1
|
Picci G, Petro NM, Casagrande CC, Ott LR, Okelberry HJ, Rice DL, Coutant AT, Ende GC, Steiner EL, Wang YP, Stephen JM, Calhoun VD, Wilson TW. Anterior pituitary gland volume mediates associations between adrenarche and changes in transdiagnostic symptoms in youth. Dev Cogn Neurosci 2025; 71:101507. [PMID: 39787639 DOI: 10.1016/j.dcn.2025.101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/05/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
The pituitary gland (PG) plays a central role in the production and secretion of pubertal hormones, with documented links to the increase in mental health symptoms during adolescence. Although literature has largely focused on examining whole PG volume, recent findings have demonstrated associations among pubertal hormone levels, including dehydroepiandrosterone (DHEA), PG subregions, and mental health symptoms during adolescence. Despite the anterior PG's role in DHEA production, studies have not yet examined potential links with transdiagnostic symptomology (i.e., dysregulation) pertinent to long-term functioning. Therefore, the current study sought examine whether anterior PG volume mediates associations between DHEA levels and changes in dysregulation symptoms in an adolescent sample (N = 114, 9 -17 years, Mage = 12.87, SD = 1.88). Following manual tracing, structural equation modeling revealed that greater anterior, not posterior, PG volume mediated the association between greater DHEA levels and increasing dysregulation symptoms across time, controlling for baseline dysregulation symptom levels. Results also showed that greater DHEA levels related to decreasing symptoms across time, suggesting potential attenuation effects. Altogether, these results provide support for separating the anterior and posterior PG by demonstrating specificity in the role of the anterior PG in adrenarcheal processes that may confer risk for adolescent psychopathology.
Collapse
Affiliation(s)
- Giorgia Picci
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA.
| | - Nathan M Petro
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Chloe C Casagrande
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Lauren R Ott
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Hannah J Okelberry
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Danielle L Rice
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Anna T Coutant
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Grace C Ende
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Erica L Steiner
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Yu-Ping Wang
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | | | - Vince D Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, GA, USA
| | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Center for Pediatric Brain Health, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| |
Collapse
|
2
|
Liu Z, Lin Z, Chen Y, Lu M, Hong W, Yu B, Liu G. Lipoteichoic Acid Rescued Age-Related Bone Loss by Enhancing Neuroendocrine and Growth Hormone Secretion Through TLR2/COX2/PGE2 Signalling Pathway. J Cell Mol Med 2024; 28:e70247. [PMID: 39622781 PMCID: PMC11611525 DOI: 10.1111/jcmm.70247] [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: 05/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The phenomenon of brain-bone crosstalk pertains to the intricate interaction and communication pathways between the central nervous system and the skeletal system. Disruption in brain-bone crosstalk, particularly in disorders such as osteoporosis, can result in skeletal irregularities. Consequently, investigating and comprehending this communication network holds paramount importance in the realm of bone disease prevention and management. In this study, we found that Staphylococcus aureus lipoteichoic acid promoted the conversion of arachidonic acid to PGE2 by interacting with TLR2 receptors acting on the surface of microglial cells in the pituitary gland, leading to the upregulation of COX-2 expression. Subsequently, PGE2 bound to the EP4 receptor of growth hormone-secreting cells and activated the intracellular CREB signalling pathway, promoting GH secretion and ameliorating age-related bone loss.
Collapse
Affiliation(s)
- Zixian Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- The Second Hospital and Clinical Medical SchoolLanzhou UniversityLanzhouChina
| | - Zexin Lin
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yingqi Chen
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Mincheng Lu
- Department of Orthopedic, Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Weisheng Hong
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Bin Yu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guanqiao Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
3
|
Khetchoumian K, Sochodolsky K, Lafont C, Gouhier A, Bemmo A, Kherdjemil Y, Kmita M, Le Tissier P, Mollard P, Christian H, Drouin J. Paracrine FGF1 signaling directs pituitary architecture and size. Proc Natl Acad Sci U S A 2024; 121:e2410269121. [PMID: 39320918 PMCID: PMC11459159 DOI: 10.1073/pnas.2410269121] [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: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Organ architecture is established during development through intricate cell-cell communication mechanisms, yet the specific signals mediating these communications often remain elusive. Here, we used the anterior pituitary gland that harbors different interdigitated hormone-secreting homotypic cell networks to dissect cell-cell communication mechanisms operating during late development. We show that blocking differentiation of corticotrope cells leads to pituitary hypoplasia with a major effect on somatotrope cells that directly contact corticotropes. Gene knockout of the corticotrope-restricted transcription factor Tpit results in fewer somatotropes, with less secretory granules and a loss of cell polarity, resulting in systemic growth retardation. Single-cell transcriptomic analyses identified FGF1 as a corticotrope-specific Tpit dosage-dependent target gene responsible for these phenotypes. Consistently, genetic ablation of FGF1 in mice phenocopies pituitary hypoplasia and growth impairment observed in Tpit-deficient mice. These findings reveal FGF1 produced by the corticotrope cell network as an essential paracrine signaling molecule participating in pituitary architecture and size.
Collapse
Affiliation(s)
- Konstantin Khetchoumian
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Kevin Sochodolsky
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Chrystel Lafont
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Arthur Gouhier
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Amandine Bemmo
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Yacine Kherdjemil
- Disease Modeling and Genome Editing platform, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Marie Kmita
- Laboratoire de recherche en génétique et développement, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Paul Le Tissier
- Centre for Integrative Physiology, University of Edinburgh, EdinburghEH8 9XD, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Helen Christian
- Department of Physiology, Anatomy and Genetics, Oxford University, OxfordOX1 3QX, United Kingdom
| | - Jacques Drouin
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| |
Collapse
|
4
|
Raz M, Milo T, Glass DS, Mayo A, Alon U. Endocrine gland size is proportional to its target tissue size. iScience 2024; 27:110625. [PMID: 39224518 PMCID: PMC11367476 DOI: 10.1016/j.isci.2024.110625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Endocrine glands secrete hormones into the circulation to target distant tissues and regulate their functions. The qualitative relationship between hormone-secreting organs and their target tissues is well established, but a quantitative approach is currently limited. Quantification is important, as it could allow us to study the endocrine system using engineering concepts of optimality and tradeoffs. In this study, we collected literature data on 24 human hormones secreted from dedicated endocrine cells. We find that the number of endocrine cells secreting a hormone is proportional to the number of its target cells. A single endocrine cell serves approximately 2,000 target cells, a relationship that spans 6 orders of magnitude of cell numbers. This suggests an economic principle of cells working near their maximal capacity, and glands that are no bigger than they need to be.
Collapse
Affiliation(s)
- Moriya Raz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tomer Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
5
|
Leng D, Zeng B, Wang T, Chen BL, Li DY, Li ZJ. Single nucleus/cell RNA-seq of the chicken hypothalamic-pituitary-ovarian axis offers new insights into the molecular regulatory mechanisms of ovarian development. Zool Res 2024; 45:1088-1107. [PMID: 39245652 PMCID: PMC11491784 DOI: 10.24272/j.issn.2095-8137.2024.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-ovarian (HPO) axis represents a central neuroendocrine network essential for reproductive function. Despite its critical role, the intrinsic heterogeneity within the HPO axis across vertebrates and the complex intercellular interactions remain poorly defined. This study provides the first comprehensive, unbiased, cell type-specific molecular profiling of all three components of the HPO axis in adult Lohmann layers and Liangshan Yanying chickens. Within the hypothalamus, pituitary, and ovary, seven, 12, and 13 distinct cell types were identified, respectively. Results indicated that the pituitary adenylate cyclase activating polypeptide (PACAP), follicle-stimulating hormone (FSH), and prolactin (PRL) signaling pathways may modulate the synthesis and secretion of gonadotropin-releasing hormone (GnRH), FSH, and luteinizing hormone (LH) within the hypothalamus and pituitary. In the ovary, interactions between granulosa cells and oocytes involved the KIT, CD99, LIFR, FN1, and ANGPTL signaling pathways, which collectively regulate follicular maturation. The SEMA4 signaling pathway emerged as a critical mediator across all three tissues of the HPO axis. Additionally, gene expression analysis revealed that relaxin 3 (RLN3), gastrin-releasing peptide (GRP), and cocaine- and amphetamine regulated transcripts (CART, also known as CARTPT) may function as novel endocrine hormones, influencing the HPO axis through autocrine, paracrine, and endocrine pathways. Comparative analyses between Lohmann layers and Liangshan Yanying chickens demonstrated higher expression levels of GRP, RLN3, CARTPT, LHCGR, FSHR, and GRPR in the ovaries of Lohmann layers, potentially contributing to their superior reproductive performance. In conclusion, this study provides a detailed molecular characterization of the HPO axis, offering novel insights into the regulatory mechanisms underlying reproductive biology.
Collapse
Affiliation(s)
- Dong Leng
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
| | - Bin-Long Chen
- College of Animal Science, Xichang University, Xichang, Sichuan 615000, China. E-mail:
| | - Di-Yan Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China. E-mail:
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China. E-mail:
| |
Collapse
|
6
|
Picci G, Petro NM, Casagrande CC, Ott LR, Okelberry HJ, Rice DL, Coutant AT, Ende GC, Steiner EL, Wang YP, Stephen JM, Calhoun VD, Wilson TW. Anterior pituitary gland volume mediates associations between pubertal hormones and changes in transdiagnostic symptoms in youth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594766. [PMID: 38798387 PMCID: PMC11118574 DOI: 10.1101/2024.05.17.594766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The pituitary gland (PG) plays a central role in the production and secretion of pubertal hormones, with documented links to the emergence and increase in mental health symptoms known to occur during adolescence. Although much of the literature has focused on examining whole PG volume, recent findings suggest that there are associations among pubertal hormone levels, including dehydroepiandrosterone (DHEA), subregions of the PG, and elevated mental health symptoms (e.g., internalizing symptoms) during adolescence. Surprisingly, studies have not yet examined associations among these factors and increasing transdiagnostic symptomology, despite DHEA being a primary output of the anterior PG. Therefore, the current study sought to fill this gap by examining whether anterior PG volume specifically mediates associations between DHEA levels and changes in dysregulation symptoms in an adolescent sample ( N = 114, 9 - 17 years, M age = 12.87, SD = 1.88). Following manual tracing of the anterior and posterior PG, structural equation modeling revealed that greater anterior, not posterior, PG volume mediated the association between greater DHEA levels and increasing dysregulation symptoms across time, controlling for baseline dysregulation symptom levels. These results suggest specificity in the role of the anterior PG in adrenarcheal processes that may confer risk for psychopathology during adolescence. This work not only highlights the importance of separately tracing the anterior and posterior PG, but also suggests that transdiagnostic factors like dysregulation are useful in parsing hormone-related increases in mental health symptoms in youth.
Collapse
|
7
|
Fazli M, Bertram R. Conversion of spikers to bursters in pituitary cell networks: Is it better to disperse for maximum exposure or circle the wagons? PLoS Comput Biol 2024; 20:e1011811. [PMID: 38289902 PMCID: PMC10826967 DOI: 10.1371/journal.pcbi.1011811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
The endocrine cells of the pituitary gland are electrically active, and in vivo they form small networks where the bidirectional cell-cell coupling is through gap junctions. Numerous studies of dispersed pituitary cells have shown that typical behaviors are tonic spiking and bursting, the latter being more effective at evoking secretion. In this article, we use mathematical modeling to examine the dynamics of small networks of spiking and bursting pituitary cells. We demonstrate that intrinsic bursting cells are capable of converting intrinsic spikers into bursters, and perform a fast/slow analysis to show why this occurs. We then demonstrate the sensitivity of network dynamics to the placement of bursting cells within the network, and demonstrate strategies that are most effective at maximizing secretion from the population of cells. This study provides insights into the in vivo behavior of cells such as the stress-hormone-secreting pituitary corticotrophs that are switched from spiking to bursting by hypothalamic neurohormones. While much is known about the electrical properties of these cells when isolated from the pituitary, how they behave when part of an electrically coupled network has been largely unstudied.
