1
|
Hashmi H, Matsumoto R, Corcoran D, Kawakami Y, Araki T. Genetic models of Cushing's disease : From cells, in vivo transgenic models to human pituitary organoids. Pituitary 2025; 28:47. [PMID: 40186634 DOI: 10.1007/s11102-025-01516-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Cushing's disease (CD) is caused by pituitary tumors that overproduce adrenocorticotropic hormone (ACTH); however, effective medical treatments remain limited, significantly impairing patients' quality of life and prognosis. Despite extensive molecular analyses, the pathogenesis of CD remains unclear. Although previous molecular studies have relied heavily on rodent-derived cells and rodent transgenic models, significant species differences exist in the tumorigenesis of CD between humans and rodents. To date, an established human CD cell model is lacking, as human CD cells are limited in availability and sustainability over time. Additionally, the gene modifications used in transgenic models do not necessarily reflect the causative genes in CD. CD tumors exhibit wide phenotypic heterogeneity, which further complicates the development of an ideal genetic model. In this review, we provide an analysis of 11 genetic models used to study CD, outlining their historical development, strengths, and limitations. Additionally, we discuss the ongoing development of human induced pluripotent stem cell (iPSC)-derived pituitary organoids and further describe various models of pituitary organoids as an emerging novel approach to studying CD. By comparing all these models, we highlight the necessity of advancing genetic models to improve our understanding and treatment of CD.
Collapse
Affiliation(s)
- Hiba Hashmi
- Division of Endocrinology & Metabolism, Department of Medicine, Western University, London, ON, Canada
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Minnesota, 420 Delaware SE, Minneapolis, MN, USA
| | - Ryusaku Matsumoto
- Center for Ips Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Dylan Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Takako Araki
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Minnesota, 420 Delaware SE, Minneapolis, MN, USA.
| |
Collapse
|
2
|
Cui R, Duan H, Hu W, Li C, Zhong S, Liang L, Chen S, Hu H, He Z, Wang Z, Guo X, Chen Z, Xu C, Zhu Y, Chen Y, Sai K, Yang Q, Guo C, Mou Y, Jiang X. Establishment of Human Pituitary Neuroendocrine Tumor Derived Organoid and Its Pilot Application for Drug Screening. J Clin Endocrinol Metab 2025; 110:e827-e840. [PMID: 38656317 DOI: 10.1210/clinem/dgae228] [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: 07/29/2023] [Indexed: 04/26/2024]
Abstract
CONTEXT Precision medicine for pituitary neuroendocrine tumors (PitNETs) is limited by the lack of reliable research models. OBJECTIVE To generate patient-derived organoids (PDOs), which could serve as a platform for personalized drug screening for PitNET patients. DESIGN From July 2019 to May 2022, a total of 32 human PitNET specimens were collected for the establishment of organoids with an optimized culture protocol. SETTING This study was conducted at Sun Yat-Sen University Cancer Center. PATIENTS PitNET patients who were pathologically confirmed were enrolled in this study. INTERVENTIONS Histological staining and whole-exome sequencing were utilized to confirm the pathologic and genomic features of PDOs. A drug response assay on PDOs was also performed. MAIN OUTCOME MEASURES PDOs retained key genetic and morphological features of their parental tumors. RESULTS PDOs were successfully established from various types of PitNET samples with an overall success rate of 87.5%. Clinical nonfunctioning PitNETs-derived organoids (22/23, 95.7%) showed a higher likelihood of successful generation compared to those from functioning PitNETs (6/9, 66.7%). Preservation of cellular structure, subtype-specific neuroendocrine profiles, mutational features, and tumor microenvironment heterogeneity from parental tumors was observed. A distinctive response profile in drug tests was observed among the organoids from patients with different subtypes of PitNETs. With the validation of key characteristics from parental tumors in histological, genomic, and microenvironment heterogeneity consistency assays, we demonstrated the predictive value of the PDOs in testing individual drugs. CONCLUSION The established PDOs, retaining typical features of parental tumors, indicate a translational significance in innovating personalized treatment for refractory PitNETs.
