1
|
Liu T, Li R, Sun L, Xu Z, Wang S, Zhou J, Wu X, Shi K. Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ. Cell Biosci 2023; 13:175. [PMID: 37740216 PMCID: PMC10517496 DOI: 10.1186/s13578-023-01119-y] [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: 06/05/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-induced liver steatosis in mice, as well as insulin resistance and type 2 diabetic phenotype. The potential link between Menin and hepatic metabolism homeostasis may provide new insights into the mechanism of fatty liver disease. RESULTS Disturbance of hepatic Menin expression impacts metabolic pathways associated with non-alcoholic fatty liver disease (NAFLD), including the FoxO signaling pathway, which is similar to that observed in both oleic acid-induced fatty hepatocytes model and biopsied fatty liver tissues, but with elevated hepatic Menin expression and inhibited FABP1. Higher levels of Menin facilitate glucose uptake while restraining fatty acid uptake. Menin targets the expression of FABP3/4/5 and also CD36 or GK, PCK by binding to their promoter regions, while recruiting and deploying the cellular localization of PPARγ and SIRT1 in the nucleus and cytoplasm. Accordingly, Menin binds to PPARγ and/or FoxO1 in hepatocytes, and orchestrates hepatic glucose and fatty acid uptake by recruiting SIRT1. CONCLUSION Menin plays an orchestration role as a transcriptional activator and/or repressor to target downstream gene expression levels involved in hepatic energy uptake by interacting with the cellular energy sensor SIRT1, PPARγ, and/or FoxO1 and deploying their translocations between the cytoplasm and nucleus, thereby maintaining metabolic homeostasis. These findings provide more evidence suggesting Menin could be targeted for the treatment of hepatic steatosis, NAFLD or metabolic dysfunction-associated fatty liver disease (MAFLD), and even other hepatic diseases.
Collapse
Affiliation(s)
- Tingjun Liu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Ranran Li
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Lili Sun
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Zhongjin Xu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Shengxuan Wang
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Jingxuan Zhou
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Xuanning Wu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Kerong Shi
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China.
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Liu N, Li R, Cao J, Song X, Ma W, Liu T, Wang L, Zou J, Zhang B, Liu Z, Liang R, Zheng R, Wang S. The inhibition of FKBP5 protects β-cell survival under inflammation stress via AKT/FOXO1 signaling. Cell Death Discov 2023; 9:247. [PMID: 37452039 PMCID: PMC10349081 DOI: 10.1038/s41420-023-01506-x] [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: 02/21/2023] [Revised: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The FK506-binding protein 51 (FKBP51, encoded by FKBP5 gene) has emerged as a critical regulator of mammalian endocrine stress responses and as a potential pharmacological target for metabolic disorders, including type 2 diabetes (T2D). However, in β cells, which secrete the only glucose-lowering hormone-insulin, the expression and function of FKBP5 has not been documented. Here, using human pancreatic tissue and primary human islets, we demonstrated the abundant expression of FKBP5 in β cells, which displayed an responsive induction upon acute inflammatory stress mimicked by in vitro treatment with a cocktail of inflammatory cytokines (IL-1β, IFN-γ, and TNF-α). To explore its function, siRNAs targeting FKBP5 and pharmacological inhibitor SAFit2 were applied both in clonal NIT-1 cells and primary human/mice islets. We found that FKBP5 inhibition promoted β-cell survival, improved insulin secretion, and upregulated β-cell functional gene expressions (MAFA and NKX6.1) in acute-inflammation stressed β cells. In primary human and mice islets, which constitutively suffer from inflammation stress during isolation and culture, FKBP5 inhibition also presented decent performance in improving islet function, in accordance with its protective effect against inflammation. Molecular studies found that FKBP5 is an important regulator for FOXO1 phosphorylation at Serine 256, and silencing of FOXO1 abrogated the protective effect of FKBP5 inhibition, suggesting that it is the key downstream effector of FKBP5 in β cells. At last, in situ detection of FKBP5 protein expression on human and mice pancreases revealed a reduction of FKBP5 expression in β cells in human T2D patients, as well as T2D mice model (db/db), which may indicate a FKBP5-inhibition-mediated pro-survival mechanism against the complex stresses in T2D milieus.
Collapse
Affiliation(s)
- Na Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, 300052, Tianjin, People's Republic of China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Rui Li
- Department of Pediatrics, Tianjin Medical University General Hospital, 300052, Tianjin, People's Republic of China
| | - Jinglin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, People's Republic of China
| | - Xinyao Song
- Department of Pediatrics, Tianjin Medical University General Hospital, 300052, Tianjin, People's Republic of China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Wenmiao Ma
- Department of Pediatrics, Tianjin Medical University General Hospital, 300052, Tianjin, People's Republic of China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Tengli Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Le Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Jiaqi Zou
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Boya Zhang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Zewen Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China.
| | - Rongxiu Zheng
- Department of Pediatrics, Tianjin Medical University General Hospital, 300052, Tianjin, People's Republic of China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, 300384, Tianjin, People's Republic of China.
| |
Collapse
|
3
|
Wang S, Liu T, Sun L, Du H, Xu Z, Li R, Yu Y, Mao Y, Shi K. Menin regulates lipid deposition in mouse hepatocytes via interacting with transcription factor FoxO1. Mol Cell Biochem 2022; 477:1555-1568. [PMID: 35182330 DOI: 10.1007/s11010-022-04392-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is rapidly being recognized as the leading cause of chronic liver disease worldwide. Men1, encoding protein of menin, is a key causative gene of multiple endocrine neoplasia type 1 syndrome including pancreatic tumor. It is known that insulin that secretes by endocrine tissue pancreatic islets plays a critical role in hepatic metabolism. Mouse model of hemizygous deletion of Men1 was shown to have severe hepatic metabolism disorders. However, the molecular function of menin on lipid deposition in hepatocytes needs to be further studied. Transcriptome sequencing does show that expression suppression of Men1 in mouse hepatocytes widely affect signaling pathways involved in hepatic metabolism, such as fatty acid metabolism, insulin response, glucose metabolism and inflammation. Further molecular studies indicates that menin overexpression inhibits expressions of the fat synthesis genes Srebp-1c, Fas, and Acc1, the fat differentiation genes Pparγ1 and Pparγ2, and the fat transport gene Cd36, thereby inhibiting the fat accumulation in hepatocytes. The biological process of menin regulating hepatic lipid metabolism was accomplished by interacting with the transcription factor FoxO1, which is also found to be critical for lipid metabolism. Moreover, menin responds to insulin in hepatocytes and mediates its regulatory effect on hepatic metabolism. Our findings suggest that menin is a crucial mediation factor in regulating the hepatic fat deposition, suggesting it could be a potential important therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Shengxuan Wang
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Tingjun Liu
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Lili Sun
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Hongxia Du
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Zhongjin Xu
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Ranran Li
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Ying Yu
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, 100093, China
| | - Yongjiang Mao
- Key Laboratory of Animal Genetics & Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, 225009, China
| | - Kerong Shi
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China.
