1
|
Zeng Z, Zhang Q, Liang T, Xiong Y, Liu Z, Zhang J, Yang P, Yang J, Lu Q, Shen D, Tian H, Zhou Z, Fang W, Zhang M, Liu Q, Gao B, Wei Y, Zhou D. Hsp70 incompletely disaggregates misfolded K488X-menin to promote tumourigenesis in a family with multiple endocrine neoplasia type 1. Cell Signal 2025; 130:111681. [PMID: 39978610 DOI: 10.1016/j.cellsig.2025.111681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is caused by germline mutations in the MEN1 gene, including nonsense mutations and missense variants, which result in the formation of truncated inactive menin protein and some of which cause degradation of mutant menin proteins. Here, we describe a c.1462 A > T (p.K488X) mutation in exon 10 of MEN1 as a potential pathogenic mutation in an extended Chinese family with MEN1. We observed that K488X-menin was degraded by ubiquitination modification resulting from the combined actions of carboxy-terminus of Hsc70-interacting protein (CHIP) and Heat Shock Protein Family 70 (Hsp70) in vitro. K488X-menin is a misfolded truncated protein that results in amyloid aggregation in live cells and affected tissues, which is promoted by Hsp70 and/or CHIP. Although Hsp70 can inhibit the aggregation of K488X-menin in vitro, it is not upregulated in the affected tissues in patients with MEN1, and thus cannot completely disaggregate the aggregated K488X-menin. Further, we found that K488X-menin triggers early tumourigenesis in a MEN1 mutant zebrafish model. Moreover, K488X-menin disaggregation was induced by Hsp70 activator and Hsp70 was upregulated in homozygous mutant zebrafish. Our findings provide a novel biophysical mechanism involving Hsp70 underlying MEN1 tumourigenesis in a Chinese family with MEN1.
Collapse
Affiliation(s)
- Zhen Zeng
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China; Shanghai Children's Medical Center GuiZhou Hospital, Shanghai Jiao Tong University School of Medicine, PR China
| | - Qianqian Zhang
- Gastroenterology Department, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Tingting Liang
- Endocrine Metabolism Department, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yu Xiong
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Zhi Liu
- Department of Dermatovenereology, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Jing Zhang
- Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Pingping Yang
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Jingye Yang
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Qingxiang Lu
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Di Shen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, PR China
| | - Hongxia Tian
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Zhongxue Zhou
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China
| | - Wen Fang
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Miao Zhang
- Endocrine Metabolism Department, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Qi Liu
- Gastroenterology Department, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Bo Gao
- Department of Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Yonghui Wei
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, PR China
| | - Ding'an Zhou
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China; Department of Clinical Biochemistry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, PR China; Key Laboratory of Medical Molecular Biology, Guizhou province; Key Laboratory of Endemic and Ethnic Disease, Ministry of Education; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| |
Collapse
|
2
|
Hinterndorfer M, Spiteri VA, Ciulli A, Winter GE. Targeted protein degradation for cancer therapy. Nat Rev Cancer 2025:10.1038/s41568-025-00817-8. [PMID: 40281114 DOI: 10.1038/s41568-025-00817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Targeted protein degradation (TPD) aims at reprogramming the target specificity of the ubiquitin-proteasome system, the major cellular protein disposal machinery, to induce selective ubiquitination and degradation of therapeutically relevant proteins. Since its conception over 20 years ago, TPD has gained a lot of attention mainly due to improvements in the design of bifunctional proteolysis targeting chimeras (PROTACs) and understanding the mechanisms underlying molecular glue degraders. Today, PROTACs are on the verge of a first clinical approval and recent structural and mechanistic insights combined with technological leaps promise to unlock the rational design of protein degraders, following the lead of lenalidomide and related clinically approved analogues. At the same time, the TPD universe is expanding at a record speed with the discovery of novel modalities beyond molecular glue degraders and PROTACs. Here we review the recent progress in the field, focusing on newly discovered degrader modalities, the current state of clinical degrader candidates for cancer therapy and upcoming design approaches.
Collapse
Affiliation(s)
- Matthias Hinterndorfer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Valentina A Spiteri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
3
|
Brunetti A, Cosso R, Vescini F, Falchetti A. Molecular Pathophysiology of Parathyroid Tumorigenesis-The Lesson from a Rare Disease: The "MEN1 Model". Int J Mol Sci 2024; 25:11586. [PMID: 39519139 PMCID: PMC11545851 DOI: 10.3390/ijms252111586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Primary hyperparathyroidism represents the third most prevalent endocrine disease in the general population, consisting of an excessive secretion of parathyroid hormone from one or, more frequently, more of the parathyroid glands, leading to a dysregulation of calcium homeostasis. Schematically, its development occurs primarily by pathophysiological events with genetic mutation, at the germline and/or somatic level, that favor the neoplastic transformation of parathyroid cells and promote their aberrant proliferation, and mutations determining the shift in the PTH "set-point", thus interfering with the normal pathways of PTH secretion and leading to a "resetting" of Ca2+-dependent PTH secretion or to a secretion of PTH insensitive to changes in extracellular Ca2+ levels. Familial syndromic and non-syndromic forms of primary hyperparathyroidism are responsible for approximately 2-5% of primary hyperparathyroidism cases and most of them are inherited forms. The history of the genetic/molecular studies of parathyroid tumorigenesis associated with multiple endocrine neoplasia type 1 syndrome (MEN1) represents an interesting model to understand genetic-epigenetic-molecular aspects underlying the pathophysiology of primary hyperparathyroidism, both in relation to syndromic and non-syndromic forms. This minireview aims to take a quick and simplified look at the MEN1-associated parathyroid tumorigenesis, focusing on the molecular underlying mechanisms. Clinical, epidemiological, and observational studies, as well as specific guidelines, molecular genetics studies, and reviews, have been considered. Only studies submitted to PubMed in the English language were included, without time constraints.
Collapse
Affiliation(s)
- Alessandro Brunetti
- SC Endocrinologia, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy; (A.B.); (F.V.)
| | | | - Fabio Vescini
- SC Endocrinologia, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy; (A.B.); (F.V.)
| | - Alberto Falchetti
- SC Endocrinologia, ASST Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore, 3, 20162 Milano, Italy
| |
Collapse
|
4
|
Boudreault J, Canaff L, Ghozlan M, Wang N, Guarnieri V, Salcuni AS, Scillitani A, Goltzman D, Ali S, Lebrun JJ. Multiple Endocrine Neoplasia Type 1 Regulates TGFβ-Mediated Suppression of Tumor Formation and Metastasis in Melanoma. Cells 2024; 13:973. [PMID: 38891107 PMCID: PMC11172218 DOI: 10.3390/cells13110973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Over the past few decades, the worldwide incidence of cutaneous melanoma, a malignant neoplasm arising from melanocytes, has been increasing markedly, leading to the highest rate of skin cancer-related deaths. While localized tumors are easily removed by excision surgery, late-stage metastatic melanomas are refractory to treatment and exhibit a poor prognosis. Consequently, unraveling the molecular mechanisms underlying melanoma tumorigenesis and metastasis is crucial for developing novel targeted therapies. We found that the multiple endocrine neoplasia type 1 (MEN1) gene product Menin is required for the transforming growth factor beta (TGFβ) signaling pathway to induce cell growth arrest and apoptosis in vitro and prevent tumorigenesis in vivo in preclinical xenograft models of melanoma. We further identified point mutations in two MEN1 family members affected by melanoma that led to proteasomal degradation of the MEN1 gene product and to a loss of TGFβ signaling. Interestingly, blocking the proteasome degradation pathway using an FDA-approved drug and RNAi targeting could efficiently restore MEN1 expression and TGFβ transcriptional responses. Together, these results provide new potential therapeutic strategies and patient stratification for the treatment of cutaneous melanoma.
Collapse
Affiliation(s)
- Julien Boudreault
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Lucie Canaff
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Mostafa Ghozlan
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Ni Wang
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Vito Guarnieri
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Antonio Stefano Salcuni
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia, 33100 Udine, Italy;
| | - Alfredo Scillitani
- Endocrinology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - David Goltzman
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Suhad Ali
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Jean-Jacques Lebrun
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| |
Collapse
|
5
|
Papadopoulou-Marketou N, Tsoli M, Chatzellis E, Alexandraki KI, Kaltsas G. Hereditary Syndromes Associated with Pancreatic and Lung Neuroendocrine Tumors. Cancers (Basel) 2024; 16:2075. [PMID: 38893191 PMCID: PMC11171219 DOI: 10.3390/cancers16112075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) and lung NETs (LNETs) represent a rare but clinically significant subgroup of neoplasms. While the majority is sporadic, approximately 17% of PanNETs and a subset of LNETs develop in the context of monogenic familial tumor syndromes, especially multiple endocrine neoplasia type 1 (MEN1) syndrome. Other inherited syndromes associated with PanNETs include MEN4, von Hippel-Lindau (VHL) syndrome, neurofibromatosis type 1 (NF1), and tuberous sclerosis complex (TSC). These syndromes are highly penetrant and their clinical manifestations may vary even among members of the same family. They are attributed to genetic mutations involving key molecular pathways regulating cell growth, differentiation, and angiogenesis. Pancreatic NETs in hereditary syndromes are often multiple, develop at a younger age compared to sporadic tumors, and are associated with endocrine and nonendocrine tumors derived from multiple organs. Lung NETs are not as common as PanNETs and are mostly encountered in MEN1 syndrome and include typical and atypical lung carcinoids. Early detection of PanNETs and LNETs related to inherited syndromes is crucial, and specific follow-up protocols need to be employed to optimize diagnosis and management. Genetic screening is recommended in childhood, and diagnostic screening starts often in adolescence, even in asymptomatic mutation carriers. Optimal management and therapeutic decisions should be made in the context of a multidisciplinary team in specialized centers, whereas specific biomarkers aiming to identify patients denoted to follow a more aggressive course need to be developed.
