1
|
Liu P, Shi C, Qiu L, Shang D, Lu Z, Tu Z, Liu H. Menin signaling and therapeutic targeting in breast cancer. Curr Probl Cancer 2024; 51:101118. [PMID: 38968834 DOI: 10.1016/j.currproblcancer.2024.101118] [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: 01/17/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
To date, mounting evidence have shown that patients with multiple endocrine neoplasia type 1 (MEN1) may face an increased risk for breast carcinogenesis. The product of the MEN1 gene, menin, was also indicated to be an important regulator in breast cancer signaling network. Menin directly interacts with MLL, EZH2, JunD, NF-κB, PPARγ, VDR, Smad3, β-catenin and ERα to modulate gene transcriptions leading to cell proliferation inhibition. Moreover, interaction of menin-FANCD2 contributes to the enhancement of BRCA1-mediated DNA repair mechanism. Ectopic expression of menin causes Bax-, Bak- and Caspase-8-dependent apoptosis. However, despite numbers of menin inhibitors were exploited in other cancers, data on the usage of menin inhibitors in breast cancer treatment remain limited. In this review, we focused on the menin associated signaling pathways and gene transcription regulations, with the aim of elucidating its molecular mechanisms and of guiding the development of novel menin targeted drugs in breast cancer therapy.
Collapse
Affiliation(s)
- Peng Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Chaowen Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Lipeng Qiu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Dongsheng Shang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Ziwen Lu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Zhigang Tu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China.
| |
Collapse
|
2
|
Wang S, Gu Z, Zhu L, Han Y, Yu H, Fang W, Han B. Genetic insights into thymic carcinomas and thymic neuroendocrine neoplasms denote prognosis signatures and pathways. Chin Med J (Engl) 2023; 136:2712-2721. [PMID: 37749819 PMCID: PMC10684125 DOI: 10.1097/cm9.0000000000002852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Thymic carcinomas (TCs) and thymic neuroendocrine neoplasms (TNENs) are two aggressive subtypes of thymic malignancy. Traditional therapy for advanced TCs and TNENs has limited outcome. New genomic profiling of TCs and TNENs might provide insights that contribute to the development of new treatment approaches. METHODS We used gene panel sequencing technologies to investigate the genetic aberrations of 32 TC patients and 15 TNEN patients who underwent surgery at Shanghai Chest Hospital between 2015 and 2017. Patient samples were sequenced using a 324-gene platform with licensed technologies. In this study, we focused on clinically relevant genomic alterations (CRGAs), which are previously proven to be pathogenic alterations, to identify the pathology-specific mutational patterns, prognostic signatures of TCs and TNENs. RESULTS The mutational profiles between TCs and TNENs were diverse. The genetic alterations that ranked highest in TCs were in CDKN2A, TP53, ASXL1, CDKN2B, PIK3C2G, PTCH1, and ROS1 , while those in TNENs were in MEN1, MLL2, APC, RB1 , and TSC2 . Prognostic analysis showed that mutations of ROS1, CDKN2A, CDKN2B, BRAF, and BAP1 were significantly associated with worse outcomes in TC patients, and that mutation of ERBB2 indicated shortened disease-free survival (DFS) and overall survival (OS) in TNEN patients. Further investigation found that the prognosis-related genes were focused on signal pathways of cell cycle control, chromatin remodeling/DNA methylation, phosphoinositide 3-kinases (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), and receptor tyrosine kinase (RTK)/RAS/mitogen-activated protein kinase (MAPK) signaling. CONCLUSION We profiled the mutational features of 47 Chinese patients with thymic malignancy of diverse pathologic phenotypes to uncover the integrated genomic landscape of these rare tumors, and identified the pathology-specific mutational patterns, prognostic signatures, and potential therapeutic targets for TCs and TNENs.
Collapse
Affiliation(s)
- Shuyuan Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhitao Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lei Zhu
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Baohui Han
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
3
|
Liu T, Li R, Sun L, Xu Z, Wang S, Zhou J, Wu X, Shi K. Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ. Cell Biosci 2023; 13:175. [PMID: 37740216 PMCID: PMC10517496 DOI: 10.1186/s13578-023-01119-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-induced liver steatosis in mice, as well as insulin resistance and type 2 diabetic phenotype. The potential link between Menin and hepatic metabolism homeostasis may provide new insights into the mechanism of fatty liver disease. RESULTS Disturbance of hepatic Menin expression impacts metabolic pathways associated with non-alcoholic fatty liver disease (NAFLD), including the FoxO signaling pathway, which is similar to that observed in both oleic acid-induced fatty hepatocytes model and biopsied fatty liver tissues, but with elevated hepatic Menin expression and inhibited FABP1. Higher levels of Menin facilitate glucose uptake while restraining fatty acid uptake. Menin targets the expression of FABP3/4/5 and also CD36 or GK, PCK by binding to their promoter regions, while recruiting and deploying the cellular localization of PPARγ and SIRT1 in the nucleus and cytoplasm. Accordingly, Menin binds to PPARγ and/or FoxO1 in hepatocytes, and orchestrates hepatic glucose and fatty acid uptake by recruiting SIRT1. CONCLUSION Menin plays an orchestration role as a transcriptional activator and/or repressor to target downstream gene expression levels involved in hepatic energy uptake by interacting with the cellular energy sensor SIRT1, PPARγ, and/or FoxO1 and deploying their translocations between the cytoplasm and nucleus, thereby maintaining metabolic homeostasis. These findings provide more evidence suggesting Menin could be targeted for the treatment of hepatic steatosis, NAFLD or metabolic dysfunction-associated fatty liver disease (MAFLD), and even other hepatic diseases.
Collapse
Affiliation(s)
- Tingjun Liu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Ranran Li
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Lili Sun
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Zhongjin Xu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Shengxuan Wang
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Jingxuan Zhou
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Xuanning Wu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Kerong Shi
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China.
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Marzyńska D, Żaba R, Lacka K. Lipomas: genetic basis of common skin lesions and their occurrence in rare diseases. Postepy Dermatol Alergol 2023; 40:481-486. [PMID: 37692275 PMCID: PMC10485760 DOI: 10.5114/ada.2023.129529] [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: 08/17/2022] [Accepted: 02/17/2023] [Indexed: 09/12/2023] Open
Abstract
Lipomas are usually sporadic, asymptomatic lesions, and their clinical and histologic presentation does not pose diagnostic difficulties. In ambiguous cases, however, knowledge of genetics is necessary. HMGA2 expression in adipose cells enables the differentiation of normal adipose tissue from lipoma and liposarcoma. Moreover, lipomas can be associated with genetic diseases, such as multiple endocrine neoplasia type 1, neurofibromatosis type 1, Wilson's disease, or mitochondrial diseases. Lipomas can run in families (familial multiple lipomatosis) or be a part of genetic syndromes such as PTEN hamartoma tumor syndrome, Proteus syndrome, and Pai syndrome. This study aims to present the genetic basis of lipomas and diseases in which these lesions occur in the clinical picture.
Collapse
Affiliation(s)
- Dorota Marzyńska
- Student’s Scientific Circle at the Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Ryszard Żaba
- Department of Dermatology and Venereology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Lacka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
5
|
Duro T, Gonzales KL. Adrenal Liposarcoma: A Novel Presentation of Multiple Endocrine Neoplasia Type 1. AACE Clin Case Rep 2022; 9:10-12. [PMID: 36654999 PMCID: PMC9837086 DOI: 10.1016/j.aace.2022.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022] Open
Abstract
Background/Objective Multiple endocrine neoplasia type 1 (MEN1) syndrome results from genetic sequence variations of the tumor suppressor MEN1 gene, which codes for the protein menin. Individuals with MEN1 are prone to developing multiple tumors involving the endocrine and nonendocrine organs. MEN1 associated with liposarcomas has not been documented previously. We highlight a case of MEN1 presenting with a metastatic adrenal liposarcoma. Case Report A 41-year-old Hispanic man with a history of nephrolithiasis and skin lesions presented to the emergency department with abdominal pain. He was found to have a right adrenal mass measuring 7.9 cm with extension into the liver and primary hyperparathyroidism. He had multiple paternal first-degree relatives with similar skin lesions, hypercalcemia, and tumors of the brain, thoracic cavity, abdomen, and thyroid. The mass was identified as a metastatic pleiomorphic adrenal liposarcoma on surgical pathology. Genetic testing revealed a germline pathogenic sequence variation of the MEN1 gene. Discussion Liposarcomas are rare malignant tumors with an annual incidence of 2.5 cases per 1 million. Although lipoma formation is a commonly described manifestation of MEN1, liposarcomas have not been associated with MEN1 previously. A potential mechanism of this association is through the role of menin in inducing adipocyte differentiation via peroxisome proliferator-activated receptor-γ activation, a highly expressed protein in liposarcomas. Conclusion Liposarcomas should be included in the differential of MEN1-related tumors.