| |
Collapse
|
4
|
Smedlund KB, Sanchez ER, Hinds TD. FKBP51 and the molecular chaperoning of metabolism. Trends Endocrinol Metab 2021; 32:862-874. [PMID: 34481731 PMCID: PMC8516732 DOI: 10.1016/j.tem.2021.08.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 01/30/2023]
Abstract
The molecular chaperone FK506-binding protein 51 (FKBP51) is gaining attention as a meaningful biomarker of metabolic dysfunction. This review examines the emerging contributions of FKBP51 in adipogenesis and lipid metabolism, myogenesis and protein catabolism, and glucocorticoid-induced skin hypoplasia and dermal adipocytes. The FKBP51 signaling mechanisms that may explain these metabolic consequences are discussed. These mechanisms are diverse, with FKBP51 independently and directly regulating phosphorylation cascades and nuclear receptors. We provide a discussion of the newly developed compounds that antagonize FKBP51, which may offer therapeutic advantages for adiposity. These observations suggest we are only beginning to uncover the complex nature of FKBP51 and its molecular chaperoning of metabolism.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Edwin R Sanchez
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Terry D Hinds
- Barnstable Brown Diabetes Center, Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA.
| |
Collapse
|
5
|
Involvement of the MEN1 Gene in Hormone-Related Cancers: Clues from Molecular Studies, Mouse Models, and Patient Investigations. ENDOCRINES 2020. [DOI: 10.3390/endocrines1020007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MEN1 mutation predisposes patients to multiple endocrine neoplasia type 1 (MEN1), a genetic syndrome associated with the predominant co-occurrence of endocrine tumors. Intriguingly, recent evidence has suggested that MEN1 could also be involved in the development of breast and prostate cancers, two major hormone-related cancers. The first clues as to its possible role arose from the identification of the physical and functional interactions between the menin protein, encoded by MEN1, and estrogen receptor α and androgen receptor. In parallel, our team observed that aged heterozygous Men1 mutant mice developed cancerous lesions in mammary glands of female and in the prostate of male mutant mice at low frequencies, in addition to endocrine tumors. Finally, observations made both in MEN1 patients and in sporadic breast and prostate cancers further confirmed the role played by menin in these two cancers. In this review, we present the currently available data concerning the complex and multifaceted involvement of MEN1 in these two types of hormone-dependent cancers.
Collapse
|
6
|
Characterization of the Menin-MLL Interaction as Therapeutic Cancer Target. Cancers (Basel) 2020; 12:cancers12010201. [PMID: 31947537 PMCID: PMC7016952 DOI: 10.3390/cancers12010201] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
Inhibiting the interaction of menin with the histone methyltransferase MLL1 (KMT2A) has recently emerged as a novel therapeutic strategy. Beneficial therapeutic effects have been postulated in leukemia, prostate, breast, liver and in synovial sarcoma models. In those indications, MLL1 recruitment by menin was described to critically regulate the expression of disease associated genes. However, most findings so far rely on single study reports. Here we independently evaluated the pathogenic functions of the menin-MLL interaction in a large set of different cancer models with a potent and selective probe inhibitor BAY-155. We characterized the inhibition of the menin-MLL interaction for anti-proliferation, gene transcription effects, and for efficacy in several in vivo xenografted tumor models. We found a specific therapeutic activity of BAY-155 primarily in AML/ALL models. In solid tumors, we observed anti-proliferative effects of BAY-155 in a surprisingly limited fraction of cell line models. These findings were further validated in vivo. Overall, our study using a novel, highly selective and potent inhibitor, shows that the menin-MLL interaction is not essential for the survival of most solid cancer models. We can confirm that disrupting the menin-MLL complex has a selective therapeutic benefit in MLL-fused leukemia. In solid cancers, effects are restricted to single models and more limited than previously claimed.
