanne) 2021; 12:720419. [PMID: 34539573 PMCID: PMC8440916 DOI: 10.3389/fendo.2021.720419] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023] Open
Abstract
The syndrome of impaired GH secretion (GH deficiency) in childhood and adolescence had been identified at the end of the 19th century. Its non-acquired variant (naGHD) is, at childhood onset, a rare syndrome of multiple etiologies, predominantly characterized by severe and permanent growth failure culminating in short stature. It is still difficult to diagnose GHD and, in particular, to ascertain impaired GH secretion in comparison to levels in normally-growing children. The debate on what constitutes an optimal diagnostic process continues. Treatment of the GH deficit via replacement with cadaveric pituitary human GH (pit-hGH) had first been demonstrated in 1958, and opened an era of therapeutic possibilities, albeit for a limited number of patients. In 1985, the era of recombinant hGH (r-hGH) began: unlimited supply meant that substantial long-term experience could be gained, with greater focus on efficacy, safety and costs. However, even today, the results of current treatment regimes indicate that there is still a substantial fraction of children who do not achieve adult height within the normal range. Renewed evaluation of height outcomes in childhood-onset naGHD is required for a better understanding of the underlying causes, whereby the role of various factors - diagnostics, treatment modalities, mode of treatment evaluation - during the important phases of child growth - infancy, childhood and puberty - are further explored.
Collapse
Affiliation(s)
- Michael B. Ranke
- Children’s Hospital, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
27
|
Obara-Moszyńska M, Budny B, Kałużna M, Zawadzka K, Jamsheer A, Rohde A, Ruchała M, Ziemnicka K, Niedziela M. CDON gene contributes to pituitary stalk interruption syndrome associated with unilateral facial and abducens nerve palsy. J Appl Genet 2021; 62:621-629. [PMID: 34235642 PMCID: PMC8571149 DOI: 10.1007/s13353-021-00649-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 11/06/2022]
Abstract
The relationship between congenital defects of the brain and facial anomalies was proven. The Hedgehog signaling pathway plays a fundamental role in normal craniofacial development in humans. Mutations in the sonic hedgehog (SHH) signaling gene CDON have been recently reported in patients with holoprosencephaly and with pituitary stalk interruption syndrome (PSIS). This study’s aim was an elucidation of an 18-year-old patient presenting PSIS, multiple pituitary hormone deficiency, and congenital unilateral facial and abducens nerve palsy. Additionally, bilateral sensorineural hearing loss, dominating at the right site, was diagnosed. From the second year of life, growth deceleration was observed, and from the age of eight, anterior pituitary hormone deficiencies were gradually confirmed and substituted. At the MRI, characteristic triad for PSIS (anterior pituitary hypoplasia, interrupted pituitary stalk and ectopic posterior lobe) was diagnosed. We performed a comprehensive genomic screening, including microarrays for structural rearrangements and whole-exome sequencing for a monogenic defect. A novel heterozygous missense variant in the CDON gene (c.1814G > T; p.Gly605Val) was identified. The variant was inherited from the mother, who, besides short stature, did not show any disease symptoms. The variant was absent in control databases and 100 healthy subjects originating from the same population. We report a novel variant in the CDON gene associated with PSIS and congenital cranial nerve palsy. The variant revealed autosomal dominant inheritance with incomplete penetrance in concordance with previous studies reporting CDON defects.
Collapse
Affiliation(s)
- Monika Obara-Moszyńska
- Department of Pediatric Endocrinology and Rheumatology, Poznan University of Medical Sciences, 27/33 Szpitalna Str, 60-572, Poznan, Poland.
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 49 Przybyszewskiego Str., 60-355, Poznan, Poland
| | - Małgorzata Kałużna
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 49 Przybyszewskiego Str., 60-355, Poznan, Poland
| | - Katarzyna Zawadzka
- MNM Diagnostics Sp. z o.o., 64 Macieja Rataja Str., 61-695, Poznan, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, 8 Rokietnicka Str, 60-806, Poznan, Poland
| | - Anna Rohde
- Department of Pediatric Endocrinology and Rheumatology, Poznan University of Medical Sciences, 27/33 Szpitalna Str, 60-572, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 49 Przybyszewskiego Str., 60-355, Poznan, Poland
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 49 Przybyszewskiego Str., 60-355, Poznan, Poland
| | - Marek Niedziela
- Department of Pediatric Endocrinology and Rheumatology, Poznan University of Medical Sciences, 27/33 Szpitalna Str, 60-572, Poznan, Poland
| |
Collapse
|
28
|
Castets S, Roucher-Boulez F, Saveanu A, Mallet-Motak D, Chabre O, Amati-Bonneau P, Bonneau D, Girardin C, Morel Y, Villanueva C, Brue T, Reynaud R, Nicolino M. Hypopituitarism in Patients with Blepharophimosis and FOXL2 Mutations. Horm Res Paediatr 2021; 93:30-39. [PMID: 32454486 DOI: 10.1159/000507249] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/15/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND FOXL2 is the gene involved in blepharophimosis, ptosis, and epicanthus inversus syndrome (BPES). There have been few single case reports of growth hormone deficiency (GHD) with this syndrome, and Foxl2 is known to be involved in pituitary development in mice. Our aim was to analyze the prevalence of FOXL2 gene alteration in a series of patients with congenital hypopituitarism and eyelid anomalies. METHODS FOXL2 was analyzed in 10 patients with hypopituitarism (ranging from isolated GHD to complete pituitary hormone deficiency) and eyelid anomalies (typical BPES in 4 patients and milder anomalies in 6 patients). In patients with an FOXL2 mutation, we ruled out other possible molecular explanations by analyzing a panel of 20 genes known to be associated with hypopituitarism, and a candidate gene approach was used for patients without an FOXL2mutation. RESULTS Three patients had an FOXL2mutation. All 3 had typical BPES. Their pituitary phenotype varied from GHD to complete pituitary hormone deficiency and their pituitary morphology ranged from normal to an interrupted pituitary stalk. No mutations were found in genes previously associated with hypopituitarism. CONCLUSION Our study shows that some patients with BPES have hypopituitarism with no molecular explanation other than FOXL2 mutation. This points toward an involvement of FOXL2 in human pituitary development.