Collapse
Affiliation(s)
- Teodor Duro
- Address correspondence to Dr Teodor Duro, University of New Mexico Health Sciences Center, Department of Endocrinology, MSC10-5550, 1 University of New Mexico, Albuquerque, NM 87131.
| | | |
Collapse
|
6
|
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
|
7
|
Gorbacheva A, Eremkina A, Goliusova D, Krupinova J, Mokrysheva N. The role of menin in bone pathology. Endocr Connect 2022; 11:EC-21-0494.R2. [PMID: 35148273 PMCID: PMC8942318 DOI: 10.1530/ec-21-0494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/11/2022] [Indexed: 12/02/2022]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is the most common cause of hereditary primary hyperparathyroidism (PHPT). Bone disorders are considered one of the key symptoms in PHPT present with the significant reduction in bone mineral density and low-energy fractures. Previously, these bone disorders were believed to be caused solely by the increase in the level of parathyroid hormone and its subsequent effect on bone resorption. The current paradigm, however, states that the mutations in the menin gene, which cause the development of MEN1, can also affect the metabolism of the cells of the osteoid lineage. This review analyzes both the proven and the potential intracellular mechanisms through which menin can affect bone metabolism.
Collapse
Affiliation(s)
- Anna Gorbacheva
- Endocrinology Research Center, Moscow, Russian Federation
- Correspondence should be addressed to A Gorbacheva:
| | - Anna Eremkina
- Endocrinology Research Center, Moscow, Russian Federation
| | | | | | | |
Collapse
|
8
|
Meister S, Hahn L, Beyer S, Paul C, Mitter S, Kuhn C, von Schönfeldt V, Corradini S, Sudan K, Schulz C, Kolben TM, Mahner S, Jeschke U, Kolben T. Regulation of Epigenetic Modifications in the Placenta during Preeclampsia: PPARγ Influences H3K4me3 and H3K9ac in Extravillous Trophoblast Cells. Int J Mol Sci 2021; 22:ijms222212469. [PMID: 34830351 PMCID: PMC8622744 DOI: 10.3390/ijms222212469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to analyze the expression of peroxisome proliferator-activated receptor γ (PPARγ) and retinoid X receptor α (RxRα), a binding heterodimer playing a pivotal role in the successful trophoblast invasion, in the placental tissue of preeclamptic patients. Furthermore, we aimed to characterize a possible interaction between PPARγ and H3K4me3 (trimethylated lysine 4 of the histone H3), respectively H3K9ac (acetylated lysine 9 of the histone H3), to illuminate the role of histone modifications in a defective trophoblast invasion in preeclampsia (PE). Therefore, the expression of PPARγ and RxRα was analyzed in 26 PE and 25 control placentas by immunohistochemical peroxidase staining, as well as the co-expression with H3K4me3 and H3K9ac by double immunofluorescence staining. Further, the effect of a specific PPARγ-agonist (Ciglitazone) and PPARγ-antagonist (T0070907) on the histone modifications H3K9ac and H3K4me3 was analyzed in vitro. In PE placentas, we found a reduced expression of PPARγ and RxRα and a reduced co-expression with H3K4me3 and H3K9ac in the extravillous trophoblast (EVT). Furthermore, with the PPARγ-antagonist treated human villous trophoblast (HVT) cells and primary isolated EVT cells showed higher levels of the histone modification proteins whereas treatment with the PPARγ-agonist reduced respective histone modifications. Our results show that the stimulation of PPARγ-activity leads to a reduction of H3K4me3 and H3K9ac in trophoblast cells, but paradoxically decreases the nuclear PPARγ expression. As the importance of PPARγ, being involved in a successful trophoblast invasion has already been investigated, our results reveal a pathophysiologic connection between PPARγ and the epigenetic modulation via H3K4me3 and H3K9ac in PE.
Collapse
Affiliation(s)
- Sarah Meister
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
- Correspondence: (S.M.); (U.J.); Tel.: +49-89-4400-54266 (S.M.); Fax: +49-89-4400-54916 (S.M.)
| | - Laura Hahn
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Susanne Beyer
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Corinna Paul
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Sophie Mitter
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Christina Kuhn
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156 Augsburg, Germany;
| | - Viktoria von Schönfeldt
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany;
| | - Kritika Sudan
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (K.S.); (C.S.)
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (K.S.); (C.S.)
| | - Theresa Maria Kolben
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Sven Mahner
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156 Augsburg, Germany;
- Correspondence: (S.M.); (U.J.); Tel.: +49-89-4400-54266 (S.M.); Fax: +49-89-4400-54916 (S.M.)
| | - Thomas Kolben
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (L.H.); (S.B.); (C.P.); (S.M.); (V.v.S.); (T.M.K.); (S.M.); (T.K.)
| |
Collapse
|
9
|
Involvement of the MEN1 Gene in Hormone-Related Cancers: Clues from Molecular Studies, Mouse Models, and Patient Investigations. ENDOCRINES 2020. [DOI: 10.3390/endocrines1020007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MEN1 mutation predisposes patients to multiple endocrine neoplasia type 1 (MEN1), a genetic syndrome associated with the predominant co-occurrence of endocrine tumors. Intriguingly, recent evidence has suggested that MEN1 could also be involved in the development of breast and prostate cancers, two major hormone-related cancers. The first clues as to its possible role arose from the identification of the physical and functional interactions between the menin protein, encoded by MEN1, and estrogen receptor α and androgen receptor. In parallel, our team observed that aged heterozygous Men1 mutant mice developed cancerous lesions in mammary glands of female and in the prostate of male mutant mice at low frequencies, in addition to endocrine tumors. Finally, observations made both in MEN1 patients and in sporadic breast and prostate cancers further confirmed the role played by menin in these two cancers. In this review, we present the currently available data concerning the complex and multifaceted involvement of MEN1 in these two types of hormone-dependent cancers.
Collapse
|
10
|
Mamedova EO, Dimitrova DA, Belaya ZE, Melnichenko GA. [The role of non-coding RNAs in the pathogenesis of multiple endocrine neoplasia syndrome type 1]. ACTA ACUST UNITED AC 2020; 66:4-12. [PMID: 33351343 DOI: 10.14341/probl12413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 06/15/2020] [Indexed: 01/03/2023]
Abstract
Changes in the expression of non-coding ribonucleic acids (ncRNAs) take part in the formation of various tumors. Multiple endocrine neoplasia syndrome type 1 (MEN1) is a rare autosomal dominant disease caused by mutations of the MEN1 gene encoding the menin protein. This syndrome is characterized by the occurrence of parathyroid tumors, gastroenteropancreatic neuroendocrine tumors, pituitary adenomas, as well as other endocrine and non-endocrine tumors. The pathogenesis of MEN-1 associated tumors due to MEN1 mutations remains unclear. In the absence of mutations of the MEN1 gene in patients with phenotypically similar features, this condition is regarded as a phenocopy of this syndrome. The cause of the combination of several MEN-1-related tumors in these patients remains unknown. The possible cause is that changes in the expression of ncRNAs affect the regulation of signaling pathways in which menin participates and may contribute to the development of MEN-1-related tumors. The identification of even a small number of agents interacting with menin makes a significant contribution to the improvement of knowledge about its pathophysiological influence and ways of developing tumors within the MEN-1 syndrome and its phenocopies.