Collapse
Affiliation(s)
- Nektaria Papadopoulou-Marketou
- Neuroendocrine Tumor Unit, EURACAN 4 and ENETS Centre of Excellence, 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.T.); (G.K.)
| | - Marina Tsoli
- Neuroendocrine Tumor Unit, EURACAN 4 and ENETS Centre of Excellence, 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.T.); (G.K.)
| | | | | | - Gregory Kaltsas
- Neuroendocrine Tumor Unit, EURACAN 4 and ENETS Centre of Excellence, 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.T.); (G.K.)
| |
Collapse
|
6
|
Duan S, Sheriff S, Elvis-Offiah UB, Witten BL, Sawyer TW, Sundaresan S, Cierpicki T, Grembecka J, Merchant JL. Clinically Defined Mutations in MEN1 Alter Its Tumor-suppressive Function Through Increased Menin Turnover. CANCER RESEARCH COMMUNICATIONS 2023; 3:1318-1334. [PMID: 37492626 PMCID: PMC10364643 DOI: 10.1158/2767-9764.crc-22-0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/02/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023]
Abstract
Loss of the tumor suppressor protein menin is a critical event underlying the formation of neuroendocrine tumors (NET) in hormone-expressing tissues including gastrinomas. While aberrant expression of menin impairs its tumor suppression, few studies explore the structure-function relationship of clinical multiple endocrine neoplasia, type 1 (MEN1) mutations in the absence of a complete LOH at both loci. Here, we determined whether clinical MEN1 mutations render nuclear menin unstable and lead to its functional inactivation. We studied the structural and functional implications of two clinical MEN1 mutations (R516fs, E235K) and a third variant (A541T) recently identified in 10 patients with gastroenteropancreatic (GEP)-NETs. We evaluated the subcellular localization and half-lives of the mutants and variant in Men1-null mouse embryo fibroblast cells and in hormone-expressing human gastric adenocarcinoma and NET cell lines. Loss of menin function was assessed by cell proliferation and gastrin gene expression assays. Finally, we evaluated the effect of the small-molecule compound MI-503 on stabilizing nuclear menin expression and function in vitro and in a previously reported mouse model of gastric NET development. Both the R516fs and E235K mutants exhibited severe defects in total and subcellular expression of menin, and this was consistent with reduced half-lives of these mutants. Mutated menin proteins exhibited loss of function in suppressing tumor cell proliferation and gastrin expression. Treatment with MI-503 rescued nuclear menin expression and attenuated hypergastrinemia and gastric hyperplasia in NET-bearing mice. Clinically defined MEN1 mutations and a germline variant confer pathogenicity by destabilizing nuclear menin expression. Significance We examined the function of somatic and germline mutations and a variant of MEN1 sequenced from gastroenteropancreatic NETs. We report that these mutations and variant promote tumor cell growth and gastrin expression by rendering menin protein unstable and prone to increased degradation. We demonstrate that the menin-MLL (mixed lineage leukemia) inhibitor MI-503 restores menin protein expression and function in vitro and in vivo, suggesting a potential novel therapeutic approach to target MEN1 GEP-NETs.
Collapse
Affiliation(s)
- Suzann Duan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Sulaiman Sheriff
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Uloma B. Elvis-Offiah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Brandon L. Witten
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Travis W. Sawyer
- Department of Optical Sciences, University of Arizona Wyant College of Optical Sciences, Tucson, Arizona
| | - Sinju Sundaresan
- Department of Physiology, Midwestern University, Downers Grove, Illinois
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Juanita L. Merchant
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
7
|
Dreijerink KMA, Ozyerli-Goknar E, Koidl S, van der Lelij EJ, van den Heuvel P, Kooijman JJ, Biniossek ML, Rodenburg KW, Nizamuddin S, Timmers HTM. Multi-omics analyses of MEN1 missense mutations identify disruption of menin-MLL and menin-JunD interactions as critical requirements for molecular pathogenicity. Epigenetics Chromatin 2022; 15:29. [PMID: 35941657 PMCID: PMC9361535 DOI: 10.1186/s13072-022-00461-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background Loss-of-function mutations of the multiple endocrine neoplasia type 1 (MEN1) gene are causal to the MEN1 tumor syndrome, but they are also commonly found in sporadic pancreatic neuroendocrine tumors and other types of cancers. The MEN1 gene product, menin, is involved in transcriptional and chromatin regulation, most prominently as an integral component of KMT2A/MLL1 and KMT2B/MLL2 containing COMPASS-like histone H3K4 methyltransferase complexes. In a mutually exclusive fashion, menin also interacts with the JunD subunit of the AP-1 and ATF/CREB transcription factors. Results Here, we applied and in silico screening approach for 253 disease-related MEN1 missense mutations in order to select a set of nine menin mutations in surface-exposed residues. The protein interactomes of these mutants were assessed by quantitative mass spectrometry, which indicated that seven of the nine mutants disrupt interactions with both MLL1/MLL2 and JunD complexes. Interestingly, we identified three missense mutations, R52G, E255K and E359K, which predominantly reduce the MLL1 and MLL2 interactions when compared with JunD. This observation was supported by a pronounced loss of binding of the R52G, E255K and E359K mutant proteins at unique MLL1 genomic binding sites with less effect on unique JunD sites. Conclusions Our results underline the effects of MEN1 gene mutations in both familial and sporadic tumors of endocrine origin on the interactions of menin with the MLL1 and MLL2 histone H3K4 methyltransferase complexes and with JunD-containing transcription factors. Menin binding pocket mutants R52G, E255K and E359K have differential effects on MLL1/MLL2 and JunD interactions, which translate into differential genomic binding patterns. Our findings encourage future studies addressing the pathophysiological relevance of the separate MLL1/MLL2- and JunD-dependent functions of menin mutants in MEN1 disease model systems.
Supplementary Information The online version contains supplementary material available at 10.1186/s13072-022-00461-8.
Collapse
Affiliation(s)
| | - Ezgi Ozyerli-Goknar
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, Medical Center -University of Freiburg, Freiburg, Germany
| | - Stefanie Koidl
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, Medical Center -University of Freiburg, Freiburg, Germany
| | | | - Priscilla van den Heuvel
- School of Life Sciences, and Research Group of Technologies of Analyses in Life Sciences (ATLS), Avans University of Applied Sciences, Breda, The Netherlands
| | - Jeffrey J Kooijman
- School of Life Sciences, and Research Group of Technologies of Analyses in Life Sciences (ATLS), Avans University of Applied Sciences, Breda, The Netherlands.,Oncolines B.V., Oss, The Netherlands
| | - Martin L Biniossek
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Kees W Rodenburg
- School of Life Sciences, and Research Group of Technologies of Analyses in Life Sciences (ATLS), Avans University of Applied Sciences, Breda, The Netherlands
| | - Sheikh Nizamuddin
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, Medical Center -University of Freiburg, Freiburg, Germany
| | - H T Marc Timmers
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Urology, Medical Center -University of Freiburg, Freiburg, Germany.
| |
Collapse
|
8
|
Complete androgen insensitivity syndrome caused by a novel mutation in the androgen receptor gene and its mechanism. Clin Chim Acta 2022; 531:94-99. [DOI: 10.1016/j.cca.2022.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/08/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
|
9
|
Høie MH, Cagiada M, Beck Frederiksen AH, Stein A, Lindorff-Larsen K. Predicting and interpreting large-scale mutagenesis data using analyses of protein stability and conservation. Cell Rep 2022; 38:110207. [PMID: 35021073 DOI: 10.1016/j.celrep.2021.110207] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/01/2021] [Accepted: 12/13/2021] [Indexed: 01/23/2023] Open
Abstract
Understanding and predicting the functional consequences of single amino acid changes is central in many areas of protein science. Here, we collect and analyze experimental measurements of effects of >150,000 variants in 29 proteins. We use biophysical calculations to predict changes in stability for each variant and assess them in light of sequence conservation. We find that the sequence analyses give more accurate prediction of variant effects than predictions of stability and that about half of the variants that show loss of function do so due to stability effects. We construct a machine learning model to predict variant effects from protein structure and sequence alignments and show how the two sources of information support one another and enable mechanistic interpretations. Together, our results show how one can leverage large-scale experimental assessments of variant effects to gain deeper and general insights into the mechanisms that cause loss of function.
Collapse
Affiliation(s)
- Magnus Haraldson Høie
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Matteo Cagiada
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Anders Haagen Beck Frederiksen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Amelie Stein
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
10
|
Duan S, Rico K, Merchant JL. Gastrin: From Physiology to Gastrointestinal Malignancies. FUNCTION (OXFORD, ENGLAND) 2021; 3:zqab062. [PMID: 35330921 PMCID: PMC8788842 DOI: 10.1093/function/zqab062] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023]
Abstract
Abetted by widespread usage of acid-suppressing proton pump inhibitors (PPIs), the mitogenic actions of the peptide hormone gastrin are being revisited as a recurring theme in various gastrointestinal (GI) malignancies. While pathological gastrin levels are intricately linked to hyperplasia of enterochromaffin-like cells leading to carcinoid development, the signaling effects exerted by gastrin on distinct cell types of the gastric mucosa are more nuanced. Indeed, mounting evidence suggests dichotomous roles for gastrin in both promoting and suppressing tumorigenesis. Here, we review the major upstream mediators of gastrin gene regulation, including inflammation secondary to Helicobacter pylori infection and the use of PPIs. We further explore the molecular biology of gastrin in GI malignancies, with particular emphasis on the regulation of gastrin in neuroendocrine neoplasms. Finally, we highlight tissue-specific transcriptional targets as an avenue for targetable therapeutics.
Collapse
Affiliation(s)
- Suzann Duan
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Karen Rico
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | | |
Collapse
|
11
|
Klementieva N, Goliusova D, Krupinova J, Yanvarev V, Panova A, Mokrysheva N, Kiselev SL. A Novel Isogenic Human Cell-Based System for MEN1 Syndrome Generated by CRISPR/Cas9 Genome Editing. Int J Mol Sci 2021; 22:12054. [PMID: 34769484 PMCID: PMC8584395 DOI: 10.3390/ijms222112054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/27/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a rare tumor syndrome that manifests differently among various patients. Despite the mutations in the MEN1 gene that commonly predispose tumor development, there are no obvious phenotype-genotype correlations. The existing animal and in vitro models do not allow for studies of the molecular genetics of the disease in a human-specific context. We aimed to create a new human cell-based model, which would consider the variability in genetic or environmental factors that cause the complexity of MEN1 syndrome. Here, we generated patient-specific induced pluripotent stem cell lines carrying the mutation c.1252G>T, D418Y in the MEN1 gene. To reduce the genetically determined variability of the existing cellular models, we created an isogenic cell system by modifying the target allele through CRISPR/Cas9 editing with great specificity and efficiency. The high potential of these cell lines to differentiate into the endodermal lineage in defined conditions ensures the next steps in the development of more specialized cells that are commonly affected in MEN1 patients, such as parathyroid or pancreatic islet cells. We anticipate that this isogenic system will be broadly useful to comprehensively study MEN1 gene function across different contexts, including in vitro modeling of MEN1 syndrome.
Collapse
Affiliation(s)
- Natalia Klementieva
- Endocrinology Research Centre, 115478 Moscow, Russia; (J.K.); (A.P.); (N.M.)
| | - Daria Goliusova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (V.Y.)
| | - Julia Krupinova
- Endocrinology Research Centre, 115478 Moscow, Russia; (J.K.); (A.P.); (N.M.)
| | - Vladislav Yanvarev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (V.Y.)
| | - Alexandra Panova
- Endocrinology Research Centre, 115478 Moscow, Russia; (J.K.); (A.P.); (N.M.)