Collapse
Affiliation(s)
- Mehran Fazli
- Department of Mathematics, Florida State University, Tallahassee, Florida, United States of America
| | - Richard Bertram
- Department of Mathematics, Florida State University, Tallahassee, Florida, United States of America
- Programs in Neuroscience and Molecular Biophysics, Florida State University, Tallahassee, Florida, United States of America
| |
Collapse
|
8
|
Merabet M, Germain N, Redouté J, Boutet C, Costes N, Ptito M, Galusca B, Schneider FC. Structure-function relationship of the pituitary gland in anorexia nervosa and intense physical activity. Brain Struct Funct 2024; 229:195-205. [PMID: 38062204 DOI: 10.1007/s00429-023-02739-3] [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: 09/21/2023] [Accepted: 11/21/2023] [Indexed: 01/31/2024]
Abstract
Patients with Anorexia Nervosa (AN) and athletes share intense physical activity and pituitary hormonal disturbances related to absolute (AN) or relative (athletes) undernutrition. Pituitary gland (PG) structure evaluations in those conditions are scarce, and did not differentiate anterior from posterior lobe. We evaluated the structure-function relationship of anterior and posterior PG in AN and athletes, and potential reversibility of this alteration in a group of weight-recovered patients (AN_Rec). Manual delineation of anterior (AP) and posterior (PP) PG was performed on T1-weighted MR images in 17 women with AN, 15 women with AN_Rec, 18 athletes women and 25 female controls. Anthropometric, hormonal, and psychometric parameters were explored and correlated with PG volumes. AP volume (APV) was lower in AN (448 ± 82 mm3), AN_Rec (505 ± 59 mm3), and athletes (540 ± 101 mm3) vs. Controls (615 ± 61 mm3, p < 0.00001, p < 0.00001 and p = 0.02, respectively); and smaller in AN vs. AN_Rec (p = 0.007). PP volume did not show any differences between the groups. APV was positively correlated with weight (R = 0.36, p = 0.011) in AN, and luteinizing hormone (R = 0.35, p = 0.014) in total group. In AN, mean growth hormone (GH) was negatively correlated with global pituitary volume (R = 0.31, p = 0.031) and APV (R = 0.29, p = 0.037). Absolute and relative undernutrition led to a decreased anterior pituitary gland volume, which was reversible with weight gain, correlated with low bodyweight, and blockade of gonadal hypothalamic-pituitary axis. Intriguing inverse correlation between anterior pituitary gland volume and GH plasma level could suggests a low storage capacity of anterior pituitary gland and increased reactivity to low insulin-like growth factor type 1.
Collapse
Affiliation(s)
- Manel Merabet
- TAPE Research Unit, EA 7423, Jean Monnet University, Saint Etienne, France
| | - Natacha Germain
- TAPE Research Unit, EA 7423, Jean Monnet University, Saint Etienne, France.
- Eating Disorders Reference Center, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France.
- Endocrinology Department, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France.
| | | | - Claire Boutet
- TAPE Research Unit, EA 7423, Jean Monnet University, Saint Etienne, France
- Radiology Department, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France
| | | | - Maurice Ptito
- École d'Optométrie, Université de Montréal, Montréal, QC, Canada
| | - Bogdan Galusca
- TAPE Research Unit, EA 7423, Jean Monnet University, Saint Etienne, France
- Eating Disorders Reference Center, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France
- Endocrinology Department, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France
| | - Fabien C Schneider
- TAPE Research Unit, EA 7423, Jean Monnet University, Saint Etienne, France
- Radiology Department, CHU Saint Etienne, 42055, Saint Etienne Cedex 2, France
| |
Collapse
|
9
|
Picci G, Casagrande CC, Ott LR, Petro NM, Christopher‐Hayes NJ, Johnson HJ, Willett MP, Okelberry HJ, Wang Y, Stephen JM, Calhoun VD, Wilson TW. Dehydroepiandrosterone mediates associations between trauma-related symptoms and anterior pituitary volume in children and adolescents. Hum Brain Mapp 2023; 44:6388-6398. [PMID: 37853842 PMCID: PMC10681633 DOI: 10.1002/hbm.26516] [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: 10/10/2022] [Revised: 09/19/2023] [Accepted: 10/01/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION The anterior pituitary gland (PG) is a potential locus of hypothalamic-pituitary-adrenal (HPA) axis responsivity to early life stress, with documented associations between dehydroepiandrosterone (DHEA) levels and anterior PG volumes. In adults, elevated anxiety/depressive symptoms are related to diminished DHEA levels, and studies have shown a positive relationship between DHEA and anterior pituitary volumes. However, specific links between responses to stress, DHEA levels, and anterior pituitary volume have not been established in developmental samples. METHODS High-resolution T1-weighted MRI scans were collected from 137 healthy youth (9-17 years; Mage = 12.99 (SD = 1.87); 49% female; 85% White, 4% Indigenous, 1% Asian, 4% Black, 4% multiracial, 2% not reported). The anterior and posterior PGs were manually traced by trained raters. We examined the mediating effects of salivary DHEA on trauma-related symptoms (i.e., anxiety, depression, and posttraumatic) and PG volumes as well as an alternative model examining mediating effects of PG volume on DHEA and trauma-related symptoms. RESULTS DHEA mediated the association between anxiety symptoms and anterior PG volume. Specifically, higher anxiety symptoms related to lower DHEA levels, which in turn were related to smaller anterior PG. CONCLUSIONS These results shed light on the neurobiological sequelae of elevated anxiety in youth and are consistent with adult findings showing suppressed levels of DHEA in those with greater comorbid anxiety and depression. Specifically, adolescents with greater subclinical anxiety may exhibit diminished levels of DHEA during the pubertal window, which may be associated with disruptions in anterior PG growth.
Collapse
Affiliation(s)
- Giorgia Picci
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
- Center for Pediatric Brain HealthBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Chloe C. Casagrande
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Lauren R. Ott
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Nathan M. Petro
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | | | - Hallie J. Johnson
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Madelyn P. Willett
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Hannah J. Okelberry
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Yu‐Ping Wang
- Department of Biomedical EngineeringTulane UniversityNew OrleansLouisianaUSA
| | | | - Vince D. Calhoun
- Tri‐Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State University, Georgia Institute of technology, and Emory UniversityAtlantaGeorgiaUSA
| | - Tony W. Wilson
- Institute for Human NeuroscienceBoys Town National Research HospitalBoys TownNebraskaUSA
- Center for Pediatric Brain HealthBoys Town National Research HospitalBoys TownNebraskaUSA
- Department of Pharmacology & NeuroscienceCreighton UniversityOmahaNebraskaUSA
| |
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
|
Tapoi DA, Popa ML, Tanase C, Derewicz D, Gheorghișan-Gălățeanu AA. Role of Tumor Microenvironment in Pituitary Neuroendocrine Tumors: New Approaches in Classification, Diagnosis and Therapy. Cancers (Basel) 2023; 15:5301. [PMID: 37958474 PMCID: PMC10649263 DOI: 10.3390/cancers15215301] [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: 09/29/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Adenohypophysal pituitary tumors account for 10-15% of all intracranial tumors, and 25-55% display signs of invasiveness. Nevertheless, oncology still relies on histopathological examination to establish the diagnosis. Considering that the classification of pituitary tumors has changed significantly in recent years, we discuss the definition of aggressive and invasive tumors and the latest molecular criteria used for classifying these entities. The pituitary tumor microenvironment (TME) is essential for neoplastic development and progression. This review aims to reveal the impact of TME characteristics on stratifying these tumors in view of finding appropriate therapeutic approaches. The role of the pituitary tumor microenvironment and its main components, non-tumoral cells and soluble factors, has been addressed. The variable display of different immune cell types, tumor-associated fibroblasts, and folliculostellate cells is discussed in relation to tumor development and aggressiveness. The molecules secreted by both tumoral and non-tumoral cells, such as VEGF, FGF, EGF, IL6, TNFα, and immune checkpoint molecules, contribute to the crosstalk between the tumor and its microenvironment. They could be considered potential biomarkers for diagnosis and the invasiveness of these tumors, together with emerging non-coding RNA molecules. Therefore, assessing this complex network associated with pituitary neuroendocrine tumors could bring a new era in diagnosing and treating this pathology.
Collapse
Affiliation(s)
- Dana Antonia Tapoi
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Maria-Linda Popa
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Cell Biology and Clinical Biochemistry, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Diana Derewicz
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pediatric Hematology and Oncology, Marie Sklodowska Curie Clinical Emergency Hospital, 041447 Bucharest, Romania
| | - Ancuța-Augustina Gheorghișan-Gălățeanu
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
| |
Collapse
|
12
|
Hymer WC, Kraemer WJ. Resistance exercise stress: theoretical mechanisms for growth hormone processing and release from the anterior pituitary somatotroph. Eur J Appl Physiol 2023; 123:1867-1878. [PMID: 37421488 DOI: 10.1007/s00421-023-05263-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/15/2023] [Indexed: 07/10/2023]
Abstract
Heavy resistance exercise (HRE) is the most effective method for inducing muscular hypertrophy and stimulating anabolic hormones, including growth hormone, into the blood. In this review, we explore possible mechanisms within the GH secretory pathway of the pituitary somatotroph, which are likely to modulate the flow of hormone synthesis and packaging as it is processed prior to exocytosis. Special emphasis is placed on the secretory granule and its possible role as a signaling hub. We also review data that summarize how HRE affects the quality and quantity of the secreted hormone. Finally, these pathway mechanisms are considered in the context of heterogeneity of the somatotroph population in the anterior pituitary.
Collapse
Affiliation(s)
- Wesley C Hymer
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - William J Kraemer
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43802, USA.
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA.
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.
| |
Collapse
|
13
|
Maita F, Maiolo L, Lucarini I, Del Rio De Vicente JI, Sciortino A, Ledda M, Mussi V, Lisi A, Convertino A. Revealing Low Amplitude Signals of Neuroendocrine Cells through Disordered Silicon Nanowires-Based Microelectrode Array. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301925. [PMID: 37357140 PMCID: PMC10460871 DOI: 10.1002/advs.202301925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Today, the key methodology to study in vitro or in vivo electrical activity in a population of electrogenic cells, under physiological or pathological conditions, is by using microelectrode array (MEA). While significant efforts have been devoted to develop nanostructured MEAs for improving the electrophysiological investigation in neurons and cardiomyocytes, data on the recording of the electrical activity from neuroendocrine cells with MEA technology are scarce owing to their weaker electrical signals. Disordered silicon nanowires (SiNWs) for developing a MEA that, combined with a customized acquisition board, successfully capture the electrical signals generated by the corticotrope AtT-20 cells as a function of the extracellular calcium (Ca2+ ) concentration are reported. The recorded signals show a shape that clearly resembles the action potential waveform by suggesting a natural membrane penetration of the SiNWs. Additionally, the generation of synchronous signals observed under high Ca2+ content indicates the occurrence of a collective behavior in the AtT-20 cell population. This study extends the usefulness of MEA technology to the investigation of the electrical communication in cells of the pituitary gland, crucial in controlling several essential human functions, and provides new perspectives in recording with MEA the electrical activity of excitable cells.