Collapse
|
7
|
Razmara M, Monazzam A, Skogseid B. Reduced menin expression impairs rapamycin effects as evidenced by an increase in mTORC2 signaling and cell migration. Cell Commun Signal 2018; 16:64. [PMID: 30285764 PMCID: PMC6167842 DOI: 10.1186/s12964-018-0278-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022] Open
Abstract
Background Mammalian target of rapamycin (mTOR) is a master regulator of various cellular responses by forming two functional complexes, mTORC1 and mTORC2. mTOR signaling is frequently dysregulated in pancreatic neuroendocrine tumors (PNETs). mTOR inhibitors have been used in attempts to treat these lesions, and prolonged progression free survival has been recorded. If this holds true also for the multiple endocrine neoplasia type 1 (MEN1) associated PNETs is yet unclear. We investigated the relationship between expression of the MEN1 protein menin and mTOR signaling in the presence or absence of the mTOR inhibitor rapamycin. Methods In addition to use of menin wild type and menin-null mouse embryonic fibroblasts (MEFs), menin was silenced by siRNA in pancreatic neuroendocrine tumor cell line BON-1. Panels of protein phosphorylation, as activation markers downstream of PI3k-mTOR-Akt pathways, as well as menin expression were evaluated by immunoblotting. The impact of menin expression in the presence and absence of rapamycin was determinate upon Wound healing, migration and proliferation in MEFs and BON1 cells. Results PDGF-BB markedly increased phosphorylation of mTORC2 substrate Akt, at serine 473 (S473) and threonine 450 (T450) in menin−/− MEFs but did not alter phosphorylation of mTORC1 substrates ribosomal protein S6 or eIF4B. Acute rapamycin treatment by mTORC1-S6 inhibition caused a greater enhancement of Akt phosphorylation on S473 in menin−/− cells as compared to menin+/+ MEFs (116% vs 38%). Chronic rapamycin treatment, which inhibits both mTORC1and 2, reduced Akt phosphorylation of S473 to a lesser extent in menin−/− MEFs than menin+/+ MEFs (25% vs 75%). Silencing of menin expression in human PNET cell line (BON1) also enhanced Akt phosphorylation at S473, but not activation of mTORC1. Interestingly, silencing menin in BON1 cells elevated S473 phosphorylation of Akt in both acute and chronic treatments with rapamycin. Finally, we show that the inhibitory effect of rapamycin on serum mediated wound healing and cell migration is impaired in menin−/− MEFs, as well as in menin-silenced BON1 cells. Conclusions Menin is involved in regulatory mechanism between the two mTOR complexes, and its reduced expression is accompanied with increased mTORC2-Akt signaling, which consequently impairs anti-migratory effect of rapamycin. Electronic supplementary material The online version of this article (10.1186/s12964-018-0278-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masoud Razmara
- Department of medical sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | - Azita Monazzam
- Department of medical sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Britt Skogseid
- Department of medical sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Khatami F, Tavangar SM. Multiple Endocrine Neoplasia Syndromes from Genetic and Epigenetic Perspectives. Biomark Insights 2018; 13:1177271918785129. [PMID: 30013307 PMCID: PMC6043927 DOI: 10.1177/1177271918785129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple endocrine neoplasia (MEN) syndromes are infrequent inherited disorders in which more than one endocrine glands develop noncancerous (benign) or cancerous (malignant) tumors or grow excessively without forming tumors. There are 3 famous and well-known forms of MEN syndromes (MEN 1, MEN 2A, and MEN 2B) and a newly documented one (MEN4). These syndromes are infrequent and occurred in all ages and both men and women. Usually, germ line mutations that can be resulted in neoplastic transformation of anterior pituitary, parathyroid glands, and pancreatic islets in addition to gastrointestinal tract can be an indicator for MEN1. The medullary thyroid cancer (MTC) in association with pheochromocytoma and/or multiple lesions of parathyroid glands with hyperparathyroidism can be pointer of MEN2 which can be subgrouped into the MEN 2A, MEN 2B, and familial MTC syndromes. There are no distinct biochemical markers that allow identification of familial versus nonfamilial forms of the tumors, but familial MTC usually happens at a younger age than sporadic MTC. The MEN1 gene (menin protein) is in charge of MEN 1 disease, CDNK1B for MEN 4, and RET proto-oncogene for MEN 2. The focus over the molecular targets can bring some hope for both diagnosis and management of MEN syndromes. In the current review, we look at this disease and responsible genes and their cell signaling pathway involved.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathology, Doctor Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Shi K, Liu X, Li H, Lin X, Yan Z, Cao Q, Zhao M, Xu Z, Wang Z. Menin Modulates Mammary Epithelial Cell Numbers in Bovine Mammary Glands Through Cyclin D1. J Mammary Gland Biol Neoplasia 2017; 22:221-233. [PMID: 29188494 PMCID: PMC5854757 DOI: 10.1007/s10911-017-9385-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/13/2017] [Indexed: 11/27/2022] Open
Abstract
Menin, the protein encoded by the MEN1 gene, is abundantly expressed in the epithelial cells of mammary glands. Here, we found MEN1/menin expression slowly decreased with advancing lactation but increased by the end of lactation. It happened that the number of bovine mammary epithelial cells decreases since lactation, suggesting a role of menin in the control of mammary epithelial cell growth. Indeed, reduction of menin expression through MEN1-specific siRNA transfection in the bovine mammary epithelial cells caused cell growth arrest in G1/S phase. Decreased mRNA and protein expression of Cyclin D1 was observed upon MEN1 knockdown. Furthermore, menin was confirmed to physically bind to the promoter region of Cyclin D1 through a ChIP assay, indicating that menin plays a regulatory role in mammary epithelial cell cycle progression. Moreover, lower expression of MEN1/menin induced increased epithelial cell apoptosis and caused extracellular matrix remodeling by down-regulating its associated genes, such as DSG2 and KRT5, suggesting that menin's role may also be involved in the control of cell-cell adhesion in normal mammary glands. Taken together, our data revealed an unknown molecular function of menin in epithelial cell proliferation, which may be important in the regulation of lactation behavior of mammary glands.
Collapse
Affiliation(s)
- Kerong Shi
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China.
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China.
| | - Xue Liu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Honghui Li
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Xueyan Lin
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Zhengui Yan
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Qiaoqiao Cao
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Meng Zhao
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Zhongjin Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China
| | - Zhonghua Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China.
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Taian, 271018, China.
| |
Collapse
|
10
|
Enhancer-Mediated Oncogenic Function of the Menin Tumor Suppressor in Breast Cancer. Cell Rep 2017; 18:2359-2372. [PMID: 28273452 DOI: 10.1016/j.celrep.2017.02.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 12/17/2016] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
While the multiple endocrine neoplasia type 1 (MEN1) gene functions as a tumor suppressor in a variety of cancer types, we explored its oncogenic role in breast tumorigenesis. The MEN1 gene product menin is involved in H3K4 trimethylation and co-activates transcription. We integrated ChIP-seq and RNA-seq data to identify menin target genes. Our analysis revealed that menin-dependent target gene promoters display looping to distal enhancers that are bound by menin, FOXA1 and GATA3. In this fashion, MEN1 co-regulates a proliferative breast cancer-specific gene expression program in ER+ cells. In primary mammary cells, MEN1 exerts an anti-proliferative function by regulating a distinct expression signature. Our findings clarify the cell-type-specific functions of MEN1 and inform the development of menin-directed treatments for breast cancer.