Collapse
|
11
|
Park U, Hwang J, Youn H, Kim E, Um S. Piperine inhibits adipocyte differentiation via dynamic regulation of histone modifications. Phytother Res 2019; 33:2429-2439. [DOI: 10.1002/ptr.6434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 06/03/2019] [Accepted: 06/08/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Ui‐Hyun Park
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| | - Jin‐Taek Hwang
- Korea Food Research InstituteResearch Group of Healthcare 245 Nongsaengmyeong‐ro Jeonju Jeonbuk 55365 Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| | - Eun‐Joo Kim
- Department of Molecular BiologyDankook University Cheonan Chungnam 31116 Korea
| | - Soo‐Jong Um
- Department of Integrative Bioscience and BiotechnologySejong University 209 Neungdong‐ro, Gwangjin‐gu Seoul 05006 Korea
| |
Collapse
|
12
|
Enhancer-Mediated Oncogenic Function of the Menin Tumor Suppressor in Breast Cancer. Cell Rep 2017; 18:2359-2372. [PMID: 28273452 DOI: 10.1016/j.celrep.2017.02.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 12/17/2016] [Accepted: 02/07/2017] [Indexed: 12/12/2022] Open
Abstract
While the multiple endocrine neoplasia type 1 (MEN1) gene functions as a tumor suppressor in a variety of cancer types, we explored its oncogenic role in breast tumorigenesis. The MEN1 gene product menin is involved in H3K4 trimethylation and co-activates transcription. We integrated ChIP-seq and RNA-seq data to identify menin target genes. Our analysis revealed that menin-dependent target gene promoters display looping to distal enhancers that are bound by menin, FOXA1 and GATA3. In this fashion, MEN1 co-regulates a proliferative breast cancer-specific gene expression program in ER+ cells. In primary mammary cells, MEN1 exerts an anti-proliferative function by regulating a distinct expression signature. Our findings clarify the cell-type-specific functions of MEN1 and inform the development of menin-directed treatments for breast cancer.
Collapse
|
13
|
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
|
14
|
Marchand L, Decaussin-Petrucci M, Giraud S, Cotton F, Thivolet C, Simon C. Hibernoma and multiple endocrine neoplasia type 1 syndrome: A non-fortuitous association? A case report and literature review. ANNALES D'ENDOCRINOLOGIE 2017; 78:194-197. [PMID: 28478946 DOI: 10.1016/j.ando.2017.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/22/2017] [Accepted: 03/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Lucien Marchand
- Department of endocrinology and diabetes, Lyon-Sud hospital, hospices civils de Lyon, 165, chemin du Grand-Revoyet, 69310 Pierre-Bénite, France.
| | | | - Sophie Giraud
- Department of genetics, Edouard-Herriot hospital, hospices civils de Lyon, 69310 Pierre-Bénite, France.
| | - François Cotton
- Department of radiology, Lyon-Sud Hospital, hospices civils de Lyon, 69310 Pierre-Bénite, France.
| | - Charles Thivolet
- Department of endocrinology and diabetes, Lyon-Sud hospital, hospices civils de Lyon, 165, chemin du Grand-Revoyet, 69310 Pierre-Bénite, France.
| | - Chantal Simon
- Department of endocrinology and diabetes, Lyon-Sud hospital, hospices civils de Lyon, 165, chemin du Grand-Revoyet, 69310 Pierre-Bénite, France.
| |
Collapse
|
15
|
Menin regulates Inhbb expression through an Akt/Ezh2-mediated H3K27 histone modification. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:427-437. [DOI: 10.1016/j.bbagrm.2017.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/24/2017] [Accepted: 02/10/2017] [Indexed: 01/02/2023]
|
16
|
Liu P, Lee S, Knoll J, Rauch A, Ostermay S, Luther J, Malkusch N, Lerner UH, Zaiss MM, Neven M, Wittig R, Rauner M, David JP, Bertolino P, Zhang CX, Tuckermann JP. Loss of menin in osteoblast lineage affects osteocyte-osteoclast crosstalk causing osteoporosis. Cell Death Differ 2017; 24:672-682. [PMID: 28106886 PMCID: PMC5384024 DOI: 10.1038/cdd.2016.165] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/08/2016] [Accepted: 12/20/2016] [Indexed: 12/21/2022] Open
Abstract
During osteoporosis bone formation by osteoblasts is reduced and/or bone resorption by osteoclasts is enhanced. Currently, only a few factors have been identified in the regulation of bone integrity by osteoblast-derived osteocytes. In this study, we show that specific disruption of menin, encoded by multiple endocrine neoplasia type 1 (Men1), in osteoblasts and osteocytes caused osteoporosis despite the preservation of osteoblast differentiation and the bone formation rate. Instead, an increase in osteoclast numbers and bone resorption was detected that persisted even when the deletion of Men1 was restricted to osteocytes. We demonstrate that isolated Men1-deficient osteocytes expressed numerous soluble mediators, such as C-X-C motif chemokine 10 (CXCL10), and that CXCL10-mediated osteoclastogenesis was reduced by CXCL10-neutralizing antibodies. Collectively, our data reveal a novel role for Men1 in osteocyte–osteoclast crosstalk by controlling osteoclastogenesis through the action of soluble factors. A role for Men1 in maintaining bone integrity and thereby preventing osteoporosis is proposed.
Collapse
Affiliation(s)
- Peng Liu
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm D-89081, Germany.,Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| | - Sooyeon Lee
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm D-89081, Germany.,Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| | - Jeanette Knoll
- Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| | - Alexander Rauch
- Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| | - Susanne Ostermay
- Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| | - Julia Luther
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Nicole Malkusch
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm D-89081, Germany
| | - Ulf H Lerner
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition at Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg SE-41345, Sweden
| | - Mario M Zaiss
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen D-91054, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Rainer Wittig
- Institute for Laser Technologies in Medicine and Metrology at Ulm University, Ulm D-89081, Germany
| | - Martina Rauner
- Division of Endocrinology and Bone Diseases, Department of Medicine III, TU Dresden, Dresden D-01307, Germany
| | - Jean-Pierre David
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen D-91054, Germany
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR5286, Université Lyon 1, Lyon F-69000, France
| | - Chang X Zhang
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR5286, Université Lyon 1, Lyon F-69000, France
| | - Jan P Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm D-89081, Germany.,Tissue-specific Hormone Action, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena D-07745, Germany
| |
Collapse
|
17
|
Son MJ, Kim WK, Oh KJ, Park A, Lee DS, Han BS, Lee SC, Bae KH. Methyltransferase and demethylase profiling studies during brown adipocyte differentiation. BMB Rep 2017; 49:388-93. [PMID: 27157542 PMCID: PMC5032007 DOI: 10.5483/bmbrep.2016.49.7.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 02/03/2023] Open
Abstract
Although brown adipose tissue is important with regard to energy balance, the molecular mechanism of brown adipocyte differentiation has not been extensively studied. Specifically, regulation factors at the level of protein modification are largely unknown. In this study, we examine the changes in the expression level of enzymes which are involved in protein lysine methylation during brown adipocyte differentiation. Several enzymes, in this case SUV420H2, PRDM9, MLL3 and JHDM1D, were found to be up-regulated. On the other hand, Set7/9 was significantly down-regulated. In the case of SUV420H2, the expression level increased sharply during brown adipocyte differentiation, whereas the expression of SUV420H2 was marginally enhanced during the white adipocyte differentiation. The knock-down of SUV420H2 caused the suppression of brown adipocyte differentiation, as compared to a scrambled control. These results suggest that SUV420H2, a methyltransferase, is involved in brown adipocyte differentiation, and that the methylation of protein lysine is important in brown adipocyte differentiation. [BMB Reports 2016; 49(7): 388-393]
Collapse
Affiliation(s)
- Min Jeong Son
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Anna Park
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Da Som Lee
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Baek Soo Han
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| |
Collapse
|
18
|