Collapse
Affiliation(s)
- Run Cui
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
- Department of Neurosurgery, Guangdong 2nd Provincial Peoples Hospital, Guangzhou, 523058 Guangdong, China
| | - Hao Duan
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Wanming Hu
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510000 Guangdong, China
| | - Chang Li
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Sheng Zhong
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Lun Liang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Siyu Chen
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Hongrong Hu
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Zhenqiang He
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Zhenning Wang
- Department of Neurosurgery, Dongguan People's Hospital, Dongguan, 523058 Guangdong, China
| | - Xiaoyu Guo
- Department of Neurosurgery, First Affiliated Hospital of Ji'nan University, Guangzhou, 510630 Guangdong, China
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510320 Guangdong, China
| | - Cong Xu
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510320 Guangdong, China
| | - Yu Zhu
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510320 Guangdong, China
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Ke Sai
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Qunying Yang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| | - Xiaobing Jiang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, S Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong, China
| |
Collapse
|
3
|
Shoji JY, Davis RP, Mummery CL, Krauss S. Global Literature Analysis of Tumor Organoid and Tumor-on-Chip Research. Cancers (Basel) 2025; 17:108. [PMID: 39796734 PMCID: PMC11719888 DOI: 10.3390/cancers17010108] [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: 11/29/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Tumor organoid and tumor-on-chip (ToC) platforms replicate aspects of the anatomical and physiological states of tumors. They, therefore, serve as models for investigating tumor microenvironments, metastasis, and immune interactions, especially for precision drug testing. To map the changing research diversity and focus in this field, we performed a quality-controlled text analysis of categorized academic publications and clinical studies. Methods: Previously, we collected metadata of academic publications on organoids or organ-on-chip platforms from PubMed, Web of Science, Scopus, EMBASE, and bioRxiv, published between January 2011 and June 2023. Here, we selected documents from this metadata corpus that were computationally determined as relevant to tumor research and analyzed them using an in-house text analysis algorithm. Additionally, we collected and analyzed metadata from ClinicalTrials.gov of clinical studies related to tumor organoids or ToC as of March 2023. Results and Discussion: From 3551 academic publications and 139 clinical trials, we identified 55 and 24 tumor classes modeled as tumor organoids and ToC models, respectively. The research was particularly active in neural and hepatic/pancreatic tumor organoids, as well as gastrointestinal, neural, and reproductive ToC models. Comparative analysis with cancer statistics showed that lung, lymphatic, and cervical tumors were under-represented in tumor organoid research. Our findings also illustrate varied research topics, including tumor physiology, therapeutic approaches, immune cell involvement, and analytical techniques. Mapping the research geographically highlighted the focus on colorectal cancer research in the Netherlands, though overall the specific research focus of countries did not reflect regional cancer prevalence. These insights not only map the current research landscape but also indicate potential new directions in tumor model research.
Collapse
Affiliation(s)
- Jun-ya Shoji
- Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| | - Richard P. Davis
- Department of Anatomy & Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christine L. Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, 7522 NB Enschede, The Netherlands
| | - Stefan Krauss
- Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
4
|
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
|
5
|
Laporte E, Vankelecom H. Organoid models of the pituitary gland in health and disease. Front Endocrinol (Lausanne) 2023; 14:1233714. [PMID: 37614709 PMCID: PMC10442803 DOI: 10.3389/fendo.2023.1233714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The pituitary gland represents the hub of our endocrine system. Its cells produce specific hormones that direct multiple vital physiological processes such as body growth, fertility, and stress. The gland also contains a population of stem cells which are still enigmatic in phenotype and function. Appropriate research models are needed to advance our knowledge on pituitary (stem cell) biology. Over the last decade, 3D organoid models have been established, either derived from the pituitary stem cells or from pluripotent stem cells, covering both healthy and diseased conditions. Here, we summarize the state-of-the-art of pituitary-allied organoid models and discuss applications of these powerful in vitro research and translational tools to study pituitary development, biology, and disease.
