1
|
Qi Y, Yuan L, Zeng J, Wang X, Ma L, Lv J. Morphological identification and distribution comparison of telocytes in pituitary gland between normal and cryptorchid yaks. BMC Vet Res 2024; 20:463. [PMID: 39394144 PMCID: PMC11468414 DOI: 10.1186/s12917-024-04307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Telocytes (TCs) is a novel type of interstitial cells in many mammals organs, which participate in the organizational metabolism, mechanical support, immunomodulation and other aspects. The aim of this study was to explore the organizational chemical characteristics of TCs in pituitary gland and their changes in cryptorchid yaks. METHODS Transmission electron microscopy (TEM), toluidine blue staining, immunofluorescence, qRT-PCR, and Western blotting may enable us to understand TCs distribution characteristics and biological functions. RESULT TEM confirmed the presence of TCs in the pituitary gland with small bodies and moniliform telopodes (Tps). The Tps extending out from the cell body to the peri-sinusoidal vessels spaces, the number of Tps is closely related to the morphology of the nucleus. The most obvious changes of TCs in the pituitary gland of cryptorchid yaks is the Tps are relatively shorter and decreased secretory vesicles. H.E. and toluidine blue staining revealed that TCs not only distributed between the sinusoidal blood vessels and the glandular cell clusters, but also present on the surface of vascular endothelial cells. The co-expression of TCs biomarkers, such as Vimentin/CD34, CD117/CD34 and α-SMA/CD34, were evaluated by immunofluorescence to further determine the phenotypic characteristics of TCs. Besides, we analyzed the mRNA and protein expression of these biomarkers to determine the characteristics of TCs changes and possible biological roles. Both the mRNA and protein expression of CD117 were significantly higher in the pituitary gland of cryptorchid yaks than in the normal (p < 0.01), the protein expression of CD34 in the cryptorchid yaks was significantly higher than the normal (p < 0.01). There were no significant difference in mRNA expression of Vimentin and α-SMA (p>0.05), while the protein expression were significantly increased in the normal yaks (p < 0.05). CONCLUSIONS In summary, this study reports for the first time that the biological characteristics of TCs in yak pituitary gland. Although there is no significant change in the distribution characteristics, the changes in biological features of TCs in cryptorchid yaks are clear, suggesting that TCs participated in alteration in the local microenvironment of the pituitary gland. Therefore, our study provides clues for further investigating the role of TCs in the pituitary gland during the occurrence of cryptorchidism in yaks.
Collapse
Affiliation(s)
- Yumei Qi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Jianlin Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaofen Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Long Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jinghan Lv
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
2
|
Respekta-Długosz N, Mlyczyńska E, Pich K, Greggio A, Ramé C, Dupont J, Rak A. The adipokine profile in the plasma and anterior pituitary of pigs during the estrous cycle. Gen Comp Endocrinol 2024; 357:114588. [PMID: 39013539 DOI: 10.1016/j.ygcen.2024.114588] [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: 05/23/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024]
Abstract
Adipokines play crucial roles in both reproductive and energy metabolic processes. This study aimed to compare the hormonal plasma profile of adiponectin, apelin, vaspin, chemerin, resistin, visfatin, and adipolin, and the expression of their receptors in the anterior pituitary (AP) between normal-weight Large White (LW) and fat Meishan (MS) pigs during different phases of the estrous cycle. We measured adipokine levels in the plasma and assessed their gene expression in the AP. We used Pearson's correlation analysis to examine potential links between adipokines levels, their receptors, and metabolic parameters (body weight; backfat thickness) and reproductive parameters (pituitary weight; age at puberty; levels of gonadotropins, steroid hormones; and gene expression of gonadotropin-releasing hormone receptor and gonadotropins in AP). The plasma levels of the evaluated adipokines fluctuated with phase and breed, except for visfatin and adipolin. Moreover, adipokine expression in AP varied significantly between breeds and estrous cycle phases, except for resistin receptor CAP1. Notably, we observed a positive correlation between plasma levels of adiponectin and its transcript in the AP only in MS pigs. Apelin gene expression correlated negatively with its receptor in MS, while we observed a breed-dependent correlation between chemerin gene expression and its receptor CMKLR1. We identified significant positive or negative correlations between adipokines or their receptor levels in plasma and AP as well as metabolic or reproductive parameters, depending on the breed. In conclusion, we have demonstrated breed-specific and estrous cycle-dependent regulation of adipokines in AP, underscoring their potential impact on metabolic and reproductive processes in swine.
Collapse
Affiliation(s)
- Natalia Respekta-Długosz
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Poland
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Poland
| | - Aleksandra Greggio
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland
| | - Christelle Ramé
- INRAE, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Joëlle Dupont
- INRAE, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Poland.
| |
Collapse
|
3
|
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
|
4
|
Stefanucci L, Moslemi C, Tomé AR, Virtue S, Bidault G, Gleadall NS, Watson LPE, Kwa JE, Burden F, Farrow S, Chen J, Võsa U, Burling K, Walker L, Ord J, Barker P, Warner J, Frary A, Renhstrom K, Ashford SE, Piper J, Biggs G, Erber WN, Hoffman GJ, Schoenmakers N, Erikstrup C, Rieneck K, Dziegiel MH, Ullum H, Azzu V, Vacca M, Aparicio HJ, Hui Q, Cho K, Sun YV, Wilson PW, Bayraktar OA, Vidal-Puig A, Ostrowski SR, Astle WJ, Olsson ML, Storry JR, Pedersen OB, Ouwehand WH, Chatterjee K, Vuckovic D, Frontini M. SMIM1 absence is associated with reduced energy expenditure and excess weight. MED 2024; 5:1083-1095.e6. [PMID: 38906141 DOI: 10.1016/j.medj.2024.05.015] [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: 08/07/2023] [Revised: 12/06/2023] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Obesity rates have nearly tripled in the past 50 years, and by 2030 more than 1 billion individuals worldwide are projected to be obese. This creates a significant economic strain due to the associated non-communicable diseases. The root cause is an energy expenditure imbalance, owing to an interplay of lifestyle, environmental, and genetic factors. Obesity has a polygenic genetic architecture; however, single genetic variants with large effect size are etiological in a minority of cases. These variants allowed the discovery of novel genes and biology relevant to weight regulation and ultimately led to the development of novel specific treatments. METHODS We used a case-control approach to determine metabolic differences between individuals homozygous for a loss-of-function genetic variant in the small integral membrane protein 1 (SMIM1) and the general population, leveraging data from five cohorts. Metabolic characterization of SMIM1-/- individuals was performed using plasma biochemistry, calorimetric chamber, and DXA scan. FINDINGS We found that individuals homozygous for a loss-of-function genetic variant in SMIM1 gene, underlying the blood group Vel, display excess body weight, dyslipidemia, altered leptin to adiponectin ratio, increased liver enzymes, and lower thyroid hormone levels. This was accompanied by a reduction in resting energy expenditure. CONCLUSION This research identified a novel genetic predisposition to being overweight or obese. It highlights the need to investigate the genetic causes of obesity to select the most appropriate treatment given the large cost disparity between them. FUNDING This work was funded by the National Institute of Health Research, British Heart Foundation, and NHS Blood and Transplant.
Collapse
Affiliation(s)
- Luca Stefanucci
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK; British Heart Foundation, Cambridge Centre for Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Camous Moslemi
- Department of Clinical Immunology, Zealand University Hospital (Roskilde University), Køge, Denmark
| | - Ana R Tomé
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Samuel Virtue
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Addenbrooke's Hospital, Cambridge, UK
| | - Nicholas S Gleadall
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura P E Watson
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Jing E Kwa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Frances Burden
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Ji Chen
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences RILD Building, Barrack Road, Exeter, UK
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Keith Burling
- NIHR Cambridge Biomedical Research Centre Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lindsay Walker
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - John Ord
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter Barker
- NIHR Cambridge Biomedical Research Centre Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James Warner
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Amy Frary
- NIHR National BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Karola Renhstrom
- NIHR National BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Sofie E Ashford
- NIHR National BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Jo Piper
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Gail Biggs
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Wendy N Erber
- Discipline of Pathology and Laboratory Science, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Gary J Hoffman
- Discipline of Pathology and Laboratory Medicine, Medical School, The University of Western Australia, Perth, WA, Australia
| | - Nadia Schoenmakers
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Klaus Rieneck
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Vian Azzu
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK; Department of Gastroenterology, Norfolk & Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Michele Vacca
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK; Interdisciplinary Department of Medicine, Università degli Studi di Bari "Aldo Moro", Bari, Italy; Roger Williams Institute of Hepatology, London, UK
| | | | - Qin Hui
- Atlanta VA Medical Center, Decatur, GA, USA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yan V Sun
- Atlanta VA Medical Center, Decatur, GA, USA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Peter W Wilson
- Atlanta VA Medical Center, Decatur, GA, USA; Emory University Schools of Medicine and Public Health, Atlanta, GA, USA
| | - Omer A Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Addenbrooke's Hospital, Cambridge, UK; Centro de Innvestigacion Principe Felipe, Valencia, Spain
| | - Sisse R Ostrowski
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - William J Astle
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK; British Heart Foundation, Cambridge Centre for Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; MRC Biostatistics Unit, East Forvie Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Martin L Olsson
- Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Lund, Sweden; Department of Laboratory Medicine, Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Jill R Storry
- Clinical Immunology and Transfusion Medicine, Office for Medical Services, Region Skåne, Lund, Sweden; Department of Laboratory Medicine, Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Ole B Pedersen
- Department of Clinical Immunology, Zealand University Hospital (Roskilde University), Køge, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK; Department of Haematology, Cambridge University Hospitals NHS Trust, CB2 0QQ Cambridge, UK; Department of Haematology, University College London Hospitals NHS Trust, NW1 2BU London, UK
| | - Krishna Chatterjee
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK; British Heart Foundation, Cambridge Centre for Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences RILD Building, Barrack Road, Exeter, UK.
| |
Collapse
|
5
|
Deng C, Qin C, Li Z, Lu L, Tong Y, Yuan J, Yin F, Cheng Y, Wu C. Diatomite-incorporated hierarchical scaffolds for osteochondral regeneration. Bioact Mater 2024; 38:305-320. [PMID: 38745590 PMCID: PMC11091463 DOI: 10.1016/j.bioactmat.2024.05.004] [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: 02/05/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Osteochondral regeneration involves the highly challenging and complex reconstruction of cartilage and subchondral bone. Silicon (Si) ions play a crucial role in bone development. Current research on Si ions mainly focuses on bone repair, by using silicate bioceramics with complex ion compositions. However, it is unclear whether the Si ions have important effect on cartilage regeneration. Developing a scaffold that solely releases Si ions to simultaneously promote subchondral bone repair and stimulate cartilage regeneration is critically important. Diatomite (DE) is a natural diatomaceous sediment that can stably release Si ions, known for its abundant availability, low cost, and environmental friendliness. Herein, a hierarchical osteochondral repair scaffold is uniquely designed by incorporating gradient DE into GelMA hydrogel. The adding DE microparticles provides a specific Si source for controlled Si ions release, which not only promotes osteogenic differentiation of rBMSCs (rabbit bone marrow mesenchymal stem cells) but also enhances proliferation and maturation of chondrocytes. Moreover, DE-incorporated hierarchical scaffolds significantly promoted the regeneration of cartilage and subchondral bone. The study suggests the significant role of Si ions in promoting cartilage regeneration and solidifies their foundational role in enhancing bone repair. Furthermore, it offers an economic and eco-friendly strategy for developing high value-added osteochondral regenerative bioscaffolds from low-value ocean natural materials.