Liu L, Lei I, Karatas H, Li Y, Wang L, Gnatovskiy L, Dou Y, Wang S, Qian L, Wang Z. Targeting Mll1 H3K4 methyltransferase activity to guide cardiac lineage specific reprogramming of fibroblasts. Cell Discov 2016; 2:16036. [PMID: 27924221 PMCID: PMC5113048 DOI: 10.1038/celldisc.2016.36] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers a great opportunity for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate. To address this issue, we attempted to identify small molecules that could potentiate the reprogramming ability towards cardiac fate by removing inhibitory roadblocks. Using mouse embryonic fibroblasts as the starting cell source, we first screened 47 cardiac development related epigenetic and transcription factors, and identified an unexpected role of H3K4 methyltransferase Mll1 and related factor Men1 in inhibiting iCM reprogramming. We then applied small molecules (MM408 and MI503) of Mll1 pathway inhibitors and observed an improved efficiency in converting embryonic fibroblasts and cardiac fibroblasts into functional cardiomyocyte-like cells. We further observed that these inhibitors directly suppressed the expression of Mll1 target gene Ebf1 involved in adipocyte differentiation. Consequently, Mll1 inhibition significantly decreased the formation of adipocytes during iCM induction. Therefore, Mll1 inhibitors likely increased iCM efficiency by suppressing alternative lineage gene expression. Our studies show that targeting Mll1 dependent H3K4 methyltransferase activity provides specificity in the process of cardiac reprogramming. These findings shed new light on the molecular mechanisms underlying cardiac conversion of fibroblasts and provide novel targets and small molecules to improve iCM reprogramming for clinical applications.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI, USA; Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hacer Karatas
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yangbing Li
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid Gnatovskiy
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Yali Dou
- Department of Pathology, University of Michigan Medical School, 1301 Catherine , Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
19
|
Cheng P, Li G, Yang SS, Liu R, Jin G, Zhou XY, Hu XG. Tumor suppressor Menin acts as a corepressor of LXRα to inhibit hepatic lipogenesis. FEBS Lett 2015; 589:3079-84. [PMID: 25962847 DOI: 10.1016/j.febslet.2015.04.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 04/03/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023]
Abstract
Menin, encoded by the MEN1 gene, was initially identified as a tumor suppressor for endocrine neoplasia. Our previous report showed that Menin enhances PPARα transactivity preventing triglyceride accumulation in the liver. Here, we further explore the role of Menin in liver steatosis. Transient transfection assays demonstrate that Menin inhibits the transcriptional activity of nuclear receptor liver X receptor α (LXRα). Accordingly, Menin overexpression results in reduced expression of LXRα target genes, such as lipogenic enzymes including SREBP-1c, FASN and SCD-1. Co-immunoprecipitation assays revealed physical interaction between Menin and LXRα. Collectively, our data suggest that Menin acts as a novel corepressor of LXRα and functions as a negative regulator of hepatic lipogenesis.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Gang Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Sheng Sheng Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Rui Liu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Xu Yu Zhou
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Xian Gui Hu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| |
Collapse
|
20
|
Parekh VI, Modali SD, Desai SS, Agarwal SK. Consequence of Menin Deficiency in Mouse Adipocytes Derived by In Vitro Differentiation. Int J Endocrinol 2015; 2015:149826. [PMID: 26229531 PMCID: PMC4503551 DOI: 10.1155/2015/149826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 12/20/2022] Open
Abstract
Lipoma in patients with the multiple endocrine neoplasia type 1 (MEN1) syndrome is a type of benign fat-cell tumor that has biallelic inactivation of MEN1 that encodes menin and could serve as a model to investigate normal and pathologic fat-cell (adipocyte) proliferation and function. The role of menin and its target genes in adipocytes is not known. We used in vitro differentiation to derive matched normal and menin-deficient adipocytes from wild type (WT) and menin-null (Men1-KO) mouse embryonic stem cells (mESCs), respectively, or 3T3-L1 cells without or with menin knockdown to investigate cell size, lipid content, and gene expression changes. Adipocytes derived from Men1-KO mESCs or after menin knockdown in 3T3-L1 cells showed a 1.5-1.7-fold increase in fat-cell size. Global gene expression analysis of mESC-derived adipocytes showed that lack of menin downregulated the expression of many differentially methylated genes including the tumor suppressor long noncoding RNA Meg3 but upregulated gene expression from the prolactin gene family locus. Our results show that menin deficiency leads to fat-cell hypertrophy and provide model systems that could be used to study the regulation of fat-cell size.
Collapse
Affiliation(s)
- Vaishali I. Parekh
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sita D. Modali
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shruti S. Desai
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sunita K. Agarwal
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- *Sunita K. Agarwal:
| |
Collapse
|
21
|
Abstract
Endocrine tumors may present as sporadic events or as part of familial endocrine syndromes. Familial endocrine syndromes (or inherited tumor/neoplasm syndromes) are characterized by multiple tumors in multiple organs. Some morphologic findings in endocrine tumor histopathology may prompt the possibility of familial endocrine syndromes, and these recognized histologic features may lead to further molecular genetic evaluation of the patient and family members. Subsequent evaluation for these syndromes in asymptomatic patients and family members may then be performed by genetic screening.
Collapse
Affiliation(s)
- Peter M Sadow
- Pathology Service, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | | | - Vania Nosé
- Pathology Service, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
MEN1 Syndrome and Hibernoma: An Uncommonly Recognised Association? Case Rep Med 2014; 2014:804580. [PMID: 25309600 PMCID: PMC4189526 DOI: 10.1155/2014/804580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/07/2014] [Indexed: 12/29/2022] Open
Abstract
MEN1 syndrome is known to classically result in parathyroid, pituitary, and pancreatic islet cell tumours. However, the potential association of MEN1 syndrome with hibernoma, a benign tumour with differentiation towards brown fat, is far less well known, despite their genetic profile both being linked to deletion of the MEN1 gene. Herein, we describe a case with its key radiological and pathological findings.
Collapse
|
23
|
Shu G, Lu NS, Zhu XT, Xu Y, Du MQ, Xie QP, Zhu CJ, Xu Q, Wang SB, Wang LN, Gao P, Xi QY, Zhang YL, Jiang QY. Phloretin promotes adipocyte differentiation in vitro and improves glucose homeostasis in vivo. J Nutr Biochem 2014; 25:1296-308. [PMID: 25283330 DOI: 10.1016/j.jnutbio.2014.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 07/03/2014] [Accepted: 07/21/2014] [Indexed: 10/24/2022]
Abstract
Adipocyte dysfunction is associated with many metabolic diseases such as obesity, insulin resistance and diabetes. Previous studies found that phloretin promotes 3T3-L1 cells differentiation, but the underlying mechanisms for phloretin's effects on adipogenesis remain unclear. In this study, we demonstrated that phloretin enhanced the lipid accumulation in porcine primary adipocytes in a time-dependent manner. Furthermore, phloretin increased the utilization of glucose and nonesterified fatty acid, while it decreased the lactate output. Microarray analysis revealed that genes associated with peroxisome proliferator-activated receptor-γ (PPARγ), mitogen-activated protein kinase and insulin signaling pathways were altered in response to phloretin. We further confirmed that phloretin enhanced expression of PPARγ, CAAT enhancer binding protein-α (C/EBPα) and adipose-related genes, such as fatty acids translocase and fatty acid synthase. In addition, phloretin activated the Akt (Thr308) and extracellular signal-regulated kinase, and therefore, inactivated Akt targets protein. Wortmannin effectively blocked the effect of phloretin on Akt activity and the protein levels of PPARγ, C/EBPα and fatty acid binding protein-4 (FABP4/aP2). Oral administration of 5 or 10 mg/kg phloretin to C57BL BKS-DB mice significantly decreased the serum glucose level and improved glucose tolerance. In conclusion, phloretin promotes the adipogenesis of porcine primary preadipocytes through Akt-associated signaling pathway. These findings suggested that phloretin might be able to increase insulin sensitivity and alleviate the metabolic diseases.