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (V.Y.)
| | - Natalia Mokrysheva
- Endocrinology Research Centre, 115478 Moscow, Russia; (J.K.); (A.P.); (N.M.)
| | - Sergey L. Kiselev
- Endocrinology Research Centre, 115478 Moscow, Russia; (J.K.); (A.P.); (N.M.)
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (D.G.); (V.Y.)
| |
Collapse
|
12
|
Therapeutic implications of menin inhibition in acute leukemias. Leukemia 2021; 35:2482-2495. [PMID: 34131281 DOI: 10.1038/s41375-021-01309-y] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 01/31/2023]
Abstract
Menin inhibitors are novel targeted agents currently in clinical development for the treatment of genetically defined subsets of acute leukemia. Menin has a tumor suppressor function in endocrine glands. Germline mutations in the gene encoding menin cause the multiple endocrine neoplasia type 1 (MEN1) syndrome, a hereditary condition associated with tumors of the endocrine glands. However, menin is also critical for leukemogenesis in subsets driven by rearrangement of the Lysine Methyltransferase 2A (KMT2A) gene, previously known as mixed-lineage leukemia (MLL), which encodes an epigenetic modifier. These seemingly opposing functions of menin can be explained by its various roles in gene regulation. Therefore, leukemias with rearrangement of KMT2A are predicted to respond to menin inhibition with early clinical data validating this proof-of-concept. These leukemias affect infants, children and adults, and lead to adverse outcomes with current standard therapies. Recent studies have identified novel targets in acute leukemia that are susceptible to menin inhibition, such as mutated Nucleophosmin 1 (NPM1), the most common genetic alteration in adult acute myeloid leukemia (AML). In addition to these alterations, other leukemia subsets with similar transcriptional dependency could be targeted through menin inhibition. This led to rationally designed clinical studies, investigating small-molecule oral menin inhibitors in relapsed acute leukemias with promising early results. Herein, we discuss the physiologic and malignant biology of menin, the mechanisms of leukemia in these susceptible subsets, and future therapeutic strategies using these inhibitors in acute leukemia.
Collapse
|
13
|
Cagiada M, Johansson KE, Valanciute A, Nielsen SV, Hartmann-Petersen R, Yang JJ, Fowler DM, Stein A, Lindorff-Larsen K. Understanding the Origins of Loss of Protein Function by Analyzing the Effects of Thousands of Variants on Activity and Abundance. Mol Biol Evol 2021; 38:3235-3246. [PMID: 33779753 PMCID: PMC8321532 DOI: 10.1093/molbev/msab095] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Understanding and predicting how amino acid substitutions affect proteins are keys to our basic understanding of protein function and evolution. Amino acid changes may affect protein function in a number of ways including direct perturbations of activity or indirect effects on protein folding and stability. We have analyzed 6,749 experimentally determined variant effects from multiplexed assays on abundance and activity in two proteins (NUDT15 and PTEN) to quantify these effects and find that a third of the variants cause loss of function, and about half of loss-of-function variants also have low cellular abundance. We analyze the structural and mechanistic origins of loss of function and use the experimental data to find residues important for enzymatic activity. We performed computational analyses of protein stability and evolutionary conservation and show how we may predict positions where variants cause loss of activity or abundance. In this way, our results link thermodynamic stability and evolutionary conservation to experimental studies of different properties of protein fitness landscapes.
Collapse
Affiliation(s)
- Matteo Cagiada
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer E Johansson
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Audrone Valanciute
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sofie V Nielsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Hartmann-Petersen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Amelie Stein
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Arakelyan J, Zohrabyan D, Philip PA. Molecular profile of pancreatic neuroendocrine neoplasms (PanNENs): Opportunities for personalized therapies. Cancer 2020; 127:345-353. [PMID: 33270905 DOI: 10.1002/cncr.33354] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are the second most common epithelial tumors of the pancreas. Despite improvements in prognostic grading and staging systems, it remains a challenge to predict the clinical behavior of panNENs and the response to specific therapies given the high degree of heterogeneity of these tumors. Most panNENs are nonfunctional and present as advanced disease. However, systemic therapies provide modest benefits. Therefore, there is a need for predictive biomarkers to develop personalized treatment and to advance new drug development. The somatostatin receptors remain the only clinically established prognostic and predictive biomarkers in panNENs. Oncogenic drivers are at a very low frequency. Commonly mutated genes in panNENs include MEN1, chromatin remodeling genes (DAXX and ATRX), and mammalian target of rapamycin pathway genes. In contrast, poorly differentiated neuroendocrine carcinomas (panNECs), which carry a very poor prognosis, have distinctive mutations in certain genes (eg, RB1 and p53). Ongoing research to integrate epigenomics will provide tremendous opportunities to improve current understanding of the clinical heterogeneity of pancreatic neuroendocrine tumors and provide invaluable insight into the biology of these tumors, new drug development, and establishing personalized therapies.
Collapse
Affiliation(s)
- Jemma Arakelyan
- Department of Oncology, Yerevan State Medical University, Yerevan, Armenia.,Adult Solid Tumor Chemotherapy Clinic, Professor Yeolan Hematology Center, Yerevan, Armenia
| | - Davit Zohrabyan
- Department of Oncology, Yerevan State Medical University, Yerevan, Armenia.,Adult Solid Tumor Chemotherapy Clinic, Professor Yeolan Hematology Center, Yerevan, Armenia
| | - Philip A Philip
- Department of Oncology, Yerevan State Medical University, Yerevan, Armenia.,Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan.,Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan.,Barbara Ann Karmanos Cancer Center, Detroit, Michigan
| |
Collapse
|
15
|
Nelakurti DD, Pappula AL, Rajasekaran S, Miles WO, Petreaca RC. Comprehensive Analysis of MEN1 Mutations and Their Role in Cancer. Cancers (Basel) 2020; 12:cancers12092616. [PMID: 32937789 PMCID: PMC7565326 DOI: 10.3390/cancers12092616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancers are characterized by accumulation of genetic mutations in key cell cycle regulators that alter or disable the function of these genes. Such mutations can be inherited or arise spontaneously during the life of the individual. The MEN1 gene prevents uncontrolled cell division and it is considered a tumor suppressor. Inherited MEN1 mutations are associated with certain parathyroid and pancreatic syndromes while spontaneous mutations have been detected in cancer cells. We investigated whether inherited mutations appear in cancer cells which would suggest that patients with parathyroid and pancreatic syndromes have a predisposition to develop cancer. We find a weak correlation between the spectrum of inherited mutations and those appearing spontaneously. Thus, inherited MEN1 mutations may not be a good predictor of tumorigenesis. Abstract MENIN is a scaffold protein encoded by the MEN1 gene that functions in multiple biological processes, including cell proliferation, migration, gene expression, and DNA damage repair. MEN1 is a tumor suppressor gene, and mutations that disrupts MEN1 function are common to many tumor types. Mutations within MEN1 may also be inherited (germline). Many of these inherited mutations are associated with a number of pathogenic syndromes of the parathyroid and pancreas, and some also predispose patients to hyperplasia. In this study, we cataloged the reported germline mutations from the ClinVar database and compared them with the somatic mutations detected in cancers from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. We then used statistical software to determine the probability of mutations being pathogenic or driver. Our data show that many confirmed germline mutations do not appear in tumor samples. Thus, most mutations that disable MEN1 function in tumors are somatic in nature. Furthermore, of the germline mutations that do appear in tumors, only a fraction has the potential to be pathogenic or driver mutations.
Collapse
Affiliation(s)
- Devi D. Nelakurti
- Biomedical Science Undergraduate Program, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Amrit L. Pappula
- Computer Science and Engineering Undergraduate Program, The Ohio State University, Columbus, OH 43210, USA;
| | - Swetha Rajasekaran
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA;
| | - Wayne O. Miles
- Department of Cancer Biology and Genetics, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Ruben C. Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, OH 43302, USA
- Correspondence:
| |
Collapse
|
16
|
Stasiak M, Dedecjus M, Zawadzka-Starczewska K, Adamska E, Tomaszewska M, Lewiński A. Novel Germline c.105_107dupGCT MEN1 Mutation in a Family with Newly Diagnosed Multiple Endocrine Neoplasia Type 1. Genes (Basel) 2020; 11:genes11090986. [PMID: 32847108 PMCID: PMC7565931 DOI: 10.3390/genes11090986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/22/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
In multiple endocrine neoplasia type 1 (MEN1), the causative MEN1 gene mutations lead to the reduced expression of menin, which is a tumor suppressor protein. In this study, we present a case of a 16-year-old woman with severe primary hyperparathyroidism and a non-functioning pituitary microadenoma. Genetic testing demonstrated a novel germline heterozygote variant c.105_107dupGCT of MEN1, leading to Leu duplication in position 37 of the menin polypeptide chain. As such a mutation was not reported before as a causative one, confirmation of its pathogenicity required showing the same mutation in a symptomatic first-degree relative. An identical mutation was found in the patient’s father, who was further diagnosed with hyperparathyroidism and a pituitary microadenoma. We observed the presence of the same MEN1-related tumors but an entirely different symptom severity. To the best of our knowledge, this is the first report of MEN1 syndrome caused by the c.105_107dupGCT MEN1 mutation. This case report demonstrates the importance of genetic evaluation towards MEN1. Genetic testing for MEN1 mutations should be performed in all patients with MEN1-related tumors, and in the young patients even with only one such tumor, despite the supposedly negative family history.
Collapse
Affiliation(s)
- Magdalena Stasiak
- Department of Endocrinology and Metabolic Diseases, Polish Mother‘s Memorial Hospital-Research Institute, 93-338 Lodz, Poland; (M.S.); (K.Z.-S.); (E.A.)
| | - Marek Dedecjus
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology (MSCNRIO), 02-781 Warsaw, Poland;
| | - Katarzyna Zawadzka-Starczewska
- Department of Endocrinology and Metabolic Diseases, Polish Mother‘s Memorial Hospital-Research Institute, 93-338 Lodz, Poland; (M.S.); (K.Z.-S.); (E.A.)
| | - Emilia Adamska
- Department of Endocrinology and Metabolic Diseases, Polish Mother‘s Memorial Hospital-Research Institute, 93-338 Lodz, Poland; (M.S.); (K.Z.-S.); (E.A.)
| | - Monika Tomaszewska
- Department of Pediatrics, Oncology, Hematology and Diabetology, Central Teaching Hospital of the Medical University of Lodz, 91-738 Lodz, Poland;
| | - Andrzej Lewiński
- Department of Endocrinology and Metabolic Diseases, Polish Mother‘s Memorial Hospital-Research Institute, 93-338 Lodz, Poland; (M.S.); (K.Z.-S.); (E.A.)