Collapse
Affiliation(s)
- Francesco Maita
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Luca Maiolo
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Ivano Lucarini
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | | | - Antonio Sciortino
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Mario Ledda
- Institute of Translational PharmacologyNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Valentina Mussi
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Antonella Lisi
- Institute of Translational PharmacologyNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| | - Annalisa Convertino
- Institute for Microelectronics and MicrosystemsNational Research CouncilVia Fosso del Cavaliere 100Rome00133Italy
| |
Collapse
|
14
|
Luigi-Sierra MG, Guan D, López-Béjar M, Casas E, Olvera-Maneu S, Gardela J, Palomo MJ, Osuagwuh UI, Ohaneje UL, Mármol-Sánchez E, Amills M. A protein-coding gene expression atlas from the brain of pregnant and non-pregnant goats. Front Genet 2023; 14:1114749. [PMID: 37519888 PMCID: PMC10382233 DOI: 10.3389/fgene.2023.1114749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background: The brain is an extraordinarily complex organ with multiple anatomical structures involved in highly specialized functions related with behavior and physiological homeostasis. Our goal was to build an atlas of protein-coding gene expression in the goat brain by sequencing the transcriptomes of 12 brain regions in seven female Murciano-Granadina goats, from which three of them were 1-month pregnant. Results: Between 14,889 (cerebellar hemisphere) and 15,592 (pineal gland) protein-coding genes were expressed in goat brain regions, and most of them displayed ubiquitous or broad patterns of expression across tissues. Principal component analysis and hierarchical clustering based on the patterns of mRNA expression revealed that samples from certain brain regions tend to group according to their position in the anterior-posterior axis of the neural tube, i.e., hindbrain (pons and medulla oblongata), midbrain (rostral colliculus) and forebrain (frontal neocortex, olfactory bulb, hypothalamus, and hippocampus). Exceptions to this observation were cerebellum and glandular tissues (pineal gland and hypophysis), which showed highly divergent mRNA expression profiles. Differential expression analysis between pregnant and non-pregnant goats revealed moderate changes of mRNA expression in the frontal neocortex, hippocampus, adenohypophysis and pons, and very dramatic changes in the olfactory bulb. Many genes showing differential expression in this organ are related to olfactory function and behavior in humans. Conclusion: With the exception of cerebellum and glandular tissues, there is a relationship between the cellular origin of sampled regions along the anterior-posterior axis of the neural tube and their mRNA expression patterns in the goat adult brain. Gestation induces substantial changes in the mRNA expression of the olfactory bulb, a finding consistent with the key role of this anatomical structure on the development of maternal behavior.
Collapse
Affiliation(s)
| | - Dailu Guan
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Manel López-Béjar
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Encarna Casas
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Sergi Olvera-Maneu
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jaume Gardela
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María Jesús Palomo
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchebuchi Ike Osuagwuh
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchechi Linda Ohaneje
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Emilio Mármol-Sánchez
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
15
|
Götz V, Qiao S, Das D, Wartenberg P, Wyatt A, Wahl V, Gamayun I, Alasmi S, Fecher-Trost C, Meyer MR, Rad R, Kaltenbacher T, Kattler K, Lipp P, Becherer U, Mollard P, Candlish M, Boehm U. Ovulation is triggered by a cyclical modulation of gonadotropes into a hyperexcitable state. Cell Rep 2023; 42:112543. [PMID: 37224016 DOI: 10.1016/j.celrep.2023.112543] [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: 09/17/2018] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Gonadotropes in the anterior pituitary gland are essential for fertility and provide a functional link between the brain and the gonads. To trigger ovulation, gonadotrope cells release massive amounts of luteinizing hormone (LH). The mechanism underlying this remains unclear. Here, we utilize a mouse model expressing a genetically encoded Ca2+ indicator exclusively in gonadotropes to dissect this mechanism in intact pituitaries. We demonstrate that female gonadotropes exclusively exhibit a state of hyperexcitability during the LH surge, resulting in spontaneous [Ca2+]i transients in these cells, which persist in the absence of any in vivo hormonal signals. L-type Ca2+ channels and transient receptor potential channel A1 (TRPA1) together with intracellular reactive oxygen species (ROS) levels ensure this state of hyperexcitability. Consistent with this, virus-assisted triple knockout of Trpa1 and L-type Ca2+ subunits in gonadotropes leads to vaginal closure in cycling females. Our data provide insight into molecular mechanisms required for ovulation and reproductive success in mammals.
Collapse
Affiliation(s)
- Viktoria Götz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Sen Qiao
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Debajyoti Das
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Philipp Wartenberg
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Amanda Wyatt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Vanessa Wahl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Igor Gamayun
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Samer Alasmi
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Claudia Fecher-Trost
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Markus R Meyer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Thorsten Kaltenbacher
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Kathrin Kattler
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken 66123, Germany
| | - Peter Lipp
- Molecular Cell Biology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Ute Becherer
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University School of Medicine, Homburg 66421, Germany
| | - Patrice Mollard
- IGF, CNRS, INSERM, University of Montpellier, Montpellier 34090, France
| | - Michael Candlish
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany
| | - Ulrich Boehm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg 66421, Germany.
| |
Collapse
|
16
|
Baker EC, San AE, Cilkiz KZ, Littlejohn BP, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Welsh TH, Riley DG. Inter-Individual Variation in DNA Methylation Patterns across Two Tissues and Leukocytes in Mature Brahman Cattle. BIOLOGY 2023; 12:biology12020252. [PMID: 36829529 PMCID: PMC9953534 DOI: 10.3390/biology12020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
Abstract
Quantifying the natural inter-individual variation in DNA methylation patterns is important for identifying its contribution to phenotypic variation, but also for understanding how the environment affects variability, and for incorporation into statistical analyses. The inter-individual variation in DNA methylation patterns in female cattle and the effect that a prenatal stressor has on such variability have yet to be quantified. Thus, the objective of this study was to utilize methylation data from mature Brahman females to quantify the inter-individual variation in DNA methylation. Pregnant Brahman cows were transported for 2 h durations at days 60 ± 5; 80 ± 5; 100 ± 5; 120 ± 5; and 140 ± 5 of gestation. A non-transport group was maintained as a control. Leukocytes, amygdala, and anterior pituitary glands were harvested from eight cows born from the non-transport group (Control) and six from the transport group (PNS) at 5 years of age. The DNA harvested from the anterior pituitary contained the greatest variability in DNA methylation of cytosine-phosphate-guanine (mCpG) sites from both the PNS and Control groups, and the amygdala had the least. Numerous variable mCpG sites were associated with retrotransposable elements and highly repetitive regions of the genome. Some of the genomic features that had high variation in DNA methylation are involved in immune responses, signaling, responses to stimuli, and metabolic processes. The small overlap of highly variable CpG sites and features between tissues and leukocytes supports the role of variable DNA methylation in regulating tissue-specific gene expression. Many of the CpG sites that exhibited high variability in DNA methylation were common between the PNS and Control groups within a tissue, but there was little overlap in genomic features with high variability. The interaction between the prenatal environment and the genome could be responsible for the differences in location of the variable DNA methylation.
Collapse
Affiliation(s)
- Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - Audrey E. San
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Texas A&M AgriLife Research, College Station, TX 77845, USA
- Texas A&M AgriLife Research & Extension Center at Overton, Overton, TX 75684, USA
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - Brittni P. Littlejohn
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Texas A&M AgriLife Research & Extension Center at Overton, Overton, TX 75684, USA
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Texas A&M AgriLife Research & Extension Center at Overton, Overton, TX 75684, USA
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Texas A&M AgriLife Research & Extension Center at Overton, Overton, TX 75684, USA
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Texas A&M AgriLife Research, College Station, TX 77845, USA
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
- Correspondence:
| |
Collapse
|
17
|
Rahmad Royan M, Siddique K, Nourizadeh-Lillabadi R, Weltzien FA, Henkel C, Fontaine R. Functional and developmental heterogeneity of pituitary lactotropes in medaka. Gen Comp Endocrinol 2023; 330:114144. [PMID: 36270338 DOI: 10.1016/j.ygcen.2022.114144] [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: 07/19/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
In fish, prolactin-producing cells (lactotropes) are located in the anterior part of the pituitary and play an essential role in osmoregulation. However, small satellite lactotrope clusters have been described in other parts of the pituitary in several species. The functional and developmental backgrounds of these satellite clusters are not known. We recently discovered two distinct prolactin-expressing cell types in Japanese medaka (Oryzias latipes), a euryhaline species, using single cell transcriptomics. In the present study, we characterize these two transcriptomically distinct lactotrope cell types and explore the hypothesis that they represent spatially distinct cell clusters, as found in other species. Single cell RNA sequencing shows that one of the two lactotrope cell types exhibits an expression profile similar to that of stem cell-like folliculo-stellate cell populations. Using in situ hybridization, we show that the medaka pituitary often develops additional small satellite lactotrope cell clusters, like in other teleost species. These satellite clusters arise early during development and grow in cell number throughout life regardless of the animal's sex. Surprisingly, our data do not show a correspondence between the stem cell-like lactotropes and these satellite lactotrope clusters. Instead, our data support a scenario in which the stem cell-like lactotropes are an intrinsic stage in the development of every spatially distinct lactotrope cluster. In addition, lactotrope activity in both spatially distinct lactotrope clusters decreases when environmental salinity increases, supporting their role in osmoregulation. However, this decrease appears weaker in the satellite lactotrope cell clusters, suggesting that these lactotropes are regulated differently.
Collapse
Affiliation(s)
- Muhammad Rahmad Royan
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Khadeeja Siddique
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | | | - Finn-Arne Weltzien
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Christiaan Henkel
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway.
| | - Romain Fontaine
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
18
|
Jiménez-Díaz E, Del-Rio D, Fiordelisio T. The Contribution of Cell Imaging to the Study of Anterior Pituitary Function and Its Regulation. Neuroendocrinology 2023; 113:179-192. [PMID: 35231920 DOI: 10.1159/000523860] [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: 07/08/2021] [Accepted: 02/18/2022] [Indexed: 11/19/2022]
Abstract
Advances in the knowledge of the neuroendocrine system are closely related to the development of cellular imaging and labeling techniques. This synergy ranges from the staining techniques that allowed the first characterizations of the anterior pituitary gland, its relationship with the hypothalamus, and the birth of neuroendocrinology; through the development of fluorescence microscopy applications, specific labeling strategies, transgenic systems, and intracellular calcium sensors that enabled the study of processes and dynamics at the cellular and tissue level; until the advent of super-resolution microscopy, miniscopes, optogenetics, fiber photometry, and other imaging methods that allowed high spatiotemporal resolution and long-term three-dimensional cellular activity recordings in living systems in a conscious and freely moving condition. In this review, we briefly summarize the main contributions of cellular imaging techniques that have allowed relevant advances in the field of neuroendocrinology and paradigm shifts that have improved our understanding of the function of the hypothalamic-pituitary axes. The development of these methods and equipment is the result of the integration of knowledge achieved by the integration of several disciplines and effort to solve scientific questions and problems of high impact on health and society that this system entails.