Collapse
Affiliation(s)
- Gang Shu
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Nai-Sheng Lu
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China; Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Xiao-Tong Zhu
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Rm 8070, Houston, TX 77030, USA
| | - Min-Qing Du
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Qiu-Ping Xie
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Can-Jun Zhu
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Qi Xu
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Song-Bo Wang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Li-Na Wang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Ping Gao
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Qian-Yun Xi
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Yong-Liang Zhang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China
| | - Qing-Yan Jiang
- College of Animal Science, South China Agricultural University, Guangzhou 510640, China; National Engineering Research Center For Breeding Swine Industry, Guangzhou, China.
| |
Collapse
|
24
|
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: 61] [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
|
25
|
Gurung B, Muhammad AB, Hua X. Menin is required for optimal processing of the microRNA let-7a. J Biol Chem 2014; 289:9902-8. [PMID: 24563463 DOI: 10.1074/jbc.m113.520692] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple endocrine neoplasia type I (MEN1) is an inherited syndrome that includes susceptibility to pancreatic islet hyperplasia. This syndrome results from mutations in the MEN1 gene, which encodes menin protein. Menin interacts with several transcription factors, including JunD, and inhibits their activities. However, the precise mechanism by which menin suppresses gene expression is not well understood. Here, we show that menin interacts with arsenite-resistant protein 2 (ARS2), a component of the nuclear RNA CAP-binding complex that is crucial for biogenesis of certain miRNAs including let-7a. The levels of primary-let-7a (pri-let-7a) are not affected by menin; however, the levels of mature let-7a are substantially decreased upon Men1 excision. Let-7a targets, including Insr and Irs2, pro-proliferative genes that are crucial for insulin-mediated signaling, are up-regulated in Men1-excised cells. Inhibition of let-7a using anti-miRNA in wild type cells is sufficient to enhance the expression of insulin receptor substrate 2 (IRS2) to levels observed in Men1-excised cells. Depletion of menin does not affect the expression of Drosha and CBP80, but substantially impairs the processing of pri-miRNA to pre-miRNA. Ars2 knockdown decreased let-7a processing in menin-expressing cells but had little impact on let-7a levels in menin-excised cells. As IRS2 is known to mediate insulin signaling and insulin/mitogen-induced cell proliferation, these findings collectively unravel a novel mechanism whereby menin suppresses cell proliferation, at least partly by promoting the processing of certain miRNAs, including let-7a, leading to suppression of Irs2 expression and insulin signaling.
Collapse
Affiliation(s)
- Buddha Gurung
- From the Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | | |
Collapse
|
26
|
Hepatic menin recruits SIRT1 to control liver steatosis through histone deacetylation. J Hepatol 2013; 59:1299-306. [PMID: 23867312 DOI: 10.1016/j.jhep.2013.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 07/02/2013] [Accepted: 07/05/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS The development and progression of non-alcoholic fatty liver disease are associated with aging, obesity, and type 2 diabetes. Understanding the precise regulatory networks of this process will contribute to novel therapeutic strategies. METHODS Hepatocyte-specific Men1 knockout mice were generated using Cre/loxP technology. Lipid and glucose metabolic phenotypes and mechanisms were investigated in aging and high-fat diet fed mice. RESULTS The expression of menin, encoded by multiple endocrine neoplasia 1 (Men1) gene, is reduced in the liver of aging mice. Hepatocyte-specific deletion of Men1 induces liver steatosis in aging mice. Menin deficiency promotes high-fat diet-induced liver steatosis in mice. Menin recruits SIRT1 to control hepatic CD36 expression and triglyceride accumulation through histone deacetylation. CONCLUSIONS Our work reveals that the adaptor protein menin is critical for the progression of hepatic steatosis during aging and metabolic imbalance.
Collapse
|
27
|
Santin JR, Daufenback Machado I, Rodrigues SFP, Teixeira S, Muscará MN, Lins Galdino S, da Rocha Pitta I, Farsky SHP. Role of an indole-thiazolidine molecule PPAR pan-agonist and COX inhibitor on inflammation and microcirculatory damage in acute gastric lesions. PLoS One 2013; 8:e76894. [PMID: 24124600 PMCID: PMC3790743 DOI: 10.1371/journal.pone.0076894] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/27/2013] [Indexed: 01/26/2023] Open
Abstract
The present study aimed to show the in vivo mechanisms of action of an indole-thiazolidine molecule peroxisome-proliferator activated receptor pan-agonist (PPAR pan) and cyclooxygenase (COX) inhibitor, LYSO-7, in an ethanol/HCl-induced (Et/HCl) gastric lesion model. Swiss male mice were treated with vehicle, LYSO-7 or Bezafibrate (p.o.) 1 hour before oral administration of Et/HCl (60%/0.03M). In another set of assays, animals were injected i.p. with an anti-granulocyte antibody, GW9962 or L-NG-nitroarginine methyl ester (L-NAME) before treatment. One hour after Et/HCl administration, neutrophils were quantified in the blood and bone marrow and the gastric microcirculatory network was studied in situ. The gastric tissue was used to quantify the percentage of damaged area, as well as myeloperoxidase (MPO), inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS) protein and PPARγ protein and gene expression. Acid secretion was evaluated by the pylorus ligation model. LYSO-7 or Bezafibrate treatment reduced the necrotic area. LYSO-7 treatment enhanced PPARγ gene and protein expression in the stomach, and impaired local neutrophil influx and stasis of the microcirculatory network caused by Et/HCl administration. The effect seemed to be due to PPARγ agonist activity, as the LYSO-7 effect was abolished in GW9962 pre-treated mice. The reversal of microcirculatory stasis, but not neutrophil influx, was mediated by nitric oxide (NO), as L-NAME pre-treatment abolished the LYSO-7-mediated reestablishment of microcirculatory blood flow. This effect may depend on enhanced eNOS protein expression in injured gastric tissue. The pH and concentration of H+ in the stomach were not modified by LYSO-7 treatment. In addition, LYSO-7 may induce less toxicity, as 28 days of oral treatment did not induce weight loss, as detected in pioglitazone treated mice. Thus, we show that LYSO-7 may be an effective treatment for gastric lesions by controlling neutrophil influx and microcirculatory blood flow mediated by NO.
Collapse
Affiliation(s)
- José Roberto Santin
- Laboratory of Experimental Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Isabel Daufenback Machado
- Laboratory of Experimental Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Stephen F. P. Rodrigues
- Laboratory of Experimental Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Simone Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suely Lins Galdino
- Department of Chemistry, Federal University of Pernambuco, Pernabumbuco, Recife, Brazil
| | - Ivan da Rocha Pitta
- Department of Chemistry, Federal University of Pernambuco, Pernabumbuco, Recife, Brazil
| | - Sandra H. P. Farsky
- Laboratory of Experimental Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
28
|
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: 175] [Impact Index Per Article: 14.6] [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
|
29
|
Kim TH, Kim MY, Jo SH, Park JM, Ahn YH. Modulation of the transcriptional activity of peroxisome proliferator-activated receptor gamma by protein-protein interactions and post-translational modifications. Yonsei Med J 2013; 54:545-59. [PMID: 23549795 PMCID: PMC3635639 DOI: 10.3349/ymj.2013.54.3.545] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a nuclear receptor superfamily; members of which play key roles in the control of body metabolism principally by acting on adipose tissue. Ligands of PPARγ, such as thiazolidinediones, are widely used in the treatment of metabolic syndromes and type 2 diabetes mellitus (T2DM). Although these drugs have potential benefits in the treatment of T2DM, they also cause unwanted side effects. Thus, understanding the molecular mechanisms governing the transcriptional activity of PPARγ is of prime importance in the development of new selective drugs or drugs with fewer side effects. Recent advancements in molecular biology have made it possible to obtain a deeper understanding of the role of PPARγ in body homeostasis. The transcriptional activity of PPARγ is subject to regulation either by interacting proteins or by modification of the protein itself. New interacting partners of PPARγ with new functions are being unveiled. In addition, post-translational modification by various cellular signals contributes to fine-tuning of the transcriptional activities of PPARγ. In this review, we will summarize recent advancements in our understanding of the post-translational modifications of, and proteins interacting with, PPARγ, both of which affect its transcriptional activities in relation to adipogenesis.