Collapse
|
11
|
Dreijerink KMA, Timmers HTM, Brown M. Twenty years of menin: emerging opportunities for restoration of transcriptional regulation in MEN1. Endocr Relat Cancer 2017; 24:T135-T145. [PMID: 28811299 PMCID: PMC5609455 DOI: 10.1530/erc-17-0281] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Since the discovery of the multiple endocrine neoplasia type 1 (MEN1) gene in 1997, elucidation of the molecular function of its protein product, menin, has been a challenge. Biochemical, proteomics, genetics and genomics approaches have identified various potential roles, which converge on gene expression regulation. The most consistent findings show that menin connects transcription factors and chromatin-modifying enzymes, in particular, the histone H3K4 methyltransferase complexes MLL1 and MLL2. Chromatin immunoprecipitation combined with next-generation sequencing has enabled studying genome-wide dynamics of chromatin binding by menin. We propose that menin regulates cell type-specific transcriptional programs by linking chromatin regulatory complexes to specific transcription factors. In this fashion, the MEN1 gene is a tumor suppressor gene in the endocrine tissues that are affected in MEN1. Recent studies have hinted at possibilities to pharmacologically restore the epigenetic changes caused by loss of menin function as therapeutic strategies for MEN1, for example, by inhibition of histone demethylases. The current lack of appropriate cellular model systems for MEN1-associated tumors is a limitation for compound testing, which needs to be addressed in the near future. In this review, we look back at the past twenty years of research on menin and the mechanism of disease of MEN1. In addition, we discuss how the current understanding of the molecular function of menin offers future directions to develop novel treatments for MEN1-associated endocrine tumors.
Collapse
Affiliation(s)
- Koen M A Dreijerink
- Department of EndocrinologyVU University Medical Center, Amsterdam, The Netherlands
| | - H T Marc Timmers
- German Cancer Consortium (DKTK) partner site FreiburgGerman Cancer Research Center (DKFZ) and Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Myles Brown
- Department of Medical OncologyDana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Agarwal SK. The future: genetics advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer 2017; 24:T119-T134. [PMID: 28899949 PMCID: PMC5679100 DOI: 10.1530/erc-17-0199] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/08/2017] [Indexed: 02/01/2023]
Abstract
The identification of the multiple endocrine neoplasia type 1 (MEN1) gene in 1997 has shown that germline heterozygous mutations in the MEN1 gene located on chromosome 11q13 predisposes to the development of tumors in the MEN1 syndrome. Tumor development occurs upon loss of the remaining normal copy of the MEN1 gene in MEN1-target tissues. Therefore, MEN1 is a classic tumor suppressor gene in the context of MEN1. This tumor suppressor role of the protein encoded by the MEN1 gene, menin, holds true in mouse models with germline heterozygous Men1 loss, wherein MEN1-associated tumors develop in adult mice after spontaneous loss of the remaining non-targeted copy of the Men1 gene. The availability of genetic testing for mutations in the MEN1 gene has become an essential part of the diagnosis and management of MEN1. Genetic testing is also helping to exclude mutation-negative cases in MEN1 families from the burden of lifelong clinical screening. In the past 20 years, efforts of various groups world-wide have been directed at mutation analysis, molecular genetic studies, mouse models, gene expression studies, epigenetic regulation analysis, biochemical studies and anti-tumor effects of candidate therapies in mouse models. This review will focus on the findings and advances from these studies to identify MEN1 germline and somatic mutations, the genetics of MEN1-related states, several protein partners of menin, the three-dimensional structure of menin and menin-dependent target genes. The ongoing impact of all these studies on disease prediction, management and outcomes will continue in the years to come.
Collapse
Affiliation(s)
- Sunita K Agarwal
- Metabolic Diseases BranchNational Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Muhammad AB, Xing B, Liu C, Naji A, Ma X, Simmons RA, Hua X. Menin and PRMT5 suppress GLP1 receptor transcript and PKA-mediated phosphorylation of FOXO1 and CREB. Am J Physiol Endocrinol Metab 2017; 313:E148-E166. [PMID: 28270438 PMCID: PMC5582886 DOI: 10.1152/ajpendo.00241.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/11/2017] [Accepted: 02/20/2017] [Indexed: 12/23/2022]
Abstract
Menin is a scaffold protein that interacts with several epigenetic mediators to regulate gene transcription, and suppresses pancreatic β-cell proliferation. Tamoxifen-inducible deletion of multiple endocrine neoplasia type 1 (MEN1) gene, which encodes the protein menin, increases β-cell mass in multiple murine models of diabetes and ameliorates diabetes. Glucagon-like-peptide-1 (GLP1) is another key physiological modulator of β-cell mass and glucose homeostasis. However, it is not clearly understood whether menin crosstalks with GLP1 signaling. Here, we show that menin and protein arginine methyltransferase 5 (PRMT5) suppress GLP1 receptor (GLP1R) transcript levels. Notably, a GLP1R agonist induces phosphorylation of forkhead box protein O1 (FOXO1) at S253, and the phosphorylation is mediated by PKA. Interestingly, menin suppresses GLP1-induced and PKA-mediated phosphorylation of both FOXO1 and cAMP response element binding protein (CREB), likely through a protein arginine methyltransferase. Menin-mediated suppression of FOXO1 and CREB phosphorylation increases FOXO1 levels and suppresses CREB target genes, respectively. A small-molecule menin inhibitor reverses menin-mediated suppression of both FOXO1 and CREB phosphorylation. In addition, ex vivo treatment of both mouse and human pancreatic islets with a menin inhibitor increases levels of proliferation marker Ki67. In conclusion, our results suggest that menin and PRMT5 suppress GLP1R transcript levels and PKA-mediated phosphorylation of FOXO1 and CREB, and a menin inhibitor may reverse this suppression to induce β-cell proliferation.
Collapse
Affiliation(s)
- Abdul Bari Muhammad
- Abramson Family Cancer Research Institute, Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Institute for Diabetes, Obesity, and Metabolism Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bowen Xing
- Shenzen University School of Medicine, Institute of Diabetes Research, Shenzhen, Guangdong, China
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Institute for Diabetes, Obesity, and Metabolism Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaosong Ma
- Shenzen University School of Medicine, Institute of Diabetes Research, Shenzhen, Guangdong, China
| | - Rebecca A Simmons
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Xianxin Hua
- Abramson Family Cancer Research Institute, Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
- Institute for Diabetes, Obesity, and Metabolism Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Li H, Liu X, Wang Z, Lin X, Yan Z, Cao Q, Zhao M, Shi K. MEN1/Menin regulates milk protein synthesis through mTOR signaling in mammary epithelial cells. Sci Rep 2017; 7:5479. [PMID: 28710500 PMCID: PMC5511157 DOI: 10.1038/s41598-017-06054-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/07/2017] [Indexed: 01/17/2023] Open
Abstract
The MEN1 gene, which encodes the protein Menin, was investigated for its regulatory role in milk protein synthesis in mammary glands. Menin responds to nutrient and hormone levels via the PI3K/Akt/mTOR pathway. Bovine mammary epithelial cells and tissues were used as experimental models in this study. The results revealed that the milk protein synthesis capacity of mammary epithelial cells could be regulated by MEN1/Menin. The overexpression of Menin caused significant suppression of factors involved in the mTOR pathway, as well as milk protein κ-casein (CSNK). In contrast, a significant increase in these factors and CSNK was observed upon MEN1/Menin knockdown. The repression of MEN1/Menin on the mTOR pathway was also observed in mammary gland tissues. Additionally, MEN1/Menin was found to elicit a negative response on prolactin (PRL) and/or insulin (INS), which caused a similar downstream impact on mTOR pathway factors and milk proteins. Collectively, our data indicate that MEN1/Menin could play a regulatory role in milk protein synthesis through mTOR signaling in the mammary gland by mediating the effects of hormones and nutrient status. The discovery of Menin's role in mammary glands suggests Menin could be potential new target for the improvement of milk performance and adjustment of lactation period of dairy cows.