Collapse
Affiliation(s)
- Sarah Castets
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, Bron, France, .,Aix-Marseille Université, AP-HM, Centre de Référence des Maladies Rares D'origine Hypophysaire HYPO, Marseille, France,
| | - Florence Roucher-Boulez
- Hospices Civils de Lyon, Groupement Hospitalier Est, Laboratoire de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Endocriniennes Rénales Musculaires et Mucoviscidose, Bron, France.,Centre de Référence du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France.,Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Alexandru Saveanu
- Aix-Marseille Université, AP-HM, Centre de Référence des Maladies Rares D'origine Hypophysaire HYPO, Marseille, France.,Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| | - Delphine Mallet-Motak
- Hospices Civils de Lyon, Groupement Hospitalier Est, Laboratoire de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Endocriniennes Rénales Musculaires et Mucoviscidose, Bron, France.,Centre de Référence du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - Olivier Chabre
- CHU de Grenoble Alpes et Université Grenoble Alpes, CS 10217 38043, Service d'Endocrinologie, Grenoble, France
| | - Patrizia Amati-Bonneau
- MitoLab Team, UMR CNRS 6015 - INSERM U1083, Institut MitoVasc, Université et Hôpital d'Angers, Angers, France.,Département de Biochimie et Génétique, Université et Hôpital d'Angers, Angers, France
| | - Dominique Bonneau
- MitoLab Team, UMR CNRS 6015 - INSERM U1083, Institut MitoVasc, Université et Hôpital d'Angers, Angers, France.,Département de Biochimie et Génétique, Université et Hôpital d'Angers, Angers, France
| | - Celine Girardin
- Hôpitaux Universitaires de Genève, Endocrinologie Pédiatrique, Genève, Switzerland
| | - Yves Morel
- Hospices Civils de Lyon, Groupement Hospitalier Est, Laboratoire de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Endocriniennes Rénales Musculaires et Mucoviscidose, Bron, France.,Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Carine Villanueva
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, Bron, France.,Centre de Référence du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - Thierry Brue
- Aix-Marseille Université, AP-HM, Centre de Référence des Maladies Rares D'origine Hypophysaire HYPO, Marseille, France.,Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| | - Rachel Reynaud
- Aix-Marseille Université, AP-HM, Centre de Référence des Maladies Rares D'origine Hypophysaire HYPO, Marseille, France.,Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| | - Marc Nicolino
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, Bron, France.,Centre de Référence du Développement Génital: du Fœtus à l'Adulte, Filière Maladies Rares Endocriniennes, Bron, France.,Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
29
|
Bosch i Ara L, Katugampola H, Dattani MT. Congenital Hypopituitarism During the Neonatal Period: Epidemiology, Pathogenesis, Therapeutic Options, and Outcome. Front Pediatr 2021; 8:600962. [PMID: 33634051 PMCID: PMC7902025 DOI: 10.3389/fped.2020.600962] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: Congenital hypopituitarism (CH) is characterized by a deficiency of one or more pituitary hormones. The pituitary gland is a central regulator of growth, metabolism, and reproduction. The anterior pituitary produces and secretes growth hormone (GH), adrenocorticotropic hormone, thyroid-stimulating hormone, follicle-stimulating hormone, luteinizing hormone, and prolactin. The posterior pituitary hormone secretes antidiuretic hormone and oxytocin. Epidemiology: The incidence is 1 in 4,000-1 in 10,000. The majority of CH cases are sporadic; however, a small number of familial cases have been identified. In the latter, a molecular basis has frequently been identified. Between 80-90% of CH cases remain unsolved in terms of molecular genetics. Pathogenesis: Several transcription factors and signaling molecules are involved in the development of the pituitary gland. Mutations in any of these genes may result in CH including HESX1, PROP1, POU1F1, LHX3, LHX4, SOX2, SOX3, OTX2, PAX6, FGFR1, GLI2, and FGF8. Over the last 5 years, several novel genes have been identified in association with CH, but it is likely that many genes remain to be identified, as the majority of patients with CH do not have an identified mutation. Clinical manifestations: Genotype-phenotype correlations are difficult to establish. There is a high phenotypic variability associated with different genetic mutations. The clinical spectrum includes severe midline developmental disorders, hypopituitarism (in isolation or combined with other congenital abnormalities), and isolated hormone deficiencies. Diagnosis and treatment: Key investigations include MRI and baseline and dynamic pituitary function tests. However, dynamic tests of GH secretion cannot be performed in the neonatal period, and a diagnosis of GH deficiency may be based on auxology, MRI findings, and low growth factor concentrations. Once a hormone deficit is confirmed, hormone replacement should be started. If onset is acute with hypoglycaemia, cortisol deficiency should be excluded, and if identified this should be rapidly treated, as should TSH deficiency. This review aims to give an overview of CH including management of this complex condition.
Collapse
Affiliation(s)
- Laura Bosch i Ara
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Harshini Katugampola
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Mehul T. Dattani
- Department of Paediatric Endocrinology, Great Ormond Street Hospital for Children, London, United Kingdom
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
30
|
Laporte E, Vennekens A, Vankelecom H. Pituitary Remodeling Throughout Life: Are Resident Stem Cells Involved? Front Endocrinol (Lausanne) 2021; 11:604519. [PMID: 33584539 PMCID: PMC7879485 DOI: 10.3389/fendo.2020.604519] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The pituitary gland has the primordial ability to dynamically adapt its cell composition to changing hormonal needs of the organism throughout life. During the first weeks after birth, an impressive growth and maturation phase is occurring in the gland during which the distinct hormonal cell populations expand. During pubertal growth and development, growth hormone (GH) levels need to peak which requires an adaptive enterprise in the GH-producing somatotrope population. At aging, pituitary function wanes which is associated with organismal decay including the somatopause in which GH levels drop. In addition to these key time points of life, the pituitary's endocrine cell landscape plastically adapts during specific (patho-)physiological conditions such as lactation (need for PRL) and stress (engagement of ACTH). Particular resilience is witnessed after physical injury in the (murine) gland, culminating in regeneration of destroyed cell populations. In many other tissues, adaptive and regenerative processes involve the local stem cells. Over the last 15 years, evidence has accumulated that the pituitary gland houses a resident stem cell compartment. Recent studies propose their involvement in at least some of the cell remodeling processes that occur in the postnatal pituitary but support is still fragmentary and not unequivocal. Many questions remain unsolved such as whether the stem cells are key players in the vivid neonatal growth phase and whether the decline in pituitary function at old age is associated with decreased stem cell fitness. Furthermore, the underlying molecular mechanisms of pituitary plasticity, in particular the stem cell-linked ones, are still largely unknown. Pituitary research heavily relies on transgenic in vivo mouse models. While having proven their value, answers to pituitary stem cell-focused questions may more diligently come from a novel powerful in vitro research model, termed organoids, which grow from pituitary stem cells and recapitulate stem cell phenotype and activation status. In this review, we describe pituitary plasticity conditions and summarize what is known on the involvement and phenotype of pituitary stem cells during these pituitary remodeling events.