Collapse
Affiliation(s)
- Tae-Hyun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-Young Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Seong-Ho Jo
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Man Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Ho Ahn
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Karrouz W, Kamoun M, Odou MF, Pigny P, Caiazzo R, Pattou F, Leteurtre E, Wémeau JL, Vantyghem MC. Hibernoma and type 1 multiple endocrine neoplasia (MEN1)? A metabolic link? ANNALES D'ENDOCRINOLOGIE 2013. [DOI: 10.1016/j.ando.2013.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
31
|
Bhan A, Hussain I, Ansari KI, Kasiri S, Bashyal A, Mandal SS. Antisense transcript long noncoding RNA (lncRNA) HOTAIR is transcriptionally induced by estradiol. J Mol Biol 2013; 425:3707-22. [PMID: 23375982 DOI: 10.1016/j.jmb.2013.01.022] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/24/2012] [Accepted: 01/17/2013] [Indexed: 12/25/2022]
Abstract
HOTAIR (HOX antisense intergenic RNA) is a long noncoding RNA (lncRNA) that is transcribed from the antisense strand of homeobox C gene locus in chromosome 12. HOTAIR coordinates with chromatin-modifying enzymes and regulates gene silencing. It is overexpressed in various carcinomas including breast cancer. Herein, we demonstrated that HOTAIR is crucial for cell growth and viability and its knockdown induced apoptosis in breast cancer cells. We also demonstrated that HOTAIR is transcriptionally induced by estradiol (E2). Its promoter contains multiple functional estrogen response elements (EREs). Estrogen receptors (ERs) along with various ER coregulators such as histone methylases MLL1 (mixed lineage leukemia 1) and MLL3 and CREB-binding protein/p300 bind to the promoter of HOTAIR in an E2-dependent manner. Level of histone H3 lysine-4 trimethylation, histone acetylation, and RNA polymerase II recruitment is enriched at the HOTAIR promoter in the presence of E2. Knockdown of ERs and MLLs downregulated the E2-induced HOTAIR expression. Thus, similar to protein-coding gene transcription, E2-induced transcription of antisense transcript HOTAIR is coordinated via ERs and ER coregulators, and this mechanism of HOTAIR overexpression potentially contributes towards breast cancer progression.
Collapse
Affiliation(s)
- Arunoday Bhan
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Menin, a product of the MEN1 gene, is related to the ontogeny of several cancers such as MEN1 and sporadic endocrine tumors, although it is considered to be a tumor suppressor. Many proteins interact with menin, and it is involved in various biological functions in several tissues. Menin plays some physiological and pathological roles related to transforming growth factor-beta (TGF-β) signaling pathway in the parathyroid, and it is implicated in the tumorigenesis of parathyroid tumors. In bone, the bone phenotype was observed in some menin-deleted mice. Menin is considered to support BMP-2- and Runx2-induced differentiation of mesenchymal cells into osteoblasts by interacting with Smad1/5, Runx2, β-catenin and LEF-1, although it has different effects on osteoblasts at later differentiation stages through TGF-β-Smad3 and AP-1 pathways. Further research is expected to shed more light on the role of menin in bone.
Collapse
Affiliation(s)
- Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan.
| |
Collapse
|
33
|
|
34
|
Araújo-Vilar D, Victoria B, González-Méndez B, Barreiro F, Fernández-Rodríguez B, Cereijo R, Gallego-Escuredo JM, Villarroya F, Pañeda-Menéndez A. Histological and molecular features of lipomatous and nonlipomatous adipose tissue in familial partial lipodystrophy caused by LMNA mutations. Clin Endocrinol (Oxf) 2012; 76:816-24. [PMID: 21883346 DOI: 10.1111/j.1365-2265.2011.04208.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Type 2 familial partial lipodystrophy (FPLD2) is a rare adipose tissue (AT) disease caused by mutations in LMNA, in which lipomas appear occasionally. In this study, we aimed to histologically characterize FPLD2-associated lipomatosis and study the expression of genes and proteins involved in cell cycle control, mitochondrial function, inflammation and adipogenesis. DESIGN AND PATIENTS One lipoma and perilipoma fat from each of four subjects with FPLD2 and 10 control subjects were analysed by optical microscopy. The presence of inflammatory cells was evaluated by immunohistochemistry. Real-time RT-PCR and Western blot were used to evaluate gene and protein levels. RESULTS Adipocytes from lipodystrophic patients were significantly larger than those of controls, in both the lipomas and perilipoma fat. Lipodystrophic AT exhibited CD68(+) macrophages and CD3(+) lymphocytes infiltration. TP53 expression was reduced in all types of lipomas. At protein level, C/EBPβ, p53 and pRb were severely disturbed in both lipodystrophic lipomas and perilipoma fat coming from lipoatrophic areas, whereas the expression of CEBPα was normal. Mitochondrial function genes were less expressed in lipoatrophic fat. In both lipomas and perilipoma fat from lipoatrophic areas, the expression of adipogenes was lower than controls. CONCLUSIONS Even in lipomas, the adipogenic machinery is impaired in lipodystrophic fat coming from lipoatrophic regions in FPLD2, although the histological phenotype is near-normal, exhibiting low-grade inflammatory features. Our results suggest that the p53 pathway and some adipogenic proteins, such as CEBPα, could contribute to the maintenance of this near normal phenotype in the remnant AT present in these patients.
Collapse
Affiliation(s)
- D Araújo-Vilar
- Thyroid and Metabolic Diseases Unit (U.E.T.eM.), Department of Medicine, University of Santiago de Compostela, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
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
|
36
|
Vermeulen M, Timmers HTM. Grasping trimethylation of histone H3 at lysine 4. Epigenomics 2012; 2:395-406. [PMID: 22121900 DOI: 10.2217/epi.10.11] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Post-translational modifications of chromatin have become a 'booming' area of biomedical research. One particularly interesting modification that is important for eukaryotic gene expression is trimethylation of histone H3 lysine 4 (H3K4me3), which is almost exclusively associated with active promoters of RNA polymerase II. In this article, we highlight the recent progress related to the biochemistry and biology of this histone mark, including its relevant 'writers' and 'readers'. We also outline the complex regulatory mechanisms that are involved in establishing H3K4me3 in health and disease. Further understanding of H3K4me3 regulation will offer both more insight into chromatin-based mechanisms of gene regulation and provide opportunities for epigenetic intervention of the diseased state.
Collapse
Affiliation(s)
- Michiel Vermeulen
- Department of Physiological Chemistry, University Medical Centre Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | |
Collapse
|
37
|
Anderson AM, Carter KW, Anderson D, Wise MJ. Coexpression of nuclear receptors and histone methylation modifying genes in the testis: implications for endocrine disruptor modes of action. PLoS One 2012; 7:e34158. [PMID: 22496781 PMCID: PMC3319570 DOI: 10.1371/journal.pone.0034158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/23/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Endocrine disruptor chemicals elicit adverse health effects by perturbing nuclear receptor signalling systems. It has been speculated that these compounds may also perturb epigenetic mechanisms and thus contribute to the early origin of adult onset disease. We hypothesised that histone methylation may be a component of the epigenome that is susceptible to perturbation. We used coexpression analysis of publicly available data to investigate the combinatorial actions of nuclear receptors and genes involved in histone methylation in normal testis and when faced with endocrine disruptor compounds. METHODOLOGY/PRINCIPAL FINDINGS The expression patterns of a set of genes were profiled across testis tissue in human, rat and mouse, plus control and exposed samples from four toxicity experiments in the rat. Our results indicate that histone methylation events are a more general component of nuclear receptor mediated transcriptional regulation in the testis than previously appreciated. Coexpression patterns support the role of a gatekeeper mechanism involving the histone methylation modifiers Kdm1, Prdm2, and Ehmt1 and indicate that this mechanism is a common determinant of transcriptional integrity for genes critical to diverse physiological endpoints relevant to endocrine disruption. Coexpression patterns following exposure to vinclozolin and dibutyl phthalate suggest that coactivity of the demethylase Kdm1 in particular warrants further investigation in relation to endocrine disruptor mode of action. CONCLUSIONS/SIGNIFICANCE This study provides proof of concept that a bioinformatics approach that profiles genes related to a specific hypothesis across multiple biological settings can provide powerful insight into coregulatory activity that would be difficult to discern at an individual experiment level or by traditional differential expression analysis methods.