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
- Correspondence: or ; Tel.: +48-42-271-11-42
| |
Collapse
|
17
|
Xin A, Qu R, Chen G, Zhang L, Chen J, Tao C, Fu J, Tang J, Ru Y, Chen Y, Peng X, Shi H, Zhang F, Sun X. Disruption in ACTL7A causes acrosomal ultrastructural defects in human and mouse sperm as a novel male factor inducing early embryonic arrest. SCIENCE ADVANCES 2020; 6:eaaz4796. [PMID: 32923619 PMCID: PMC7455188 DOI: 10.1126/sciadv.aaz4796] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 07/15/2020] [Indexed: 06/02/2023]
Abstract
Early embryonic arrest is a challenge for in vitro fertilization (IVF). No genetic factors were previously revealed in the sperm-derived arrest of embryonic development. Here, we reported two infertile brothers presenting normal in conventional semen analysis, but both couples had no embryos for transfer after several IVF and intracytoplasmic sperm injection (ICSI). Whole-exome sequencing identified a homozygous missense mutation of ACTL7A in both brothers. This mutation is deleterious and causes sperm acrosomal ultrastructural defects. The Actl7a knock-in mouse model was generated, and male mutated mice showed sperm acrosomal defects, which were completely consistent with the observations in patients. Furthermore, the sperm from ACTL7A/Actl7a-mutated men and mice showed reduced expression and abnormal localization of PLCζ as a potential cause of embryonic arrest and failure of fertilization. Artificial oocyte activation could successfully overcome the Actl7a-mutated sperm-derived infertility, which is meaningful in the future practice of IVF/ICSI for the ACTL7A-associated male infertility.
Collapse
Affiliation(s)
- Aijie Xin
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Pharmacy, Fudan University, Shanghai, China
| | - Ronggui Qu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Guowu Chen
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Ling Zhang
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Junling Chen
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Chengqiu Tao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jianan Tang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Pharmacy, Fudan University, Shanghai, China
| | - Yanfei Ru
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Pharmacy, Fudan University, Shanghai, China
| | - Ying Chen
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiandong Peng
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Huijuan Shi
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Pharmacy, Fudan University, Shanghai, China
| | - Feng Zhang
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Pharmacy, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- Department of Endocrinology and Reproductive Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Wild-type menin is rapidly degraded via the ubiquitin-proteasome pathway in a rat insulinoma cell line. Biosci Rep 2019; 39:220725. [PMID: 31652443 PMCID: PMC6822493 DOI: 10.1042/bsr20190471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 09/18/2019] [Accepted: 10/02/2019] [Indexed: 11/17/2022] Open
Abstract
Menin is encoded by multiple endocrine neoplasia type 1 (MEN1) gene, the germ line mutations of which are the main cause of pancreatic neuroendocrine tumors (PNETs). To date, a large number of frameshift, nonsense and missense mutations of MEN1 have been identified to be responsible for part of MEN1-defficient PNETs patients due to truncation or rapid degradation of menin protein. However, the stability of the wild-type (WT) menin in PNETs is totally unknown. In the present study, we observed ubiquitination of WT menin in 293T cells by transfection of ectopic WT menin and HA-ubiquitin. As expected, either endogenous or ectopic WT menin is stable in 293T cells, whereas in INS-1 cells, a rat insulinoma cell line derived from PNETs, either endogenous or ectopic WT menin is rapidly degraded through ubiquitin-proteasome pathway. Furthermore, the degradation of WT menin is more rapid in the presence of serum. Our findings suggest that in part of PNETs patients with WT MEN1, a ubiquitin-proteasome system targeting menin is untimely activated.
Collapse
|
19
|
Wu Y, Doepner M, Hojnacki T, Feng Z, Katona BW, He X, Ma J, Cao Y, Busino L, Zhou F, Hua X. Disruption of the menin-MLL interaction triggers menin protein degradation via ubiquitin-proteasome pathway. Am J Cancer Res 2019; 9:1682-1694. [PMID: 31497350 PMCID: PMC6726985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023] Open
Abstract
Menin, a protein encoded by the MEN1 gene, suppresses cancers associated with multiple endocrine neoplasia type 1 (MEN1), but promotes the development of a subset of leukemia induced by mixed lineage leukemia (MLL)-derived fusion proteins (MLL-FPs). The crystal structure of menin indicates that it acts as a scaffold protein to bind the N-terminus of MLL via a central pocket. Small molecule menin-MLL inhibitors (MIs) bind the menin pocket to disrupt the menin/MLL interaction, resulting in suppression of MLL-FP-transformed acute myeoloid leukemia (AML). It is thought that MIs suppress the MLL-FP-induced leukemia by blocking the menin/MLL interaction and menin/MLL-induced HOX gene transcription. However, it is not clear whether MIs also affect other aspects of menin biology beyond disruption of the menin/MLL interaction. Here we show for the first time that MIs reduced menin protein levels and decreased the half-life of menin protein but have no effect on mRNA level in MLL-FP-expressing leukemia cells, and proteasome or E1 ligase inhibitor rescued the MI-induced menin degradation. Notably, the MI-induced reduction of H3K4m3 and HOXA9 expression was rescued with a proteasome inhibitor that blocks MI-induced menin protein degradation. Mechanistically, MIs promote the interaction of menin with Hsp70-associated ubiquitin ligase CHIP, resulting in increased menin ubiquitination, leading to increased menin degradation. Together, these findings uncover a novel mechanism whereby small molecule MIs increase menin degradation by triggering the Hsp70/CHIP-mediated ubiquitin-proteasome pathway that ultimately leads to the reduction in HOXA9 gene expression and leukemia suppression.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan UniversityWuhan 430071, China
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430071, China
| | - Miriam Doepner
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Taylor Hojnacki
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Bryson W Katona
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Yan Cao
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Luca Busino
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan UniversityWuhan 430071, China
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania421 Curie Blvd., Philadelphia 19014, PA, USA
| |
Collapse
|
20
|
Candida Barisson Villares Fragoso M, Pontes Cavalcante I, Meneses Ferreira A, Marinho de Paula Mariani B, Ferini Pacicco Lotfi C. Genetics of primary macronodular adrenal hyperplasia. Presse Med 2018; 47:e139-e149. [PMID: 30075949 DOI: 10.1016/j.lpm.2018.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advances in molecular genetics investigations of primary macronodular adrenal hyperplasia (PMAH) have been providing new insights for the research on this issue. The cAMP-dependent pathway is physiologically triggered by ACTH and its receptor, MC2-R, in adrenocortical cells. Different mechanisms of this cascade may be altered in some functioning adrenal cortical disorders. Activating somatic mutations of the GNAS gene (known as gsp oncogene) which encodes the stimulatory G protein alpha-subunit (Gsα) have been found in a small number of adrenocortical secreting adenomas and rarely in PMAH. Lately, ARMC5 was linked to the cyclic AMP signaling pathway, which could be implicated in all of mechanisms of cortisol-secreting by macronodules adrenal hyperplasia and the molecular defects in: G protein aberrant receptors; MC2R; GNAS; PRKAR1A; PDE11A; PDE8B. Around 50 % of patient's relatives with PMAH and 30 % of apparently sporadic hypercortisolism carried ARMC5 mutations. Therefore, PMAH is genetically determined more frequently than previously believed. This review summarizes the most important molecular mechanisms involved in PMAH.
Collapse
Affiliation(s)
| | - Isadora Pontes Cavalcante
- University of Sao Paulo, Adrenal Unit, Service of Endocrinology and Metabolism, 03178-200 Sao Paulo, Brazil; University of Sao Paulo, Institute of Biomedical Sciences, Department of Anatomy, 03178-200 Sao Paulo, Brazil
| | - Amanda Meneses Ferreira
- University of Sao Paulo, Adrenal Unit, Service of Endocrinology and Metabolism, 03178-200 Sao Paulo, Brazil
| | | | | |
Collapse
|
21
|
Dreijerink KMA. Re: Genotype-phenotype pancreatic neuroendocrine tumor relationship in multiple endocrine neoplasia type 1 patients: a 23-year experience at a single institution. Surgery 2018; 163:1325-1329. [PMID: 29338880 DOI: 10.1016/j.surg.2017.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 12/05/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Koen M A Dreijerink
- Department of Endocrinology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Neuroendocrine tumors (NETs) were initially identified as a separate entity in the early 1900s as a unique malignancy that secretes bioactive amines. GI-NETs are the most frequent type and represent a unique subset of NETs, because at least 75% of these tumors represent gastrin stimulation of the enterochromaffin-like cell located in the body of the stomach. The purpose of this review is to understand the specific role of gastrin in the generation of Gastric NETs (G-NETs). RECENT FINDINGS We review here the origin of enterochromaffin cells gut and the role of hypergastrinemia in gastric enteroendocrine tumorigenesis. We describe generation of the first genetically engineered mouse model of gastrin-driven G-NETs that mimics the human phenotype. The common mechanism observed in both the hypergastrinemic mouse model and human carcinoids is translocation of the cyclin-dependent inhibitor p27kip to the cytoplasm and its subsequent degradation by the proteasome. Therapies that block degradation of p27kip, the CCKBR2 gastrin receptor, or gastrin peptide are likely to facilitate treatment.
Collapse
Affiliation(s)
- Sinju Sundaresan
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, BSRB, 2051, 109 Zina Pitcher PL, Ann Arbor, MI, 48109-2200, USA
| | - Anthony J Kang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, BSRB, 2051, 109 Zina Pitcher PL, Ann Arbor, MI, 48109-2200, USA
| | - Juanita L Merchant
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, BSRB, 2051, 109 Zina Pitcher PL, Ann Arbor, MI, 48109-2200, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Sundaresan S, Meininger CA, Kang AJ, Photenhauer AL, Hayes MM, Sahoo N, Grembecka J, Cierpicki T, Ding L, Giordano TJ, Else T, Madrigal DJ, Low MJ, Campbell F, Baker AM, Xu H, Wright NA, Merchant JL. Gastrin Induces Nuclear Export and Proteasome Degradation of Menin in Enteric Glial Cells. Gastroenterology 2017; 153:1555-1567.e15. [PMID: 28859856 PMCID: PMC5705278 DOI: 10.1053/j.gastro.2017.08.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/31/2017] [Accepted: 08/13/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS The multiple endocrine neoplasia, type 1 (MEN1) locus encodes the nuclear protein and tumor suppressor menin. MEN1 mutations frequently cause neuroendocrine tumors such as gastrinomas, characterized by their predominant duodenal location and local metastasis at time of diagnosis. Diffuse gastrin cell hyperplasia precedes the appearance of MEN1 gastrinomas, which develop within submucosal Brunner's glands. We investigated how menin regulates expression of the gastrin gene and induces generation of submucosal gastrin-expressing cell hyperplasia. METHODS Primary enteric glial cultures were generated from the VillinCre:Men1FL/FL:Sst-/- mice or C57BL/6 mice (controls), with or without inhibition of gastric acid by omeprazole. Primary enteric glial cells from C57BL/6 mice were incubated with gastrin and separated into nuclear and cytoplasmic fractions. Cells were incubated with forskolin and H89 to activate or inhibit protein kinase A (a family of enzymes whose activity depends on cellular levels of cyclic AMP). Gastrin was measured in blood, tissue, and cell cultures using an ELISA. Immunoprecipitation with menin or ubiquitin was used to demonstrate post-translational modification of menin. Primary glial cells were incubated with leptomycin b and MG132 to block nuclear export and proteasome activity, respectively. We obtained human duodenal, lymph node, and pancreatic gastrinoma samples, collected from patients who underwent surgery from 1996 through 2007 in the United States or the United Kingdom. RESULTS Enteric glial cells that stained positive for glial fibrillary acidic protein (GFAP+) expressed gastrin de novo through a mechanism that required PKA. Gastrin-induced nuclear export of menin via cholecystokinin B receptor (CCKBR)-mediated activation of PKA. Once exported from the nucleus, menin was ubiquitinated and degraded by the proteasome. GFAP and other markers of enteric glial cells (eg, p75 and S100B), colocalized with gastrin in human duodenal gastrinomas. CONCLUSIONS MEN1-associated gastrinomas, which develop in the submucosa, might arise from enteric glial cells through hormone-dependent PKA signaling. This pathway disrupts nuclear menin function, leading to hypergastrinemia and associated sequelae.