Collapse
Affiliation(s)
- Cuijun Deng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, PR China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Zhenguang Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Laiya Lu
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200032, PR China
| | - Yifan Tong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Jiaqi Yuan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200032, PR China
| | - Yu Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| |
Collapse
|
6
|
Yang S, Deng C, Pu C, Bai X, Tian C, Chang M, Feng M. Single-Cell RNA Sequencing and Its Applications in Pituitary Research. Neuroendocrinology 2024; 114:875-893. [PMID: 39053437 PMCID: PMC11460981 DOI: 10.1159/000540352] [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: 03/10/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Mounting evidence underscores the significance of cellular diversity within the endocrine system and the intricate interplay between different cell types and tissues, essential for preserving physiological balance and influencing disease trajectories. The pituitary gland, a central player in the endocrine orchestra, exemplifies this complexity with its assortment of hormone-secreting and nonsecreting cells. SUMMARY The pituitary gland houses several types of cells responsible for hormone production, alongside nonsecretory cells like fibroblasts and endothelial cells, each playing a crucial role in the gland's function and regulatory mechanisms. Despite the acknowledged importance of these cellular interactions, the detailed mechanisms by which they contribute to pituitary gland physiology and pathology remain largely uncharted. The last decade has seen the emergence of groundbreaking technologies such as single-cell RNA sequencing, offering unprecedented insights into cellular heterogeneity and interactions. However, the application of this advanced tool in exploring the pituitary gland's complexities has been scant. This review provides an overview of this methodology, highlighting its strengths and limitations, and discusses future possibilities for employing it to deepen our understanding of the pituitary gland and its dysfunction in disease states. KEY MESSAGE Single-cell RNA sequencing technology offers an unprecedented means to study the heterogeneity and interactions of pituitary cells, though its application has been limited thus far. Further utilization of this tool will help uncover the complex physiological and pathological mechanisms of the pituitary, advancing research and treatment of pituitary diseases.
Collapse
Affiliation(s)
- Shuangjian Yang
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Congcong Deng
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Changqin Pu
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xuexue Bai
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Chenxin Tian
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Mengqi Chang
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Feng
- Department of Neurosurgery, China Pituitary Disease Registry Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Asaad W, Utkina M, Shcherbakova A, Popov S, Melnichenko G, Mokrysheva N. scRNA sequencing technology for PitNET studies. Front Endocrinol (Lausanne) 2024; 15:1414223. [PMID: 39114291 PMCID: PMC11303145 DOI: 10.3389/fendo.2024.1414223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) are common, most likely benign tumors with complex clinical characteristics related to hormone hypersecretion and/or growing sellar tumor mass. PitNET types are classified according to their expression of specific transcriptional factors (TFs) and hormone secretion levels. Some types show aggressive, invasive, and reoccurrence behavior. Current research is being conducted to understand the molecular mechanisms regulating these high-heterogeneous neoplasms originating from adenohypophysis, and single-cell RNA sequencing (scRNA-seq) technology is now playing an essential role in these studies due to its remarkable resolution at the single-cell level. This review describes recent studies on human PitNETs performed with scRNA-seq technology, highlighting the potential of this approach in revealing these tumor pathologies, behavior, and regulatory mechanisms.
Collapse
Affiliation(s)
| | - Marina Utkina
- Department of General, Molecular and Population Genetics, Endocrinology Research Centre, Moscow, Russia
| | | | | | | | | |
Collapse
|
8
|
Qian Q, Li M, Zhang Z, Davis SW, Rahmouni K, Norris AW, Cao H, Ding WX, Hotamisligil GS, Yang L. Obesity disrupts the pituitary-hepatic UPR communication leading to NAFLD progression. Cell Metab 2024; 36:1550-1565.e9. [PMID: 38718793 PMCID: PMC11222033 DOI: 10.1016/j.cmet.2024.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/05/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024]
Abstract
Obesity alters levels of pituitary hormones that govern hepatic immune-metabolic homeostasis, dysregulation of which leads to nonalcoholic fatty liver disease (NAFLD). However, the impact of obesity on intra-pituitary homeostasis is largely unknown. Here, we uncovered a blunted unfolded protein response (UPR) but elevated inflammatory signatures in pituitary glands of obese mice and humans. Furthermore, we found that obesity inflames the pituitary gland, leading to impaired pituitary inositol-requiring enzyme 1α (IRE1α)-X-box-binding protein 1 (XBP1) UPR branch, which is essential for protecting against pituitary endocrine defects and NAFLD progression. Intriguingly, pituitary IRE1-deletion resulted in hypothyroidism and suppressed the thyroid hormone receptor B (THRB)-mediated activation of Xbp1 in the liver. Conversely, activation of the hepatic THRB-XBP1 axis improved NAFLD in mice with pituitary UPR defect. Our study provides the first evidence and mechanism of obesity-induced intra-pituitary cellular defects and the pathophysiological role of pituitary-liver UPR communication in NAFLD progression.
Collapse
Affiliation(s)
- Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Shannon W Davis
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Andrew W Norris
- Division of Endocrinology and Diabetes, Department of Pediatrics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, Division of Biostatistics and Computational Biology, Department of Endodontics, University of Iowa College of Dentistry, Iowa City, IA 52242, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. School of Public Health, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02141, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
9
|
Yang H, Chen M, Xu H, Zhen Y, Zhang Y, Wang L, Duan L, Gong F, Zhu H, Pan H. Bone mineral density in adults growth hormone deficiency with different ages of onset: a real-world retrospective study. Endocrine 2024; 85:347-355. [PMID: 38536548 DOI: 10.1007/s12020-024-03786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/12/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE Bone mineral density (BMD) impairment is one of the critical factors for long-term quality of life in adults growth hormone deficiency (AGHD). This study aims to investigate the annual changes in BMD in AGHD patients with different ages of onset and to identify predicting factors that influence BMD. METHODS AGHD patients (n = 160) with available data for 4 years follow-up from a major tertiary medical center in China were retrospectively included (110 [68.8%] childhood-onset, 119 [74.4%] male). BMD of the axial bone (including total hip, neck of femur, and L1-4) derived from dual X-ray absorptiometry and final height were investigated at the first visit, 12 months, 24 months, 36 months, and 48 months thereafter. Low BMD was defined as Z-score ≤ -2. RESULTS The prevalence of low BMD was 30.0% at baseline and 12.5% at 4 years of follow-up. The CO AGHD group presented a significantly lower BMD than the AO AGHD group at the baseline (P = 0.009). In contrast, the CO AGHD group had significantly greater median annual BMD change than the AO AGHD group (0.044 vs. -0.0003 g/cm2/year in L1-4, P < 0.001), indicating a significant difference in the overall BMD trend between CO and AO groups. Childhood-onset (odds ratio [OR] 0.326, P = 0.012), low serum testosterone (OR 0.847; P = 0.004) and FT4 (OR 0.595; P = 0.039) level were independent risk factors for BMD loss. CONCLUSION The annual changes of BMD show a different pattern in AGHD patients with varying ages of onset. Patients with CO AGHD have a lower bone mass, and in general, appropriate replacement therapy is necessary for long-term bone health in AGHD patients.
Collapse
Affiliation(s)
- Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Meiping Chen
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Hanyuan Xu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yunfeng Zhen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yuelun Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Lian Duan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China.
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China.
| |
Collapse
|
10
|
Wang W, Cen Y, Lu Z, Xu Y, Sun T, Xiao Y, Liu W, Li JJ, Wang C. scCDC: a computational method for gene-specific contamination detection and correction in single-cell and single-nucleus RNA-seq data. Genome Biol 2024; 25:136. [PMID: 38783325 PMCID: PMC11112958 DOI: 10.1186/s13059-024-03284-w] [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: 01/30/2023] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
In droplet-based single-cell and single-nucleus RNA-seq assays, systematic contamination of ambient RNA molecules biases the quantification of gene expression levels. Existing methods correct the contamination for all genes globally. However, there lacks specific evaluation of correction efficacy for varying contamination levels. Here, we show that DecontX and CellBender under-correct highly contaminating genes, while SoupX and scAR over-correct lowly/non-contaminating genes. Here, we develop scCDC as the first method to detect the contamination-causing genes and only correct expression levels of these genes, some of which are cell-type markers. Compared with existing decontamination methods, scCDC excels in decontaminating highly contaminating genes while avoiding over-correction of other genes.
Collapse
Affiliation(s)
- Weijian Wang
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China
| | - Yihui Cen
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China
| | - Zezhen Lu
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China
| | - Yueqing Xu
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China
| | - Tianyi Sun
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095, USA
| | - Ying Xiao
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, China
| | - Wanlu Liu
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China
| | - Jingyi Jessica Li
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095, USA.
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, International Campus, ZJU-UoE Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, 314400, China.
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, China.
- Biomedical and Health Translational Research Centre, Zhejiang University, Haining, Zhejiang, 314400, China.
| |
Collapse
|
11
|
Xiao Y, Xia L, Jiang W, Qin J, Zhao L, Li Z, Huang L, Li K, Yu P, Wei L, Jiang X, Chen Z, Yu X. Cardiopulmonary progenitors facilitate cardiac repair via exosomal transfer of miR-27b-3p targeting the SIK1-CREB1 axis. Cell Prolif 2024; 57:e13593. [PMID: 38185757 PMCID: PMC11056695 DOI: 10.1111/cpr.13593] [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: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemic heart disease, especially myocardial infarction (MI), is one of the leading causes of death worldwide, and desperately needs effective treatments, such as cell therapy. Cardiopulmonary progenitors (CPPs) are stem cells for both heart and lung, but their repairing role in damaged heart is still unknown. Here, we obtained CPPs from E9.5 mouse embryos, maintained their stemness while expanding, and identified their characteristics by scRNA-seq, flow cytometry, quantitative reverse transcription-polymerase chain reaction, and differentiation assays. Moreover, we employed mouse MI model to investigate whether CPPs could repair the injured heart. Our data identified that CPPs exhibit hybrid fibroblastic, endothelial, and mesenchymal state, and they could differentiate into cell lineages within the cardiopulmonary system. Moreover, intramyocardial injection of CPPs improves cardiac function through CPPs exosomes (CPPs-Exo) by promotion of cardiomyocytic proliferation and vascularization. To uncover the underlying mechanism, we used miRNA-seq, bulk RNA-seq, and bioinformatic approaches, and found the highly expressed miR-27b-3p in CPPs-Exo and its target gene Sik1, which can influence the transcriptional activity of CREB1. Therefore, we postulate that CPPs facilitate cardiac repair partially through the SIK1-CREB1 axis via exosomal miR-27b-3p. Our study offers a novel insight into the role of CPPs-Exo in heart repair and highlights the potential of CPPs-Exo as a promising therapeutic strategy for MI.
Collapse
Affiliation(s)
- Ying‐Ying Xiao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Luo‐Xing Xia
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Wen‐Jing Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jian‐Feng Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Xin Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhan Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li‐Juan Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Ke‐Xin Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Peng‐Jiu Yu
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Li Wei
- Department of Pharmacy, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xue‐Yan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for BiotechnologySt. John's UniversityQueensNew YorkUSA
| | - Xi‐Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital & the First Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
12
|
Huang QH, Zhao GK, Wang HQ, Wei FH, Zhang JY, Zhang JB, Gao F, Yuan B. Single-Cell Transcriptional Profile Construction of Rat Pituitary Glands before and after Sexual Maturation and Identification of Novel Marker Spp1 in Gonadotropes. Int J Mol Sci 2024; 25:4694. [PMID: 38731915 PMCID: PMC11083676 DOI: 10.3390/ijms25094694] [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: 03/07/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
The mammalian pituitary gland drives highly conserved physiological processes such as somatic cell growth, pubertal transformation, fertility, and metabolism by secreting a variety of hormones. Recently, single-cell transcriptomics techniques have been used in pituitary gland research. However, more studies have focused on adult pituitary gland tissues from different species or different sexes, and no research has yet resolved cellular differences in pituitary gland tissue before and after sexual maturation. Here, we identified a total of 15 cell clusters and constructed single-cell transcriptional profiles of rats before and after sexual maturation. Furthermore, focusing on the gonadotrope cluster, 106 genes were found to be differentially expressed before and after sexual maturation. It was verified that Spp1, which is specifically expressed in gonadotrope cells, could serve as a novel marker for this cell cluster and has a promotional effect on the synthesis and secretion of follicle-stimulating hormone. The results provide a new resource for further resolving the regulatory mechanism of pituitary gland development and pituitary hormone synthesis and secretion.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (Q.-H.H.); (G.-K.Z.); (H.-Q.W.); (F.-H.W.); (J.-Y.Z.); (J.-B.Z.)