Collapse
Affiliation(s)
| | | | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven (University of Leuven), Leuven, Belgium
| |
Collapse
|
31
|
Sanders K, Galac S, Meij BP. Pituitary tumour types in dogs and cats. Vet J 2021; 270:105623. [PMID: 33641809 DOI: 10.1016/j.tvjl.2021.105623] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/25/2022]
Abstract
Pituitary tumours are common in dogs and are being increasingly recognized in cats. Pituitary tumours are usually classified as adenomas and should only be classified as carcinomas when there is evidence of metastatic spread of the tumour, which is rare. Despite the benign nature of most pituitary tumours, they can still compress or invade neighbouring tissues. Pituitary tumours can be functional (hormonally active) or non-functional (hormonally silent). The aim of this review was to provide an overview of the different pituitary tumour types in dogs and cats that have been reported in the literature. In dogs, the most common pituitary tumour type is the corticotroph adenoma, which can cause pituitary-dependent hypercortisolism. In cats, the most common pituitary tumour is the somatotroph adenoma, which can cause hypersomatotropism, and the second-most common is the corticotroph adenoma. A lactotroph adenoma has been described in one dog, while gonadotroph, thyrotroph and null cell adenomas have not been described in dogs or cats. Hormonally silent adenomas are likely underdiagnosed because they do not result in an endocrine syndrome. Tools used to classify pituitary tumours in humans, particularly immunohistochemistry for lineage-specific transcription factors, are likely to be useful to classify canine and feline pituitary tumours of unknown origin. Future studies are required to better understand the full range of pituitary adenoma pathology in dogs and cats and to determine whether certain adenoma subtypes behave more aggressively than others. Currently, the mechanisms that underlie pituitary tumorigenesis in dogs and cats are still largely unknown. A better understanding of the molecular background of these tumours could help to identify improved pituitary-targeted therapeutics.
Collapse
Affiliation(s)
- K Sanders
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands.
| | - S Galac
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands
| | - B P Meij
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 108, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
32
|
Abstract
Pituitary stalk interruption syndrome (PSIS) is a distinct developmental defect of the pituitary gland identified by magnetic resonance imaging and characterized by a thin, interrupted, attenuated or absent pituitary stalk, hypoplasia or aplasia of the adenohypophysis, and an ectopic posterior pituitary. The precise etiology of PSIS still remains elusive or incompletely confirmed in most cases. Adverse perinatal events, including breech delivery and hypoxia, were initially proposed as the underlying mechanism affecting the hypothalamic-pituitary axis. Nevertheless, recent findings have uncovered a wide variety of PSIS-associated molecular defects in genes involved in pituitary development, holoprosencephaly (HPE), neural development, and other important cellular processes such as cilia function. The application of whole exome sequencing (WES) in relatively large cohorts has identified an expanded pool of potential candidate genes, mostly related to the Wnt, Notch, and sonic hedgehog signaling pathways that regulate pituitary growth and development during embryogenesis. Importantly, WES has revealed coexisting pathogenic variants in a significant number of patients; therefore, pointing to a multigenic origin and inheritance pattern of PSIS. The disorder is characterized by inter- and intrafamilial variability and incomplete or variable penetrance. Overall, PSIS is currently viewed as a mild form of an expanded HPE spectrum. The wide and complex clinical manifestations include evolving pituitary hormone deficiencies (with variable timing of onset and progression) and extrapituitary malformations. Severe and life-threatening symptomatology is observed in a subset of patients with complete pituitary hormone deficiency during the neonatal period. Nevertheless, most patients are referred later in childhood for growth retardation. Prompt and appropriate hormone substitution therapy constitutes the cornerstone of treatment. Further studies are needed to uncover the etiopathogenesis of PSIS.
Collapse
Affiliation(s)
- Antonis Voutetakis
- Department of Pediatrics, School of Medicine, Democritus University of Thrace, Alexandroupolis, Thrace, Greece.
| |
Collapse
|
33
|
Abstract
Growth is the task of children. We review the normal process of linear growth from the fetus through adolescence and note that growth is the result of age- and gender-dependent interactions among key genetic, environmental, dietary, socioeconomic, developmental, behavioral, nutritional, metabolic, biochemical, and hormonal factors. We then define the wide range of normative data at each stage of growth and note that a pattern within this range is generally indicative of good general health and that growth significantly slower than this range may lead to growth faltering and subsequent short stature. Although not often emphasized, we detail how to properly measure infants and children because height velocity is usually determined from two height measurements (both relatively large values) to calculate the actual height velocity (a relatively much smaller number in comparison). Traditionally the physiology of growth has been taught from an endocrine-centric point-of-view. Here we review the hypothalamic-pituitary-end organ axes for the GH/IGF-1 and gonadal steroid hormones (hypothalamic-pituitary-gonadal axis), both during "mini"-puberty as well as at puberty. However, over the past few decades much more emphasis has been placed on the growth plate and its many interactions with the endocrine system but also with its own intrinsic physiology and gene mutations. These latter, whether individually (large effect size) or in combination with many others including endocrine system-based, may account in toto for meaningful differences in adult height. The clinical assessment of children with short stature includes medical, social and family history, physical exam and importantly proper interpretation of the growth curve. This analysis should lead to judicious use of screening laboratory and imaging tests depending on the pre-test probability (Bayesian inference) of a particular diagnosis in that child. In particular for those with no pathological features in the history and physical exam and a low, but normal height velocity, may lead only to a bone age exam and reevaluation (re-measurement), perhaps 6 months later. he next step depends on the comfort level of the primary care physician, the patient, and the parent; that is, whether to continue with the evaluation with more directed, more sophisticated testing, again based on Bayesian inference or to seek consultation with a subspecialist pediatrician based on the data obtained. This is not necessarily an endocrinologist. The newest area and the one most in flux is the role for genetic testing, given that growth is a complex process with large effect size for single genes but smaller effect sizes for multiple other genes which in the aggregate may be relevant to attained adult height. Genetics is a discipline that is rapidly changing, especially as the cost of exome or whole gene sequencing diminishes sharply. Within a decade it is quite likely that a genetic approach to the evaluation of children with short stature will become the standard, truncating the diagnostic odyssey and be cost effective as fewer biochemical and imaging studies are required to make a proper diagnosis.