Collapse
Affiliation(s)
- Sinju Sundaresan
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Cameron A Meininger
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Anthony J Kang
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Amanda L Photenhauer
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Michael M Hayes
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Nirakar Sahoo
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Lin Ding
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan
| | - Thomas J Giordano
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Tobias Else
- Division of Metabolism Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - David J Madrigal
- Endocrine Oncology Program, University of Michigan, Ann Arbor, Michigan
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Fiona Campbell
- Department of Pathology, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Ann-Marie Baker
- Center for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Nicholas A Wright
- Center for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Juanita L Merchant
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
24
|
Pieterman CRC, de Laat JM, Twisk JWR, van Leeuwaarde RS, de Herder WW, Dreijerink KMA, Hermus ARMM, Dekkers OM, van der Horst-Schrivers ANA, Drent ML, Bisschop PH, Havekes B, Borel Rinkes IHM, Vriens MR, Valk GD. Long-Term Natural Course of Small Nonfunctional Pancreatic Neuroendocrine Tumors in MEN1-Results From the Dutch MEN1 Study Group. J Clin Endocrinol Metab 2017; 102:3795-3805. [PMID: 28938468 DOI: 10.1210/jc.2017-00372] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 08/01/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (pNETs) are highly prevalent in patients with multiple endocrine neoplasia type 1 (MEN1), and metastatic disease is an important cause of MEN1-related mortality. Especially small nonfunctional (NF) pNETs pose a challenge to the treating physician and more information is needed regarding their natural course. We assessed long-term natural history of small NF-pNETs and its modifiers in the Dutch MEN1 population. PATIENTS AND METHODS Retrospective longitudinal observational cohort study of patients with small (<2 cm) NF-pNETs from the Dutch national MEN1 database, which includes >90% of the Dutch MEN1 population. Modifiers of long-term natural course were analyzed using linear mixed-models analysis. RESULTS Growth rate of the 115 included small NF-pNETs from 99 patients was slow (0.4 mm/y; 95% confidence interval, 0.15 to 0.59). Seventy percent of the tumors was stable and a subgroup of 30% of the tumors was growing (1.6 mm/y; 95% confidence interval, 1.1 to 2.0). No differences in clinical characteristics were identified between growing and stable tumors. Within the subgroup of growing tumors, germline missense mutations were significantly associated with accelerated growth compared with nonsense and frameshift mutations. CONCLUSION The majority of small NF-pNETs are stable at long-term follow-up, irrespective of the underlying MEN1 genotype. A subgroup of tumors is slowly growing but cannot be identified on clinical grounds. In this subgroup, tumors with missense mutations exhibited faster growth. Additional events appear necessary for pNETs to progress. Future studies should be aimed at identifying these molecular driving events, which could be used as potential biomarkers.
Collapse
Affiliation(s)
- Carolina R C Pieterman
- Department of Endocrine Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Joanne M de Laat
- Department of Endocrine Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Jos W R Twisk
- Department of Clinical Epidemiology and Biostatistics, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
- Department of Health Sciences, VU University, 1007 MB Amsterdam, The Netherlands
| | - Rachel S van Leeuwaarde
- Department of Endocrine Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Wouter W de Herder
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Koen M A Dreijerink
- Department of Endocrine Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Ad R M M Hermus
- Department of Endocrinology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Olaf M Dekkers
- Departments of Endocrinology and Metabolism and Clinical Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Madeleine L Drent
- Department of Internal Medicine, Section of Endocrinology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Academic Medical Center, 1100 DD Amsterdam, The Netherlands
| | - Bastiaan Havekes
- Department of Internal Medicine, Division of Endocrinology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
| | - Inne H M Borel Rinkes
- Department of Endocrine Surgical Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Menno R Vriens
- Department of Endocrine Surgical Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Gerlof D Valk
- Department of Endocrine Oncology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
25
|
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: 4.4] [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
|
26
|
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: 6.5] [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
|
27
|
Feng Z, Ma J, Hua X. Epigenetic regulation by the menin pathway. Endocr Relat Cancer 2017; 24:T147-T159. [PMID: 28811300 PMCID: PMC5612327 DOI: 10.1530/erc-17-0298] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [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: 08/15/2017] [Indexed: 02/06/2023]
Abstract
There is a trend of increasing prevalence of neuroendocrine tumors (NETs), and the inherited multiple endocrine neoplasia type 1 (MEN1) syndrome serves as a genetic model to investigate how NETs develop and the underlying mechanisms. Menin, encoded by the MEN1 gene, at least partly acts as a scaffold protein by interacting with multiple partners to regulate cellular homeostasis of various endocrine organs. Menin has multiple functions including regulation of several important signaling pathways by controlling gene transcription. Here, we focus on reviewing the recent progress in elucidating the key biochemical role of menin in epigenetic regulation of gene transcription and cell signaling, as well as posttranslational regulation of menin itself. In particular, we will review the progress in studying structural and functional interactions of menin with various histone modifiers and transcription factors such as MLL, PRMT5, SUV39H1 and other transcription factors including c-Myb and JunD. Moreover, the role of menin in regulating cell signaling pathways such as TGF-beta, Wnt and Hedgehog, as well as miRNA biogenesis and processing will be described. Further, the regulation of the MEN1 gene transcription, posttranslational modifications and stability of menin protein will be reviewed. These various modes of regulation by menin as well as regulation of menin by various biological factors broaden the view regarding how menin controls various biological processes in neuroendocrine organ homeostasis.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Ma
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary BiotechnologyHarbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xianxin Hua
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Kampmeyer C, Nielsen SV, Clausen L, Stein A, Gerdes AM, Lindorff-Larsen K, Hartmann-Petersen R. Blocking protein quality control to counter hereditary cancers. Genes Chromosomes Cancer 2017; 56:823-831. [PMID: 28779490 DOI: 10.1002/gcc.22487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/29/2017] [Accepted: 08/01/2017] [Indexed: 12/28/2022] Open
Abstract
Inhibitors of molecular chaperones and the ubiquitin-proteasome system have already been clinically implemented to counter certain cancers, including multiple myeloma and mantle cell lymphoma. The efficacy of this treatment relies on genomic alterations in cancer cells causing a proteostatic imbalance, which makes them more dependent on protein quality control (PQC) mechanisms than normal cells. Accordingly, blocking PQC, e.g. by proteasome inhibitors, may cause a lethal proteotoxic crisis in cancer cells, while leaving normal cells unaffected. Evidence, however, suggests that the PQC system operates by following a better-safe-than-sorry principle and is thus prone to target proteins that are only slightly structurally perturbed, but still functional. Accordingly, implementing PQC inhibitors may also, through an entirely different mechanism, hold potential for other cancers. Several inherited cancer susceptibility syndromes, such as Lynch syndrome and von Hippel-Lindau disease, are caused by missense mutations in tumor suppressor genes, and in some cases, the resulting amino acid substitutions in the encoded proteins cause the cellular PQC system to target them for degradation, although they may still retain function. As a consequence of this over-meticulous PQC mechanism, the cell may end up with an insufficient amount of the abnormal, but functional, protein, which in turn leads to a loss-of-function phenotype and manifestation of the disease. Increasing the amounts of such proteins by stabilizing with chemical chaperones, or by targeting molecular chaperones or the ubiquitin-proteasome system, may thus avert or delay the disease onset. Here, we review the potential of targeting the PQC system in hereditary cancer susceptibility syndromes.
Collapse
Affiliation(s)
- Caroline Kampmeyer
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| | - Sofie V Nielsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| | - Lene Clausen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| | - Amelie Stein
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, Copenhagen, DK-2100, Denmark
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| | - Rasmus Hartmann-Petersen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen, DK-2200, Denmark
| |
Collapse
|
29
|
De Sousa SMC, McCabe MJ, Wu K, Roscioli T, Gayevskiy V, Brook K, Rawlings L, Scott HS, Thompson TJ, Earls P, Gill AJ, Cowley MJ, Dinger ME, McCormack AI. Germline variants in familial pituitary tumour syndrome genes are common in young patients and families with additional endocrine tumours. Eur J Endocrinol 2017; 176:635-644. [PMID: 28220018 DOI: 10.1530/eje-16-0944] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/08/2017] [Accepted: 02/17/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Familial pituitary tumour syndromes (FPTS) account for 5% of pituitary adenomas. Multi-gene analysis via next-generation sequencing (NGS) may unveil greater prevalence and inform clinical care. We aimed to identify germline variants in selected patients with pituitary adenomas using a targeted NGS panel. DESIGN We undertook a nationwide cross-sectional study of patients with pituitary adenomas with onset ≤40 years of age and/or other personal/family history of endocrine neoplasia. A custom NGS panel was performed on germline DNA to interrogate eight FPTS genes. Genome data were analysed via a custom bioinformatic pipeline, and validation was performed by Sanger sequencing. Multiplex ligation-dependent probe amplification (MLPA) was performed in cases with heightened suspicion for MEN1, CDKN1B and AIP mutations. The main outcomes were frequency and pathogenicity of rare variants in AIP, CDKN1B, MEN1, PRKAR1A, SDHA, SDHB, SDHC and SDHD. RESULTS Forty-four patients with pituitary tumours, 14 of whom had a personal history of other endocrine tumours and/or a family history of pituitary or other endocrine tumours, were referred from endocrine tertiary-referral centres across Australia. Eleven patients (25%) had a rare variant across the eight FPTS genes tested: AIP (p.A299V, p.R106C, p.F269F, p.R304X, p.K156K, p.R271W), MEN1 (p.R176Q), SDHB (p.A2V, p.S8S), SDHC (p.E110Q) and SDHD (p.G12S), with two patients harbouring dual variants. Variants were classified as pathogenic or of uncertain significance in 9/44 patients (20%). No deletions/duplications were identified in MEN1, CDKN1B or AIP. CONCLUSIONS A high yield of rare variants in genes implicated in FPTS can be found in selected patients using an NGS panel. It may also identify individuals harbouring more than one rare variant.