Collapse
Affiliation(s)
- Edgar Jiménez-Díaz
- Laboratorio de Neuroendocrinología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Diana Del-Rio
- Laboratorio de Neuroendocrinología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Tatiana Fiordelisio
- Laboratorio de Neuroendocrinología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia LaNSBioDyT, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
19
|
Willis TL, Lodge EJ, Andoniadou CL, Yianni V. Cellular interactions in the pituitary stem cell niche. Cell Mol Life Sci 2022; 79:612. [PMID: 36451046 PMCID: PMC9712314 DOI: 10.1007/s00018-022-04612-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022]
Abstract
Stem cells in the anterior pituitary gland can give rise to all resident endocrine cells and are integral components for the appropriate development and subsequent maintenance of the organ. Located in discreet niches within the gland, stem cells are involved in bi-directional signalling with their surrounding neighbours, interactions which underpin pituitary gland homeostasis and response to organ challenge or physiological demand. In this review we highlight core signalling pathways that steer pituitary progenitors towards specific endocrine fate decisions throughout development. We further elaborate on those which are conserved in the stem cell niche postnatally, including WNT, YAP/TAZ and Notch signalling. Furthermore, we have collated a directory of single cell RNA sequencing studies carried out on pituitaries across multiple organisms, which have the potential to provide a vast database to study stem cell niche components in an unbiased manner. Reviewing published data, we highlight that stem cells are one of the main signalling hubs within the anterior pituitary. In future, coupling single cell sequencing approaches with genetic manipulation tools in vivo, will enable elucidation of how previously understudied signalling pathways function within the anterior pituitary stem cell niche.
Collapse
Affiliation(s)
- Thea L Willis
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Val Yianni
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
20
|
Fontaine R, Rahmad Royan M, Henkel C, Hodne K, Ager-Wick E, Weltzien FA. Pituitary multi-hormone cells in mammals and fish: history, origin, and roles. Front Neuroendocrinol 2022; 67:101018. [PMID: 35870647 DOI: 10.1016/j.yfrne.2022.101018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022]
Abstract
The vertebrate pituitary is a dynamic organ, capable of adapting its hormone secretion to different physiological demands. In this context, endocrinologists have debated for the past 40 years if endocrine cells are mono- or multi-hormonal. Since its establishment, the dominant "one cell, one hormone" model has been continuously challenged. In mammals, the use of advanced multi-staining approaches, sensitive gene expression techniques, and the analysis of tumor tissues have helped to quickly demonstrate the existence of pituitary multi-hormone cells. In fishes however, only recent advances in imaging and transcriptomics have enabled the identification of such cells. In this review, we first describe the history of the discovery of cells producing multiple hormones in mammals and fishes. We discuss the technical limitations that have led to uncertainties and debates. Then, we present the current knowledge and hypotheses regarding their origin and biological role, which provides a comprehensive review of pituitary plasticity.
Collapse
Affiliation(s)
- Romain Fontaine
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway.
| | - Muhammad Rahmad Royan
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Christiaan Henkel
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Kjetil Hodne
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Eirill Ager-Wick
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Finn-Arne Weltzien
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
21
|
Le Tissier PR, Murray JF, Mollard P. A New Perspective on Regulation of Pituitary Plasticity: The Network of SOX2-Positive Cells May Coordinate Responses to Challenge. Endocrinology 2022; 163:6609891. [PMID: 35713880 PMCID: PMC9273012 DOI: 10.1210/endocr/bqac089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Indexed: 11/19/2022]
Abstract
Plasticity of function is required for each of the anterior pituitary endocrine axes to support alterations in the demand for hormone with physiological status and in response to environmental challenge. This plasticity is mediated at the pituitary level by a change in functional cell mass resulting from a combination of alteration in the proportion of responding cells, the amount of hormone secreted from each cell, and the total number of cells within an endocrine cell population. The functional cell mass also depends on its organization into structural and functional networks. The mechanisms underlying alteration in gland output depend on the strength of the stimulus and are axis dependent but in all cases rely on sensing of output of the functional cell mass and its regulation. Here, we present evidence that the size of pituitary cell populations is constrained and suggest this is mediated by a form of quorum sensing. We propose that pituitary cell quorum sensing is mediated by interactions between the networks of endocrine cells and hormone-negative SOX2-positive (SOX2+ve) cells and speculate that the latter act as both a sentinel and actuator of cell number. Evidence for a role of the network of SOX2+ve cells in directly regulating secretion from multiple endocrine cell networks suggests that it also regulates other aspects of the endocrine cell functional mass. A decision-making role of SOX2+ve cells would allow precise coordination of pituitary axes, essential for their appropriate response to physiological status and challenge, as well as prioritization of axis modification.
Collapse
Affiliation(s)
- Paul R Le Tissier
- Correspondence: Paul R. Le Tissier, PhD, Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Bldg, 15 George Square, Edinburgh EH8 9XD, UK.
| | - Joanne F Murray
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Patrice Mollard
- Correspondence: Patrice Mollard, PhD, Institute of Functional Genomics, University of Montpellier, 141 rue de la Cardonille, F-34093, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
22
|
Zhang J, Zhou Y, Guo J, Li L, Liu H, Lu C, Jiang Y, Cui S. MicroRNA-7a2 is required for the development of pituitary stem cells. Stem Cells Dev 2022; 31:357-368. [PMID: 35652338 DOI: 10.1089/scd.2022.0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The pituitary gland is inhabited by a subpopulation of SOX2+ stem cells. However, the regulatory mechanisms underlying pituitary stem cell development remain poorly understood. Here, we demonstrate that microRNA-7a (miR-7a) is enriched in the developing pituitary and is spatiotemporally expressed in the pituitary stem cells. Constitutive deletion of miR-7a2 in mice results in pituitary dysplasia emerging during birth, which is primarily manifested as malformed anterior lobes. Using immunofluorescence, immunohistochemistry or in situ hybridization, we observe that the specification of hormone-expressing cells is not impeded post miR-7a2 deletion at birth, although the terminal differentiation of gonadotropes is inhibited. Further investigation of neonatal and adult pituitaries in miR-7a2 knockout mice reveals an expansion of the SOX2+ pituitary stem cell compartment. The inhibition of epithelial-mesenchymal like transition seems to be responsible for this phenotype, rather than abnormal proliferation or apoptosis. Furthermore, our data suggest that Gli3 and Ckap4 are potential targets of miR-7a in pituitary stem cells. In summary, our results identify miR-7a2 as a crucial factor involved in pituitary stem cell development.
Collapse
Affiliation(s)
- Jinglin Zhang
- Yangzhou University, 38043, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China;
| | - Yewen Zhou
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China;
| | - Jiajia Guo
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Liuhui Li
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Hui Liu
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Chenyang Lu
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Ying Jiang
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Sheng Cui
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China;
| |
Collapse
|
23
|
Churilov AN, Milton JG. Modeling pulsativity in the hypothalamic-pituitary-adrenal hormonal axis. Sci Rep 2022; 12:8480. [PMID: 35589935 PMCID: PMC9120490 DOI: 10.1038/s41598-022-12513-w] [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: 09/24/2021] [Accepted: 05/04/2022] [Indexed: 11/18/2022] Open
Abstract
A new mathematical model for biological rhythms in the hypothalamic–pituitary–adrenal (HPA) axis is proposed. This model takes the form of a system of impulsive time-delay differential equations which include pulsatile release of adrenocorticotropin (ACTH) by the pituitary gland and a time delay for the release of glucocorticoid hormones by the adrenal gland. Numerical simulations demonstrate that the model’s response to periodic and circadian inputs from the hypothalamus are consistent with those generated by recent models which do not include a pulsatile pituitary. In contrast the oscillatory phenomena generated by the impulsive delay equation mode occur even if the time delay is zero. The observation that the time delay merely introduces a small phase shift suggesting that the effects of the adrenal gland are “downstream” to the origin of pulsativity. In addition, the model accounts for the occurrence of ultradian oscillations in an isolated pituitary gland. These observations suggest that principles of pulse modulated control, familiar to control engineers, may have an increasing role to play in understanding the HPA axis.
Collapse
Affiliation(s)
- Alexander N Churilov
- Faculty of Mathematics and Mechanics, Saint Petersburg State University, Saint Petersburg, Russia
| | - John G Milton
- W. M. Keck Science Center, The Claremont Colleges, Claremont, CA, USA.
| |
Collapse
|
24
|
Nunes B, Pópulo H, Lopes JM, Reis M, Nascimento G, Nascimento AG, Fernandes J, Faria M, de Carvalho DP, Soares P, Miranda-Alves L. Connexin Expression in Pituitary Adenomas and the Effects of Overexpression of Connexin 43 in Pituitary Tumor Cell Lines. Genes (Basel) 2022; 13:genes13040674. [PMID: 35456480 PMCID: PMC9032236 DOI: 10.3390/genes13040674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Gap junction intercellular communication (GJIC) is considered a key mechanism in the regulation of tissue homeostasis. GJIC structures are organized in two transmembrane channels, with each channel formed by connexins (Cxs). GJIC and Cxs expression alterations are related to the process of tumorigenesis in different cell types. Pituitary neuroendocrine tumors (PitNETs) represent 15–20% of intracranial neoplasms, and usually display benign behavior. Nevertheless, some may have aggressive behavior, invading adjacent tissues, and featuring a high proliferation rate. We aimed to assess the expression and relevance of GJIC and Cxs proteins in PitNETs. We evaluated the mRNA expression levels of Cx26, 32, and 43, and the protein expression of Cx43 in a series of PitNETs. In addition, we overexpressed Cx43 in pituitary tumor cell lines. At the mRNA level, we observed variable expression of all the connexins in the tumor samples. Cx43 protein expression was absent in most of the pituitary tumor samples that were studied. Moreover, in vitro studies revealed that the overexpression of Cx43 decreases cell growth and induces apoptosis in pituitary tumor cell lines. Our results indicate that the downregulation of Cx43 protein might be involved in the tumorigenesis of most pituitary adenomas and have a potential therapeutic value for pituitary tumor therapy.
Collapse
Affiliation(s)
- Bruno Nunes
- Laboratory of Experimental Endocrinology—LEEx, Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (B.N.); (D.P.d.C.); (L.M.-A.)
- Postgraduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Laboratory of Endocrine Physiology, Doris Rosenthal, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena Pópulo
- Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (H.P.); (J.M.L.); (M.R.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)—Cancer Signalling & Metabolism, 4200-135 Porto, Portugal
- Department of Pathology, Medical Faculty of the University of Porto, 4200-319 Porto, Portugal
| | - José Manuel Lopes
- Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (H.P.); (J.M.L.); (M.R.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)—Cancer Signalling & Metabolism, 4200-135 Porto, Portugal
- Department of Pathology, Medical Faculty of the University of Porto, 4200-319 Porto, Portugal
| | - Marta Reis
- Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (H.P.); (J.M.L.); (M.R.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)—Cancer Signalling & Metabolism, 4200-135 Porto, Portugal
| | - Gilvan Nascimento
- Centre of Clinical Research (CEPEC), President Dutra Hospital of Federal University of Maranhão (UFMA), São Luís 65020-600, Brazil; (G.N.); (M.F.)
- Endocrinology Service, President Dutra Hospital of Federal University of Maranhão (UFMA), São Luís 65060-600, Brazil
| | - Ana Giselia Nascimento
- Pathology Service, President Dutra Hospital of Federal University of Maranhão (UFMA), São Luís 65020-070, Brazil;
| | - Janaína Fernandes
- NUPEX, Polo Duque de Caxias, Universidade Federal do Rio de Janeiro, Rio de Janeiro 25240-005, Brazil;
| | - Manuel Faria
- Centre of Clinical Research (CEPEC), President Dutra Hospital of Federal University of Maranhão (UFMA), São Luís 65020-600, Brazil; (G.N.); (M.F.)