Collapse
Affiliation(s)
- Roberto Bogarín
- Department of Pediatric Endocrinology, National Children's Hospital, San José, Costa Rica
| | - Erick Richmond
- Department of Pediatric Endocrinology, National Children's Hospital, San José, Costa Rica
| | - Alan D Rogol
- Department of Pediatric Endocrinology, University of Virginia, Charlottesville, VA, USA -
| |
Collapse
|
34
|
Budny B, Karmelita-Katulska K, Stajgis M, Żemojtel T, Ruchała M, Ziemnicka K. Copy Number Variants Contributing to Combined Pituitary Hormone Deficiency. Int J Mol Sci 2020; 21:ijms21165757. [PMID: 32796691 PMCID: PMC7461210 DOI: 10.3390/ijms21165757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/25/2022] Open
Abstract
Combined pituitary hormone deficiency represents a disorder with complex etiology. For many patients, causes of the disease remain unexplained, despite usage of advanced genetic testing. Although major and common transcription factors were identified two decades ago, we still struggle with identification of rare inborn factors contributing to pituitary function. In this report, we follow up genomic screening of CPHD patient cohort that were previously tested for changes in a coding sequences of genes with the use of the whole exome. We aimed to find contribution of rare copy number variations (CNVs). As a result, we identified genomic imbalances in 7 regions among 12 CPHD patients. Five out of seven regions showed copy gains whereas two presented losses of genomic fragment. Three regions with detected gains encompassed known CPHD genes namely LHX4, HESX1, and OTX2. Among new CPHD loci, the most interesting seem to be the region covering SIX3 gene, that is abundantly expressed in developing brain, and together with HESX1 contributes to pituitary organogenesis as it was evidenced before in functional studies. In conclusion, with the use of broadened genomic approach we identified copy number imbalances for 12 CPHD patients. Although further functional studies are required in order to estimate its true impact on expression pattern during pituitary organogenesis and CPHD etiology.
Collapse
Affiliation(s)
- Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
- Correspondence: ; Tel.: +48-691-814-330
| | - Katarzyna Karmelita-Katulska
- Department of General Radiology and Neuroradiology, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (K.K.-K.); (M.S.)
| | - Marek Stajgis
- Department of General Radiology and Neuroradiology, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (K.K.-K.); (M.S.)
| | - Tomasz Żemojtel
- Genomics Platform, Berlin Institute of Health, 10117 Berlin, Germany;
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 60-569 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (M.R.); (K.Z.)
| |
Collapse
|
35
|
David O, Eskin-Schwartz M, Ling G, Dolgin V, Kristal E, Benkowitz E, Osyntsov L, Gradstein L, Birk OS, Loewenthal N, Yerushalmi B. Pituitary stalk interruption syndrome broadens the clinical spectrum of the TTC26 ciliopathy. Clin Genet 2020; 98:303-307. [PMID: 32617964 DOI: 10.1111/cge.13805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/27/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022]
Abstract
Ciliopathies are a heterogeneous group of disorders, related to abnormal ciliary function. Severe biliary ciliopathy, caused by bi-allelic mutations in TTC26, has been recently described in the context of a syndrome of polydactyly and severe neonatal cholestasis, with brain, kidney and heart involvement. Pituitary involvement has not been previously reported for patients with this condition. Pituitary stalk interruption syndrome (PSIS) is a congenital anomaly of the pituitary gland, diagnosed by characteristic MRI findings. We now describe four patients with TTC26 ciliopathy due to a homozygous c.695A>G p.Asn232Ser mutation and delineate PSIS as a novel clinical feature of this disorder, highlighting an important role of TTC26 in pituitary development.
Collapse
Affiliation(s)
- Odeya David
- Pediatric Endocrinology Unit, Soroka University Medical Center, Beer-Sheva, Israel.,Saban Pediatric Medical Center for Israel, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Marina Eskin-Schwartz
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Genetics Institute at Soroka University Medical Center and the Morris Kahn Laboratory of Human Genetics, National Center for Rare Diseases, at the Faculty of Health Sciences and National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Galina Ling
- Saban Pediatric Medical Center for Israel, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Gastroenterology Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Vadim Dolgin
- Genetics Institute at Soroka University Medical Center and the Morris Kahn Laboratory of Human Genetics, National Center for Rare Diseases, at the Faculty of Health Sciences and National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Kristal
- Saban Pediatric Medical Center for Israel, Soroka University Medical Center, Beer-Sheva, Israel
| | - Ela Benkowitz
- Radiology Department, Soroka Medical Center, Beer-Sheva, Israel
| | - Lidia Osyntsov
- Institute of Pathology, Soroka Medical Center, Beer-Sheva, Israel
| | - Libe Gradstein
- Genetics Institute at Soroka University Medical Center and the Morris Kahn Laboratory of Human Genetics, National Center for Rare Diseases, at the Faculty of Health Sciences and National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Ophthalmology Clinic, Southern District, Clalit Health Services, Beer-sheva, Israel
| | - Ohad S Birk
- Genetics Institute at Soroka University Medical Center and the Morris Kahn Laboratory of Human Genetics, National Center for Rare Diseases, at the Faculty of Health Sciences and National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Neta Loewenthal
- Pediatric Endocrinology Unit, Soroka University Medical Center, Beer-Sheva, Israel.,Saban Pediatric Medical Center for Israel, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Baruch Yerushalmi
- Saban Pediatric Medical Center for Israel, Soroka University Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Gastroenterology Unit, Soroka University Medical Center, Beer-Sheva, Israel
| |
Collapse
|
36
|
Ho Y, Hu P, Peel MT, Chen S, Camara PG, Epstein DJ, Wu H, Liebhaber SA. Single-cell transcriptomic analysis of adult mouse pituitary reveals sexual dimorphism and physiologic demand-induced cellular plasticity. Protein Cell 2020; 11:565-583. [PMID: 32193873 PMCID: PMC7381518 DOI: 10.1007/s13238-020-00705-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/26/2020] [Indexed: 12/23/2022] Open
Abstract
The anterior pituitary gland drives highly conserved physiologic processes in mammalian species. These hormonally controlled processes are central to somatic growth, pubertal transformation, fertility, lactation, and metabolism. Current cellular models of mammalian anteiror pituitary, largely built on candidate gene based immuno-histochemical and mRNA analyses, suggest that each of the seven hormones synthesized by the pituitary is produced by a specific and exclusive cell lineage. However, emerging evidence suggests more complex relationship between hormone specificity and cell plasticity. Here we have applied massively parallel single-cell RNA sequencing (scRNA-seq), in conjunction with complementary imaging-based single-cell analyses of mRNAs and proteins, to systematically map both cell-type diversity and functional state heterogeneity in adult male and female mouse pituitaries at single-cell resolution and in the context of major physiologic demands. These quantitative single-cell analyses reveal sex-specific cell-type composition under normal pituitary homeostasis, identify an array of cells associated with complex complements of hormone-enrichment, and undercover non-hormone producing interstitial and supporting cell-types. Interestingly, we also identified a Pou1f1-expressing cell population that is characterized by a unique multi-hormone gene expression profile. In response to two well-defined physiologic stresses, dynamic shifts in cellular diversity and transcriptome profiles were observed for major hormone producing and the putative multi-hormone cells. These studies reveal unanticipated cellular complexity and plasticity in adult pituitary, and provide a rich resource for further validating and expanding our molecular understanding of pituitary gene expression programs and hormone production.