Collapse
Affiliation(s)
- Sunita M C De Sousa
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- Endocrine and Metabolic UnitRoyal Adelaide Hospital, Adelaide, Australia
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
- School of MedicineUniversity of Adelaide, Adelaide, Australia
| | - Mark J McCabe
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Kathy Wu
- Familial Cancer ServiceWestmead Hospital, Westmead, Australia
- School of MedicineUniversity of Sydney, Sydney, Australia
| | - Tony Roscioli
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
- Department of Medical GeneticsSydney Children's Hospital, Sydney, Australia
| | - Velimir Gayevskiy
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
| | - Katelyn Brook
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
| | - Lesley Rawlings
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
| | - Hamish S Scott
- Department of Genetics and Molecular PathologyCentre for Cancer Biology, an SA Pathology and UniSA alliance, Adelaide, Australia
- School of MedicineUniversity of Adelaide, Adelaide, Australia
- ACRF Cancer Genomics FacilityCentre for Cancer Biology, SA Pathology, Adelaide, Australia
- School of Biological SciencesUniversity of Adelaide, Adelaide, Australia
- School of Pharmacy and Medical SciencesUniversity of South Australia, Adelaide, Australia
| | - Tanya J Thompson
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
| | - Peter Earls
- School of Pharmacy and Medical SciencesUniversity of South Australia, Adelaide, Australia
| | - Anthony J Gill
- School of MedicineUniversity of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology GroupKolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
- Sydney Vital Translational Cancer Research CentreRoyal North Shore Hospital and University of Sydney, Sydney, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Marcel E Dinger
- Kinghorn Centre for Clinical GenomicsGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
| | - Ann I McCormack
- Hormones and Cancer GroupGarvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical SchoolUniversity of New South Wales, Sydney, Australia
- Department of EndocrinologySt Vincent's Hospital, Sydney, Australia
| |
Collapse
|
30
|
Tonelli F, Marini F, Giusti F, Brandi ML. Gastro-entero-pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1: a therapy update. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2017. [DOI: 10.2217/ije-2016-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) are the second most common tumors in multiple endocrine neoplasia type 1 (MEN1), mainly occurring in pancreatic islets and duodenum, usually as multiple tumors. They can manifest as both nonfunctioning and functioning tumors. Currently, surgical removal of GEP-NETs in MEN1 represents the gold standard curative approach. Conventional medical therapies for sporadic GEP-NETs showed to be effective also in a percentage of MEN1 patients. Innovative medical therapies, that have demonstrated to be effective on sporadic GEP-NETs, still need to be evaluated on MEN1 patients in prospective clinical trials and long-term follow-up. This review resumes current knowledge of MEN1 GEP-NETs, discussing surgical and medical approaches, genetic and molecular bases of tumorigenesis, and presenting novel possible drug therapies.
Collapse
Affiliation(s)
- Francesco Tonelli
- Department of Surgery & Translational Medicine, University of Florence, Florence, Italy
| | - Francesca Marini
- Department of Surgery & Translational Medicine, University of Florence, Florence, Italy
| | - Francesca Giusti
- Department of Surgery & Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery & Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
31
|
Gantenbein N, Haybaeck J. Neuroendocrine Tumorigenesis. MECHANISMS OF MOLECULAR CARCINOGENESIS – VOLUME 2 2017:141-146. [DOI: 10.1007/978-3-319-53661-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
32
|
He X, Wang L, Yan J, Yuan C, Witze ES, Hua X. Menin localization in cell membrane compartment. Cancer Biol Ther 2016; 17:114-22. [PMID: 26560942 DOI: 10.1080/15384047.2015.1108497] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Menin is encoded by the MEN1 gene, which is mutated in an inherited human syndrome, multiple endocrine neoplasia type 1(MEN1). Menin is primarily nuclear protein, acting as a tumor suppressor in endocrine organs, but as an oncogenic factor in the mixed lineage leukemia, in a tissue-specific manner. Recently, the crystal structures of menin with different binding partners reveal menin as a key scaffold protein that functionally interacts with various partners to regulate gene transcription in the nucleus. However, outside the nucleus, menin also regulates multiple signaling pathways that traverse the cell surface membrane. The precise nature regarding to how menin associates with the membrane fraction is poorly understood. Here we show that a small fraction of menin associates with the cell membrane fraction likely via serine palmitoylation. Moreover, the majority of the membrane-associated menin may reside inside membrane vesicles, as menin is protected from trypsin-mediated proteolysis, but disruption of the membrane fraction using detergent abolishes the detection. Consistently, cellular staining for menin also reveals the distribution of menin in the cell membrane and the punctate-like cell organelles. Our findings suggest that part of intracellular menin associates with the cell membrane peripherally as well as resides within the membrane vesicles.
Collapse
Affiliation(s)
- Xin He
- a Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine , 421 Curie Blvd., Philadelphia , PA 19104 , USA
| | - Lei Wang
- a Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine , 421 Curie Blvd., Philadelphia , PA 19104 , USA.,d Department of Urology , Renmin Hospital of Wuhan University , Wuhan 430060 , Hubei , China
| | - Jizhou Yan
- b Department of Biology and Biotechnology , Shanghai Ocean University , 999 Hucheng Ring Rd Lingang New City, Shanghai , 201306 , China
| | - Chaoxing Yuan
- c The Proteomics and Systems Facility, Department of Pharmacology, University of Pennsylvania Perelman School of Medicine , Philadelphia, 421 Curie Blvd., Philadelphia , PA 19104 , USA
| | - Eric S Witze
- a Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine , 421 Curie Blvd., Philadelphia , PA 19104 , USA
| | - Xianxin Hua
- a Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine , 421 Curie Blvd., Philadelphia , PA 19104 , USA
| |
Collapse
|
33
|
Joshi V, Amanullah A, Upadhyay A, Mishra R, Kumar A, Mishra A. A Decade of Boon or Burden: What Has the CHIP Ever Done for Cellular Protein Quality Control Mechanism Implicated in Neurodegeneration and Aging? Front Mol Neurosci 2016; 9:93. [PMID: 27757073 PMCID: PMC5047891 DOI: 10.3389/fnmol.2016.00093] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/20/2016] [Indexed: 01/13/2023] Open
Abstract
Cells regularly synthesize new proteins to replace old and abnormal proteins for normal cellular functions. Two significant protein quality control pathways inside the cellular milieu are ubiquitin proteasome system (UPS) and autophagy. Autophagy is known for bulk clearance of cytoplasmic aggregated proteins, whereas the specificity of protein degradation by UPS comes from E3 ubiquitin ligases. Few E3 ubiquitin ligases, like C-terminus of Hsc70-interacting protein (CHIP) not only take part in protein quality control pathways, but also plays a key regulatory role in other cellular processes like signaling, development, DNA damage repair, immunity and aging. CHIP targets misfolded proteins for their degradation through proteasome, as well as autophagy; simultaneously, with the help of chaperones, it also regulates folding attempts for misfolded proteins. The broad range of CHIP substrates and their associations with multiple pathologies make it a key molecule to work upon and focus for future therapeutic interventions. E3 ubiquitin ligase CHIP interacts and degrades many protein inclusions formed in neurodegenerative diseases. The presence of CHIP at various nodes of cellular protein-protein interaction network presents this molecule as a potential candidate for further research. In this review, we have explored a wide range of functionality of CHIP inside cells by a detailed presentation of its co-chaperone, E3 and E4 enzyme like functions, with central focus on its protein quality control roles in neurodegenerative diseases. We have also raised many unexplored but expected fundamental questions regarding CHIP functions, which generate hopes for its future applications in research, as well as drug discovery.
Collapse
Affiliation(s)
- Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Amit Kumar
- Centre for Biosciences and Biomedical Engineering, Indian Institute of Technology Indore Madhya Pradesh, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| |
Collapse
|
34
|
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: 5.0] [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
|
35
|
Familial syndromes associated with neuroendocrine tumours. Contemp Oncol (Pozn) 2015; 19:176-83. [PMID: 26557756 PMCID: PMC4631294 DOI: 10.5114/wo.2015.52710] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine tumours may be associated with familial syndromes. At least eight inherited syndromes predisposing to endocrine neoplasia have been identified. Two of these are considered to be major factors predisposing to benign and malignant endocrine tumours, designated multiple endocrine neoplasia type 1 and type 2 (MEN1 and MEN2). Five other autosomal dominant diseases show more heterogeneous clinical patterns, such as the Carney complex, hyperparathyroidism-jaw tumour syndrome, Von Hippel-Lindau syndrome (VHL), neurofibromatosis type 1 (NF1) and tuberous sclerosis. The molecular and cellular interactions underlying the development of most endocrine cells and related organs represent one of the more complex pathways not yet to be deciphered. Almost all endocrine cells are derived from the endoderm and neuroectoderm. It is suggested that within the first few weeks of human development there are complex interactions between, firstly, the major genes involved in the initiation of progenitor-cell differentiation, secondly, factors secreted by the surrounding mesenchyme, and thirdly, a series of genes controlling cell differentiation, proliferation and migration. Together these represent a formula for the harmonious development of endocrine glands and tissue.
Collapse
|
36
|
129-Derived Mouse Strains Express an Unstable but Catalytically Active DNA Polymerase Iota Variant. Mol Cell Biol 2015; 35:3059-70. [PMID: 26124279 DOI: 10.1128/mcb.00371-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/18/2015] [Indexed: 01/08/2023] Open
Abstract
Mice derived from the 129 strain have a nonsense codon mutation in exon 2 of the polymerase iota (Polι) gene and are therefore considered Polι deficient. When we amplified Polι mRNA from 129/SvJ or 129/Ola testes, only a small fraction of the full-length cDNA contained the nonsense mutation; the major fraction corresponded to a variant Polι isoform lacking exon 2. Polι mRNA lacking exon 2 contains an open reading frame, and the corresponding protein was detected using a polyclonal antibody raised against the C terminus of the murine Polι protein. The identity of the corresponding protein was further confirmed by mass spectrometry. Although the variant protein was expressed at only 5 to 10% of the level of wild-type Polι, it retained de novo DNA synthesis activity, the capacity to form replication foci following UV irradiation, and the ability to rescue UV light sensitivity in Polι(-/-) embryonic fibroblasts derived from a new, fully deficient Polι knockout (KO) mouse line. Furthermore, in vivo treatment of 129-derived male mice with Velcade, a drug that inhibits proteasome function, stabilized and restored a substantial amount of the variant Polι in these animals, indicating that its turnover is controlled by the proteasome. An analysis of two xeroderma pigmentosum-variant (XPV) cases corresponding to missense mutants of Polη, a related translesion synthesis (TLS) polymerase in the same family, similarly showed a destabilization of the catalytically active mutant protein by the proteasome. Collectively, these data challenge the prevailing hypothesis that 129-derived strains of mice are completely deficient in Polι activity. The data also document, both for 129-derived mouse strains and for some XPV patients, new cases of genetic defects corresponding to the destabilization of an otherwise functional protein, the phenotype of which is reversible by proteasome inhibition.