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (Q.-H.H.); (G.-K.Z.); (H.-Q.W.); (F.-H.W.); (J.-Y.Z.); (J.-B.Z.)
| |
Collapse
|
13
|
Lin S, Dai Y, Han C, Han T, Zhao L, Wu R, Liu J, Zhang B, Huang N, Liu Y, Lai S, Shi J, Wang Y, Lou M, Xie J, Cheng Y, Tang H, Yao H, Fang H, Zhang Y, Wu X, Shen L, Ye Y, Xue L, Wu ZB. Single-cell transcriptomics reveal distinct immune-infiltrating phenotypes and macrophage-tumor interaction axes among different lineages of pituitary neuroendocrine tumors. Genome Med 2024; 16:60. [PMID: 38658971 PMCID: PMC11040908 DOI: 10.1186/s13073-024-01325-4] [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: 10/14/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNETs) are common gland neoplasms demonstrating distinctive transcription factors. Although the role of immune cells in PitNETs has been widely recognized, the precise immunological environment and its control over tumor cells are poorly understood. METHODS The heterogeneity, spatial distribution, and clinical significance of macrophages in PitNETs were analyzed using single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, spatial transcriptomics, immunohistochemistry, and multiplexed quantitative immunofluorescence (QIF). Cell viability, cell apoptosis assays, and in vivo subcutaneous xenograft experiments have confirmed that INHBA-ACVR1B influences the process of tumor cell apoptosis. RESULTS The present study evaluated scRNA-seq data from 23 PitNET samples categorized into 3 primary lineages. The objective was to explore the diversity of tumors and the composition of immune cells across these lineages. Analyzed data from scRNA-seq and 365 bulk RNA sequencing samples conducted in-house revealed the presence of three unique subtypes of tumor immune microenvironment (TIME) in PitNETs. These subtypes were characterized by varying levels of immune infiltration, ranging from low to intermediate to high. In addition, the NR5A1 lineage is primarily associated with the subtype characterized by limited infiltration of immune cells. Tumor-associated macrophages (TAMs) expressing CX3CR1+, C1Q+, and GPNMB+ showed enhanced contact with tumor cells expressing NR5A1 + , TBX19+, and POU1F1+, respectively. This emphasizes the distinct interaction axes between TAMs and tumor cells based on their lineage. Moreover, the connection between CX3CR1+ macrophages and tumor cells via INHBA-ACVR1B regulates tumor cell apoptosis. CONCLUSIONS In summary, the different subtypes of TIME and the interaction between TAM and tumor cells offer valuable insights into the control of TIME that affects the development of PitNET. These findings can be utilized as prospective targets for therapeutic interventions.
Collapse
Affiliation(s)
- Shaojian Lin
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changxi Han
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Han
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linfeng Zhao
- Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renyan Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyue Liu
- Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Zhang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanting Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujing Lai
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jintong Shi
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijun Cheng
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Tang
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Yao
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Wu
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Shen
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Youqiong Ye
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Li Xue
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, China.
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Neurosurgery, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
14
|
Mitra S, Malik R, Wong W, Rahman A, Hartemink AJ, Pritykin Y, Dey KK, Leslie CS. Single-cell multi-ome regression models identify functional and disease-associated enhancers and enable chromatin potential analysis. Nat Genet 2024; 56:627-636. [PMID: 38514783 PMCID: PMC11018525 DOI: 10.1038/s41588-024-01689-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
We present a gene-level regulatory model, single-cell ATAC + RNA linking (SCARlink), which predicts single-cell gene expression and links enhancers to target genes using multi-ome (scRNA-seq and scATAC-seq co-assay) sequencing data. The approach uses regularized Poisson regression on tile-level accessibility data to jointly model all regulatory effects at a gene locus, avoiding the limitations of pairwise gene-peak correlations and dependence on peak calling. SCARlink outperformed existing gene scoring methods for imputing gene expression from chromatin accessibility across high-coverage multi-ome datasets while giving comparable to improved performance on low-coverage datasets. Shapley value analysis on trained models identified cell-type-specific gene enhancers that are validated by promoter capture Hi-C and are 11× to 15× and 5× to 12× enriched in fine-mapped eQTLs and fine-mapped genome-wide association study (GWAS) variants, respectively. We further show that SCARlink-predicted and observed gene expression vectors provide a robust way to compute a chromatin potential vector field to enable developmental trajectory analysis.
Collapse
Affiliation(s)
- Sneha Mitra
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Wilfred Wong
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, New York City, NY, USA
| | - Afsana Rahman
- Hunter College, City University of New York, New York City, NY, USA
| | - Alexander J Hartemink
- Department of Computer Science, Duke University, Durham, NC, USA
- Program in Computational Biology and Bioinformatics, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Yuri Pritykin
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kushal K Dey
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
15
|
Li S, Zhao B, Yang H, Dai K, Cai Y, Xu H, Chen P, Wang F, Zhang Y. Comprehensive transcriptomic analysis revealing the regulatory dynamics and networks of the pituitary-testis axis in sheep across developmental stages. Front Vet Sci 2024; 11:1367730. [PMID: 38440388 PMCID: PMC10909840 DOI: 10.3389/fvets.2024.1367730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Spermatogenesis is a complex process intricately regulated by the hypothalamic-pituitary-testis (HPT) axis. However, research on the regulatory factors governing the HPT axis remains limited. This study addresses this gap by conducting a comprehensive analysis of transcriptomes from the pituitary and testis tissues across various developmental stages, encompassing embryonic day (E120), neonatal period (P0), pre-puberty (P90), and post-puberty day (P270). Utilizing edgeR and WGCNA, we identified stage-specific genes in both the pituitary and testis throughout the four developmental stages. Notably, 380, 242, 34, and 479 stage-specific genes were identified in the pituitary, while 886, 297, 201, and 3,678 genes were identified in the testis. Subsequent analyses unveiled associations between these stage-specific genes and crucial pathways such as the cAMP signaling pathway, GnRH secretion, and male gamete generation. Furthermore, leveraging single-cell data from the pituitary and testis, we identified some signaling pathways involving BMP, HGF, IGF, and TGF-β, highlighting mutual regulation between the pituitary and testis at different developmental stages. This study sheds light on the pivotal role of the pituitary-testis axis in the reproductive process of sheep across four distinct developmental stages. Additionally, it delves into the intricate regulatory networks governing reproduction, offering novel insights into the dynamics of the pituitary-testis axis within the reproductive system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
16
|
Rubinfeld H, Cohen ZR, Bendavid U, Fichman-Horn S, Levy-Barda A, David C, Melamed P, Shimon I. Erythropoietin-producing hepatocellular receptor B6 is highly expressed in non-functioning pituitary neuroendocrine tumors and its expression correlates with tumor size. Mol Biol Rep 2024; 51:297. [PMID: 38341842 PMCID: PMC10859332 DOI: 10.1007/s11033-023-09186-7] [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: 10/24/2023] [Accepted: 12/19/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND Erythropoietin-producing hepatocellular (EPH) receptors are the largest known family of receptor tyrosine kinases characterized in humans. These proteins are involved in tissue organization, synaptic plasticity, vascular development and the progression of various diseases including cancer. The Erythropoietin-producing hepatocellular receptor tyrosine kinase member EphB6 is a pseudokinase which has not attracted an equivalent amount of interest as its enzymatically-active counterparts. The aim of this study was to assess the expression of EphB6 in pituitary tumors. METHODS AND RESULTS Human normal pituitaries and pituitary tumors were examined for EphB6 mRNA expression using real-time PCR and for EphB6 protein by immunohistochemistry and Western blotting. EphB6 was highly expressed in non-functioning pituitary neuroendocrine tumors (NF-PitNETs) versus the normal pituitary and GH-secreting PitNETs. EphB6 mRNA expression was correlated with tumor size. CONCLUSIONS Our results suggest EphB6 aberrant expression in NF-PitNETs. Future studies are warranted to determine the role and significance of EphB6 in NF-PitNETs tumorigenesis.
Collapse
Affiliation(s)
- Hadara Rubinfeld
- Institute of Endocrinology, Diabetes & Metabolism and Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zvi R Cohen
- Department of Neurosurgery, Sheba Medical Center, Tel-Hashomer, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uzi Bendavid
- Department of Neurosurgery, Rabin Medical Center, Petah Tikva, Israel
| | | | - Adva Levy-Barda
- Biobank, Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Cfir David
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Philippa Melamed
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ilan Shimon
- Institute of Endocrinology, Diabetes & Metabolism and Felsenstein Medical Research Center, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.
- School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
17
|
Pérez Millán MI, Cheung LYM, Mercogliano F, Camilletti MA, Chirino Felker GT, Moro LN, Miriuka S, Brinkmeier ML, Camper SA. Pituitary stem cells: past, present and future perspectives. Nat Rev Endocrinol 2024; 20:77-92. [PMID: 38102391 PMCID: PMC10964491 DOI: 10.1038/s41574-023-00922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 12/17/2023]
Abstract
Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.
Collapse
Affiliation(s)
- María Inés Pérez Millán
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Florencia Mercogliano
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Maria Andrea Camilletti
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Gonzalo T Chirino Felker
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Lucia N Moro
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Santiago Miriuka
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Cai Y, Liu S, Zhao X, Ren L, Liu X, Gang X, Wang G. Pathogenesis, clinical features, and treatment of plurihormonal pituitary adenoma. Front Neurosci 2024; 17:1323883. [PMID: 38260014 PMCID: PMC10800528 DOI: 10.3389/fnins.2023.1323883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Plurihormonal pituitary adenoma (PPA) is a type of pituitary tumor capable of producing two or more hormones and usually presents as an aggressive, large adenoma. As yet, its pathogenesis remains unclear. This is the first study to systematically summarize the underlying pathogenesis of PPA. The pathogenesis is related to plurihormonal primordial stem cells, co-transcription factors, hormone co-expression, differential gene expression, and cell transdifferentiation. We conducted a literature review of PPA and analyzed its clinical characteristics. We found that the average age of patients with PPA was approximately 40 years, and most showed only one clinical symptom. The most common manifestation was acromegaly. Currently, PPA is treated with surgical resection. However, recent studies suggest that immunotherapy may be a potentially effective treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Yan N, Xie W, Wang D, Fang Q, Guo J, Chen Y, Li X, Gong L, Wang J, Guo W, Zhang X, Zhang Y, Gu J, Li C. Single-cell transcriptomic analysis reveals tumor cell heterogeneity and immune microenvironment features of pituitary neuroendocrine tumors. Genome Med 2024; 16:2. [PMID: 38167466 PMCID: PMC10759356 DOI: 10.1186/s13073-023-01267-3] [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: 03/10/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNETs) are one of the most common types of intracranial tumors. Currently, the cellular characteristics of normal pituitary and various other types of PitNETs are still not completely understood. METHODS We performed single-cell RNA sequencing (scRNA-seq) on 4 normal samples and 24 PitNET samples for comprehensive bioinformatics analysis. Findings regarding the function of PBK in the aggressive tumor cells were validated by siRNA knockdown, overexpression, and transwell experiments. RESULTS We first constructed a reference cell atlas of the human pituitary. Subsequent scRNA-seq analysis of PitNET samples, representing major tumor subtypes, shed light on the intrinsic cellular heterogeneities of the tumor cells and tumor microenvironment (TME). We found that the expression of hormone-encoding genes defined the major variations of the PIT1-lineage tumor cell transcriptomic heterogeneities. A sub-population of TPIT-lineage tumor cells highly expressing GZMK suggested a novel subtype of corticotroph tumors. In immune cells, we found two clusters of tumor-associated macrophages, which were both highly enriched in PitNETs but with distinct functional characteristics. In PitNETs, the stress response pathway was significantly activated in T cells. While a majority of these tumors are benign, our study unveils a common existence of aggressive tumor cells in the studied samples, which highly express a set of malignant signature genes. The following functional experiments confirmed the oncogenic role of selected up-regulated genes. The over-expression of PBK could promote both tumor cell proliferation and migration, and it was also significantly associated with poor prognosis in PitNET patients. CONCLUSIONS Our data and analysis manifested the basic cell types in the normal pituitary and inherent heterogeneity of PitNETs, identified several features of the tumor immune microenvironments, and found a novel epithelial cell sub-population with aggressive signatures across all the studied cases.