Collapse
Affiliation(s)
- Yugong Ho
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Peng Hu
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael T Peel
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sixing Chen
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pablo G Camara
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas J Epstein
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hao Wu
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Stephen A Liebhaber
- Departments of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Departments of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
37
|
Lu X, Arbab AAI, Zhang Z, Fan Y, Han Z, Gao Q, Sun Y, Yang Z. Comparative Transcriptomic Analysis of the Pituitary Gland between Cattle Breeds Differing in Growth: Yunling Cattle and Leiqiong Cattle. Animals (Basel) 2020; 10:E1271. [PMID: 32722439 PMCID: PMC7460210 DOI: 10.3390/ani10081271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The hypothalamic-pituitary-thyroid (HPT) axis hormones regulate the growth and development of ruminants, and the pituitary gland plays a decisive role in this process. In order to identify pivotal genes in the pituitary gland that could affect the growth of cattle by regulating the secretion of hormones, we detected the content of six HPT hormones related to growth in the plasma of two cattle breeds (Yunling and Leiqiong cattle, both also known as the zebu cattle) with great differences in growth and compared the transcriptome data of their pituitary glands. Our study found that the contents of GH, IGF, TSH, thyroxine, triiodothyronine, and insulin were significantly different between the two breeds, which was the main cause of the difference in growth; 175 genes were identified as differentially expressed genes (DEGs). Functional association analyses revealed that DEGs were mainly involved in the process of transcription and signal transduction. Combining the enrichment analysis and protein interaction analysis, eight DEGs were predicted to control the growth of cattle by affecting the expression of growth-related hormones in the pituitary gland. In summary, our results suggested that SLC38A1, SLC38A3, DGKH, GNB4, GNAQ, ESR1, NPY, and GAL are candidates in the pituitary gland for regulating the growth of Yunling and Leiqiong cattle by regulating the secretion of growth-related hormones. This study may help researchers further understand the growth mechanisms and improve the artificial selection of zebu cattle.
Collapse
Affiliation(s)
- Xubin Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Abdelaziz Adam Idriss Arbab
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Zhipeng Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Yongliang Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Ziyin Han
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Qisong Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| | - Yujia Sun
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China;
| | - Zhangping Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.L.); (A.A.I.A.); (Z.Z.); (Y.F.); (Z.H.); (Q.G.)
| |
Collapse
|
38
|
Abstract
The development of the anterior pituitary gland occurs in distinct sequential developmental steps, leading to the formation of a complex organ containing five different cell types secreting six different hormones. During this process, the temporal and spatial expression of a cascade of signaling molecules and transcription factors plays a crucial role in organ commitment, cell proliferation, patterning, and terminal differentiation. The morphogenesis of the gland and the emergence of distinct cell types from a common primordium are governed by complex regulatory networks involving transcription factors and signaling molecules that may be either intrinsic to the developing pituitary or extrinsic, originating from the ventral diencephalon, the oral ectoderm, and the surrounding mesenchyme. Endocrine cells of the pituitary gland are organized into structural and functional networks that contribute to the coordinated response of endocrine cells to stimuli; these cellular networks are formed during embryonic development and are maintained or may be modified in adulthood, contributing to the plasticity of the gland. Abnormalities in any of the steps of pituitary development may lead to congenital hypopituitarism that includes a spectrum of disorders from isolated to combined hormone deficiencies including syndromic disorders such as septo-optic dysplasia. Over the past decade, the acceleration of next-generation sequencing has allowed for rapid analysis of the patient genome to identify novel mutations and novel candidate genes associated with hypothalmo-pituitary development. Subsequent functional analysis using patient fibroblast cells, and the generation of stem cells derived from patient cells, is fast replacing the need for animal models while providing a more physiologically relevant characterization of novel mutations. Furthermore, CRISPR-Cas9 as the method for gene editing is replacing previous laborious and time-consuming gene editing methods that were commonly used, thus yielding knockout cell lines in a fraction of the time. © 2020 American Physiological Society. Compr Physiol 10:389-413, 2020.
Collapse
Affiliation(s)
- Kyriaki S Alatzoglou
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Louise C Gregory
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Mehul T Dattani
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| |
Collapse
|
39
|
Klingler C, Ashley J, Shi K, Stiefvater A, Kyba M, Sinnreich M, Aihara H, Kinter J. DNA aptamers against the DUX4 protein reveal novel therapeutic implications for FSHD. FASEB J 2020; 34:4573-4590. [PMID: 32020675 PMCID: PMC7079142 DOI: 10.1096/fj.201902696] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/17/2020] [Indexed: 01/13/2023]
Abstract
Aberrant expression of the transcription factor double homeobox protein 4 (DUX4) can lead to a number of diseases including facio‐scapulo‐humeral muscular dystrophy (FSHD), acute lymphoblastic leukemia, and sarcomas. Inhibition of DUX4 may represent a therapeutic strategy for these diseases. By applying Systematic Evolution of Ligands by EXponential Enrichment (SELEX), we identified aptamers against DUX4 with specific secondary structural elements conveying high affinity to DUX4 as assessed by fluorescence resonance energy transfer and fluorescence polarization techniques. Sequences analysis of these aptamers revealed the presence of two consensus DUX4 motifs in a reverse complementary fashion forming hairpins interspersed with bulge loops at distinct positions that enlarged the binding surface with the DUX4 protein, as determined by crystal structure analysis. We demonstrate that insertion of specific structural elements into transcription factor binding oligonucleotides can enhance specificity and affinity.