- Endocrinology Service, President Dutra Hospital of Federal University of Maranhão (UFMA), São Luís 65060-600, Brazil
| | - Denise Pires de Carvalho
- Laboratory of Experimental Endocrinology—LEEx, Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (B.N.); (D.P.d.C.); (L.M.-A.)
- Postgraduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Laboratory of Endocrine Physiology, Doris Rosenthal, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Paula Soares
- Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (H.P.); (J.M.L.); (M.R.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)—Cancer Signalling & Metabolism, 4200-135 Porto, Portugal
- Department of Pathology, Medical Faculty of the University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology—LEEx, Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (B.N.); (D.P.d.C.); (L.M.-A.)
- Postgraduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Laboratory of Endocrine Physiology, Doris Rosenthal, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Postgraduate Program in Pharmacology and Medicinal Chemistry, Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
25
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
26
|
Xiong J, Zhang H, Wang Y, Cheng Y, Luo J, Chen T, Xi Q, Sun J, Zhang Y. Rno_circ_0001004 Acts as a miR-709 Molecular Sponge to Regulate the Growth Hormone Synthesis and Cell Proliferation. Int J Mol Sci 2022; 23:ijms23031413. [PMID: 35163336 PMCID: PMC8835962 DOI: 10.3390/ijms23031413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: As a novel type of non-coding RNA with a stable closed-loop structure, circular RNA (circRNA) can interact with microRNA (miRNA) and influence the expression of miRNA target genes. However, circRNA involved in pituitary growth hormone (GH) regulation is poorly understood. Our previous study revealed protein kinase C alpha (PRKCA) as the target gene of miR-709. Currently, the expression and function of rno_circRNA_0001004 in the rat pituitary gland is not clarified; (2) Methods: In this study, both bioinformatics analysis and dual-luciferase report assays showed a target relationship between rno_circRNA_0001004 and miR-709. Furthermore, the rno_circRNA_0001004 overexpression vector and si-circ_0001004 were constructed and transfected into GH3 cells; (3) Results: We found that rno_circRNA_0001004 expression was positively correlated with the PRKCA gene and GH expression levels, while it was negatively correlated with miR-709. In addition, overexpression of rno-circ_0001004 also promoted proliferation and relieved the inhibition of miR-709 in GH3 cells; (4) Conclusions: Our findings show that rno_circ_0001004 acts as a novel sponge for miR-709 to regulate GH synthesis and cell proliferation, and are the first case of discovery of the regulatory role of circRNA_0001004 in pituitary GH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jiajie Sun
- Correspondence: (J.S.); (Y.Z.); Tel.: +86-139-2515-8841 (J.S.); +86-135-2780-3004 (Y.Z.)
| | - Yongliang Zhang
- Correspondence: (J.S.); (Y.Z.); Tel.: +86-139-2515-8841 (J.S.); +86-135-2780-3004 (Y.Z.)
| |
Collapse
|
27
|
Wang TY, Xia FY, Gong JW, Xu XK, Lv MC, Chatoo M, Shamsi BH, Zhang MC, Liu QR, Liu TX, Zhang DD, Lu XJ, Zhao Y, Du JZ, Chen XQ. CRHR1 mediates the transcriptional expression of pituitary hormones and their receptors under hypoxia. Front Endocrinol (Lausanne) 2022; 13:893238. [PMID: 36147561 PMCID: PMC9487150 DOI: 10.3389/fendo.2022.893238] [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: 03/10/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Hypothalamus-pituitary-adrenal (HPA) axis plays critical roles in stress responses under challenging conditions such as hypoxia, via regulating gene expression and integrating activities of hypothalamus-pituitary-targets cells. However, the transcriptional regulatory mechanisms and signaling pathways of hypoxic stress in the pituitary remain to be defined. Here, we report that hypoxia induced dynamic changes in the transcription factors, hormones, and their receptors in the adult rat pituitary. Hypoxia-inducible factors (HIFs), oxidative phosphorylation, and cAMP signaling pathways were all differentially enriched in genes induced by hypoxic stress. In the pituitary gene network, hypoxia activated c-Fos and HIFs with specific pituitary transcription factors (Prop1), targeting the promoters of hormones and their receptors. HIF and its related signaling pathways can be a promising biomarker during acute or constant hypoxia. Hypoxia stimulated the transcription of marker genes for microglia, chemokines, and cytokine receptors of the inflammatory response. Corticotropin-releasing hormone receptor 1 (CRHR1) mediated the transcription of Pomc, Sstr2, and Hif2a, and regulated the function of HPA axis. Together with HIF, c-Fos initiated and modulated dynamic changes in the transcription of hormones and their receptors. The receptors were also implicated in the regulation of functions of target cells in the pituitary network under hypoxic stress. CRHR1 played an integrative role in the hypothalamus-pituitary-target axes. This study provides new evidence for CRHR1 involved changes of hormones, receptors, signaling molecules and pathways in the pituitary induced by hypoxia.
Collapse
Affiliation(s)
- Tong Ying Wang
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Research and Development, Jiuyuan Gene Engineering, Hangzhou, China
| | - Fang Yuan Xia
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jing Wen Gong
- Department of Pathology, and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Kang Xu
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Min Chao Lv
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Mahanand Chatoo
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Bilal Haider Shamsi
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Meng Chen Zhang
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Qian Ru Liu
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Tian Xing Liu
- Department of Cell and System Biology, University of Toronto, St. George, NB, Canada
| | - Dan Dan Zhang
- Department of Pathology, and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Jiang Lu
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Zhao
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ji Zeng Du
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xue Qun Chen
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
28
|
Fazli M, Bertram R. Network Properties of Electrically Coupled Bursting Pituitary Cells. Front Endocrinol (Lausanne) 2022; 13:936160. [PMID: 35872987 PMCID: PMC9299381 DOI: 10.3389/fendo.2022.936160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/01/2022] [Indexed: 12/03/2022] Open
Abstract
The endocrine cells of the anterior pituitary gland are electrically active when stimulated or, in some cases, when not inhibited. The activity pattern thought to be most effective in releasing hormones is bursting, which consists of depolarization with small spikes that are much longer than single spikes. Although a majority of the research on cellular activity patterns has been performed on dispersed cells, the environment in situ is characterized by networks of coupled cells of the same type, at least in the case of somatotrophs and lactotrophs. This produces some degree of synchronization of their activity, which can be greatly increased by hormones and changes in the physiological state. In this computational study, we examine how electrical coupling among model cells influences synchronization of bursting oscillations among the population. We focus primarily on weak electrical coupling, since strong coupling leads to complete synchronization that is not characteristic of pituitary cell networks. We first look at small networks to point out several unexpected behaviors of the coupled system, and then consider a larger random scale-free network to determine what features of the structural network formed through gap junctional coupling among cells produce a high degree of functional coupling, i.e., clusters of synchronized cells. We employ several network centrality measures, and find that cells that are closely related in terms of their closeness centrality are most likely to be synchronized. We also find that structural hubs (cells with extensive coupling to other cells) are typically not functional hubs (cells synchronized with many other cells). Overall, in the case of weak electrical coupling, it is hard to predict the functional network that arises from a structural network, or to use a functional network as a means for determining the structural network that gives rise to it.
Collapse
Affiliation(s)
- Mehran Fazli
- Department of Mathematics, Florida State University, Tallahassee, FL, United States
| | - Richard Bertram
- Department of Mathematics, Florida State University, Tallahassee, FL, United States
- Programs in Neuroscience and Molecular Biophysics, Florida State University, Tallahassee, FL, United States
- *Correspondence: Richard Bertram,
| |
Collapse
|
29
|
Xiong J, Zhang H, Zeng B, Liu J, Luo J, Chen T, Sun J, Xi Q, Zhang Y. An Exploration of Non-Coding RNAs in Extracellular Vesicles Delivered by Swine Anterior Pituitary. Front Genet 2021; 12:772753. [PMID: 34912377 PMCID: PMC8667663 DOI: 10.3389/fgene.2021.772753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/01/2021] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles are lipid bilayer-delimited particles carrying proteins, lipids, and small RNAs. Previous studies have demonstrated that they had regulatory functions both physiologically and pathologically. However, information remains inadequate on extracellular vesicles from the anterior pituitary, a key endocrine organ in animals and humans. In this study, we separated and identified extracellular vesicles from the anterior pituitary of the Duroc swine model. Total RNA was extracted and RNA-seq was performed, followed by a comprehensive analysis of miRNAs, lncRNAs, and circRNAs. Resultantly, we obtained 416 miRNAs, 16,232 lncRNAs, and 495 circRNAs. Furthermore, GO and KEGG enrichment analysis showed that the ncRNAs in extracellular vesicles may participate in regulating intracellular signal transduction, cellular component organization or biogenesis, small molecule binding, and transferase activity. The cross-talk between them also suggested that they may play an important role in the signaling process and biological regulation. This is the first report of ncRNA data in the anterior pituitary extracellular vesicles from the duroc swine breed, which is a fundamental resource for exploring detailed functions of extracellular vesicles from the anterior pituitary.
Collapse
Affiliation(s)
- Jiali Xiong
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Haojie Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Bin Zeng
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Jie Liu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Junyi Luo
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Jiajie Sun
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| |
Collapse
|
30
|
Claudin-9 constitutes tight junctions of folliculo-stellate cells in the anterior pituitary gland. Sci Rep 2021; 11:21642. [PMID: 34737342 PMCID: PMC8568902 DOI: 10.1038/s41598-021-01004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/20/2021] [Indexed: 12/27/2022] Open
Abstract
The anterior pituitary gland regulates growth, metabolism, and reproduction by secreting hormones. Folliculo-stellate (FS) cells are non-endocrine cells located among hormone-producing cells in the anterior pituitary glands. They form follicular lumens, which are sealed by tight junctions (TJs). Although FS cells are hypothesized to contribute to fine-tuning of endocrine cells, little is known about the exact roles of FS cells. Here, we investigated the molecular composition of TJs in FS cells. We demonstrated that occludin is a good marker for TJs in the pituitary gland and examined the structure of the lumens surrounded by FS cells. We also found that claudin-9 is a major component of TJs in the FS cells. In immunoelectron microscopy, claudin-9 was specifically localized at TJs of the FS cells. The expression of claudin-9 was gradually increased in the pituitary gland after birth, suggesting that claudin-9 is developmentally regulated and performs some specific functions on the paracellular barrier of follicles in the pituitary gland. Furthermore, we found that angulin-1, angulin-2, and tricellulin are localized at the tricellular contacts of the FS cells. Our findings provide a first comprehensive molecular profile of TJs in the FS cells, and may lead us towards unveiling the FS cell functions.
Collapse
|
31
|
Fazli M, Vo T, Bertram R. Fast-slow analysis of a stochastic mechanism for electrical bursting. CHAOS (WOODBURY, N.Y.) 2021; 31:103128. [PMID: 34717336 DOI: 10.1063/5.0059338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/03/2021] [Indexed: 06/13/2023]
Abstract
Electrical bursting oscillations in neurons and endocrine cells are activity patterns that facilitate the secretion of neurotransmitters and hormones and have been the focus of study for several decades. Mathematical modeling has been an extremely useful tool in this effort, and the use of fast-slow analysis has made it possible to understand bursting from a dynamic perspective and to make testable predictions about changes in system parameters or the cellular environment. It is typically the case that the electrical impulses that occur during the active phase of a burst are due to stable limit cycles in the fast subsystem of equations or, in the case of so-called "pseudo-plateau bursting," canards that are induced by a folded node singularity. In this article, we show an entirely different mechanism for bursting that relies on stochastic opening and closing of a key ion channel. We demonstrate, using fast-slow analysis, how the short-lived stochastic channel openings can yield a much longer response in which single action potentials are converted into bursts of action potentials. Without this stochastic element, the system is incapable of bursting. This mechanism can describe stochastic bursting in pituitary corticotrophs, which are small cells that exhibit a great deal of noise as well as other pituitary cells, such as lactotrophs and somatotrophs that exhibit noisy bursts of electrical activity.