Collapse
Affiliation(s)
- Nan Yan
- MOE Key Laboratory of Bioinformatics, Department of Automation, BNRIST Bioinformatics Division, Tsinghua University, Beijing, 100084, China
| | - Weiyan Xie
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Dongfang Wang
- Biomedical Pioneering Innovative Center, Peking University, Beijing, 100871, China
| | - Qiuyue Fang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jing Guo
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Yiyuan Chen
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Xinqi Li
- MOE Key Laboratory of Bioinformatics, Department of Automation, BNRIST Bioinformatics Division, Tsinghua University, Beijing, 100084, China
| | - Lei Gong
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jialin Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Wenbo Guo
- MOE Key Laboratory of Bioinformatics, Department of Automation, BNRIST Bioinformatics Division, Tsinghua University, Beijing, 100084, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics, Department of Automation, BNRIST Bioinformatics Division, Tsinghua University, Beijing, 100084, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, Department of Automation, BNRIST Bioinformatics Division, Tsinghua University, Beijing, 100084, China.
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
20
|
Øystese KA, Olarescu NC, Lindskog C, Xheka F, Berg-Johnsen J, Petter Berg J, Bollerslev J, Casar-Borota O. Stem cell-associated transcription factors in non-functioning pituitary neuroendocrine tumours. FREE NEUROPATHOLOGY 2024; 5:5-14. [PMID: 38845811 PMCID: PMC11155689 DOI: 10.17879/freeneuropathology-2024-5396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Background: Cells with stem cell features have been described in pituitary neuroendocrine tumours (PitNETs). Transcription factors SOX2 and SOX9 are stem cell-associated markers while the pituitary progenitor marker PROP1 is involved in anterior pituitary development. We characterised the presence of these markers known to be present in the human pituitary in non-functioning (NF) PitNETs. Methods: We investigated the pituitary transcription factors SOX2, SOX9 and PROP1 by immunohistochemistry (IHC) (N = 125) and RT-qPCR (N = 78) in a retrospective cohort of clinically NF-PitNETs. The markers were scored based on the percentage of immunolabeled cells. IHC staining scores were compared to reintervention rates for the whole cohort, and to expression of FSH, LH or ER in gonadotroph NF-PitNETs. Results: Most tumours showed no or few cells positive for SOX2, SOX9 and PROP1. More patients with SOX2-negative tumours went through reintervention (40 % vs 19 %, p = 0.03). SOX2, SOX9 and PROP1 staining correlated positively to each other (SOX2 and SOX9 rs = 0.666, SOX2 and PROP1 rs = 0.704, SOX9 and PROP1 rs = 0.570, and p < 0.001 for all). In gonadotroph NF-PitNETs, staining for SOX2 and PROP1 was positively associated to FSHβ staining (p < 0.001 for both). Staining for SOX2, SOX9 and PROP1 was positively associated with gene expression of Estrogen Receptor 1 (ESR1) (p < 0.001, p = 0.004 and p < 0.001) and IHC staining for ERα (p = 0.001, p = 0.03 and p = 0.05, respectively). Conclusion: SOX2, SOX9 and PROP1 were present at low levels in NF-PitNETs. Absence of SOX2 staining was associated with a higher reintervention rate. The stem cell markers correlated positively with markers of gonadotroph differentiation in gonadotroph NF-PitNETs. SOX2 and SOX9 were frequently coexpressed and showed positivity in intratumoural cells with epithelial features, however without coexpression of pituitary transcription factors.
Collapse
Affiliation(s)
- Kristin Astrid Øystese
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Nicoleta Cristina Olarescu
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Cecilia Lindskog
- Cancer precision medicine, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fabjola Xheka
- Department of Clinical Pathology, Uppsala University Hospital, Uppsala, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Jon Berg-Johnsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Jens Petter Berg
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jens Bollerslev
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olivera Casar-Borota
- Department of Clinical Pathology, Uppsala University Hospital, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Chen Q, Leshkowitz D, Li H, van Impel A, Schulte-Merker S, Amit I, Rizzoti K, Levkowitz G. Neural plate progenitors give rise to both anterior and posterior pituitary cells. Dev Cell 2023; 58:2652-2665.e6. [PMID: 37683631 DOI: 10.1016/j.devcel.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
The pituitary is the master neuroendocrine gland, which regulates body homeostasis. It consists of the anterior pituitary/adenohypophysis harboring hormones producing cells and the posterior pituitary/neurohypophysis, which relays the passage of hormones from the brain to the periphery. It is accepted that the adenohypophysis originates from the oral ectoderm (Rathke's pouch), whereas the neural ectoderm contributes to the neurohypophysis. Single-cell transcriptomics of the zebrafish pituitary showed that cyp26b1-positive astroglial pituicytes of the neurohypophysis and prop1-positive adenohypophyseal progenitors expressed common markers implying lineage relatedness. Genetic tracing identifies that, in contrast to the prevailing dogma, neural plate precursors of zebrafish (her4.3+) and mouse (Sox1+) contribute to both neurohypophyseal and a subset of adenohypophyseal cells. Pituicyte-derived retinoic-acid-degrading enzyme Cyp26b1 fine-tunes differentiation of prop1+ progenitors into hormone-producing cells. These results challenge the notion that adenohypophyseal cells are exclusively derived from non-neural ectoderm and demonstrate that crosstalk between neuro- and adeno-hypophyseal cells affects differentiation of pituitary cells.
Collapse
Affiliation(s)
- Qiyu Chen
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Hanjie Li
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Present address: CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Andreas van Impel
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, UK
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, PO Box 26, Rehovot 7610001, Israel.
| |
Collapse
|
22
|
Armeni E, Alexandraki KI, Roncaroli F, Grossman AB. Primary Pituitary Carcinoids Do Not Exist: A Reappraisal in the Era of Pituitary Neuroendocrine Tumours. Arch Med Res 2023; 54:102841. [PMID: 37394342 DOI: 10.1016/j.arcmed.2023.102841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023]
Abstract
The World Health Organization classification of pituitary tumours, published in 2022, supported a change in the terminology from "pituitary adenoma" to "pituitary neuroendocrine tumour" (PitNET). The neuroendocrine cells represent an integral part of the diffuse neuroendocrine system, including, among others, thyroid C cells, the parathyroid chief cells, and the anterior pituitary. Normal and neoplastic adenohypophyseal neuroendocrine cells have light microscopic, ultrastructural features and an immunoprofile compatible with the neuroendocrine cells and neuroendocrine tumours from other organs. Moreover, neuroendocrine cells of pituitary origin express transcription factors which indicate their cell-lineage origin. Thus, pituitary tumours are now considered as a continuum with other neuroendocrine tumours. PitNETs may occasionally be aggressive. In this context, the term "pituitary carcinoid" has no specific meaning: it either represents a PitNET, or a metastasis to the pituitary gland of a neuroendocrine tumour (NET). An accurate pathological evaluation, combined where necessary with functional radionuclide imaging, can define the origin of the tumour. We recommend that clinicians liaise with patient groups to understand the terminology to define primary tumours of adenohypophyseal cells. It is incumbent upon the responsible clinician to explain the use of the word "tumour" in a given clinical context.
Collapse
Affiliation(s)
- Eleni Armeni
- Department of Endocrinology and NET Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - Krystallenia I Alexandraki
- 2(nd) Department of Surgery, Endocrine Unit, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Ashley B Grossman
- Department of Endocrinology and NET Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK; Barts and the London School of Medicine, London, UK; Green Templeton College, University of Oxford, UK.
| |
Collapse
|
23
|
Yan T, Wang R, Yao J, Luo M. Single-cell transcriptomic analysis reveals rich pituitary-Immune interactions under systemic inflammation. PLoS Biol 2023; 21:e3002403. [PMID: 38109308 PMCID: PMC10727439 DOI: 10.1371/journal.pbio.3002403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/26/2023] [Indexed: 12/20/2023] Open
Abstract
The pituitary represents an essential hub in the hypothalamus-pituitary-adrenal (HPA) axis. Pituitary hormone-producing cells (HPCs) release several hormones to regulate fundamental bodily functions under normal and stressful conditions. It is well established that the pituitary endocrine gland modulates the immune system by releasing adrenocorticotropic hormone (ACTH) in response to neuronal activation in the hypothalamus. However, it remains unclear how systemic inflammation regulates the transcriptomic profiles of pituitary HPCs. Here, we performed single-cell RNA-sequencing (scRNA-seq) of the mouse pituitary and revealed that upon inflammation, all major pituitary HPCs respond robustly in a cell type-specific manner, with corticotropes displaying the strongest reaction. Systemic inflammation also led to the production and release of noncanonical bioactive molecules, including Nptx2 by corticotropes, to modulate immune homeostasis. Meanwhile, HPCs up-regulated the gene expression of chemokines that facilitated the communication between the HPCs and immune cells. Together, our study reveals extensive interactions between the pituitary and immune system, suggesting multifaceted roles of the pituitary in mediating the effects of inflammation on many aspects of body physiology.
Collapse
Affiliation(s)
- Ting Yan
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Ruiyu Wang
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
- PTN Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Jingfei Yao
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Minmin Luo
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, China
- New Cornerstone Science Laboratory, Shenzhen, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Nazaret A, Fan JL, Lavallée VP, Cornish AE, Kiseliovas V, Masilionis I, Chun J, Bowman RL, Eisman SE, Wang J, Shi L, Levine RL, Mazutis L, Blei D, Pe'er D, Azizi E. Deep generative model deciphers derailed trajectories in acute myeloid leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566719. [PMID: 38014231 PMCID: PMC10680623 DOI: 10.1101/2023.11.11.566719] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Single-cell genomics has the potential to map cell states and their dynamics in an unbiased way in response to perturbations like disease. However, elucidating the cell-state transitions from healthy to disease requires analyzing data from perturbed samples jointly with unperturbed reference samples. Existing methods for integrating and jointly visualizing single-cell datasets from distinct contexts tend to remove key biological differences or do not correctly harmonize shared mechanisms. We present Decipher, a model that combines variational autoencoders with deep exponential families to reconstruct derailed trajectories ( https://github.com/azizilab/decipher ). Decipher jointly represents normal and perturbed single-cell RNA-seq datasets, revealing shared and disrupted dynamics. It further introduces a novel approach to visualize data, without the need for methods such as UMAP or TSNE. We demonstrate Decipher on data from acute myeloid leukemia patient bone marrow specimens, showing that it successfully characterizes the divergence from normal hematopoiesis and identifies transcriptional programs that become disrupted in each patient when they acquire NPM1 driver mutations.