Collapse
Affiliation(s)
- Christian Klingler
- Neuromuscular Research Group, Department of Neurology, University Hospital Basel, Basel, Switzerland.,Neuromuscular Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Jon Ashley
- Neuromuscular Research Group, Department of Neurology, University Hospital Basel, Basel, Switzerland.,Neuromuscular Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.,Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Adeline Stiefvater
- Neuromuscular Research Group, Department of Neurology, University Hospital Basel, Basel, Switzerland.,Neuromuscular Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Michael Sinnreich
- Neuromuscular Research Group, Department of Neurology, University Hospital Basel, Basel, Switzerland.,Neuromuscular Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jochen Kinter
- Neuromuscular Research Group, Department of Neurology, University Hospital Basel, Basel, Switzerland.,Neuromuscular Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
40
|
Pradilla Dieste A, Chenlo M, Perez-Romero S, Garcia-Rendueles ÁR, Suarez-Fariña M, Garcia-Lavandeira M, Bernabeu I, Cameselle-Teijeiro JM, Alvarez CV. GFRα 1-2-3-4 co-receptors for RET Are co-expressed in Pituitary Stem Cells but Individually Retained in Some Adenopituitary Cells. Front Endocrinol (Lausanne) 2020; 11:631. [PMID: 33071961 PMCID: PMC7543094 DOI: 10.3389/fendo.2020.00631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/04/2020] [Indexed: 11/26/2022] Open
Abstract
The RET tyrosine kinase receptor is expressed by the endocrine somatotroph cells of the pituitary where it has important functions regulating survival/apoptosis. However, RET is also expressed by the GPS pituitary stem cells localized in a niche between the adenopituitary and the intermediate lobe. To bind any of its four ligands, RET needs one of four co-receptors called GFRα1-4. It has been previously shown that GFRα1 is expressed by somatotroph cells and acromegaly tumors. GFRα2 was shown to be expressed by pituitary stem cells. GFRα4 was proposed as not expressed in the pituitary. Here we study the RNA and protein expression of the four GFRα co-receptors for RET in rat and human pituitary. The four co-receptors were abundantly expressed at the RNA level both in rat and human pituitary, although GFRα4 was the less abundant. Multiple immunofluorescence for each co-receptor and β-catenin, a marker of stem cell niche was performed. The four GFRα co-receptors were co-expressed by the GPS cells at the niche colocalizing with β-catenin. Isolated individual scattered cells positive for one or other receptor could be found through the adenopituitary with low β-catenin expression. Some of them co-express GFRα1 and PIT1. Immunohistochemistry in normal human pituitary confirmed the data. Our data suggest that the redundancy of GFRα co-expression is a self-supportive mechanism which ensures niche maintenance and proper differentiation.
Collapse
Affiliation(s)
- Alberto Pradilla Dieste
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Miguel Chenlo
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Sihara Perez-Romero
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Ángela R. Garcia-Rendueles
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Maria Suarez-Fariña
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Montserrat Garcia-Lavandeira
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Ignacio Bernabeu
- Department of Endocrinology, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Instituto de Investigación Sanitaria de Santiago (IDIS), USC, Santiago de Compostela, Spain
| | - José Manuel Cameselle-Teijeiro
- Department of Pathology, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Clara V. Alvarez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Instituto de Investigación Sanitaria (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- *Correspondence: Clara V. Alvarez
| |
Collapse
|
41
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
42
|
Chen J, Zhang XX, Wu XC, Li J. [Clinical and genetic characteristics of a young child with combined pituitary hormone deficiency type I caused by POU1F1 gene variation]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:685-689. [PMID: 31315769 PMCID: PMC7389113 DOI: 10.7499/j.issn.1008-8830.2019.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/30/2019] [Indexed: 06/10/2023]
Abstract
This paper reports the clinical and genetic characteristics of a case of combined pituitary hormone deficiency type I (CPHD1) caused by POU domain, class 1, transcription factor 1 (POU1F1) gene variation. A 2 years and 3 months old girl mainly presented with short stature, special facial features of prominent forehead, enophthalmos, and short mandible, loose skin, central hypothyroidism, complete growth hormone deficiency, and anterior pituitary hypoplasia. Gene analysis identified a novel heterozygous mutation, c.889C>T (p.R297W), in POU1F1 gene, and this locus of her parents was wild-type. This mutation was analyzed as a possible pathogenic variant according to the guidelines of the American College of Medical Genetics and Genomics, which has not been previously reported in the literature and conforms to the autosomal dominant inheritance. This child was diagnosed with CPHD1. Her height increased by 19.8 cm and showed a catch-up growth trend after one year of combined treatment with growth hormone and euthyrox. This study enriches the mutation spectrum of POU1F1 gene and has important significance for the diagnosis and classification of combined pituitary hormone deficiency.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pediatrics, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | | | | | | |
Collapse
|
43
|
Chukwurah E, Osmundsen A, Davis SW, Lizarraga SB. All Together Now: Modeling the Interaction of Neural With Non-neural Systems Using Organoid Models. Front Neurosci 2019; 13:582. [PMID: 31293366 PMCID: PMC6598414 DOI: 10.3389/fnins.2019.00582] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022] Open
Abstract
The complex development of the human nervous system has been traditionally studied using a combination of animal models, human post-mortem brain tissue, and human genetics studies. However, there has been a lack of experimental human cellular models that would allow for a more precise elucidation of the intricate dynamics of early human brain development. The development of stem cell technologies, both embryonic and induced pluripotent stem cells (iPSCs), has given neuroscientists access to the previously inaccessible early stages of human brain development. In particular, the recent development of three-dimensional culturing methodologies provides a platform to study the differentiation of stem cells in both normal development and disease states in a more in vivo like context. Three-dimensional neural models or cerebral organoids possess an innate advantage over two-dimensional neural cultures as they can recapitulate tissue organization and cell type diversity that resemble the developing brain. Brain organoids also provide the exciting opportunity to model the integration of different brain regions in vitro. Furthermore, recent advances in the differentiation of non-neuronal tissue from stem cells provides the opportunity to study the interaction between the developing nervous system and other non-neuronal systems that impact neuronal function. In this review, we discuss the potential and limitations of the organoid system to study in vitro neurological diseases that arise in the neuroendocrine and the enteric nervous system or from interactions with the immune system.
Collapse
Affiliation(s)
- Evelyn Chukwurah
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States
| | - Allison Osmundsen
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States
| | - Shannon W. Davis
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States
| | - Sofia B. Lizarraga
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
44
|
Li X, Li C, Wureli H, Ni W, Zhang M, Li H, Xu Y, Rizabek K, Bolatkhan M, Askar D, Gulzhan K, Hou X, Hu S. Screening and evaluating of long non-coding RNAs in prenatal and postnatal pituitary gland of sheep. Genomics 2019; 112:934-942. [PMID: 31200027 DOI: 10.1016/j.ygeno.2019.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/18/2022]
Abstract
Long non-coding RNAs are transcribed into RNA molecules that are >200 nucleotides in length. However, the expression and function analysis of lncRNAs in the sheep pituitary gland are still lacking. In this study, we identified 1755 lncRNAs (545 annotated lncRNAs and 1210 novel lncRNAs) from RNA-seq data in the pituitary gland of embryonic and adult sheep. A total of 235 lncRNAs were differentially expressed between embryonic and adult group. We verified the presence of some lncRNAs using RT-PCR and DNA sequencing, and identified some differentially expressed lncRNAs using qPCR. We also investigated the role of cis-acting lncRNAs on target genes. GO and KEGG enrichment analysis revealed that the target genes of lncRNAs were involved in the regulation of hormones secretion and some signaling pathways in the sheep pituitary gland. Our study provides comprehensive expression profiles of lncRNAs and valuable resource for understanding their function in the pituitary gland.