Collapse
Affiliation(s)
- Mehran Fazli
- Department of Mathematics, Florida State University, Tallahassee, Florida 32306, USA
| | - Theodore Vo
- School of Mathematics, Monash University, Clayton, Victoria 3800, Australia
| | - Richard Bertram
- Department of Mathematics, Florida State University, Tallahassee, Florida 32306, USA
| |
Collapse
|
32
|
Royan MR, Siddique K, Csucs G, Puchades MA, Nourizadeh-Lillabadi R, Bjaalie JG, Henkel CV, Weltzien FA, Fontaine R. 3D Atlas of the Pituitary Gland of the Model Fish Medaka ( Oryzias latipes). Front Endocrinol (Lausanne) 2021; 12:719843. [PMID: 34497587 PMCID: PMC8419251 DOI: 10.3389/fendo.2021.719843] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/12/2021] [Indexed: 12/23/2022] Open
Abstract
In vertebrates, the anterior pituitary plays a crucial role in regulating several essential physiological processes via the secretion of at least seven peptide hormones by different endocrine cell types. Comparative and comprehensive knowledge of the spatial distribution of those endocrine cell types is required to better understand their physiological functions. Using medaka as a model and several combinations of multi-color fluorescence in situ hybridization, we present the first 3D atlas revealing the gland-wide distribution of seven endocrine cell populations: lactotropes, thyrotropes, Lh and Fsh gonadotropes, somatotropes, and pomca-expressing cells (corticotropes and melanotropes) in the anterior pituitary of a teleost fish. By combining in situ hybridization and immunofluorescence techniques, we deciphered the location of corticotropes and melanotropes within the pomca-expressing cell population. The 3D localization approach reveals sexual dimorphism of tshba-, pomca-, and lhb-expressing cells in the adult medaka pituitary. Finally, we show the existence of bi-hormonal cells co-expressing lhb-fshb, fshb-tshba and lhb-sl using single-cell transcriptomics analysis and in situ hybridization. This study offers a solid basis for future comparative studies of the teleost pituitary and its functional plasticity.
Collapse
Affiliation(s)
- Muhammad Rahmad Royan
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Khadeeja Siddique
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Gergely Csucs
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maja A. Puchades
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Jan G. Bjaalie
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christiaan V. Henkel
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Finn-Arne Weltzien
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Romain Fontaine
- Physiology Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
33
|
Ouyang Q, Hu S, Li L, Ran M, Zhu J, Zhao Y, Hu B, Hu J, He H, Li L, Wang J. Integrated mRNA and miRNA transcriptome analysis provides novel insights into the molecular mechanisms underlying goose pituitary development during the embryo-to-hatchling transition. Poult Sci 2021; 100:101380. [PMID: 34358958 PMCID: PMC8350522 DOI: 10.1016/j.psj.2021.101380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 11/25/2022] Open
Abstract
It is well established that the endocrine system plays a pivotal role in preparing the avian embryos for the abrupt switch from chorioallantoic to pulmonary respiration during the critical embryo-to-hatchling transition. However, as the master gland of the endocrine system, there has been little research focusing on the molecular mechanisms controlling the development and function of the pituitary gland during the peri-hatch period in birds. In the present study, we aimed to determine the genome-wide mRNA and miRNA transcriptome profiles of the pituitary during the embryo-to-hatchling transition period from embryonic day 22 (E22) to post-hatching day 6 (P6) in the goose (Anser cygnoides). Of note, expression of Anser_cygnoides_newGene_32456 and LOC106031011 were significantly different among these 4 stages (i.e., E22, E26, P2, and P6). Meanwhile, the neuroactive ligand-receptor interaction pathway was significantly enriched by the DEGs commonly identified among three pairwise comparisons. At the miRNA transcriptome level, there were not commonly identified DE miRNAs among these 4 stages, while the 418 of their predicted target genes were mutually shared. Both the target genes of DE miRNAs in each comparison and these 418 shared target genes were significantly enriched in the ECM-receptor interaction and focal adhesion pathways. In the predicted miRNA-mRNA interaction networks of these 2 pathways, novel_miRNA_467, novel_miRNA_154, and novel_miRNA_340 were the hub miRNAs. In addition, multiple DE miRNAs also showed predicted target relationships with the DEGs associated with extracellular matrix (ECM) components. Among them, expression of novel_miR_120, tgu-miR-92-3p, and novel_miR_398 was significantly negatively correlated with that of LAMC3 (laminin subunit gamma3), suggesting that these miRNAs may regulate pituitary tissue remodeling and functional changes through targeting LAMC3 during development. These identified DE mRNAs and miRNAs as well as their predicted interaction networks involved in regulation of tissue remodeling and cellular functions were most likely to play critical roles in facilitating the embryo-to-hatchling transition. These results provide novel insights into the early developmental process of avian pituitary gland and will help better understand the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingxia Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiaran Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yiting Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| |
Collapse
|
34
|
Zhang J, Lv C, Mo C, Liu M, Wan Y, Li J, Wang Y. Single-Cell RNA Sequencing Analysis of Chicken Anterior Pituitary: A Bird's-Eye View on Vertebrate Pituitary. Front Physiol 2021; 12:562817. [PMID: 34267669 PMCID: PMC8276247 DOI: 10.3389/fphys.2021.562817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 05/21/2021] [Indexed: 01/08/2023] Open
Abstract
It is well-established that anterior pituitary contains multiple endocrine cell populations, and each of them can secrete one/two hormone(s) to regulate vital physiological processes of vertebrates. However, the gene expression profiles of each pituitary cell population remains poorly characterized in most vertebrate groups. Here we analyzed the transcriptome of each cell population in adult chicken anterior pituitaries using single-cell RNA sequencing technology. The results showed that: (1) four out of five known endocrine cell clusters have been identified and designated as the lactotrophs, thyrotrophs, corticotrophs, and gonadotrophs, respectively. Somatotrophs were not analyzed in the current study. Each cell cluster can express at least one known endocrine hormone, and novel marker genes (e.g., CD24 and HSPB1 in lactotrophs, NPBWR2 and NDRG1 in corticotrophs; DIO2 and SOUL in thyrotrophs, C5H11ORF96 and HPGDS in gonadotrophs) are identified. Interestingly, gonadotrophs were shown to abundantly express five peptide hormones: FSH, LH, GRP, CART and RLN3; (2) four non-endocrine/secretory cell types, including endothelial cells (expressing IGFBP7 and CFD) and folliculo-stellate cells (FS-cells, expressing S100A6 and S100A10), were identified in chicken anterior pituitaries. Among them, FS-cells can express many growth factors, peptides (e.g., WNT5A, HBEGF, Activins, VEGFC, NPY, and BMP4), and progenitor/stem cell-associated genes (e.g., Notch signaling components, CDH1), implying that the FS-cell cluster may act as a paracrine/autocrine signaling center and enrich pituitary progenitor/stem cells; (3) sexually dimorphic expression of many genes were identified in most cell clusters, including gonadotrophs and lactotrophs. Taken together, our data provides a bird's-eye view on the diverse aspects of anterior pituitaries, including cell composition, heterogeneity, cell-to-cell communication, and gene expression profiles, which facilitates our comprehensive understanding of vertebrate pituitary biology.
Collapse
Affiliation(s)
- Jiannan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Can Lv
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Chunheng Mo
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Meng Liu
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yiping Wan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yajun Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Santiago-Andres Y, Golan M, Fiordelisio T. Functional Pituitary Networks in Vertebrates. Front Endocrinol (Lausanne) 2021; 11:619352. [PMID: 33584547 PMCID: PMC7873642 DOI: 10.3389/fendo.2020.619352] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The pituitary is a master endocrine gland that developed early in vertebrate evolution and therefore exists in all modern vertebrate classes. The last decade has transformed our view of this key organ. Traditionally, the pituitary has been viewed as a randomly organized collection of cells that respond to hypothalamic stimuli by secreting their content. However, recent studies have established that pituitary cells are organized in tightly wired large-scale networks that communicate with each other in both homo and heterotypic manners, allowing the gland to quickly adapt to changing physiological demands. These networks functionally decode and integrate the hypothalamic and systemic stimuli and serve to optimize the pituitary output into the generation of physiologically meaningful hormone pulses. The development of 3D imaging methods and transgenic models have allowed us to expand the research of functional pituitary networks into several vertebrate classes. Here we review the establishment of pituitary cell networks throughout vertebrate evolution and highlight the main perspectives and future directions needed to decipher the way by which pituitary networks serve to generate hormone pulses in vertebrates.
Collapse
Affiliation(s)
- Yorgui Santiago-Andres
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Matan Golan
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Rishon Lezion, Israel
| | - Tatiana Fiordelisio
- Laboratorio de Neuroendocrinología Comparada, Departamento de Ecología y Recursos Naturales, Biología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| |
Collapse
|
36
|
Querat B. Unconventional Actions of Glycoprotein Hormone Subunits: A Comprehensive Review. Front Endocrinol (Lausanne) 2021; 12:731966. [PMID: 34671318 PMCID: PMC8522476 DOI: 10.3389/fendo.2021.731966] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/06/2021] [Indexed: 01/17/2023] Open
Abstract
The glycoprotein hormones (GPH) are heterodimers composed of a common α subunit and a specific β subunit. They act by activating specific leucine-rich repeat G protein-coupled receptors. However, individual subunits have been shown to elicit responses in cells devoid of the receptor for the dimeric hormones. The α subunit is involved in prolactin production from different tissues. The human chorionic gonadotropin β subunit (βhCG) plays determinant roles in placentation and in cancer development and metastasis. A truncated form of the thyrotropin (TSH) β subunit is also reported to have biological effects. The GPH α- and β subunits are derived from precursor genes (gpa and gpb, respectively), which are expressed in most invertebrate species and are still represented in vertebrates as GPH subunit paralogs (gpa2 and gpb5, respectively). No specific receptor has been found for the vertebrate GPA2 and GPB5 even if their heterodimeric form is able to activate the TSH receptor in mammals. Interestingly, GPA and GPB are phylogenetically and structurally related to cysteine-knot growth factors (CKGF) and particularly to a group of antagonists that act independently on any receptor. This review article summarizes the observed actions of individual GPH subunits and presents the current hypotheses of how these actions might be induced. New approaches are also proposed in light of the evolutionary relatedness with antagonists of the CKGF family of proteins.