Collapse
Affiliation(s)
- Honghui Li
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Xue Liu
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Zhonghua Wang
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Xueyan Lin
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Zhengui Yan
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Qiaoqiao Cao
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Meng Zhao
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China
| | - Kerong Shi
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, 271018, P. R. China.
| |
Collapse
|
15
|
Ehrlich L, Hall C, Meng F, Lairmore T, Alpini G, Glaser S. A Review of the Scaffold Protein Menin and its Role in Hepatobiliary Pathology. Gene Expr 2017; 17:251-263. [PMID: 28485270 PMCID: PMC5765438 DOI: 10.3727/105221617x695744] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a familial cancer syndrome with neuroendocrine tumorigenesis of the parathyroid glands, pituitary gland, and pancreatic islet cells. The MEN1 gene codes for the canonical tumor suppressor protein, menin. Its protein structure has recently been crystallized, and it has been investigated in a multitude of other tissues. In this review, we summarize recent advancements in understanding the structure of the menin protein and its function as a scaffold protein in histone modification and epigenetic gene regulation. Furthermore, we explore its role in hepatobiliary autoimmune diseases, cancers, and metabolic diseases. In particular, we discuss how menin expression and function are regulated by extracellular signaling factors and nuclear receptor activation in various hepatic cell types. How the many signaling pathways and tissue types affect menin's diverse functions is not fully understood. We show that small-molecule inhibitors affecting menin function can shed light on menin's broad role in pathophysiology and elucidate distinct menin-dependent processes. This review reveals menin's often dichotomous function through analysis of its role in multiple disease processes and could potentially lead to novel small-molecule therapies in the treatment of cholangiocarcinoma or biliary autoimmune diseases.
Collapse
Affiliation(s)
- Laurent Ehrlich
- *Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
| | - Chad Hall
- †Department of Surgery, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
| | - Fanyin Meng
- *Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
- ‡Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
| | - Terry Lairmore
- †Department of Surgery, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
| | - Gianfranco Alpini
- *Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
- ‡Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
| | - Shannon Glaser
- *Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, USA
- ‡Research, Central Texas Veterans Health Care System, Temple, TX, USA
- §Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
| |
Collapse
|
16
|
Bonnavion R, Teinturier R, Gherardi S, Leteurtre E, Yu R, Cordier-Bussat M, Du R, Pattou F, Vantyghem MC, Bertolino P, Lu J, Zhang CX. Foxa2, a novel protein partner of the tumour suppressor menin, is deregulated in mouse and human MEN1 glucagonomas. J Pathol 2017; 242:90-101. [PMID: 28188614 DOI: 10.1002/path.4885] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 11/10/2022]
Abstract
Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rémy Bonnavion
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France
| | - Romain Teinturier
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France
| | - Samuele Gherardi
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France
| | - Emmanuelle Leteurtre
- Institut de Pathologie, CHRU de Lille, Lille, France.,Department of Endocrinology and Metabolism, Univ. Lille 2, INSERM UMR 1190, Lille, France
| | - Run Yu
- Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Martine Cordier-Bussat
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France
| | - Rui Du
- The E-Institute of Shanghai, Sino-French Life Science and Genomic Centre, Ruijin Hospital, Shanghai, PR China.,Shanghai Clinical Centre for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao-Tong University, Shanghai, PR China
| | - François Pattou
- Department of Endocrinology and Metabolism, Univ. Lille 2, INSERM UMR 1190, Lille, France.,CHRU Lille, Endocrine Surgery, Lille, France
| | - Marie-Christine Vantyghem
- Department of Endocrinology and Metabolism, Univ. Lille 2, INSERM UMR 1190, Lille, France.,CHRU Lille, Endocrinology, Lille, France
| | - Philippe Bertolino
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France
| | - Jieli Lu
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France.,The E-Institute of Shanghai, Sino-French Life Science and Genomic Centre, Ruijin Hospital, Shanghai, PR China.,Shanghai Clinical Centre for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao-Tong University, Shanghai, PR China
| | - Chang Xian Zhang
- INSERM U1052, Lyon, France.,CNRS UMR5286, Lyon, France.,Université de Lyon, Lyon, France.,The E-Institute of Shanghai, Sino-French Life Science and Genomic Centre, Ruijin Hospital, Shanghai, PR China
| |
Collapse
|
17
|
Abstract
Despite its identification in 1997, the functions of the MEN1 gene-the main gene underlying multiple endocrine neoplasia type 1 syndrome-are not yet fully understood. In addition, unlike the RET-MEN2 causative gene-no hot-spot mutational areas or genotype-phenotype correlations have been identified. More than 1,300 MEN1 gene mutations have been reported and are mostly "private" (family specific). Even when mutations are shared at an intra- or inter-familial level, the spectrum of clinical presentation is highly variable, even in identical twins. Despite these inherent limitations for genetic counseling, identifying MEN1 mutations in individual carriers offers them the opportunity to have lifelong clinical surveillance schemes aimed at revealing MEN1-associated tumors and lesions, dictates the timing and scope of surgical procedures, and facilitates specific mutation analysis of relatives to define presymptomatic carriers.