Collapse
Affiliation(s)
- Xiaoyue Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Cunyuan Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China; College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Hazi Wureli
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Wei Ni
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Mengdan Zhang
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Huixiang Li
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yueren Xu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Kadyken Rizabek
- Department of Food Engineering, Kazakh National Agrarian University, Almaty Province 050010, Kazakhstan
| | - Makhatov Bolatkhan
- Department of Technology and Biological Resources, Kazakh National Agrarian University, Almaty Province 050010, Kazakhstan
| | - Dzhunysov Askar
- Department of Technology and Biological Resources, Kazakh National Agrarian University, Almaty Province 050010, Kazakhstan
| | - Kulmanova Gulzhan
- Department of Technology and Biological Resources, Kazakh National Agrarian University, Almaty Province 050010, Kazakhstan
| | - Xiaoxu Hou
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Shengwei Hu
- College of Life Sciences, Shihezi University, Shihezi, Xinjiang 832003, China.
| |
Collapse
|
45
|
Tabor J, Collins R, Debert CT, Shultz SR, Mychasiuk R. Neuroendocrine Whiplash: Slamming the Breaks on Anabolic-Androgenic Steroids Following Repetitive Mild Traumatic Brain Injury in Rats May Worsen Outcomes. Front Neurol 2019; 10:481. [PMID: 31133974 PMCID: PMC6517549 DOI: 10.3389/fneur.2019.00481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/23/2019] [Indexed: 11/13/2022] Open
Abstract
Sport-related concussion is an increasingly common injury among adolescents, with repetitive mild traumatic brain injuries (RmTBI) being a significant risk factor for long-term neurobiological and psychological consequences. It is not uncommon for younger professional athletes to consume anabolic-androgenic steroids (AAS) in an attempt to enhance their performance, subjecting their hormonally sensitive brains to potential impairment during neurodevelopment. Furthermore, RmTBI produces acute neuroendocrine dysfunction, specifically in the anterior pituitary, disrupting the hypothalamic-pituitary adrenal axis, lowering cortisol secretion that is needed to appropriately respond to injury. Some AAS users exhibit worse symptoms post-RmTBI if they quit their steroid regime. We sought to examine the pathophysiological outcomes associated with the abrupt cessation of the commonly abused AAS, Metandienone (Met) on RmTBI outcomes in rats. Prior to injury, adolescent male rats received either Met or placebo, and exercise. Rats were then administered RmTBIs or sham injuries, followed by steroid and exercise cessation (SEC) or continued treatment. A behavioral battery was conducted to measure outcomes consistent with clinical representations of post-concussion syndrome and chronic AAS exposure, followed by analysis of serum hormone levels, and qRT-PCR for mRNA expression and telomere length. RmTBI increased loss of consciousness and anxiety-like behavior, while also impairing balance and short-term working memory. SEC induced hyperactivity while Met treatment alone increased depressive-like behavior. There were cumulative effects whereby RmTBI and SEC exacerbated anxiety and short-term memory outcomes. mRNA expression in the prefrontal cortex, amygdala, hippocampus, and pituitary were modified in response to Met and SEC. Analysis of telomere length revealed the negative impact of SEC while Met and SEC produced changes in serum levels of testosterone and corticosterone. We identified robust changes in mRNA to serotonergic circuitry, neuroinflammation, and an enhanced stress response. Interestingly, Met treatment promoted glucocorticoid secretion after injury, suggesting that maintained AAS may be more beneficial than abstaining after mTBI.
Collapse
Affiliation(s)
- Jason Tabor
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Reid Collins
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Chantel T Debert
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Division of Physical Medicine and Rehabilitation, Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Wright TA, Page RC, Konkolewicz D. Polymer conjugation of proteins as a synthetic post-translational modification to impact their stability and activity. Polym Chem 2019; 10:434-454. [PMID: 31249635 PMCID: PMC6596429 DOI: 10.1039/c8py01399c] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
For more than 40 years, protein-polymer conjugates have been widely used for many applications, industrially and biomedically. These bioconjugates have been shown to modulate the activity and stability of various proteins while introducing reusability and new activities that can be used for drug delivery, improve pharmacokinetic ability, and stimuli-responsiveness. Techniques such as RDRP, ROMP and "click" have routinely been utilized for development of well-defined bioconjugate and polymeric materials. Synthesis of bioconjugate materials often take advantage of natural amino acids present within protein and peptide structures for a host of coupling chemistries. Polymer modification may elicit increased or decreased activity, activity retention under harsh conditions, prolonged activity in vivo and in vitro, and introduce stimuli responsiveness. Bioconjugation has resulted to modulated thermal stability, chemical stability, storage stability, half-life and reusability. In this review we aim to provide a brief state of the field, highlight a wide range of behaviors caused by polymer conjugation, and provide areas of future work.
Collapse
Affiliation(s)
- Thaiesha A Wright
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| | - Richard C Page
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| | - Dominik Konkolewicz
- Department of Chemistry and Biochemistry, Miami University Oxford, Ohio 45056, United States
| |
Collapse
|
47
|
Li M, Lu X, Xia H, Zhang C, Wang X, Chen Z, Zhang H, Qu K, Huang B, Moore S, Mao Y, Yang Z. In-depth characterization of the pituitary transcriptome in Simmental and Chinese native cattle. Domest Anim Endocrinol 2019; 66:35-42. [PMID: 30391830 DOI: 10.1016/j.domaniend.2018.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/10/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022]
Abstract
The pituitary gland is a key endocrine organ responsible for growth and development. To get closer to understanding important molecular mechanisms at work in the bovine pituitary and identifying the core genes controlling growth, in the present study we have analyzed the transcriptome profiling of the pituitary glands of two cattle breeds (Wenshan and Simmental cattle) with extreme growth differences using high-throughput RNA sequencing. Our study revealed that the overall gene expression levels are quite similar between the two breeds. A total of 18,065 annotated genes were identified, which accounted for 85% of the annotated genes in cattle. The five most abundant hormone genes (GH, PRL, POMC, CGA, and LHB) were relatively stable in both breeds, indicating their pivotal roles in maintaining the basic functions of the pituitary. In addition, 105 genes were identified as differentially expressed between Wenshan and Simmental pituitary glands, including 83 known and 22 novel genes. Functional association analyses of the differentially expressed genes between the breeds revealed 60 enriched gene ontology terms and 3 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Interestingly, the most enriched KEGG pathway, cell adhesion molecules, can modulate pituitary hormone secretion by cell-cell contact. Our findings demonstrated that SYTL2, SLC38A4, and NCAM2 are new candidates for crucial functions in the secretory pathways of the pituitary gland. These results will further understanding of the important molecular mechanisms at work in the bovine pituitary.