Collapse
|
25
|
Rizzoti K, Chakravarty P, Sheridan D, Lovell-Badge R. SOX9-positive pituitary stem cells differ according to their position in the gland and maintenance of their progeny depends on context. SCIENCE ADVANCES 2023; 9:eadf6911. [PMID: 37792947 PMCID: PMC10550238 DOI: 10.1126/sciadv.adf6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Stem cell (SC) differentiation and maintenance of resultant progeny underlie cell turnover in many organs, but it is difficult to pinpoint the contribution of either process. In the pituitary, a central regulator of endocrine axes, adult SCs undergo activation after target organ ablation, providing a well-characterized paradigm to study an adaptative response in a multi-organ system. Here, we used single-cell technologies to characterize SC heterogeneity and mobilization together with lineage tracing. We show that SC differentiation occurs more frequently than thought previously. In adaptative conditions, differentiation increases and is more diverse than demonstrated by the lineage tracing experiments. Detailed examination of SC progeny suggests that maintenance of selected nascent cells underlies SC output, highlighting a trophic role for the microenvironment. Analyses of cell trajectories further predict pathways and potential regulators. Our model provides a valuable system to study the influence of evolving states on the mechanisms of SC mobilization.
Collapse
Affiliation(s)
- Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Daniel Sheridan
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
26
|
Gonigam RL, Weis KE, Ge X, Yao B, Zhang Q, Raetzman LT. Characterization of Somatotrope Cell Expansion in Response to GHRH in the Neonatal Mouse Pituitary. Endocrinology 2023; 164:bqad131. [PMID: 37616545 PMCID: PMC11009787 DOI: 10.1210/endocr/bqad131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
In humans and mice, loss-of-function mutations in growth hormone-releasing hormone receptor (GHRHR) cause isolated GH deficiency. The mutant GHRHR mouse model, GhrhrLit/Lit (LIT), exhibits loss of serum GH, but also fewer somatotropes. However, how loss of GHRH signaling affects expansion of stem and progenitor cells giving rise to GH-producing cells is unknown. LIT mice and wild-type littermates were examined for differences in proliferation and gene expression of pituitary lineage markers by quantitative reverse transcription polymerase chain reaction and immunohistochemistry at postnatal day 5 (p5) and 5 weeks. At p5, the LIT mouse shows a global decrease in pituitary proliferation measured by proliferation marker Ki67 and phospho-histone H3. This proliferative defect is seen in a pituitary cell expressing POU1F1 with or without GH. SOX9-positive progenitors show no changes in proliferation in p5 LIT mice. Additionally, the other POU1F1 lineage cells are not decreased in number; rather, we observe an increase in lactotrope cell population as well as messenger RNA for Tshb and Prl. In the 5-week LIT pituitary, the proliferative deficit in POU1F1-expressing cells observed neonatally persists, while the number and proliferative proportion of SOX9 cells do not appear changed. Treatment of cultured pituitary explants with GHRH promotes proliferation of POU1F1-expressing cells, but not GH-positive cells, in a mitogen-activated protein kinase-dependent manner. These findings indicate that hypothalamic GHRH targets proliferation of a POU1F1-positive cell, targeted to the somatotrope lineage, to fine tune their numbers.
Collapse
Affiliation(s)
- Richard L Gonigam
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Karen E Weis
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiyu Ge
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Boyuan Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
27
|
He T, Guo W, Yang G, Su H, Dou A, Chen L, Ma T, Su J, Liu M, Su B, Qi W, Li H, Mao W, Wang X, Li X, Yang Y, Song Y, Cao G. A Single-Cell Atlas of an Early Mongolian Sheep Embryo. Vet Sci 2023; 10:543. [PMID: 37756065 PMCID: PMC10536297 DOI: 10.3390/vetsci10090543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Cell types have been established during organogenesis based on early mouse embryos. However, our understanding of cell types and molecular mechanisms in the early embryo development of Mongolian sheep has been hampered. This study presents the first comprehensive single-cell transcriptomic characterization at E16 in Ujumqin sheep and Hulunbuir short-tailed sheep. Thirteen major cell types were identified at E16 in Ujumqin sheep, and eight major cell types were identified at E16 in Hulunbuir short-tailed sheep. Function enrichment analysis showed that several pathways were significantly enriched in the TGF-beta signaling pathway, the Hippo signaling pathway, the platelet activation pathway, the riboflavin metabolism pathway, the Wnt signaling pathway, regulation of the actin cytoskeleton, and the insulin signaling pathway in the notochord cluster. Glutathione metabolism, glyoxylate, and dicarboxylate metabolism, the citrate cycle, thyroid hormone synthesis, pyruvate metabolism, cysteine and methionine metabolism, thermogenesis, and the VEGF signaling pathway were significantly enriched in the spinal cord cluster. Steroid biosynthesis, riboflavin metabolism, the cell cycle, the Hippo signaling pathway, the Hedgehog signaling pathway, the FoxO signaling pathway, the JAK-STAT signaling pathway, and the Wnt signaling pathway were significantly enriched in the paraxial mesoderm cluster. The notochord cluster, spinal cord cluster, and paraxial mesoderm cluster were found to be highly associated with tail development. Pseudo-time analysis demonstrated that the mesenchyme can translate to the notochord in Ujumqin sheep. Molecular assays revealed that the Hippo signaling pathway was enriched in Ujumqin sheep. This comprehensive single-cell map revealed previously unrecognized signaling pathways that will further our understanding of the mechanism of short-tailed sheep formation.
Collapse
Affiliation(s)
- Tingyi He
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Huhhot 010031, China
| | - Wenrui Guo
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Guang Yang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (G.Y.); (X.L.)
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Science, Inner Mongolia University, Hohhot 010020, China
| | - Hong Su
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Aolei Dou
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Lu Chen
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Teng Ma
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Jie Su
- Department of Medical Neurobiology, Inner Mongolia Medical University, Huhhot 010030, China;
| | - Moning Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Budeng Su
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Wangmei Qi
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Haijun Li
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Wei Mao
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Xiumei Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (G.Y.); (X.L.)
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Science, Inner Mongolia University, Hohhot 010020, China
| | - Yanyan Yang
- Institute of Animal Husbandry, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Huhhot 010031, China
| | - Yongli Song
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010020, China; (G.Y.); (X.L.)
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Science, Inner Mongolia University, Hohhot 010020, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Key Laboratory of Animal Embryo, and Development Engineering Autonomous Region, Inner Mongolia Agricultural University, Hohhot 010018, China; (T.H.); (W.G.); (H.S.); (A.D.); (L.C.); (T.M.); (M.L.); (B.S.); (W.Q.); (H.L.); (W.M.); (X.W.)
| |
Collapse
|
28
|
McDonald R, Larsen M, Liu Z, Southekal S, Eudy J, Guda C, Kumar TR. RNA-seq analysis identifies age-dependent changes in expression of mRNAs - encoding N-glycosylation pathway enzymes in mouse gonadotropes. Mol Cell Endocrinol 2023; 574:111971. [PMID: 37301504 PMCID: PMC10528389 DOI: 10.1016/j.mce.2023.111971] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/01/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Follicle-stimulating hormone (FSH) is a glycoprotein that is assembled as a heterodimer of α/β subunits in gonadotropes. Each subunit contains two N-glycan chains. Our previous in vivo genetic studies identified that at least one N-glycan chain must be present on the FSHβ subunit for efficient FSH dimer assembly and secretion. Moreover, macroheterogeneity observed uniquely on human FSHβ results in ratiometric changes in age-specific FSH glycoforms, particularly during menopausal transition. Despite the recognition of many prominent roles of sugars on FSH including dimer assembly and secretion, serum half-life, receptor binding and signal transduction, the N-glycosylation machinery in gonadotropes has never been defined. Here, we used a mouse model in which gonadotropes are GFP-labeled in vivo and achieved rapid purification of GFP+ gonadotropes from pituitaries of female mice at reproductively young, middle, and old ages. We identified by RNA-seq analysis 52 mRNAs encoding N-glycosylation pathway enzymes expressed in 3- and 8-10-month-old mouse gonadotropes. We hierarchically mapped and localized the enzymes to distinct subcellular organelles within the N-glycosylation biosynthetic pathway. Of the 52 mRNAs, we found 27 mRNAs are differentially expressed between the 3- and 8-10-month old mice. We subsequently selected 8 mRNAs which showed varying changes in expression for confirmation of abundance in vivo via qPCR analysis, using more expanded aging time points with distinct 8-month and 14-month age groups. Real time qPCR analysis indicated dynamic changes in expression of N-glycosylation pathway enzyme-encoding mRNAs across the life span. Notably, computational analysis predicted the promoters of genes encoding these 8 mRNAs contain multiple high probability binding sites for estrogen receptor-1 and progesterone receptor. Collectively, our studies define the N-glycome and identify age-specific dynamic changes in mRNAs encoding N-glycosylation pathway enzymes in mouse gonadotropes. Our studies suggest the age-related decline in ovarian steroids may regulate expression of N-glycosylation enzymes in mouse gonadotropes and explain the age-related N-glycosylation shift previously observed on human FSHβ subunit in pituitaries of women.
Collapse
Affiliation(s)
- Rosemary McDonald
- Garduate Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Siddesh Southekal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - James Eudy
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - T Rajendra Kumar
- Garduate Program in Integrated Physiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
29
|
Bian Y, Hahn H, Uhmann A. The hidden hedgehog of the pituitary: hedgehog signaling in development, adulthood and disease of the hypothalamic-pituitary axis. Front Endocrinol (Lausanne) 2023; 14:1219018. [PMID: 37476499 PMCID: PMC10355329 DOI: 10.3389/fendo.2023.1219018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023] Open
Abstract
Hedgehog signaling plays pivotal roles in embryonic development, adult homeostasis and tumorigenesis. However, its engagement in the pituitary gland has been long underestimated although Hedgehog signaling and pituitary embryogenic development are closely linked. Thus, deregulation of this signaling pathway during pituitary development results in malformation of the gland. Research of the last years further implicates a regulatory role of Hedgehog signaling in the function of the adult pituitary, because its activity is also interlinked with homeostasis, hormone production, and most likely also formation of neoplasms of the gland. The fact that this pathway can be efficiently targeted by validated therapeutic strategies makes it a promising candidate for treating pituitary diseases. We here summarize the current knowledge about the importance of Hedgehog signaling during pituitary development and review recent data that highlight the impact of Hedgehog signaling in the healthy and the diseased adult pituitary gland.
Collapse
|
30
|
Cheung LYM, Menage L, Rizzoti K, Hamilton G, Dumontet T, Basham K, Daly AZ, Brinkmeier ML, Masser BE, Treier M, Cobb J, Delogu A, Lovell-Badge R, Hammer GD, Camper SA. Novel Candidate Regulators and Developmental Trajectory of Pituitary Thyrotropes. Endocrinology 2023; 164:bqad076. [PMID: 37183548 PMCID: PMC10227867 DOI: 10.1210/endocr/bqad076] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations.
Collapse
Affiliation(s)
- Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucy Menage
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Greg Hamilton
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Typhanie Dumontet
- Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kaitlin Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation: Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandre Z Daly
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation is Vanguard, Valley Forge, PA 19482, USA
| | | | - Bailey E Masser
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mathias Treier
- Max Delbrϋck Center for Molecular Medicine (MDC), 13092 Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - John Cobb
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Alessio Delogu
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
Lyu L, Jiang Y, Ma W, Li H, Liu X, Li L, Shen A, Yu Y, Jiang S, Li H, Zhou P, Yin S. Single-cell sequencing of PIT1-positive pituitary adenoma highlights the pro-tumour microenvironment mediated by IFN-γ-induced tumour-associated fibroblasts remodelling. Br J Cancer 2023; 128:1117-1133. [PMID: 36631635 PMCID: PMC10006201 DOI: 10.1038/s41416-022-02126-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND PIT1-positive pituitary adenoma (PIT1-PA) is one of the most important lineages of pituitary adenoma (PA), which causes systematic endocrine disorders and a worse prognosis. Tumour-associated fibroblast (TAF) is a crucial stroma cell type in the tumour microenvironment (TME). However, cellular and functional heterogeneity of TAF and immune cells in PIT1-PA have not been fully investigated. METHODS By single-cell RNA sequencing of four PIT1-PAs and further analyses, we characterised the molecular and functional profiles of 28 different cell subtypes. RESULTS PA stem cells in PIT1/SF1-positve PA were in a hybrid epithelial/mesenchymal state, and differentiated along the PIT1- and SF- dependent branches. C1Q was overwhelmingly expressed in tumour-associated macrophages, indicating its pro-tumoral functionality. PIT1-PA progression was characterised by lower cell-cell communication strength and higher cell adhesion-associated signals, indicating the immunosuppressive but pro-invasive microenvironment. IFN-γ signal repressed functional remodelling of myofibroblastic TAF (mTAF) towards inflammatory TAF/antigen-presenting TAF. IFN-γ inhibited mTAF phenotypes and N-cadherin expression through STAT3 signal axis. CDH2 knockdown in TAFs abrogated their pro-tumour function in PAs. CONCLUSIONS Our study builds up a cellular landscape of PIT1-PA TME and highlights anti-tumour function of IFN-γ mediated TAF remodelling, which benefits clinical treatments and drug development.