Collapse
|
37
|
Chen X, Sun S, Wang C, Chen D, Chen H, Ran X. Novel mutation 928G>C of MEN1 gene in a familial multiple endocrine neoplasia type 1 case (MEN1) with co-existence of insulinoma and glucagonoma. J Diabetes 2015; 7:426-429. [PMID: 25047095 DOI: 10.1111/1753-0407.12199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/29/2014] [Accepted: 07/02/2014] [Indexed: 11/30/2022] Open
Affiliation(s)
- Xiang Chen
- Laboratory of Endocrinology and Metabolism, Sichuan University, Chengdu, China
| | | | | | | | | | | |
Collapse
|
38
|
Hall MJ, Innocent J, Rybak C, Veloski C, Scott WJ, Wu H, Ridge JA, Hoffman JP, Borghaei H, Turaka A, Daly MB. Bilateral granulosa cell tumors: a novel malignant manifestation of multiple endocrine neoplasia 1 syndrome found in a patient with a rare menin in-frame deletion. APPLICATION OF CLINICAL GENETICS 2015; 8:69-73. [PMID: 25733923 PMCID: PMC4337709 DOI: 10.2147/tacg.s72223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction Multiple endocrine neoplasia 1 (MEN1) is a cancer syndrome resulting from mutations of the MEN1 gene. The syndrome is characterized by neoplasia of the parathyroid and pituitary glands, and malignant tumors of the endocrine pancreas. Other manifestations include benign lipomas, angiofibromas, and carcinoid tumors commonly originating in the colon, thymus, and lung. This is the first report of MEN1 syndrome manifesting as bilateral granulosa cell ovarian tumors, and which is associated with a rare intronic mutation of the MEN1 gene. Case report A 41-year-old woman presented with abdominal pain, increasing abdominal girth, and dysmenorrhea. Ultrasound demonstrated enlarged ovaries and uterine fibroids. After an exploratory laparotomy, she subsequently underwent bilateral salpingo–oophorectomy with hysterectomy where the pathology revealed bilateral cystic granulosa cell tumors of the ovaries. Additional workup including computed tomography imaging discovered a thymic mass, which the pathology showed was malignant, along with a pancreatic mass suspicious for a neuroendocrine tumor. Hyperparathyroidism was also discovered and was found to be secondary to a parathyroid adenoma. Genetic testing revealed an exceedingly rare mutation in the MEN1 gene (c.654 + 1 G>A). Discussion Mutations of the menin gene leading to MEN1 syndrome are classically nonsense or missense mutations producing a dysfunctional protein product. Recently, researchers described a novel mutation of MEN1 (c.654 + 1 G>A) in a male proband meeting the criteria for clinical MEN1 syndrome. Functional analysis performed on the stable mutant protein showed selective disruption of the transforming growth factor beta signaling pathway, yet it maintained its wild-type ability to inhibit nuclear factor kappa B and to suppress JunD transcriptional activity. Conclusion To our knowledge, this is the first report of MEN1 syndrome associated with bilateral granulosa cell malignancy. We postulate that this presentation may be due to the novel menin gene mutation recently described.
Collapse
Affiliation(s)
- Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Julie Innocent
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Christina Rybak
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Colleen Veloski
- Department of Internal Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Walter J Scott
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hong Wu
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John A Ridge
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John P Hoffman
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hossein Borghaei
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aruna Turaka
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
39
|
Pieterman CRC, Conemans EB, Dreijerink KMA, de Laat JM, Timmers HTM, Vriens MR, Valk GD. Thoracic and duodenopancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1: natural history and function of menin in tumorigenesis. Endocr Relat Cancer 2014; 21:R121-42. [PMID: 24389729 DOI: 10.1530/erc-13-0482] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mutations of the multiple endocrine neoplasia type 1 (MEN1) gene lead to loss of function of its protein product menin. In keeping with its tumor suppressor function in endocrine tissues, the majority of the MEN1-related neuroendocrine tumors (NETs) show loss of heterozygosity (LOH) on chromosome 11q13. In sporadic NETs, MEN1 mutations and LOH are also reported, indicating common pathways in tumor development. Prevalence of thymic NETs (thNETs) and pulmonary carcinoids in MEN1 patients is 2-8%. Pulmonary carcinoids may be underreported and research on natural history is limited, but disease-related mortality is low. thNETs have a high mortality rate. Duodenopancreatic NETs (dpNETs) are multiple, almost universally found at pathology, and associated with precursor lesions. Gastrinomas are usually located in the duodenal submucosa while other dpNETs are predominantly pancreatic. dpNETs are an important determinant of MEN1-related survival, with an estimated 10-year survival of 75%. Survival differs between subtypes and apart from tumor size there are no known prognostic factors. Natural history of nonfunctioning pancreatic NETs needs to be redefined because of increased detection of small tumors. MEN1-related gastrinomas seem to behave similar to their sporadic counterparts, while insulinomas seem to be more aggressive. Investigations into the molecular functions of menin have led to new insights into MEN1-related tumorigenesis. Menin is involved in gene transcription, both as an activator and repressor. It is part of chromatin-modifying protein complexes, indicating involvement of epigenetic pathways in MEN1-related NET development. Future basic and translational research aimed at NETs in large unbiased cohorts will clarify the role of menin in NET tumorigenesis and might lead to new therapeutic options.
Collapse
Affiliation(s)
- C R C Pieterman
- Division of Internal Medicine and Dermatology, Department of Internal Medicine, University Medical Center Utrecht, Internal post number L.00.408, PO Box 85500, 3508 GA Utrecht, The Netherlands Division of Biomedical Genetics, Department of Molecular Cancer Research Division of Surgical Specialties, Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Cierpicki T, Grembecka J. Challenges and opportunities in targeting the menin-MLL interaction. Future Med Chem 2014; 6:447-62. [PMID: 24635524 PMCID: PMC4138051 DOI: 10.4155/fmc.13.214] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Menin is an essential co-factor of oncogenic MLL fusion proteins and the menin-MLL interaction is critical for development of acute leukemia in vivo. Targeting the menin-MLL interaction with small molecules represents an attractive strategy to develop new anticancer agents. Recent developments, including determination of menin crystal structure and development of potent small molecule and peptidomimetic inhibitors, demonstrate the feasibility of targeting the menin-MLL interaction. On the other hand, biochemical and structural studies revealed that MLL binds to menin in a complex bivalent mode engaging two MLL motifs, and therefore inhibition of this protein-protein interaction represents a challenge. This review summarizes the most recent achievements in targeting the menin-MLL interaction as well as discusses potential benefits of blocking menin in cancer.
Collapse
Affiliation(s)
- Tomasz Cierpicki
- Author for correspondence: Tel.: +1 734 615 9324, Fax: +1 734 615 0688,
| | | |
Collapse
|
41
|
Hamze Z, Vercherat C, Bernigaud-Lacheretz A, Bazzi W, Bonnavion R, Lu J, Calender A, Pouponnot C, Bertolino P, Roche C, Stein R, Scoazec JY, Zhang CX, Cordier-Bussat M. Altered MENIN expression disrupts the MAFA differentiation pathway in insulinoma. Endocr Relat Cancer 2013; 20:833-48. [PMID: 24157940 PMCID: PMC3841063 DOI: 10.1530/erc-13-0164] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The protein MENIN is the product of the multiple endocrine neoplasia type I (MEN1) gene. Altered MENIN expression is one of the few events that are clearly associated with foregut neuroendocrine tumours (NETs), classical oncogenes or tumour suppressors being not involved. One of the current challenges is to understand how alteration of MENIN expression contributes to the development of these tumours. We hypothesised that MENIN might regulate factors maintaining endocrine-differentiated functions. We chose the insulinoma model, a paradigmatic example of well-differentiated pancreatic NETs, to study whether MENIN interferes with the expression of v-MAF musculoaponeurotic fibrosarcoma oncogene homologue A (MAFA), a master glucose-dependent transcription factor in differentiated β-cells. Immunohistochemical analysis of a series of human insulinomas revealed a correlated decrease in both MENIN and MAFA. Decreased MAFA expression resulting from targeted Men1 ablation was also consistently observed in mouse insulinomas. In vitro analyses using insulinoma cell lines showed that MENIN regulated MAFA protein and mRNA levels, and bound to Mafa promoter sequences. MENIN knockdown concomitantly decreased mRNA expression of both Mafa and β-cell differentiation markers (Ins1/2, Gck, Slc2a2 and Pdx1) and, in parallel, increased the proliferation rate of tumours as measured by bromodeoxyuridine incorporation. Interestingly, MAFA knockdown alone also increased proliferation rate but did not affect the expression of candidate proliferation genes regulated by MENIN. Finally, MENIN variants with missense mutations detected in patients with MEN1 lost the WT MENIN properties to regulate MAFA. Together, our findings unveil a previously unsuspected MENIN/MAFA connection regarding control of the β-cell differentiation/proliferation balance, which could contribute to tumorigenesis.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Apoptosis
- Blotting, Western
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/pathology
- Cell Differentiation
- Cell Proliferation
- Chromatin Immunoprecipitation
- Female
- Glucose/pharmacology
- Humans
- Immunoenzyme Techniques
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulinoma/genetics
- Insulinoma/metabolism
- Insulinoma/pathology
- Maf Transcription Factors, Large/antagonists & inhibitors
- Maf Transcription Factors, Large/genetics
- Maf Transcription Factors, Large/metabolism
- Male
- Mice
- Mice, Knockout
- Middle Aged
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Rats
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Z Hamze
- INSERM U1052/CNRS UMR5286/Université de Lyon, Lyon1 UMR-S1052, Cancer Research Center of Lyon, Lyon F-69008, France Service de Génétique Moléculaire et Clinique, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon F-69437, France UMR 3347/CNRS, U1021/INSERM, Institut Curie, Orsay F-91405, France Service Central d'Anatomie et Cytologie Pathologiques, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon F-69437, France Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
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: 196] [Impact Index Per Article: 16.3] [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
|
43
|
Feng ZJ, Gurung B, Jin GH, Yang XL, Hua XX. SUMO modification of menin. Am J Cancer Res 2013; 3:96-106. [PMID: 23359867 PMCID: PMC3555195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/27/2012] [Indexed: 06/01/2023] Open
Abstract
Menin acts as contextual a tumor suppressor and a tumor promoter, partly via epigenetic regulation of gene transcription. While menin is phosphorylated, it remains unclear whether wild type menin has other post-translational modifications. Here, we report that menin is SUMOylated by SUMO1 in vivo and in vitro, and the SUMOylation is reduced by a SUMO protease. Lysine 591 of menin was covalently modified by SUMO1 and K591R mutation in menin blocked SUMOylation of the C-terminal part of menin in transfected cells. Full-length menin with K591 mutation was still SUMOylated in vivo, suggesting the existence of multiple SUMOylation sites. Menin K591R mutant or menin-SUMO fusion protein still retains the ability to regulate cell proliferation and the expression of the examined menin target genes.