Collapse
Affiliation(s)
- Alberto Falchetti
- EndOsMet Unit, Villa Donatello, Piazzale Donatello 2, Florence 50100, Italy; Hercolani Clinical Center, Via D'Azeglio 46, Bologna 40136, Italy
| |
Collapse
|
18
|
Gao Z, Zhang L, Xie W, Wang S, Bao X, Guo Y, Zhang H, Hu Q, Chen Y, Wang Z, Xue M, Jin G. Male Men1 heterozygous mice exhibit fasting hyperglycemia in the early stage of MEN1. J Endocrinol 2016; 230:347-55. [PMID: 27432891 DOI: 10.1530/joe-16-0090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/18/2016] [Indexed: 01/29/2023]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant inherited syndrome characterized by multiple tumors in the parathyroid glands, endocrine pancreas and anterior pituitary. Recent clinical studies have revealed a strong association between MEN1 syndrome and the risk of developing diabetes mellitus; however, the underlying mechanisms remain unknown. In this study, heterozygous Men1 knockout (Men1(+/-)) mice were used as MEN1 models to investigate MEN1-associated glucose metabolic phenotypes and mechanisms. Heterozygous deficiency of Men1 in 12-month-old male mice induced fasting hyperglycemia, along with increased serum insulin levels. However, male Men1(+/-) mice did not show insulin resistance, as evidenced by Akt activation in hepatic tissues and an insulin tolerance test. Increased glucose levels following pyruvate challenge and expression of key gluconeogenic genes suggested increased hepatic glucose output in the male Men1(+/-) mice. This effect could be partly due to higher basal serum glucagon levels, which resulted from pancreatic islet cell proliferation induced by heterozygous loss of Men1 Taken together, our results indicate that fasted male Men1(+/-) mice, in the early stage of development of MEN1, display glucose metabolic disorders. These disorders are caused not by direct induction of insulin resistance, but via increased glucagon secretion and the consequent stimulation of hepatic glucose production.
Collapse
Affiliation(s)
- Zhongxiuzi Gao
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Li Zhang
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Wenting Xie
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Siqi Wang
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Xiaorui Bao
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Yuli Guo
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Houjian Zhang
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Qingzhong Hu
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Yi Chen
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Zeen Wang
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Maoqiang Xue
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| | - Guanghui Jin
- Department of Basic Medical SciencesMedical College, Xiamen University, Xiamen, China
| |
Collapse
|
19
|
Schernthaner-Reiter MH, Trivellin G, Stratakis CA. MEN1, MEN4, and Carney Complex: Pathology and Molecular Genetics. Neuroendocrinology 2016; 103:18-31. [PMID: 25592387 PMCID: PMC4497946 DOI: 10.1159/000371819] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/31/2014] [Indexed: 12/17/2022]
Abstract
Pituitary adenomas are a common feature of a subset of endocrine neoplasia syndromes, which have otherwise highly variable disease manifestations. We provide here a review of the clinical features and human molecular genetics of multiple endocrine neoplasia (MEN) type 1 and 4 (MEN1 and MEN4, respectively) and Carney complex (CNC). MEN1, MEN4, and CNC are hereditary autosomal dominant syndromes that can present with pituitary adenomas. MEN1 is caused by inactivating mutations in the MEN1 gene, whose product menin is involved in multiple intracellular pathways contributing to transcriptional control and cell proliferation. MEN1 clinical features include primary hyperparathyroidism, pancreatic neuroendocrine tumours and prolactinomas as well as other pituitary adenomas. A subset of patients with pituitary adenomas and other MEN1 features have mutations in the CDKN1B gene; their disease has been called MEN4. Inactivating mutations in the type 1α regulatory subunit of protein kinase A (PKA; the PRKAR1A gene), that lead to dysregulation and activation of the PKA pathway, are the main genetic cause of CNC, which is clinically characterised by primary pigmented nodular adrenocortical disease, spotty skin pigmentation (lentigines), cardiac and other myxomas and acromegaly due to somatotropinomas or somatotrope hyperplasia.
Collapse
Affiliation(s)
- Marie Helene Schernthaner-Reiter
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Md., USA
| | | | | |
Collapse
|
20
|
Menin immunoreactivity in secretory granules of human pancreatic islet cells. Appl Immunohistochem Mol Morphol 2015; 22:748-55. [PMID: 25153502 DOI: 10.1097/pai.0000000000000046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The protein product of the Multiple Endocrine Neoplasia Type I (MEN1) gene is thought to be involved in predominantly nuclear functions; however, immunohistochemical (IHC) analysis data on cellular localization are conflicting. To further investigate menin expression, we analyzed human pancreas (an MEN1 target organ) using IHC analyses and 6 antibodies raised against full-length menin or its peptides. In 10 normal pancreas specimens, 2 independently raised antibodies showed unexpected cytoplasmic immunoreactivity in peripheral cells in each islet examined (over 100 total across all 10 patients). The staining exhibited a distinct punctate pattern and subsequent immunoelectron microscopy indicated the target antigen was in secretory granules. Exocrine pancreas and pancreatic stroma were not immunoreactive. In MEN1 patients, unaffected islets stained similar to those in normal samples but with a more peripheral location of positive cells, whereas hyperplastic islets and tumorlets showed increased and diffuse cytoplasmic staining, respectively. Endocrine tumors from MEN1 patients were negative for menin, consistent with a 2-hit loss of a tumor suppressor gene. Secretory granule localization of menin in a subset of islet cells suggests a function of the protein unique to a target organ of familial endocrine neoplasia, although the IHC data must be interpreted with some caution because of the possibility of antibody cross-reaction. The identity, cellular trafficking, and role of this putative secretory granule-form of menin warrant additional investigation.