Collapse
Affiliation(s)
- Liang Lyu
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China
| | - Yong Jiang
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
- Department of Neurosurgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Weichao Ma
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
- Department of Neurosurgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Haiyan Li
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China
| | - Xiaoling Liu
- Departments of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ao Shen
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yang Yu
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
| | - Shu Jiang
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China
| | - Huihui Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Clinical Medical School, Sichuan University, Chengdu, China.
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Peizhi Zhou
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China.
| | - Senlin Yin
- Department of Neurosurgery, Pituitary Adenoma Multidisciplinary Center, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
32
|
Kolnes AJ, Øystese KAB, Sjöstedt E, Olarescu NC, Heck A, Pahnke J, Dahlberg D, Berg-Johnsen J, Ringstad G, Casar-Borota O, Bollerslev J, Jørgensen AP. TGFBR3L is associated with gonadotropin production in non-functioning gonadotroph pituitary neuroendocrine tumours. Pituitary 2023:10.1007/s11102-023-01310-x. [PMID: 36952069 DOI: 10.1007/s11102-023-01310-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE Transforming growth factor-beta receptor 3-like (TGFBR3L) is a pituitary enriched membrane protein selectively detected in gonadotroph cells. TGFBR3L is named after transforming growth factor-beta receptor 3 (TGFBR3), an inhibin A co-receptor in mice, due to sequence identity to the C-terminal region. We aimed to characterize TGFBR3L detection in a well-characterized, prospectively collected cohort of non-functioning pituitary neuroendocrine tumours (NF-PitNETs) and correlate it to clinical data. METHODS 144 patients operated for clinically NF-PitNETs were included. Clinical, radiological and biochemical data were recorded. Immunohistochemical (IHC) staining for FSHβ and LHβ was scored using the immunoreactive score (IRS), TGFBR3L and TGFBR3 were scored by the percentage of positive stained cells. RESULTS TGFBR3L staining was selectively present in 52% of gonadotroph tumours. TGFBR3L was associated to IRS of LHβ (median 2 [IQR 0-3] in TGFBR3L negative and median 6 [IQR 3-9] in TGFBR3L positive tumours, p < 0.001), but not to the IRS of FSHβ (p = 0.32). The presence of TGFBR3L was negatively associated with plasma gonadotropin concentrations in males (P-FSH median 5.5 IU/L [IQR 2.9-9.6] and median 3.0 [IQR 1.8-5.6] in TGFBR3L negative and positive tumours respectively, p = 0.008) and P-LH (median 2.8 IU/L [IQR 1.9-3.7] and median 1.8 [IQR 1.1-3.0] in TGFBR3L negative and positive tumours respectively, p = 0.03). TGFBR3 stained positive in 22% (n = 25) of gonadotroph tumours with no correlation to TGFBR3L. CONCLUSION TGFBR3L was selectively detected in half (52%) of gonadotroph NF-PitNETs. The association to LHβ staining and plasma gonadotropins suggests that TGFBR3L may be involved in hormone production in gonadotroph NF-PitNETs.
Collapse
Affiliation(s)
- Anders Jensen Kolnes
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Kristin Astrid Berland Øystese
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway.
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway.
| | - Evelina Sjöstedt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nicoleta Cristina Olarescu
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Ansgar Heck
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Jens Pahnke
- Department of Pathology, Section of Neuropathology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Pahnke lab (Drug Discovery and Chemical Biology), Lübeck Institute of Dermatology, LIED, University of Lübeck, Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Rīga, Latvia
| | - Daniel Dahlberg
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Jon Berg-Johnsen
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Olivera Casar-Borota
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Clinical Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Jens Bollerslev
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Sognsvannsveien 20, 0372, Oslo, Norway
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Anders Palmstrøm Jørgensen
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
33
|
Intra-pituitary follicle-stimulating hormone signaling regulates hepatic lipid metabolism in mice. Nat Commun 2023; 14:1098. [PMID: 36841874 PMCID: PMC9968338 DOI: 10.1038/s41467-023-36681-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/09/2023] [Indexed: 02/27/2023] Open
Abstract
Inter-organ communication is a major hallmark of health and is often orchestrated by hormones released by the anterior pituitary gland. Pituitary gonadotropes secrete follicle-stimulating hormone (FSH) and luteinizing hormone (LH) to regulate gonadal function and control fertility. Whether FSH and LH also act on organs other than the gonads is debated. Here, we find that gonadotrope depletion in adult female mice triggers profound hypogonadism, obesity, glucose intolerance, fatty liver, and bone loss. The absence of sex steroids precipitates these phenotypes, with the notable exception of fatty liver, which results from ovary-independent actions of FSH. We uncover paracrine FSH action on pituitary corticotropes as a mechanism to restrain the production of corticosterone and prevent hepatic steatosis. Our data demonstrate that functional communication of two distinct hormone-secreting cell populations in the pituitary regulates hepatic lipid metabolism.
Collapse
|
34
|
Zhang Q, Yao B, Long X, Chen Z, He M, Wu Y, Qiao N, Ma Z, Ye Z, Zhang Y, Yao S, Wang Y, Cheng H, Chen H, Ye H, Wang Y, Li Y, Chen J, Zhang Z, Guo F, Zhao Y. Single-cell sequencing identifies differentiation-related markers for molecular classification and recurrence prediction of PitNET. Cell Rep Med 2023; 4:100934. [PMID: 36754052 PMCID: PMC9975294 DOI: 10.1016/j.xcrm.2023.100934] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/29/2022] [Accepted: 01/13/2023] [Indexed: 02/10/2023]
Abstract
Pituitary neuroendocrine tumor (PitNET) is one of the most common intracranial tumors with variable recurrence rate. Currently, the recurrence prediction is unsatisfying and can be improved by understanding the cellular origins and differentiation status. Here, to comprehensively reveal the origin of PitNET, we perform comparative analysis of single-cell RNA sequencing data from 3 anterior pituitary glands and 21 PitNETs. We identify distinct genes representing major subtypes of well and poorly differentiated PitNETs in each lineage. To further verify the predictive value of differentiation biomarkers, we include an independent cohort of 800 patients with an average follow-up of 7.2 years. In both PIT1 and TPIT lineages, poorly differentiated groups show significantly higher recurrence rates while well-differentiated groups show higher recurrence rates in SF1 lineage. Our findings reveal the possible origin and differentiation status of PitNET based on which new differentiation classification is proposed and verified to predict tumor recurrence.
Collapse
Affiliation(s)
- Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Boyuan Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Xin Long
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengyuan Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Min He
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yue Wu
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Zengyi Ma
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Shun Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Ye Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Haixia Cheng
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Hong Chen
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Hongying Ye
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Yimin Li
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianhua Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaoyun Zhang
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.
| | - Fan Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai 200040, China; Neurosurgical Institute of Fudan University, Shanghai 200040, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Shanghai Clinical Medical Center of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
35
|
Identification and gene expression profiling of human gonadotrophic pituitary adenoma stem cells. Acta Neuropathol Commun 2023; 11:24. [PMID: 36750863 PMCID: PMC9906881 DOI: 10.1186/s40478-023-01517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Gonadotrophic pituitary adenoma is a major subtype of pituitary adenoma in the sellar region, but it is rarely involved in the hypersecretion of hormones into blood; thus, it is commonly regarded as "non-functioning." Its tumorigenic mechanisms remain unknown. The aim of this study was to identify human gonadotrophic pituitary adenoma stem cells (hPASCs) and explore the underlying gene expression profiles. In addition, the potential candidate genes involved in the invasive properties of pituitary adenoma were examined. METHODS The hPASCs from 14 human gonadotrophic pituitary adenoma clinical samples were cultured and verified via immunohistochemistry. Genetic profiling of hPASCs and the matched tumor cells was performed through RNA-sequencing and subjected to enrichment analysis. By aligning the results with public databases, the candidate genes were screened and examined in invasive and non-invasive gonadotrophic pituitary adenomas using Real-time polymerase chain reaction. RESULTS The hPASCs were successfully isolated and cultured from gonadotrophic pituitary adenoma in vitro, which were identified as positive for generic stem cell markers (Sox2, Oct4, Nestin and CD133) via immunohistochemical staining. The hPASCs could differentiate into the tumor cells expressing follicle-stimulating hormone in the presence of fetal bovine serum in the culture medium. Through RNA-sequencing, 1352 differentially expressed genes were screened and identified significantly enriched in various gene ontologies and important pathways. The expression levels of ANXA2, PMAIP1, SPRY2, C2CD4A, APOD, FGF14 and FKBP10 were significantly upregulated while FNDC5 and MAP3K4 were downregulated in the invasive gonadotrophic pituitary adenomas compared to the non-invasive ones. CONCLUSION Genetic profiling of hPASCs may explain the tumorigenesis and invasiveness of gonadotrophic pituitary adenoma. ANXA2 may serve as a potential therapeutic target for the treatment of gonadotrophic pituitary adenoma.
Collapse
|
36
|
Fletcher PA, Smiljanic K, Prévide RM, Constantin S, Sherman AS, Coon SL, Stojilkovic SS. The astroglial and stem cell functions of adult rat folliculostellate cells. Glia 2023; 71:205-228. [PMID: 36093576 PMCID: PMC9772113 DOI: 10.1002/glia.24267] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
The mammalian pituitary gland is a complex organ consisting of hormone-producing cells, anterior lobe folliculostellate cells (FSCs), posterior lobe pituicytes, vascular pericytes and endothelial cells, and Sox2-expressing stem cells. We present single-cell RNA sequencing and immunohistofluorescence analyses of pituitary cells of adult female rats with a focus on the transcriptomic profiles of nonhormonal cell types. Samples obtained from whole pituitaries and separated anterior and posterior lobe cells contained all expected pituitary resident cell types and lobe-specific vascular cell subpopulations. FSCs and pituicytes expressed S100B, ALDOC, EAAT1, ALDH1A1, and VIM genes and proteins, as well as other astroglial marker genes, some common and some cell type-specific. We also found that the SOX2 gene and protein were expressed in ~15% of pituitary cells, including FSCs, pituicytes, and a fraction of hormone-producing cells, arguing against its stem cell specificity. FSCs comprised two Sox2-expressing subclusters; FS1 contained more cells but lower genetic diversity, while FS2 contained proliferative cells, shared genes with hormone-producing cells, and expressed genes consistent with stem cell niche formation, regulation of cell proliferation and stem cell pluripotency, including the Hippo and Wnt pathways. FS1 cells were randomly distributed in the anterior and intermediate lobes, while FS2 cells were localized exclusively in the marginal zone between the anterior and intermediate lobes. These data indicate the identity of the FSCs as anterior pituitary-specific astroglia, with FS1 cells representing differentiated cells equipped for classical FSC roles and FS2 cells exhibiting additional stem cell-like features.