Collapse
Affiliation(s)
- Alison M Anderson
- Computer Science and Software Engineering, University of Western Australia, Perth, Australia.
| | | | | | | |
Collapse
|
38
|
Auriemma LB, Shah S, Linden LM, Henriksen MA. Knockdown of menin affects pre-mRNA processing and promoter fidelity at the interferon-gamma inducible IRF1 gene. Epigenetics Chromatin 2012; 5:2. [PMID: 22240255 PMCID: PMC3271985 DOI: 10.1186/1756-8935-5-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 01/12/2012] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The tumor suppressor menin (MEN1) is mutated in the inherited disease multiple endocrine neoplasia type I, and has several documented cellular roles, including the activation and repression of transcription effected by several transcription factors. As an activator, MEN1 is a component of the Set1-like mixed lineage leukemia (MLL) MLL1/MLL2 methyltransferase complex that methylates histone H3 lysine 4 (H3K4). MEN1 is localized to the signal transducer and activator of transcription 1 (STAT1)-dependent gene, interferon regulatory factor 1 (IRF1), and is further recruited when IRF1 transcription is triggered by interferon-γ signaling. RESULTS RNAi-mediated knockdown of MEN1 alters the H3K4 dimethylation and H3 acetylation profiles, and the localization of histone deacetylase 3, at IRF1. While MEN1 knockdown does not impact the rate of transcription, IRF1 heteronuclear transcripts become enriched in MEN1-depleted cells. The processed mRNA and translated protein product are concomitantly reduced, and the antiviral state is attenuated. Additionally, the transcription start site at the IRF1 promoter is disrupted in the MEN1-depleted cells. The H3K4 demethylase, lysine specific demethylase 1, is also associated with IRF1, and its inhibition alters H3K4 methylation and disrupts the transcription start site as well. CONCLUSIONS Taken together, the data indicate that MEN1 contributes to STAT1-activated gene expression in a novel manner that includes defining the transcription start site and RNA processing.
Collapse
Affiliation(s)
- Lauren B Auriemma
- Department of Biology, The University of Virginia, 485 McCormick Road, Charlottesville, VA 22903, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
The Peutz-Jeghers syndrome (PJS) culprit kinase LKB1 phosphorylates and activates multiple intracellular kinases regulating cell metabolism and polarity. The relevance of each of these pathways is highly variable depending on the tissue type, but typically represents functions of differentiated cells. These include formation and maintenance of specialized cell compartments in nerve axons, swift refunneling of metabolites and restructuring of cell architecture in response to environmental cues in committed lymphocytes, and ensuring energy-efficient oxygen-based energy expenditure. Such features are often lost or reduced in cancer cells, and indeed LKB1 defects in PJS-associated and sporadic cancers and even the benign PJS polyps lead to differentiation defects, including expansion of partially differentiated epithelial cells in PJS polyps and epithelial-to-mesenchymal transition in carcinomas. This review focuses on the involvement of LKB1 in the differentiation of epithelial, mesenchymal, hematopoietic and germinal lineages.
Collapse
Affiliation(s)
- Lina Udd
- Institute of Biotechnology and Genome-Scale Biology Research Program, University of Helsinki, P.O. Box 56 (Biocenter 1), 00014, Helsinki, Finland
| | | |
Collapse
|
40
|
Kanungo J, Chandrasekharappa SC. Menin induces endodermal differentiation in aggregated P19 stem cells by modulating the retinoic acid receptors. Mol Cell Biochem 2012; 359:95-104. [PMID: 21833538 PMCID: PMC3412628 DOI: 10.1007/s11010-011-1003-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/19/2011] [Indexed: 10/17/2022]
Abstract
Menin, a ubiquitously expressed protein, is the product of the multiple endocrine neoplasia type I (Men1) gene, mutations of which cause tumors primarily of the parathyroid, endocrine pancreas, and anterior pituitary. Menin-null mice display early embryonic lethality, and thus imply a critical role for menin in early development. In this study, using the P19 embryonic carcinoma stem cells, we studied menin's role in cell differentiation. Menin expression is induced in P19 cell aggregates by retinoic acid (RA). Menin over-expressing stable clones proliferated in a significantly reduced rate compared to the empty vector harboring cells. RA induced cell death in aggregated menin over-expressing cells. However, in the absence of RA, specific populations of the aggregated menin over-expressing cells displayed the characteristic of an endodermal phenotype by the acquisition of cytokeratin Endo A expression (TROMA-1), a marker for the primitive endoderm, with a concomitant loss of the stem cell marker SSEA-1. Menin's ability to induce endodermal differentiation in specific populations of the aggregated cells in the absence of RA implied that menin could substitute RA by inducing a set of target genes that are RA responsive. Menin over-expressing cells upon aggregation showed a robust expression of RA receptors (RAR), RARα, β, and γ relative to the empty vector-harboring cells. Moreover, endodermal differentiation was inhibited by the pan-RAR antagonist Ro41-5253, suggesting that menin could induce endodermal differentiation of uncommitted cells by functionally modulating the RARs.
Collapse
Affiliation(s)
- Jyotshnabala Kanungo
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, 50 South Dr, Bldg 50, Room 5232, Bethesda, MD 20892, USA.
| | | |
Collapse
|
41
|
van Wijk JPH, Dreijerink KMA, Pieterman CRC, Lips CJM, Zelissen PMJ, Valk GD. Increased prevalence of impaired fasting glucose in MEN1 gene mutation carriers. Clin Endocrinol (Oxf) 2012; 76:67-71. [PMID: 21726266 DOI: 10.1111/j.1365-2265.2011.04166.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Multiple endocrine neoplasia type 1 (MEN1) is a hereditary syndrome characterized by parathyroid, gastroenteropancreatic, pituitary and adrenal tumours. Cardiovascular disease has been identified as an important cause of death in MEN1 patients. Menin, the product of the MEN1 gene, is a co-activator for peroxisome proliferator-activated receptor-γ and the vitamin D receptor, which are involved in glucose metabolism. We aimed to compare insulin sensitivity and prevalence of impaired fasting glucose and diabetes mellitus between MEN1 patients and controls. DESIGN Cross-sectional study. PATIENTS Sixty-three MEN1 gene mutation carriers (44% men, mean age 41 years) from 22 kindreds and 126 unrelated controls matched for gender, age and BMI. MEASUREMENTS Fasting glucose levels were categorized and compared using WHO criteria. Homeostasis model assessment (HOMA) was used as a measure of insulin resistance. RESULTS Homeostasis model assessment was significantly increased in MEN1 patients compared with controls (3·0 ± 2·0 vs 2·0 ± 1·0, P < 0·05). In MEN1 patients, HOMA was associated with BMI, but not with age, calcium and gastrin levels. Using logistic regression analysis, the presence of hyperparathyroidism, pancreatic lesions and various other manifestations was not associated with HOMA. Impaired fasting glucose was more prevalent in MEN1 compared with controls (17%vs 6%, P < 0·05). Three MEN1 patients (5%) compared with four controls (3%) were diabetic (not significant). CONCLUSIONS Multiple endocrine neoplasia type 1 patients had decreased insulin sensitivity and higher prevalence of impaired fasting glucose compared with controls, which was unrelated to MEN1 manifestations. Impaired glucose metabolism may result in increased risk of cardiovascular disease in MEN1 patients.