Collapse
Affiliation(s)
- Zi-Jie Feng
- Department of Basic Medical Sciences, Medical College, Xiamen University Xiamen, Fujian, China 361005 ; Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine 421 Curie Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
44
|
Arlow T, Scott K, Wagenseller A, Gammie A. Proteasome inhibition rescues clinically significant unstable variants of the mismatch repair protein Msh2. Proc Natl Acad Sci U S A 2013; 110:246-51. [PMID: 23248292 PMCID: PMC3538226 DOI: 10.1073/pnas.1215510110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
MSH2 is required for DNA mismatch repair recognition in eukaryotes. Deleterious mutations in human MSH2 account for approximately half of the alleles associated with a common hereditary cancer syndrome. Previously, we characterized clinically identified MSH2 missense mutations, using yeast as a model system, and found that the most common cause of defective DNA mismatch repair was low levels of the variant Msh2 proteins. Here, we show that increased protein turnover is responsible for the reduced cellular levels. Increasing gene dosage of more than half of the missense alleles fully restored function. A titration experiment revealed that raising the expression level of one variant to less than wild-type levels restored mismatch repair, suggesting that overexpression is not always required to regain function. We found that the ubiquitin-mediated proteasome degradation pathway is the major mechanism for increased turnover of the Msh2 variants and identified the primary ubiquitin ligase as San1. Deletion of San1 restored protein levels for all but one variant, but did not elevate wild-type Msh2 levels. The unstable variants interacted with San1, whereas wild-type Msh2 did not. Additionally, san1Δ suppressed the mismatch repair defect of unstable variants. Of medical significance, the clinically approved drug Bortezomib partially restored protein levels and mismatch repair function for low-level variants and reversed the resistance to cisplatin, a common chemotherapeutic. Our results provide the foundation for an innovative therapeutic regime for certain mismatch-repair-defective cancers that are refractory to conventional chemotherapies.
Collapse
Affiliation(s)
- Tim Arlow
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Kristan Scott
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Aubrey Wagenseller
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Alison Gammie
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
45
|
Carroll RW. Multiple endocrine neoplasia type 1 (MEN1). Asia Pac J Clin Oncol 2012; 9:297-309. [DOI: 10.1111/ajco.12046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Richard W Carroll
- Endocrine, Diabetes and Research Centre; Wellington Regional Hospital; Wellington New Zealand
| |
Collapse
|
46
|
Wu Y, Feng ZJ, Gao SB, Matkar S, Xu B, Duan HB, Lin X, Li SH, Hua X, Jin GH. Interplay between menin and K-Ras in regulating lung adenocarcinoma. J Biol Chem 2012; 287:40003-11. [PMID: 23027861 DOI: 10.1074/jbc.m112.382416] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
MEN1, which encodes the nuclear protein menin, acts as a tumor suppressor in lung cancer and is often inactivated in human primary lung adenocarcinoma. Here, we show that the inactivation of MEN1 is associated with increased DNA methylation at the MEN1 promoter by K-Ras. On one hand, the activated K-Ras up-regulates the expression of DNA methyltransferases and enhances the binding of DNA methyltransferase 1 to the MEN1 promoter, leading to increased DNA methylation at the MEN1 gene in lung cancer cells; on the other hand, menin reduces the level of active Ras-GTP at least partly by preventing GRB2 and SOS1 from binding to Ras, without affecting the expression of GRB2 and SOS1. In human lung adenocarcinoma samples, we further demonstrate that reduced menin expression is associated with the enhanced expression of Ras (p < 0.05). Finally, excision of the Men1 gene markedly accelerates the K-Ras(G12D)-induced tumor formation in the Men1(f/f);K-Ras(G12D/+);Cre ER mouse model. Together, these findings uncover a previously unknown link between activated K-Ras and menin, an important interplay governing tumor activation and suppression in the development of lung cancer.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Basic Medical Sciences, Medical College, Zhongshan Hospital, Xiamen University, 361005 Fujian, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lips CJ, Dreijerink KM, Links TP, Höppener JW. Recent results of basic and clinical research in MEN1: opportunities to improve early detection and treatment. Expert Rev Endocrinol Metab 2012; 7:331-344. [PMID: 30780845 DOI: 10.1586/eem.12.22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Due to the variable expression of multiple endocrine neoplasia type 1 (MEN1), it is difficult to predict the course of the disease. However, knowledge about the normal function of the MEN1 gene product, together with the effects of cellular derangement by subsequent genetic events, has increased considerably. At first, the possible existence of a genotype-phenotype correlation is discussed. Thus, mild- and late-onset phenotypes may be distinguished from more malignant phenotypes depending on the character of the primary MEN1 disease gene mutation. Subsequently, tumor-promoting factors such as gender, additional genetic mutations and ecogenetic factors may contribute to the course of the disease. New developments in management are based on the knowledge and experience of the multidisciplinary teams involved. Finally, the metabolic effects of MEN1 mutations in aged patients are discussed. Early identification of predisposition to the disease, together with knowledge about the natural history of specific mutations, risks of additional mutations and periodic clinical monitoring, allow early treatment and may improve life expectancy and quality of life.
Collapse
Affiliation(s)
- Cornelis Jm Lips
- a Department of Internal Medicine and Endocrinology, University Medical Center, Utrecht & The Hague, The Netherlands
- d Department of Internal Medicine and Endocrinology, University Medical Center, Utrecht & The Hague, The Netherlands.
| | - Koen Ma Dreijerink
- a Department of Internal Medicine and Endocrinology, University Medical Center, Utrecht & The Hague, The Netherlands
| | - Thera P Links
- b Department of Endocrinology, University Medical Center Groningen, The Netherlands
| | - Jo Wm Höppener
- c Department of Metabolic Diseases, University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
48
|
Canaff L, Vanbellinghen JF, Kanazawa I, Kwak H, Garfield N, Vautour L, Hendy GN. Menin missense mutants encoded by the MEN1 gene that are targeted to the proteasome: restoration of expression and activity by CHIP siRNA. J Clin Endocrinol Metab 2012; 97:E282-91. [PMID: 22090276 DOI: 10.1210/jc.2011-0241] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CONTEXT In multiple endocrine neoplasia type 1 (MEN1) characterized by tumors of parathyroid, enteropancreas, and anterior pituitary, missense mutations in the MEN1 gene product, menin, occur in a subset of cases. The mutant proteins are degraded by the proteasome. However, whether their expression and activity can be restored is not known. OBJECTIVE Our objective was to functionally characterize a panel of 16 menin missense mutants, including W423R and S443Y identified in new MEN1 families, with respect to protein stability, targeting to the proteasome and restoration of expression by proteasome inhibitors and expression and function by small interfering RNA technology. METHODS Flag-tagged wild-type (WT) and missense menin mutant expression vectors were transiently transfected in human embryonic kidney (HEK293) and/or rat insulinoma (Rin-5F) cells. RESULTS The majority of mutants were short-lived, whereas WT menin was stable. Proteasome inhibitors MG132 and PS-341 and inhibition of the chaperone, heat-shock protein 70 (Hsp70), or the ubiquitin ligase, COOH terminus of Hsp70-interacting protein (CHIP), by specific small interfering RNA, restored the levels of the mutants, whereas that of WT menin was largely unaffected. Inhibition of CHIP restored the ability of mutants to mediate normal functions of menin: TGF-β up-regulation of the promoters of its target genes, the cyclin-dependent kinase inhibitors p15 and p21 as well as TGF-β inhibition of cell numbers. CONCLUSION When the levels of missense menin mutants that are targeted to the proteasome are normalized they may function similarly to WT menin. Potentially, targeting specific components of the proteasome chaperone pathway could be beneficial in treating a subset of MEN1 cases.
Collapse
Affiliation(s)
- Lucie Canaff
- Department of Medicine, Calcium Research Laboratory, and Hormones and Cancer Research Unit, Royal Victoria Hospital, McGill University, Montreal, Quebec H3A 1A1, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Canaff L, Vanbellinghen JF, Kaji H, Goltzman D, Hendy GN. Impaired transforming growth factor-β (TGF-β) transcriptional activity and cell proliferation control of a menin in-frame deletion mutant associated with multiple endocrine neoplasia type 1 (MEN1). J Biol Chem 2012; 287:8584-97. [PMID: 22275377 DOI: 10.1074/jbc.m112.341958] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is characterized by tumors of the parathyroid, enteropancreas, and anterior pituitary. The MEN1 gene encodes the tumor suppressor menin of 610 amino acids that has multiple protein partners and activities. The particular pathways that, when lost, lead to tumorigenesis are not known. We demonstrated that members of a three-generation MEN1 kindred are heterozygous for a donor splice site mutation at the beginning of intron 3 (IVS3 + 1G→A). Lymphoblastoid cells of a mutant gene carrier had, in addition to the wild-type menin transcript, an aberrant transcript resulting from use of a cryptic splice site within exon III that splices to the start of exon IV. The predicted menin Δ(184-218) mutant has an in-frame deletion of 35 amino acids but is otherwise of wild-type sequence. The transfected menin Δ(184-218) mutant was well expressed and fully able to mediate the normal inhibition of the activity of the transcriptional regulators JunD and NF-κB. However, it was defective in mediating TGF-β-stimulated Smad3 action in promoter-reporter assays in insulinoma cells. Importantly, lymphoblastoid cells from an individual heterozygous for the mutation had reduced TGF-β-induced (Smad3) transcriptional activity but normal JunD and NF-κB function. In addition, the mutant gene carrier lymphoblastoid cells proliferated faster and were less responsive to the cytostatic effects of TGF-β than cells from an unaffected family member. In conclusion, the menin mutant exhibits selective loss of the TGF-β signaling pathway and loss of cell proliferation control contributing to the development of MEN1.
Collapse
Affiliation(s)
- Lucie Canaff
- Department of Medicine, Royal Victoria Hospital, McGill University, Montreal, Quebec H3A 1A1, Canada
| | | | | | | | | |
Collapse
|
50
|
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: 29] [Impact Index Per Article: 2.2] [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
|