Collapse
|
21
|
Cheng P, Li G, Yang SS, Liu R, Jin G, Zhou XY, Hu XG. Tumor suppressor Menin acts as a corepressor of LXRα to inhibit hepatic lipogenesis. FEBS Lett 2015; 589:3079-84. [PMID: 25962847 DOI: 10.1016/j.febslet.2015.04.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 04/03/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023]
Abstract
Menin, encoded by the MEN1 gene, was initially identified as a tumor suppressor for endocrine neoplasia. Our previous report showed that Menin enhances PPARα transactivity preventing triglyceride accumulation in the liver. Here, we further explore the role of Menin in liver steatosis. Transient transfection assays demonstrate that Menin inhibits the transcriptional activity of nuclear receptor liver X receptor α (LXRα). Accordingly, Menin overexpression results in reduced expression of LXRα target genes, such as lipogenic enzymes including SREBP-1c, FASN and SCD-1. Co-immunoprecipitation assays revealed physical interaction between Menin and LXRα. Collectively, our data suggest that Menin acts as a novel corepressor of LXRα and functions as a negative regulator of hepatic lipogenesis.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Gang Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Sheng Sheng Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Rui Liu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Xu Yu Zhou
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Xian Gui Hu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| |
Collapse
|
22
|
Flynn N, Getz A, Visser F, Janes TA, Syed NI. Menin: a tumor suppressor that mediates postsynaptic receptor expression and synaptogenesis between central neurons of Lymnaea stagnalis. PLoS One 2014; 9:e111103. [PMID: 25347295 PMCID: PMC4210270 DOI: 10.1371/journal.pone.0111103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/27/2014] [Indexed: 11/19/2022] Open
Abstract
Neurotrophic factors (NTFs) support neuronal survival, differentiation, and even synaptic plasticity both during development and throughout the life of an organism. However, their precise roles in central synapse formation remain unknown. Previously, we demonstrated that excitatory synapse formation in Lymnaea stagnalis requires a source of extrinsic NTFs and receptor tyrosine kinase (RTK) activation. Here we show that NTFs such as Lymnaea epidermal growth factor (L-EGF) act through RTKs to trigger a specific subset of intracellular signalling events in the postsynaptic neuron, which lead to the activation of the tumor suppressor menin, encoded by Lymnaea MEN1 (L-MEN1) and the expression of excitatory nicotinic acetylcholine receptors (nAChRs). We provide direct evidence that the activation of the MAPK/ERK cascade is required for the expression of nAChRs, and subsequent synapse formation between pairs of neurons in vitro. Furthermore, we show that L-menin activation is sufficient for the expression of postsynaptic excitatory nAChRs and subsequent synapse formation in media devoid of NTFs. By extending our findings in situ, we reveal the necessity of EGFRs in mediating synapse formation between a single transplanted neuron and its intact presynaptic partner. Moreover, deficits in excitatory synapse formation following EGFR knock-down can be rescued by injecting synthetic L-MEN1 mRNA in the intact central nervous system. Taken together, this study provides the first direct evidence that NTFs functioning via RTKs activate the MEN1 gene, which appears sufficient to regulate synapse formation between central neurons. Our study also offers a novel developmental role for menin beyond tumour suppression in adult humans.
Collapse
Affiliation(s)
- Nichole Flynn
- Department of Cell Biology and Anatomy, and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Canada
| | - Angela Getz
- Department of Cell Biology and Anatomy, and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Canada
| | - Frank Visser
- Department of Cell Biology and Anatomy, and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Canada
| | - Tara A. Janes
- Department of Cell Biology and Anatomy, and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Canada
| | - Naweed I. Syed
- Department of Cell Biology and Anatomy, and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
23
|
Gurung B, Muhammad AB, Hua X. Menin is required for optimal processing of the microRNA let-7a. J Biol Chem 2014; 289:9902-8. [PMID: 24563463 DOI: 10.1074/jbc.m113.520692] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple endocrine neoplasia type I (MEN1) is an inherited syndrome that includes susceptibility to pancreatic islet hyperplasia. This syndrome results from mutations in the MEN1 gene, which encodes menin protein. Menin interacts with several transcription factors, including JunD, and inhibits their activities. However, the precise mechanism by which menin suppresses gene expression is not well understood. Here, we show that menin interacts with arsenite-resistant protein 2 (ARS2), a component of the nuclear RNA CAP-binding complex that is crucial for biogenesis of certain miRNAs including let-7a. The levels of primary-let-7a (pri-let-7a) are not affected by menin; however, the levels of mature let-7a are substantially decreased upon Men1 excision. Let-7a targets, including Insr and Irs2, pro-proliferative genes that are crucial for insulin-mediated signaling, are up-regulated in Men1-excised cells. Inhibition of let-7a using anti-miRNA in wild type cells is sufficient to enhance the expression of insulin receptor substrate 2 (IRS2) to levels observed in Men1-excised cells. Depletion of menin does not affect the expression of Drosha and CBP80, but substantially impairs the processing of pri-miRNA to pre-miRNA. Ars2 knockdown decreased let-7a processing in menin-expressing cells but had little impact on let-7a levels in menin-excised cells. As IRS2 is known to mediate insulin signaling and insulin/mitogen-induced cell proliferation, these findings collectively unravel a novel mechanism whereby menin suppresses cell proliferation, at least partly by promoting the processing of certain miRNAs, including let-7a, leading to suppression of Irs2 expression and insulin signaling.
Collapse
Affiliation(s)
- Buddha Gurung
- From the Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | | |
Collapse
|
24
|
Hepatic menin recruits SIRT1 to control liver steatosis through histone deacetylation. J Hepatol 2013; 59:1299-306. [PMID: 23867312 DOI: 10.1016/j.jhep.2013.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 07/02/2013] [Accepted: 07/05/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS The development and progression of non-alcoholic fatty liver disease are associated with aging, obesity, and type 2 diabetes. Understanding the precise regulatory networks of this process will contribute to novel therapeutic strategies. METHODS Hepatocyte-specific Men1 knockout mice were generated using Cre/loxP technology. Lipid and glucose metabolic phenotypes and mechanisms were investigated in aging and high-fat diet fed mice. RESULTS The expression of menin, encoded by multiple endocrine neoplasia 1 (Men1) gene, is reduced in the liver of aging mice. Hepatocyte-specific deletion of Men1 induces liver steatosis in aging mice. Menin deficiency promotes high-fat diet-induced liver steatosis in mice. Menin recruits SIRT1 to control hepatic CD36 expression and triglyceride accumulation through histone deacetylation. CONCLUSIONS Our work reveals that the adaptor protein menin is critical for the progression of hepatic steatosis during aging and metabolic imbalance.