Collapse
|
37
|
Pavlović M, Jovanović I, Ugrenović S, Stojanović V, Živković V, Kundalić B, Trandafilović M, Antić M, Čukuranović-Kokoris J. Human anterior pituitary's ACTH cells during the aging process: immunohistochemic and morphometric study. Anat Sci Int 2020; 96:250-257. [PMID: 33098535 DOI: 10.1007/s12565-020-00583-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/10/2020] [Indexed: 11/28/2022]
Abstract
Corticotrophs produce a hormone that stimulates the adrenal gland cortex to secrete glucocorticoids, which in turn have effects on carbohydrate and protein metabolism. Quantification, morphological characteristics, and distribution of corticotrophs in the anterior pituitary and changes in the number and shape of the cells during aging have been examined using immunohistochemical and morphometric methods. The material consisted of 14 anterior pituitaries taken from cadavers at routine autopsy. The tissue was processed by standard histological procedure and the obtained slices were stained by the monoclonal anti-ACTH antibody for corticotrophs identification. Digital images of stained histological sections were analyzed using the morphometric method with the Image J system. The volume density of ACTH positive cells was determined. The cases were classified into three age groups. One-way ANOVA showed that the volume density of the corticotrophs was significantly higher in the second and third group in relation to the first group. The difference in the volume densities of the corticotrophs between the genders was not significant. Morphometric and statistical analyses demonstrated a significant positive correlation between the corticotrophs volume densities and the age of the evaluated cases. Linear regression showed that age significantly predicts corticotrophs volume density. Corticotrophs significantly increase during the life span.
Collapse
Affiliation(s)
- Miljana Pavlović
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia.
| | - Ivan Jovanović
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Slađana Ugrenović
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Vesna Stojanović
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Vladimir Živković
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Braca Kundalić
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Milena Trandafilović
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Milorad Antić
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| | - Jovana Čukuranović-Kokoris
- Department of Anatomy, Faculty of Medicine, University of Niš, Bul. dr Zorana Đinđića 81, 18000, Niš, Serbia
| |
Collapse
|
38
|
O'Hara L, Christian HC, Jeffery N, Le Tissier P, Smith LB. Characterisation of a mural cell network in the murine pituitary gland. J Neuroendocrinol 2020; 32:e12903. [PMID: 32959418 DOI: 10.1111/jne.12903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 08/03/2020] [Accepted: 08/21/2020] [Indexed: 11/29/2022]
Abstract
The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRβ) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRβ+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRβ+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.
Collapse
Affiliation(s)
- Laura O'Hara
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Helen C Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nathan Jeffery
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Paul Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
39
|
Trudeau VL, Somoza GM. Multimodal hypothalamo-hypophysial communication in the vertebrates. Gen Comp Endocrinol 2020; 293:113475. [PMID: 32240708 DOI: 10.1016/j.ygcen.2020.113475] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/21/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022]
Abstract
The vertebrate pituitary is arguably one of the most complex endocrine glands from the evolutionary, anatomical and functional perspectives. The pituitary plays a master role in endocrine physiology for the control of growth, metabolism, reproduction, water balance, and the stress response, among many other key processes. The synthesis and secretion of pituitary hormones are under the control of neurohormones produced by the hypothalamus. Under this conceptual framework, the communication between the hypophysiotropic brain and the pituitary gland is at the foundation of our understanding of endocrinology. The anatomy of the connections between the hypothalamus and the pituitary gland has been described in different vertebrate classes, revealing diverse modes of communication together with varying degrees of complexity. In this context, the evolution and variation in the neuronal, neurohemal, endocrine and paracrine modes will be reviewed in light of recent discoveries, and a re-evaluation of earlier observations. There appears to be three main hypothalamo-pituitary communication systems: 1. Diffusion, best exemplified by the agnathans; 2. Direct innervation of the adenohypophysis, which is most developed in teleost fish, and 3. The median eminence/portal blood vessel system, most conspicuously developed in tetrapods, showing also considerable variation between classes. Upon this basic classification, there exists various combinations possible, giving rise to taxon and species-specific, multimodal control over major physiological processes. Intrapituitary paracrine regulation and communication between folliculostellate cells and endocrine cells are additional processes of major importance. Thus, a more complex evolutionary picture of hypothalamo-hypophysial communication is emerging. There is currently little direct evidence to suggest which neuroendocrine genes may control the evolution of one communication system versus another. However, studies at the developmental and intergenerational timescales implicate several genes in the angiogenesis and axonal guidance pathways that may be important.
Collapse
Affiliation(s)
- Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada.
| | - Gustavo M Somoza
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Buenos Aires B7130IWA, Argentina.
| |
Collapse
|
40
|
Pituitary Hyperplasia, Hormonal Changes and Prolactinoma Development in Males Exposed to Estrogens-An Insight From Translational Studies. Int J Mol Sci 2020; 21:ijms21062024. [PMID: 32188093 PMCID: PMC7139613 DOI: 10.3390/ijms21062024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/21/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Estrogen signaling plays an important role in pituitary development and function. In sensitive rat or mice strains of both sexes, estrogen treatments promote lactotropic cell proliferation and induce the formation of pituitary adenomas (dominantly prolactin or growth-hormone-secreting ones). In male patients receiving estrogen, treatment does not necessarily result in pituitary hyperplasia, hyperprolactinemia or adenoma development. In this review, we comprehensively analyze the mechanisms of estrogen action upon their application in male animal models comparing it with available data in human subjects. Sex-specific molecular targets of estrogen action in lactotropic (PRL) cells are highlighted in the context of their proliferative and secretory activity. In addition, putative effects of estradiol on the cellular/tumor microenvironment and the contribution of postnatal pituitary progenitor/stem cells and transdifferentiation processes to prolactinoma development have been analyzed. Finally, estrogen-induced morphological and hormone-secreting changes in pituitary thyrotropic (TSH) and adrenocorticotropic (ACTH) cells are discussed, as well as the putative role of the thyroid and/or glucocorticoid hormones in prolactinoma development, based on the current scarce literature.
Collapse
|
41
|
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
|
42
|
Hymer WC, Kennett MJ, Maji SK, Gosselink KL, McCall GE, Grindeland RE, Post EM, Kraemer WJ. Bioactive growth hormone in humans: Controversies, complexities and concepts. Growth Horm IGF Res 2020; 50:9-22. [PMID: 31809882 DOI: 10.1016/j.ghir.2019.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 10/07/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To revisit a finding, first described in 1978, which documented existence of a pituitary growth factor that escaped detection by immunoassay, but which was active in the established rat tibia GH bioassay. METHODS We present a narrative review of the evolution of growth hormone complexity, and its bio-detectability, from a historical perspective. RESULTS In humans under the age of 60, physical training (i.e. aerobic endurance and resistance training) are stressors which preferentially stimulate release of bioactive GH (bGH) into the blood. Neuroanatomical studies indicate a) that nerve fibers directly innervate the human anterior pituitary and b) that hind limb muscle afferents, in both humans and rats, also modulate plasma bGH. In the pituitary gland itself, molecular variants of GH, somatotroph heterogeneity and cell plasticity all appear to play a role in regulation of this growth factor. CONCLUSION This review considers more recent findings on this often forgotten/neglected subject. Comparison testing of a) human plasma samples, b) sub-populations of separated rat pituitary somatotrophs or c) purified human pituitary peptides by GH bioassay vs immunoassay consistently yield conflicting results.
Collapse
Affiliation(s)
- Wesley C Hymer
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Mary J Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States of America
| | - Samir K Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 4000076, India
| | - Kristin L Gosselink
- Department of Physiology and Pathology, Burrell College of Osteopathic Medicine, Las Cruces, NM 88001, United States of America
| | - Gary E McCall
- Department of Exercise Science Exercise and Neuroscience Program, University of Puget Sound, Tacoma, WA 98416, United States of America
| | - Richard E Grindeland
- Life Science Division, NASA-Ames Research Center, Moffett Field, CA 94035, United States of America
| | - Emily M Post
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, United States of America
| | - William J Kraemer
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, United States of America.
| |
Collapse
|
43
|
Basaran R, Gundogan D, Senol M, Bozdogan C, Gezen F, Sav A. THE EXPRESSION OF STEM CELL MARKERS (CD133, NESTIN, OCT4, SOX2) IN INVASIVE PITUITARY ADENOMAS. ACTA ENDOCRINOLOGICA-BUCHAREST 2020; 16:303-310. [PMID: 33363651 DOI: 10.4183/aeb.2020.303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction The pituitary gland serves as the center of the endocrine system. Stem cells are typically found in a specialized microenvironment of the tissue, called the niche, which regulates their maintenance, self-renewal, fate determination, and reaction to external influences. The aim of this study is to elucidate the role of stem cells in the initiation, invasion, and progression of pituitary adenomas. Materials and methods All specimens were collected between January 2007 and April 2015. Radiological classification (invasiveness) for all cases was performed according to the Wilson-Hardy classification system. Immunohistochemical staining was performed to all specimens for CD133, Oct4, Sox2 and nestin. Results The study included 48 patients. Of 48 patients, 17 (35.4%) were male and 31 (64.6%) were female. Mean age is 47.10±14.14 (17-86 yrs.). According to the Wilson-Hardy classification system, 27 (56.3%) were non-invasive adenomas. There was no statistical significance between the expression of pituitary stem cell markers (CD133, OCT4, SOX2, nestin) and invasiveness. Conclusion All stem cell markers are stained extensively in pituitary adenomas, except for SOX2 which was stained weakly. However, there is no effect of stem cells on invasiveness of pituitary adenomas because we cannot find a difference of the staining level between invasive and non-invasive adenomas. Nestin was stained extensively in functional adenomas, especially for GH, PRL, and gonadotropin secreting adenomas. SOX2 was stained extensively for ACTH-secreting adenomas.
Collapse
Affiliation(s)
- R Basaran
- University of Medical Sciences, Sancaktepe Education and Research Hospital - Dept. of Neurosurgery, Istanbul, Turkey
| | - D Gundogan
- Istanbul Surgery Hospital - Dept. of Neurosurgery, Istanbul, Turkey
| | - M Senol
- Erzurum Bolge Education and Research Hospital - Dept. of Neurosurgery, Istanbul, Turkey
| | - C Bozdogan
- Aydin State Hospital - Neurosurgery, Aydin, Turkey
| | - F Gezen
- Medeniyet University Faculty of Medicine - Dept. of Neurosurgery, Istanbul, Turkey
| | - A Sav
- Yeditepe University - Pathology, Istanbul, Turkey
| |
Collapse
|
44
|
Clay CM, Cherrington BD, Navratil AM. Plasticity of Anterior Pituitary Gonadotrope Cells Facilitates the Pre-Ovulatory LH Surge. Front Endocrinol (Lausanne) 2020; 11:616053. [PMID: 33613451 PMCID: PMC7890248 DOI: 10.3389/fendo.2020.616053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/15/2020] [Indexed: 01/04/2023] Open
Abstract
Gonadotropes cells located in the anterior pituitary gland are critical for reproductive fitness. A rapid surge in the serum concentration of luteinizing hormone (LH) secreted by anterior pituitary gonadotropes is essential for stimulating ovulation and is thus required for a successful pregnancy. To meet the requirements to mount the LH surge, gonadotrope cells display plasticity at the cellular, molecular and morphological level. First, gonadotrope cells heighten their sensitivity to an increasing frequency of hypothalamic GnRH pulses by dynamically elevating the expression of the GnRH receptor (GnRHR). Following ligand binding, GnRH initiates highly organized intracellular signaling cascades that ultimately promote the synthesis of LH and the trafficking of LH vesicles to the cell periphery. Lastly, gonadotrope cells display morphological plasticity, where there is directed mobilization of cytoskeletal processes towards vascular elements to facilitate rapid LH secretion into peripheral circulation. This mini review discusses the functional and organizational plasticity in gonadotrope cells including changes in sensitivity to GnRH, composition of the GnRHR signaling platform within the plasma membrane, and changes in cellular morphology. Ultimately, multimodal plasticity changes elicited by gonadotropes are critical for the generation of the LH surge, which is required for ovulation.