Collapse
Affiliation(s)
- Mingxun Li
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Xubin Lu
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Hailei Xia
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Chenglong Zhang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Xiaolong Wang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Zhi Chen
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Huimin Zhang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Kaixing Qu
- Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan 650212, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan 650212, China
| | - Stephen Moore
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Australia
| | - Yongjiang Mao
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China
| | - Zhangping Yang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu province, Yangzhou University, Yangzhou 225002, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225002, China.
| |
Collapse
|
48
|
Ando M, Goto M, Hojo M, Kita A, Kitagawa M, Ohtsuka T, Kageyama R, Miyamoto S. The proneural bHLH genes Mash1, Math3 and NeuroD are required for pituitary development. J Mol Endocrinol 2018; 61:127-138. [PMID: 30307165 DOI: 10.1530/jme-18-0090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple signaling molecules and transcription factors are required for pituitary development. Activator-type bHLH genes Mash1, Math, NeuroD (Neurod) and Neurogenin (Neurog) are well known as key molecules in neural development. Although analyses of targeted mouse mutants have demonstrated involvement of these bHLH genes in pituitary development, studies with single-mutant mice could not elucidate their exact functions, because they cooperatively function and compensate each other. The aim of this study was to elucidate the roles of Mash1, Math3 and NeuroD in pituitary development. Mash1;Math3;NeuroD triple-mutant mice were analyzed by immunohistochemistry and quantitative real-time RT-PCR. Misexpression studies with retroviruses in pituisphere cultures were also performed. The triple-mutant adenohypophysis was morphologically normal, though the lumen of the neurohypophysis remained unclosed. However, in triple-mutant pituitaries, somatotropes, gonadotropes and corticotropes were severely decreased, whereas lactotropes were increased. Misexpression of Mash1 alone with retrovirus could not induce generation of hormonal cells, though Mash1 was involved in differentiation of pituitary progenitor cells. These data suggest that Mash1, Math3 and NeuroD cooperatively control the timing of pituitary progenitor cell differentiation and that they are also required for subtype specification of pituitary hormonal cells. Mash1 is necessary for corticotroph and gonadotroph differentiation, and compensated by Math3 and NeuroD. Math3 is necessary for somatotroph differentiation, and compensated by Mash1 and NeuroD. Neurog2 may compensate Mash1, Math3 and NeuroD during pituitary development. Furthermore, Mash1, Math3 and NeuroD are required for neurohypophysis development. Thus, Mash1, Math3 and NeuroD are required for pituitary development, and compensate each other.
Collapse
Affiliation(s)
- Mitsushige Ando
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masanori Goto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masato Hojo
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Neurosurgery, Shiga Medical Center for Adults, Shiga, Japan
| | - Aya Kita
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masashi Kitagawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiyuki Ohtsuka
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
49
|
Youngblood JL, Coleman TF, Davis SW. Regulation of Pituitary Progenitor Differentiation by β-Catenin. Endocrinology 2018; 159:3287-3305. [PMID: 30085028 DOI: 10.1210/en.2018-00563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
The pituitary gland is a critical organ that is necessary for many physiological processes, including growth, reproduction, and stress response. The secretion of pituitary hormones from specific cell types regulates these essential processes. Pituitary hormone cell types arise from a common pool of pituitary progenitors, and mutations that disrupt the formation and differentiation of pituitary progenitors result in hypopituitarism. Canonical WNT signaling through CTNNB1 (β-catenin) is known to regulate the formation of the POU1F1 lineage of pituitary cell types. When β-catenin is deleted during the initial formation of the pituitary progenitors, Pou1f1 is not transcribed, which leads to the loss of the POU1F1 lineage. However, when β-catenin is deleted after lineage specification, there is no observable effect. Similarly, the generation of a β-catenin gain-of-function allele in early pituitary progenitors or stem cells results in the formation of craniopharyngiomas, whereas stimulating β-catenin in differentiated cell types has no effect. PROP1 is a pituitary-specific transcription factor, and the peak of PROP1 expression coincides with a critical time point in pituitary organogenesis-that is, after pituitary progenitor formation but before lineage specification. We used a Prop1-cre to conduct both loss- and gain-of-function studies on β-catenin during this critical time point. Our results demonstrate that pituitary progenitors remain sensitive to both loss and gain of β-catenin at this time point, and that either manipulation results in hypopituitarism.
Collapse
Affiliation(s)
- Julie L Youngblood
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Tanner F Coleman
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
50
|
Gounden V, Rampursat YD, Jialal I. Secretory tumors of the pituitary gland: a clinical biochemistry perspective. ACTA ACUST UNITED AC 2018; 57:150-164. [DOI: 10.1515/cclm-2018-0552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
Abstract
The pituitary gland is responsible for the production and/or secretion of various hormones that play a vital role in regulating endocrine function within the body. Secretory tumors of the anterior pituitary predominantly, pituitary adenomas, collectively account for 10%–25% of central nervous system tumors requiring surgical treatment. The most common secretory tumors are prolactinomas, which can be diagnosed by basal prolactin levels. Acromegaly can be diagnosed by basal insulin growth-like factor 1 levels and the failure of growth hormone (GH) to suppress during an oral glucose tolerance test. Cushing disease can be diagnosed by demonstrating hypercortisolemia evidenced by increased salivary cortisol levels in the evening, increased urine free cortisol excretion and failure of plasma cortisol to suppress following oral dexamethasone given overnight (1.0 mg). We also discuss the diagnosis of the rarer thyroid-stimulating hormone and gonadotrophin secretory tumors. Morbidity is associated with tumor occurrence, clinical sequelae as well as the related medical, surgical and radiological management. This review focuses on the pathogenesis of secretory tumors of the anterior pituitary with emphasis on molecular mechanisms associated with tumorigenesis and the major role of the clinical chemistry laboratory in diagnosis and management of these tumors.
Collapse
Affiliation(s)
- Verena Gounden
- Department of Chemical Pathology , University of KwaZulu Natal and National Health Laboratory Services, Inkosi Albert Luthuli Central Hospital , Durban , South Africa
| | - Yashna D. Rampursat
- Department of Chemical Pathology , University of KwaZulu Natal and National Health Laboratory Services, Inkosi Albert Luthuli Central Hospital , Durban , South Africa
| | - Ishwarlal Jialal
- California North-State University, College of Medicine , Elk Grove, CA 95757 , USA
- Director, Section of Clinical Chemistry, VA Medical Center , Sacramento, CA , USA
| |
Collapse
|