Collapse
Affiliation(s)
- Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Rafael M. Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Steven L. Coon
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, 20892
| | - Stanko S. Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| |
Collapse
|
37
|
Yan CY, Wu FY, Sun F, Fang Y, Zhang RJ, Zhang CR, Zhang CX, Wang Z, Yang RM, Yang L, Dong M, Zhang QY, Ye XP, Song HD, Zhao SX. The isl2a transcription factor regulates pituitary development in zebrafish. Front Endocrinol (Lausanne) 2023; 14:920548. [PMID: 36824359 PMCID: PMC9941339 DOI: 10.3389/fendo.2023.920548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND ISL LIM homeobox 2, also known as insulin gene enhancer protein ISL-2 (ISL2), is a transcription factor gene that participates in a wide range of developmental events. However, the role of ISL2 in the hypothalamus-pituitary-thyroid axis is largely unknown. In the present study, we characterized the expression patterns of ISL2 and revealed its regulative role during embryogenesis using zebrafish. METHODS We used the CRISPR/Cas9 system to successfully establish homozygous ISL2-orthologue (isl2a and isl2b) knockout zebrafish. Moreover, we utilized these knockout zebrafish to analyze the pituitary and thyroid phenotypes in vivo. For further molecular characterization, in situ hybridization and immunofluorescence were performed. RESULTS The isl2a mutant zebrafish presented with thyroid hypoplasia, reduced whole-body levels of thyroid hormones, increased early mortality, gender imbalance, and morphological retardation during maturity. Additionally, thyrotropes, a pituitary cell type, was notably decreased during development. Importantly, the transcriptional levels of pituitary-thyroid axis hormones-encoding genes, such as tshba, cga, and tg, were significantly decreased in isl2a mutants. Finally, the thyroid dysplasia in isl2a mutant larvae may be attributed to a reduction in proliferation rather than changes in apoptosis. CONCLUSIONS In summary, isl2a regulates the transcriptional levels of marker genes in hypothalamus-pituitary-thyroid axis, and isl2a knockout causing low thyroid hormone levels in zebrafish. Thus, isl2a identified by the present study, is a novel regulator for pituitary cell differentiation in zebrafish, resulting in thyroid gland hypoplasia and phenotypes of hypothyroidism.
Collapse
Affiliation(s)
- Chen-Yan Yan
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Feng Sun
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Ya Fang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Rui-Jia Zhang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Chang-Run Zhang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Cao-Xu Zhang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Rui-Meng Yang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Liu Yang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Mei Dong
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Qian-Yue Zhang
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Xiao-Ping Ye
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Huai-Dong Song
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
- *Correspondence: Shuang-Xia Zhao, ; Huai-Dong Song,
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics and Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
- *Correspondence: Shuang-Xia Zhao, ; Huai-Dong Song,
| |
Collapse
|
38
|
Transcriptomic Profiles of Normal Pituitary Cells and Pituitary Neuroendocrine Tumor Cells. Cancers (Basel) 2022; 15:cancers15010110. [PMID: 36612109 PMCID: PMC9817686 DOI: 10.3390/cancers15010110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
The pituitary gland is one of the most cellularly diverse regions of the brain. Recent advancements in transcriptomic biology, such as single-cell RNA sequencing, bring an unprecedented glimpse into the molecular composition of the pituitary, both in its normal physiological state and in disease. Deciphering the normal pituitary transcriptomic signatures provides a better insight into the ontological origin and development of five types of endocrine cells, a process involving complex cascades of transcription factors that are still being established. In parallel with these observations about normal pituitary development, recent transcriptomic findings on pituitary neuroendocrine tumors (PitNETs) demonstrate both preservations and changes in transcription factor expression patterns compared to those seen during gland development. Furthermore, recent studies also identify differentially expressed genes that drive various tumor behaviors, including hormone hypersecretion and tumor aggression. Understanding the comprehensive multiomic profiles of PitNETs is essential in developing molecular profile-based therapies for PitNETs not curable with current treatment modalities and could eventually help align PitNETs with the breakthroughs being made in applying precision medicine to other tumors.
Collapse
|
39
|
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: 3.0] [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
|
40
|
Wen L, Li G, Huang T, Geng W, Pei H, Yang J, Zhu M, Zhang P, Hou R, Tian G, Su W, Chen J, Zhang D, Zhu P, Zhang W, Zhang X, Zhang N, Zhao Y, Cao X, Peng G, Ren X, Jiang N, Tian C, Chen ZJ. Single-cell technologies: From research to application. Innovation (N Y) 2022; 3:100342. [PMID: 36353677 PMCID: PMC9637996 DOI: 10.1016/j.xinn.2022.100342] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
In recent years, more and more single-cell technologies have been developed. A vast amount of single-cell omics data has been generated by large projects, such as the Human Cell Atlas, the Mouse Cell Atlas, the Mouse RNA Atlas, the Mouse ATAC Atlas, and the Plant Cell Atlas. Based on these single-cell big data, thousands of bioinformatics algorithms for quality control, clustering, cell-type annotation, developmental inference, cell-cell transition, cell-cell interaction, and spatial analysis are developed. With powerful experimental single-cell technology and state-of-the-art big data analysis methods based on artificial intelligence, the molecular landscape at the single-cell level can be revealed. With spatial transcriptomics and single-cell multi-omics, even the spatial dynamic multi-level regulatory mechanisms can be deciphered. Such single-cell technologies have many successful applications in oncology, assisted reproduction, embryonic development, and plant breeding. We not only review the experimental and bioinformatics methods for single-cell research, but also discuss their applications in various fields and forecast the future directions for single-cell technologies. We believe that spatial transcriptomics and single-cell multi-omics will become the next booming business for mechanism research and commercial industry.
Collapse
Affiliation(s)
- Lu Wen
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Guoqiang Li
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Geng
- School of Chemical Engineering and Technology, Sun Yat-Sen University, Zhuhai 519082, China
| | - Hao Pei
- Mozhuo Biotech (Zhejiang) Co., Ltd., Tongxiang, Jiaxing 314500, China
| | | | - Miao Zhu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Rui Hou
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Geng Tian
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Wentao Su
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jian Chen
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, China
| | - Pingan Zhu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Wei Zhang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xiuxin Zhang
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yunlong Zhao
- Advanced Technology Institute, University of Surrey, Guildford, Surrey, GU2 7XH, UK
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, UK
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guangdun Peng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Caihuan Tian
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
41
|
Chakrabarti J, Pandey R, Churko JM, Eschbacher J, Mallick S, Chen Y, Hermes B, Mallick P, Stansfield BN, Pond KW, Thorne CA, Yuen KCJ, Little AS, Zavros Y. Development of Human Pituitary Neuroendocrine Tumor Organoids to Facilitate Effective Targeted Treatments of Cushing's Disease. Cells 2022; 11:3344. [PMID: 36359740 PMCID: PMC9659185 DOI: 10.3390/cells11213344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 08/25/2023] Open
Abstract
(1) Background: Cushing's disease (CD) is a serious endocrine disorder caused by an adrenocorticotropic hormone (ACTH)-secreting pituitary neuroendocrine tumor (PitNET) that stimulates the adrenal glands to overproduce cortisol. Chronic exposure to excess cortisol has detrimental effects on health, including increased stroke rates, diabetes, obesity, cognitive impairment, anxiety, depression, and death. The first-line treatment for CD is pituitary surgery. Current surgical remission rates reported in only 56% of patients depending on several criteria. The lack of specificity, poor tolerability, and low efficacy of the subsequent second-line medical therapies make CD a medical therapeutic challenge. One major limitation that hinders the development of specific medical therapies is the lack of relevant human model systems that recapitulate the cellular composition of PitNET microenvironment. (2) Methods: human pituitary tumor tissue was harvested during transsphenoidal surgery from CD patients to generate organoids (hPITOs). (3) Results: hPITOs generated from corticotroph, lactotroph, gonadotroph, and somatotroph tumors exhibited morphological diversity among the organoid lines between individual patients and amongst subtypes. The similarity in cell lineages between the organoid line and the patient's tumor was validated by comparing the neuropathology report to the expression pattern of PitNET specific markers, using spectral flow cytometry and exome sequencing. A high-throughput drug screen demonstrated patient-specific drug responses of hPITOs amongst each tumor subtype. Generation of induced pluripotent stem cells (iPSCs) from a CD patient carrying germline mutation CDH23 exhibited dysregulated cell lineage commitment. (4) Conclusions: The human pituitary neuroendocrine tumor organoids represent a novel approach in how we model complex pathologies in CD patients, which will enable effective personalized medicine for these patients.
Collapse
Affiliation(s)
- Jayati Chakrabarti
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Ritu Pandey
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
- Center for Biomedical Informatics and Biostatistics, University of Arizona Health Sciences, Tucson, AZ 85721, USA
| | - Jared M. Churko
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Jennifer Eschbacher
- Department of Neuropathology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Saptarshi Mallick
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Yuliang Chen
- University of Arizona Cancer Center Bioinformatics Core, Tucson, AZ 85721, USA
| | - Beth Hermes
- Department of Neuropathology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Palash Mallick
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Ben N. Stansfield
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Kelvin W. Pond
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Curtis A. Thorne
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Kevin C. J. Yuen
- Department of Neuroendocrinology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Andrew S. Little
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Yana Zavros
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| |
Collapse
|
42
|
Lin YF, Schang G, Buddle ERS, Schultz H, Willis TL, Ruf-Zamojski F, Zamojski M, Mendelev N, Boehm U, Sealfon SC, Andoniadou CL, Bernard DJ. Steroidogenic Factor 1 Regulates Transcription of the Inhibin B Coreceptor in Pituitary Gonadotrope Cells. Endocrinology 2022; 163:6661776. [PMID: 35957608 PMCID: PMC9761571 DOI: 10.1210/endocr/bqac131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 11/19/2022]
Abstract
The inhibins control reproduction by suppressing follicle-stimulating hormone synthesis in pituitary gonadotrope cells. The newly discovered inhibin B coreceptor, TGFBR3L, is selectively and highly expressed in gonadotropes in both mice and humans. Here, we describe our initial characterization of mechanisms controlling cell-specific Tgfbr3l/TGFBR3L transcription. We identified two steroidogenic factor 1 (SF-1 or NR5A1) cis-elements in the proximal Tgfbr3l promoter in mice. SF-1 induction of murine Tgfbr3l promoter-reporter activity was inhibited by mutations in one or both sites in heterologous cells. In homologous cells, mutation of these cis-elements or depletion of endogenous SF-1 similarly decreased reporter activity. We observed nearly identical results when using a human TGFBR3L promoter-reporter. The Tgfbr3l gene was tightly compacted and Tgfbr3l mRNA expression was essentially absent in gonadotropes of SF-1 (Nr5a1) conditional knockout mice. During murine embryonic development, Tgfbr3l precedes Nr5a1 expression, though the two transcripts are fully colocalized by embryonic day 18.5 and thereafter. Collectively, these data indicate that SF-1 directly regulates Tgfbr3l/TGFBR3L transcription and is required for postnatal expression of the gene in gonadotropes.
Collapse
Affiliation(s)
- Yeu-Farn Lin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Gauthier Schang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Evan R S Buddle
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Hailey Schultz
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Thea L Willis
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 1UL, UK
| | - Frederique Ruf-Zamojski
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michel Zamojski
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Natalia Mendelev
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Stuart C Sealfon
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 1UL, UK
| | - Daniel J Bernard
- Correspondence: Daniel J. Bernard, PhD, Department of Pharmacology and Therapeutics, 3655 Promenade Sir William Osler, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
43
|
Giannakopoulos A, Sertedaki A, Chrysis D. A human paradigm of LHX4 and NR5A1 developmental gene interaction in the pituitary gland and ovary? Eur J Hum Genet 2022; 30:1191-1194. [PMID: 35277652 PMCID: PMC9553932 DOI: 10.1038/s41431-022-01076-z] [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: 09/12/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
The pituitary gland, as a nodal component of the endocrine system, is responsible for the regulation of growth, reproduction, metabolism, and homeostasis. Although pituitary formation though the hierarchical action of different transcription factors is well studied in mouse models, there is little evidence of the analogous developmental processes in humans. Herein, we present a female patient with a phenotype that includes blepharoptosis-ptosis-epicanthus syndrome and premature ovarian failure. Clinical exome sequencing revealed two heterozygous variants in two genes, LHX4 (pathogenic) and NR5A1 (VUS) genes and no mutation in FOXL2 gene. We propose a model of genetic interaction between LHX4 and NR5A1 during pituitary and ovarian development that may lead to a similar phenotype mediated by reduced FOXL2 expression.