Collapse
Affiliation(s)
- Colin M. Clay
- Department of Biomedical Science, Colorado State University, Fort Collins, CO, United States
| | - Brian D. Cherrington
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Amy M. Navratil
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
- *Correspondence: Amy M. Navratil,
| |
Collapse
|
45
|
Rojo-Ruiz J, Navas-Navarro P, Nuñez L, García-Sancho J, Alonso MT. Imaging of Endoplasmic Reticulum Ca 2+ in the Intact Pituitary Gland of Transgenic Mice Expressing a Low Affinity Ca 2+ Indicator. Front Endocrinol (Lausanne) 2020; 11:615777. [PMID: 33664709 PMCID: PMC7921146 DOI: 10.3389/fendo.2020.615777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
The adenohypophysis contains five secretory cell types (somatotrophs, lactotrophs, thyrotrophs, corticotrophs, and gonadotrophs), each secreting a different hormone, and controlled by different hypothalamic releasing hormones (HRHs). Exocytic secretion is regulated by cytosolic Ca2+ signals ([Ca2+]C), which can be generated either by Ca2+ entry through the plasma membrane and/or by Ca2+ release from the endoplasmic reticulum (ER). In addition, Ca2+ entry signals can eventually be amplified by ER release via calcium-induced calcium release (CICR). We have investigated the contribution of ER Ca2+ release to the action of physiological agonists in pituitary gland. Changes of [Ca2+] in the ER ([Ca2+]ER) were measured with the genetically encoded low-affinity Ca2+ sensor GAP3 targeted to the ER. We used a transgenic mouse strain that expressed erGAP3 driven by a ubiquitous promoter. Virtually all the pituitary cells were positive for the sensor. In order to mimick the physiological environment, intact pituitary glands or acute slices from the transgenic mouse were used to image [Ca2+]ER. [Ca2+]C was measured simultaneously with Rhod-2. Luteinizing hormone-releasing hormone (LHRH) or thyrotropin releasing hormone (TRH), two agonists known to elicit intracellular Ca2+ mobilization, provoked robust decreases of [Ca2+]ER and concomitant rises of [Ca2+]C. A smaller fraction of cells responded to thyrotropin releasing hormone (TRH). By contrast, depolarization with high K+ triggered a rise of [Ca2+]C without a decrease of [Ca2+]ER, indicating that the calcium-induced calcium-release (CICR) via ryanodine receptor amplification mechanism is not present in these cells. Our results show the potential of transgenic ER Ca2+ indicators as novel tools to explore intraorganellar Ca2+ dynamics in pituitary gland in situ.
Collapse
|
46
|
Mirczuk SM, Lessey AJ, Catterick AR, Perrett RM, Scudder CJ, Read JE, Lipscomb VJ, Niessen SJ, Childs AJ, McArdle CA, McGonnell IM, Fowkes RC. Regulation and Function of C-Type Natriuretic Peptide (CNP) in Gonadotrope-Derived Cell Lines. Cells 2019; 8:cells8091086. [PMID: 31540096 PMCID: PMC6769446 DOI: 10.3390/cells8091086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/02/2019] [Accepted: 09/12/2019] [Indexed: 01/29/2023] Open
Abstract
C-type natriuretic peptide (CNP) is the most conserved member of the mammalian natriuretic peptide family, and is implicated in the endocrine regulation of growth, metabolism and reproduction. CNP is expressed throughout the body, but is particularly abundant in the central nervous system and anterior pituitary gland. Pituitary gonadotropes are regulated by pulsatile release of gonadotropin releasing hormone (GnRH) from the hypothalamus, to control reproductive function. GnRH and CNP reciprocally regulate their respective signalling pathways in αT3-1 gonadotrope cells, but effects of pulsatile GnRH stimulation on CNP expression has not been explored. Here, we examine the sensitivity of the natriuretic peptide system in LβT2 and αT3-1 gonadotrope cell lines to continuous and pulsatile GnRH stimulation, and investigate putative CNP target genes in gonadotropes. Multiplex RT-qPCR assays confirmed that primary mouse pituitary tissue express Nppc,Npr2 (encoding CNP and guanylyl cyclase B (GC-B), respectively) and Furin (a CNP processing enzyme), but failed to express transcripts for Nppa or Nppb (encoding ANP and BNP, respectively). Pulsatile, but not continuous, GnRH stimulation of LβT2 cells caused significant increases in Nppc and Npr2 expression within 4 h, but failed to alter natriuretic peptide gene expression in αT3-1 cells. CNP enhanced expression of cJun, Egr1, Nr5a1 and Nr0b1, within 8 h in LβT2 cells, but inhibited Nr5a1 expression in αT3-1 cells. Collectively, these data show the gonadotrope natriuretic peptide system is sensitive to pulsatile GnRH signalling, and gonadotrope transcription factors are putative CNP-target genes. Such findings represent additional mechanisms by which CNP may regulate reproductive function.
Collapse
Affiliation(s)
- Samantha M Mirczuk
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Andrew J Lessey
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Alice R Catterick
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Rebecca M Perrett
- Laboratories for Integrative Neuroscience and Endocrinology, Department of Clinical Sciences at South Bristol, University of Bristol, Whitson Street, Bristol, BS13NY, UK.
| | - Christopher J Scudder
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Jordan E Read
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Victoria J Lipscomb
- Clinical Science and Services, Royal Veterinary College, AL9 7TA Hertfordshire, UK.
| | - Stijn J Niessen
- Clinical Science and Services, Royal Veterinary College, AL9 7TA Hertfordshire, UK.
| | - Andrew J Childs
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Craig A McArdle
- Laboratories for Integrative Neuroscience and Endocrinology, Department of Clinical Sciences at South Bristol, University of Bristol, Whitson Street, Bristol, BS13NY, UK.
| | - Imelda M McGonnell
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| | - Robert C Fowkes
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, Royal College Street, NW1 0TU London, UK.
| |
Collapse
|
47
|
Anastassiadis C, Jones SL, Pruessner JC. Imaging the pituitary in psychopathologies: a review of in vivo magnetic resonance imaging studies. Brain Struct Funct 2019; 224:2587-2601. [DOI: 10.1007/s00429-019-01942-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 08/13/2019] [Indexed: 12/17/2022]
|
48
|
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: 5.7] [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
|
49
|
Momiji H, Hassall KL, Featherstone K, McNamara AV, Patist AL, Spiller DG, Christian HC, White MRH, Davis JRE, Finkenstädt BF, Rand DA. Disentangling juxtacrine from paracrine signalling in dynamic tissue. PLoS Comput Biol 2019; 15:e1007030. [PMID: 31194728 PMCID: PMC6592563 DOI: 10.1371/journal.pcbi.1007030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/25/2019] [Accepted: 04/15/2019] [Indexed: 11/18/2022] Open
Abstract
Prolactin is a major hormone product of the pituitary gland, the central endocrine regulator. Despite its physiological importance, the cell-level mechanisms of prolactin production are not well understood. Having significantly improved the resolution of real-time-single-cell-GFP-imaging, the authors recently revealed that prolactin gene transcription is highly dynamic and stochastic yet shows space-time coordination in an intact tissue slice. However, it still remains an open question as to what kind of cellular communication mediates the observed space-time organization. To determine the type of interaction between cells we developed a statistical model. The degree of similarity between two expression time series was studied in terms of two distance measures, Euclidean and geodesic, the latter being a network-theoretic distance defined to be the minimal number of edges between nodes, and this was used to discriminate between juxtacrine from paracrine signalling. The analysis presented here suggests that juxtacrine signalling dominates. To further determine whether the coupling is coordinating transcription or post-transcriptional activities we used stochastic switch modelling to infer the transcriptional profiles of cells and estimated their similarity measures to deduce that their spatial cellular coordination involves coupling of transcription via juxtacrine signalling. We developed a computational model that involves an inter-cell juxtacrine coupling, yielding simulation results that show space-time coordination in the transcription level that is in agreement with the above analysis. The developed model is expected to serve as the prototype for the further study of tissue-level organised gene expression for epigenetically regulated genes, such as prolactin.
Collapse
Affiliation(s)
- Hiroshi Momiji
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom, Mathematics Institute, University of Warwick, Coventry, United Kingdom
- * E-mail: (HM); (MRHW); (JRED); (BFF); (DAR)
| | - Kirsty L. Hassall
- Department of Statistics, University of Warwick, Coventry, United Kingdom
| | - Karen Featherstone
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Anne V. McNamara
- Systems Microscopy Centre, University of Manchester, Manchester, United Kingdom
| | - Amanda L. Patist
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - David G. Spiller
- Systems Microscopy Centre, University of Manchester, Manchester, United Kingdom
| | - Helen C. Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Michael R. H. White
- Systems Microscopy Centre, University of Manchester, Manchester, United Kingdom
- * E-mail: (HM); (MRHW); (JRED); (BFF); (DAR)
| | - Julian R. E. Davis
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
- * E-mail: (HM); (MRHW); (JRED); (BFF); (DAR)
| | - Bärbel F. Finkenstädt
- Department of Statistics, University of Warwick, Coventry, United Kingdom
- * E-mail: (HM); (MRHW); (JRED); (BFF); (DAR)
| | - David A. Rand
- Zeeman Institute for Systems Biology & Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom, Mathematics Institute, University of Warwick, Coventry, United Kingdom
- * E-mail: (HM); (MRHW); (JRED); (BFF); (DAR)
| |
Collapse
|
50
|
Czaja W, Nakamura YK, Li N, Eldridge JA, DeAvila DM, Thompson TB, Rodgers BD. Myostatin regulates pituitary development and hepatic IGF1. Am J Physiol Endocrinol Metab 2019; 316:E1036-E1049. [PMID: 30888862 PMCID: PMC6620572 DOI: 10.1152/ajpendo.00001.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating myostatin-attenuating agents are being developed to treat muscle-wasting disease despite their potential to produce serious off-target effects, as myostatin/activin receptors are widely distributed among many nonmuscle tissues. Our studies suggest that the myokine not only inhibits striated muscle growth but also regulates pituitary development and growth hormone (GH) action in the liver. Using a novel myostatin-null label-retaining model (Jekyll mice), we determined that the heterogeneous pool of pituitary stem, transit-amplifying, and progenitor cells in Jekyll mice depletes more rapidly after birth than the pool in wild-type mice. This correlated with increased levels of GH, prolactin, and the cells that secrete these hormones, somatotropes and lactotropes, respectively, in Jekyll pituitaries. Recombinant myostatin also stimulated GH release and gene expression in pituitary cell cultures although inhibiting prolactin release. In primary hepatocytes, recombinant myostatin blocked GH-stimulated expression of two key mediators of growth, insulin-like growth factor (IGF)1 and the acid labile subunit and increased expression of an inhibitor, IGF-binding protein-1. The significance of these findings was demonstrated by smaller muscle fiber size in a model lacking myostatin and liver IGF1 expression (LID-o-Mighty mice) compared with that in myostatin-null (Mighty) mice. These data together suggest that myostatin may regulate pituitary development and function and that its inhibitory actions in muscle may be partly mediated by attenuating GH action in the liver. They also suggest that circulating pharmacological inhibitors of myostatin could produce unintended consequences in these and possibly other tissues.
Collapse
Affiliation(s)
- Wioletta Czaja
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- Department of Biochemistry and Molecular Biology, University of Georgia , Athens, Georgia
| | - Yukiko K Nakamura
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Naisi Li
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Jennifer A Eldridge
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - David M DeAvila
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati , Cincinnati, Ohio
| | - Buel D Rodgers
- Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University , Pullman, Washington
- AAVogen, Incorporated, Rockville, Maryland
| |
Collapse
|