Collapse
Affiliation(s)
- Emma Laporte
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Mallick S, Chakrabarti J, Eschbacher J, Moraitis AG, Greenstein AE, Churko J, Pond KW, Livolsi A, Thorne CA, Little AS, Yuen KCJ, Zavros Y. Genetically engineered human pituitary corticotroph tumor organoids exhibit divergent responses to glucocorticoid receptor modulators. Transl Res 2023; 256:56-72. [PMID: 36640905 PMCID: PMC11345864 DOI: 10.1016/j.trsl.2023.01.002] [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] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023]
Abstract
Cushing's disease (CD) is a serious endocrine disorder attributed to an adrenocorticotropic hormone (ACTH)-secreting pituitary neuroendocrine tumor (PitNET) that that subsequently leads to chronic hypercortisolemia. PitNET regression has been reported following treatment with the investigational selective glucocorticoid receptor (GR) modulator relacorilant, but the mechanisms behind that effect remain unknown. Human PitNET organoid models were generated from induced human pluripotent stem cells (iPSCs) or fresh tissue obtained from CD patient PitNETs (hPITOs). Genetically engineered iPSC derived organoids were used to model the development of corticotroph PitNETs expressing USP48 (iPSCUSP48) or USP8 (iPSCUSP8) somatic mutations. Organoids were treated with the GR antagonist mifepristone or the GR modulator relacorilant with or without somatostatin receptor (SSTR) agonists pasireotide or octreotide. In iPSCUSP48 and iPSCUSP8 cultures, mifepristone induced a predominant expression of SSTR2 with a concomitant increase in ACTH secretion and tumor cell proliferation. Relacorilant predominantly induced SSTR5 expression and tumor cell apoptosis with minimal ACTH induction. Hedgehog signaling mediated the induction of SSTR2 and SSTR5 in response to mifepristone and relacorilant. Relacorilant sensitized PitNET organoid responsiveness to pasireotide. Therefore, our study identified the potential therapeutic use of relacorilant in combination with somatostatin analogs and demonstrated the advantages of relacorilant over mifepristone, supporting its further development for use in the treatment of Cushing's disease patients.
Collapse
Affiliation(s)
- Saptarshi Mallick
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Jayati Chakrabarti
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Jennifer Eschbacher
- Department of Neuropathology, Barrow Neurological Institute, Phoenix, Arizona
| | | | | | - Jared Churko
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Kelvin W Pond
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | | | - Curtis A Thorne
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Andrew S Little
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona
| | - Kevin C J Yuen
- Department of Neuroendocrinology, Barrow Neurological Institute, Phoenix, Arizona
| | - Yana Zavros
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona.
| |
Collapse
|
7
|
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: 3.5] [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
|
8
|
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: 2.3] [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
|
9
|
Zhang D, Hugo W, Bergsneider M, Wang MB, Kim W, Vinters HV, Heaney AP. Single-cell RNA sequencing in silent corticotroph tumors confirms impaired POMC processing and provides new insights into their invasive behavior. Eur J Endocrinol 2022; 187:49-64. [PMID: 35521707 PMCID: PMC9248914 DOI: 10.1530/eje-21-1183] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/21/2022] [Indexed: 11/08/2022]
Abstract
Objective Provide insights into the defective POMC processing and invasive behavior in silent pituitary corticotroph tumors. Design and methods Single-cell RNAseq was used to compare the cellular makeup and transcriptome of silent and active corticotroph tumors. Results A series of transcripts related to hormone processing peptidases and genes involved in the structural organization of secretory vesicles were reduced in silent compared to active corticotroph tumors. Most relevant to their invasive behavior, silent corticotroph tumors exhibited several features of epithelial-to-mesenchymal transition, with increased expression of mesenchymal genes along with the loss of transcripts that regulate hormonal biogenesis and secretion. Silent corticotroph tumor vascular smooth muscle cell and pericyte stromal cell populations also exhibited plasticity in their mesenchymal features. Conclusions Our findings provide novel insights into the mechanisms of impaired POMC processing and invasion in silent corticotroph tumors and suggest that a common transcriptional reprogramming mechanism simultaneously impairs POMC processing and activates tumor invasion.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Willy Hugo
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Marvin Bergsneider
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Marilene B. Wang
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Won Kim
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Harry V. Vinters
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Anthony P. Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
10
|
Fuentes-Fayos AC, Luque RM. A novel human tumoroid 3D model of sustained ACTH-secreting cell cultures to study critically needed therapies for Cushing's disease. EBioMedicine 2021; 67:103368. [PMID: 33993049 PMCID: PMC8206815 DOI: 10.1016/j.ebiom.2021.103368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.
| |
Collapse
|