Collapse
|
25
|
Menin: a scaffold protein that controls gene expression and cell signaling. Trends Biochem Sci 2013; 38:394-402. [PMID: 23850066 DOI: 10.1016/j.tibs.2013.05.005] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/22/2013] [Accepted: 05/31/2013] [Indexed: 12/22/2022]
Abstract
The protein menin is encoded by the MEN1 gene, which is mutated in patients with multiple endocrine neoplasia type 1 (MEN1) syndrome. Although menin acts as a tumor suppressor in endocrine organs, it is required for leukemic transformation in mouse models. Menin possesses these dichotomous functions probably because it can both positively and negatively regulate gene expression, as well as interact with a multitude of proteins with diverse functions. Here, we review the recent progress in understanding the molecular mechanisms by which menin functions. The crystal structures of menin with different binding partners reveal that menin is a key scaffold protein that functionally crosstalks with various partners to regulate gene transcription and interplay with multiple signaling pathways.
Collapse
|
26
|
Thevenon J, Bourredjem A, Faivre L, Cardot-Bauters C, Calender A, Murat A, Giraud S, Niccoli P, Odou MF, Borson-Chazot F, Barlier A, Lombard-Bohas C, Clauser E, Tabarin A, Parfait B, Chabre O, Castermans E, Beckers A, Ruszniewski P, Le Bras M, Delemer B, Bouchard P, Guilhem I, Rohmer V, Goichot B, Caron P, Baudin E, Chanson P, Groussin L, Du Boullay H, Weryha G, Lecomte P, Penfornis A, Bihan H, Archambeaud F, Kerlan V, Duron F, Kuhn JM, Vergès B, Rodier M, Renard M, Sadoul JL, Binquet C, Goudet P. Higher risk of death among MEN1 patients with mutations in the JunD interacting domain: a Groupe d’étude des Tumeurs Endocrines (GTE) cohort study. Hum Mol Genet 2013; 22:1940-8. [DOI: 10.1093/hmg/ddt039] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
27
|
Abstract
Background and Aims: Ingestion of food stimulates the secretion of incretin peptides glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 to ensure the proper absorption and storage of nutrients. Menin is the 67 kDa protein product of the MEN1 gene recently reported to have a role in metabolism. In this study, we will determine the regulation of menin in the proximal duodenum by food intake and diet in correlation with GIP levels in the proximal duodenum of mice after an 18 h fast followed by 4 and 7 h refeeding and 3 months of high-fat diet. Methods: A dual luciferase assay was used to determine GIP promoter activity and ELISA was used to measure the levels of GIP after inhibition of menin through small interfering RNA (siRNA) and exposure to MAPK and AKT inhibitors. Colocalization of menin and GIP were determined by immunofluorescence. Results: Menin and GIP expression are regulated by fasting, refeeding and diet in the proximal duodenum. Overexpression of menin in STC-1 cells significantly inhibited GIP mRNA and promoter activity, whereas menin siRNA upregulated GIP levels. Inhibition of GIP expression by the PI3/AKT inhibitor, LY294002, was abrogated in STC-1 cells with reduced menin levels, whereas the MAPK inhibitor, UO126, inhibited the expression of GIP independent of menin. Exposure of STC-1 cells to GIP reduced menin expression in a dose-dependent manner via PI3K-AKT signaling. Conclusion: Feeding and diet regulates the expression of menin, which inversely correlates with GIP levels in the proximal duodenum. In vitro assays indicate that menin is a negative regulator of GIP via inhibition of PI3K-AKT signaling. We show menin colocalizing with GIP in K cells of the proximal gut and hypothesize that downregulation of menin may serve as a mechanism by which GIP is regulated in response to food intake and diet.
Collapse
|
28
|
Menin liver-specific hemizygous mice challenged with high fat diet show increased weight gain and markers of metabolic impairment. Nutr Diabetes 2012; 2:e34. [PMID: 23168387 PMCID: PMC3366066 DOI: 10.1038/nutd.2012.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE: The menin tumor suppressor protein is abundantly expressed in the liver, although no function has been identified because of lack of tumor development in multiple endocrine neoplasia type 1 (Men1) null livers. We examine the phenotype of mice lacking one functional allele of Men1 (consistent with the phenotype in humans with MEN1 syndrome) challenged with high fat diet (HFD) to elucidate a metabolic function for hepatic menin. METHODS: In this study, we challenged mice harboring a liver-specific hemizygous deletion of Men1 (HETs) alongside wild-type (WT) counterparts with HFD for 3 months and monitored the severity of metabolic changes. We demonstrate that the HET mice challenged with HFD for 3 months show an increased weight gain with decreased glucose tolerance compared with WT counterparts. Along with these changes, there was a more severe serum hormone profile involving increased serum insulin, glucose and glucagon, all hallmarks of the type 2 diabetic phenotype. In concert with increased serum hormones, we found that these mice have significantly increased liver triglycerides coupled with increased liver steatosis and inflammatory markers. Quantitative real-time PCR and western blotting studies show increases in enzymes involved with lipogenesis and hepatic glucose production. CONCLUSION: We conclude that hepatic menin is required for regulation of diet-induced metabolism, and our studies indicate a protective role for the Men1 gene in the liver when challenged with HFD.
Collapse
|
29
|
Lips CJ, Dreijerink KM, Höppener JW. Variable clinical expression in patients with a germline MEN1 disease gene mutation: clues to a genotype-phenotype correlation. Clinics (Sao Paulo) 2012; 67 Suppl 1:49-56. [PMID: 22584706 PMCID: PMC3328827 DOI: 10.6061/clinics/2012(sup01)10] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple endocrine neoplasia type 1 is an inherited endocrine tumor syndrome, predominantly characterized by tumors of the parathyroid glands, gastroenteropancreatic tumors, pituitary adenomas, adrenal adenomas, and neuroendocrine tumors of the thymus, lungs or stomach. Multiple endocrine neoplasia type 1 is caused by germline mutations of the multiple endocrine neoplasia type 1 tumor suppressor gene. The initial germline mutation, loss of the wild-type allele, and modifying genetic and possibly epigenetic and environmental events eventually result in multiple endocrine neoplasia type 1 tumors. Our understanding of the function of the multiple endocrine neoplasia type 1 gene product, menin, has increased significantly over the years. However, to date, no clear genotype-phenotype correlation has been established. In this review we discuss reports on exceptional clinical presentations of multiple endocrine neoplasia type 1, which may provide more insight into the pathogenesis of this disorder and offer clues for a possible genotype-phenotype correlation.
Collapse
Affiliation(s)
- Cornelis J Lips
- Department of Internal Medicine & Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|