Collapse
Affiliation(s)
- Aristeidis Giannakopoulos
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, Greece.
| | - Amalia Sertedaki
- Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dionisios Chrysis
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, Greece
| |
Collapse
|
44
|
Constantin S, Bjelobaba I, Stojilkovic SS. Pituitary gonadotroph-specific patterns of gene expression and hormone secretion. Curr Opin Pharmacol 2022; 66:102274. [PMID: 35994915 PMCID: PMC9509429 DOI: 10.1016/j.coph.2022.102274] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Pituitary gonadotrophs play a key role in reproductive functions by secreting luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The LH secretory activity of gonadotroph is controlled by hypothalamic gonadotropin-releasing hormone (GnRH) via GnRH receptors and is accompanied by only minor effects on high basal Lhb gene expression. The secretory profiles of GnRH and LH are highly synchronized, with the latter reflecting a depletion of prestored LH in secretory vesicles by regulated exocytosis. In contrast, FSH is predominantly released by constitutive exocytosis, and secretory activity reflects the kinetics of Fshb gene expression controlled by GnRH, activin, and inhibin. Here is a review of recent data to improve the understanding of multiple patterns of gonadotroph gene expression and hormone secretion.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivana Bjelobaba
- Department for Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000, Belgrade, Serbia
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
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: 1.0] [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
|
46
|
Asuzu DT, Alvarez R, Fletcher PA, Mandal D, Johnson K, Wu W, Elkahloun A, Clavijo P, Allen C, Maric D, Ray-Chaudhury A, Rajan S, Abdullaev Z, Nwokoye D, Aldape K, Nieman LK, Stratakis C, Stojilkovic SS, Chittiboina P. Pituitary adenomas evade apoptosis via noxa deregulation in Cushing's disease. Cell Rep 2022; 40:111223. [PMID: 36001971 PMCID: PMC9527711 DOI: 10.1016/j.celrep.2022.111223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/03/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Sporadic pituitary adenomas occur in over 10% of the population. Hormone-secreting adenomas, including those causing Cushing’s disease (CD), cause severe morbidity and early mortality. Mechanistic studies of CD are hindered by a lack of in vitro models and control normal human pituitary glands. Here, we surgically annotate adenomas and adjacent normal glands in 25 of 34 patients. Using single-cell RNA sequencing (RNA-seq) analysis of 27594 cells, we identify CD adenoma transcriptomic signatures compared with adjacent normal cells, with validation by bulk RNA-seq, DNA methylation, qRT-PCR, and immunohistochemistry. CD adenoma cells include a subpopulation of proliferating, terminally differentiated corticotrophs. In CD adenomas, we find recurrent promoter hypomethylation and transcriptional upregulation of PMAIP1 (encoding pro-apoptotic BH3-only bcl-2 protein noxa) but paradoxical noxa downregulation. Using primary CD adenoma cell cultures and a corticotroph-enriched mouse cell line, we find that selective proteasomal inhibition with bortezomib stabilizes noxa and induces apoptosis, indicating its utility as an anti-tumor agent. Asuzu et al. perform single-cell transcriptomic profiling in Cushing’s disease (CD) adenomas and find overexpression and DNA hypomethylation of PMAIP1, which encodes the pro-apoptotic protein noxa. Noxa is degraded by the proteasome. Proteasomal inhibition rescues noxa and induces apoptosis in CD.
Collapse
Affiliation(s)
- David T Asuzu
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD 20892, USA; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Reinier Alvarez
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD 20892, USA; Florida International University Herbert Wertheim College of Medicine, Miami, FL, USA
| | - Patrick A Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Debjani Mandal
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD 20892, USA
| | - Kory Johnson
- DIR Bioinformatics Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Weiwei Wu
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Abdel Elkahloun
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Paul Clavijo
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA
| | - Clint Allen
- Translational Tumor Immunology Program, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Sharika Rajan
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Zied Abdullaev
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Diana Nwokoye
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD 20892, USA
| | - Kenneth Aldape
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Lynnette K Nieman
- Section on Translational Endocrinology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Constantine Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Prashant Chittiboina
- Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD 20892, USA; Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
47
|
Stojilkovic SS, Previde RM, Sherman AS, Fletcher PA. Pituitary corticotroph identity and receptor-mediated signaling: A transcriptomics perspective. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25. [PMID: 36177190 PMCID: PMC9514143 DOI: 10.1016/j.coemr.2022.100364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent single-cell RNA sequencing has offered an unprecedented view of pituitary cell transcriptomic profiles. In this review, these new data are briefly discussed and compared with the classical literature, focusing on pituitary corticotrophs. These cells are introduced by discussing their marker genes, followed by a review of G protein-coupled receptor gene expression, heterotrimeric G protein genes, and genes encoding signaling pathways downstream of G proteins: adenylate cyclases, phosphodiesterases, phospholipases, and protein kinases. The expression patterns of enzyme-linked plasma membrane and nuclear hormone receptor genes was also analyzed. The overview of these selected groups of genes sheds new light on corticotrophic receptors and their signaling pathways and provides guidance for further basic and clinical research by identifying genes that not been studied so far.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence: Stanko S. Stojilkovic ()
| | - Rafael M. Previde
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Directed evolution of adeno-associated virus for efficient gene delivery to microglia. Nat Methods 2022; 19:976-985. [PMID: 35879607 DOI: 10.1038/s41592-022-01547-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/10/2022] [Indexed: 12/12/2022]
Abstract
As the resident immune cells in the central nervous system (CNS), microglia orchestrate immune responses and dynamically sculpt neural circuits in the CNS. Microglial dysfunction and mutations of microglia-specific genes have been implicated in many diseases of the CNS. Developing effective and safe vehicles for transgene delivery into microglia will facilitate the studies of microglia biology and microglia-associated disease mechanisms. Here, we report the discovery of adeno-associated virus (AAV) variants that mediate efficient in vitro and in vivo microglial transduction via directed evolution of the AAV capsid protein. These AAV-cMG and AAV-MG variants are capable of delivering various genetic payloads into microglia with high efficiency, and enable sufficient transgene expression to support fluorescent labeling, Ca2+ and neurotransmitter imaging and genome editing in microglia in vivo. Furthermore, single-cell RNA sequencing shows that the AAV-MG variants mediate in vivo transgene delivery without inducing microglia immune activation. These AAV variants should facilitate the use of various genetically encoded sensors and effectors in the study of microglia-related biology.
Collapse
|
49
|
Chen Z, Jia Q, Zhao Z, Zhang Q, Chen Y, Qiao N, Ye Z, Ji C, Zhang Y, He W, Shi C, Cai Y, Yao B, Han R, Wang Y, Shou X, Shen M, Cao X, Zhou X, Cheng H, Zhu J, Hu Y, Zhang Z, Ye H, Li Y, Li S, Wang Y, Ma Z, Ni T, Zhao Y. Transcription Factor ASCL1 Acts as a Novel Potential Therapeutic Target for the Treatment of the Cushing's Disease. J Clin Endocrinol Metab 2022; 107:2296-2306. [PMID: 35521682 DOI: 10.1210/clinem/dgac280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The pathogenesis of Cushing's disease (CD) is still not adequately understood despite the identification of somatic driver mutations in USP8, BRAF, and USP48. In this multiomics study, we combined RNA sequencing (RNA-seq) with Sanger sequencing to depict transcriptional dysregulation under different gene mutation backgrounds. Furthermore, we evaluated the potential of achaete-scute complex homolog 1 (ASCL1), a pioneer transcription factor, as a novel therapeutic target for treatment of CD and its possible downstream pathway. METHODS RNA-seq was adopted to investigate the gene expression profile of CD, and Sanger sequencing was adopted to detect gene mutations. Bioinformatics analysis was used to depict transcriptional dysregulation under different gene mutation backgrounds. The function of ASCL1 in hormone secretion, cell proliferation, and apoptosis were studied in vitro. The effectiveness of an ASCL1 inhibitor was evaluated in primary CD cells, and the clinical relevance of ASCL1 was examined in 68 patients with CD. RNA-seq in AtT-20 cells on Ascl1 knockdown combined with published chromatin immunoprecipitation sequencing data and dual luciferase assays were used to explore downstream pathways. RESULTS ASCL1 was exclusively overexpressed in USP8-mutant and wild-type tumors. Ascl1 promoted adrenocorticotrophin hormone overproduction and tumorigenesis and directly regulated Pomc in AtT-20 cells. An ASCL1 inhibitor presented promising efficacy in both AtT-20 and primary CD cells. ASCL1 overexpression was associated with a larger tumor volume and higher adrenocorticotrophin secretion in patients with CD. CONCLUSION Our findings help to clarify the pathogenesis of CD and suggest that ASCL1 is a potential therapeutic target the treatment of CD. SUMMARY The pathogenesis of Cushing's disease (CD) is still not adequately understood despite the identification of somatic driver mutations in USP8, BRAF, and USP48. Moreover, few effective medical therapies are currently available for the treatment of CD. Here, using a multiomics approach, we first report the aberrant overexpression of the transcription factor gene ASCL1 in USP8-mutant and wild-type tumors of CD. Ascl1 promoted adrenocorticotrophin hormone overproduction and tumorigenesis and directly regulated Pomc in mouse AtT-20 cells. Notably, an ASCL1 inhibitor presented promising efficacy in both AtT-20 and primary CD cells. Importantly, ASCL1 overexpression was associated with a larger tumor volume and higher adrenocorticotrophin secretion in patients with CD. Thus, our findings improve understanding of CD pathogenesis and suggest that ASCL1 is a potential therapeutic target the treatment of CD.
Collapse
Affiliation(s)
- Zhengyuan Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Qi Jia
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Zhaozhao Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Qilin Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Yu Chen
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Nidan Qiao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Zhao Ye
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Chenxing Ji
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Yichao Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Wenqiang He
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Chengzhang Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Yixin Cai
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Boyuan Yao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Rui Han
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Ye Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Xuefei Shou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Ming Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Xiaoyun Cao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Xiang Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Haixia Cheng
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingjing Zhu
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yao Hu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Shiqi Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Zengyi Ma
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai,, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, 201100, China
| |
Collapse
|
50
|
Duan S, Sawyer TW, Sontz RA, Wieland BA, Diaz AF, Merchant JL. GFAP-directed Inactivation of Men1 Exploits Glial Cell Plasticity in Favor of Neuroendocrine Reprogramming. Cell Mol Gastroenterol Hepatol 2022; 14:1025-1051. [PMID: 35835391 PMCID: PMC9490044 DOI: 10.1016/j.jcmgh.2022.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/16/2022] [Accepted: 06/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.
Collapse
Affiliation(s)
- Suzann Duan
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Travis W. Sawyer
- Wyant College of Optical Sciences, University of Arizona, Tucson, Arizona
| | - Ricky A. Sontz
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Bradley A. Wieland
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Andres F. Diaz
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona
| | - Juanita L. Merchant
- University of Arizona College of Medicine, Department of Medicine, Division of Gastroenterology, Tucson, Arizona,Correspondence Address correspondence to: Dr Juanita L. Merchant, University of Arizona, 1515 N. Campbell Ave, Tucson, AZ 85724; tel: (520) 626-7897; fax: (520) 626-1291.
| |
Collapse
|