Collapse
Affiliation(s)
- J P H van Wijk
- Department of Internal Medicine and Endocrinology, University Medical Center, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Liu W, Lau F, Liu K, Wood HB, Zhou G, Chen Y, Li Y, Akiyama TE, Castriota G, Einstein M, Wang C, McCann ME, Doebber TW, Wu M, Chang CH, McNamara L, McKeever B, Mosley RT, Berger JP, Meinke PT. Benzimidazolones: a new class of selective peroxisome proliferator-activated receptor γ (PPARγ) modulators. J Med Chem 2011; 54:8541-54. [PMID: 22070604 DOI: 10.1021/jm201061j] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of benzimidazolone carboxylic acids and oxazolidinediones were designed and synthesized in search of selective PPARγ modulators (SPPARγMs) as potential therapeutic agents for the treatment of type II diabetes mellitus (T2DM) with improved safety profiles relative to rosiglitazone and pioglitazone, the currently marketed PPARγ full agonist drugs. Structure-activity relationships of these potent and highly selective SPPARγMs were studied with a focus on their unique profiles as partial agonists or modulators. A variety of methods, such as X-ray crystallographic analysis, PPARγ transactivation coactivator profiling, gene expression profiling, and mutagenesis studies, were employed to reveal the differential interactions of these new analogues with PPARγ receptor in comparison to full agonists. In rodent models of T2DM, benzimidazolone analogues such as (5R)-5-(3-{[3-(5-methoxybenzisoxazol-3-yl)benzimidazol-1-yl]methyl}phenyl)-5-methyloxazolidinedione (51) demonstrated efficacy equivalent to that of rosiglitazone. Side effects, such as fluid retention and heart weight gain associated with PPARγ full agonists, were diminished with 51 in comparison to rosiglitazone based on studies in two independent animal models.
Collapse
Affiliation(s)
- Weiguo Liu
- Merck Research Laboratories, P.O. Box 2000, Rahway, New Jersey 07065, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cheng P, Yang SS, Hu XG, Zhou XY, Zhang YJ, Jin G, Zhou YQ. Menin prevents liver steatosis through co-activation of peroxisome proliferator-activated receptor alpha. FEBS Lett 2011; 585:3403-8. [PMID: 22001208 DOI: 10.1016/j.febslet.2011.09.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 09/29/2011] [Accepted: 09/30/2011] [Indexed: 12/21/2022]
Abstract
Fatty liver is strongly associated with metabolic syndrome. Here, we show that the impaired hepatic expression of menin, the product of the MEN1 (multiple endocrine neoplasia type 1) tumor suppressor gene, represents a common feature of several fatty liver mouse models. The liver specific ablation of MEN1 gene expression in healthy mice induced hepatic steatosis under high-fat dietary conditions. Moreover, overexpression of menin in livers of steatotic db/db mice reduced liver triglyceride accumulation. At the molecular level, we found that menin acts synergistically with the nuclear receptor PPARα to control gene expression of fatty acid oxidation. Collectively, these data suggest a crucial role for menin as an integrator of the complex transcriptional network controlling hepatic steatosis.
Collapse
Affiliation(s)
- Peng Cheng
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
44
|
Alvelos MI, Mendes M, Soares P. Molecular alterations in sporadic primary hyperparathyroidism. GENETICS RESEARCH INTERNATIONAL 2011; 2011:275802. [PMID: 22567348 PMCID: PMC3335633 DOI: 10.4061/2011/275802] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 07/03/2011] [Accepted: 07/11/2011] [Indexed: 12/25/2022]
Abstract
Primary hyperparathyroidism (PHPT) is a frequent endocrine disorder
characterized by an excessive autonomous production and release of
parathyroid hormone (PTH) by the parathyroid glands. This
endocrinopathy may result from the development of a benign lesion
(adenoma or hyperplasia) or from a carcinoma. Most of the PHPT cases
occur sporadically; however, approximately 10% of the patients
present a familial form of the disease. The molecular mechanisms
underlying the pathogenesis of sporadic PHPT are incompletely
understood, even though somatic alterations in MEN1
gene and CCND1 protein overexpression are frequently observed. The
MEN1 gene is mutated in about 30% of the
parathyroid tumours and the protooncogene CCND1 is
implicated in parathyroid neoplasia by rearrangements, leading to an
overexpression of CCND1 protein in parathyroid cells. The aim of this
work is to briefly update the molecular alterations underlying
sporadic primary hyperparathyroidism.
Collapse
Affiliation(s)
- Maria Inês Alvelos
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-465 Porto, Portugal
| | | | | |
Collapse
|
45
|
Sugii S, Evans RM. Epigenetic codes of PPARγ in metabolic disease. FEBS Lett 2011; 585:2121-8. [PMID: 21605560 PMCID: PMC3129683 DOI: 10.1016/j.febslet.2011.05.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 04/29/2011] [Accepted: 05/02/2011] [Indexed: 01/03/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a ligand-regulated nuclear hormone receptor, plays critical roles in metabolism and adipogenesis. PPARγ ligands such as thiazolidinediones (TZDs) exert insulin sensitizing and anti-inflammatory effects primarily through action on adipocytes, and are thus widely used to treat metabolic syndrome, especially type II diabetes. A number of PPARγ interacting partners have been identified, many of which are known epigenetic regulators, including enzymes for histone acetylation/deacetylation and histone methylation/demethylation. However, their functional roles in the PPARγ transcriptional pathway are not well defined. Recent advances in ChIP-based and deep sequencing technology are revealing previously underappreciated epigenomic mechanisms and therapeutic potentials of this nuclear receptor pathway.
Collapse
Affiliation(s)
- Shigeki Sugii
- Howard Hughes Medical Institute and Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA 92037, USA
- Singapore Bioimaging Consortium and Duke-NUS Graduate Medical School, 11 Biopolis Way #02-02, Singapore 138667
| | - Ronald M. Evans
- Howard Hughes Medical Institute and Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA 92037, USA
- Corresponding author. Fax #: +1-858-455-1349.
| |
Collapse
|
46
|
Wu T, Hua X. Menin represses tumorigenesis via repressing cell proliferation. Am J Cancer Res 2011; 1:726-739. [PMID: 22016823 PMCID: PMC3195934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 05/08/2011] [Indexed: 05/31/2023] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) results from mutations in the tumor suppressor gene, MEN1, which encodes nuclear protein menin. Menin is important for suppressing tumorigenesis in various endocrine and certain non-endocrine tissues. Although menin suppresses MEN1 through a variety of mechanisms including regulating apoptosis and DNA repair, the role of menin in regulating cell proliferation is one of the best-studied functions. Here, we focus on reviewing various mechanisms underlying menin-mediated inhibition of cell proliferation. Menin inhibits cell proliferation to repress MEN1 through multiple mechanisms. 1) Menin interacts with various histonemodifying enzymes, such as MLL, EZH2 and HDACs, to affect gene transcription, leading to repression of cell proliferation. 2) Menin also interacts with various transcription factors, such as JunD, NF-κB, PPARγ and VDR, to induce or suppress gene transcription. As these various transcription factors are known to regulate cell proliferation, their interaction with menin may be relevant to menin's role in inhibiting cell proliferation. 3) Menin inhibits cell proliferation via TGF-β signaling and Wnt/β-catenin signaling pathways. 4) Menin represses certain pro-proliferative factors involved in endocrine tumors such as IGFBP-2, IGF2 and PTHrP to repress cell proliferation. 5) Menin affects cell cycle progression to inhibit cell proliferation. This review is helpful in our understanding of the comprehensive mechanisms whereby menin represses MEN1 through inhibiting cell proliferation.
Collapse
Affiliation(s)
- Ting Wu
- Department of Basic Medical Sciences, Medical College, Xiamen UniversityXiamen, China
| | - Xianxin Hua
- Abramson Family Cancer Research Institute, Department of Cancer Biology, the University of Pennsylvania, School of Medicine421 Curie Blvd., Philadelphia, PA 19104, USA
| |
Collapse
|
47
|
Concomitant deletions of tumor suppressor genes MEN1 and AIP are essential for the pathogenesis of the brown fat tumor hibernoma. Proc Natl Acad Sci U S A 2010; 107:21122-7. [PMID: 21078971 DOI: 10.1073/pnas.1013512107] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hibernomas are benign tumors with morphological features resembling brown fat. They consistently display cytogenetic rearrangements, typically translocations, involving chromosome band 11q13. Here we demonstrate that these aberrations are associated with concomitant deletions of AIP and MEN1, tumor suppressor genes that are located 3 Mb apart and that underlie the hereditary syndromes pituitary adenoma predisposition and multiple endocrine neoplasia type I. MEN1 and AIP displayed a low expression in hibernomas whereas the expression of genes up-regulated in brown fat--PPARA, PPARG, PPARGC1A, and UCP1--was high. Thus, loss of MEN1 and AIP is likely to be pathogenetically essential for hibernoma development. Simultaneous loss of two tumor suppressor genes has not previously been shown to result from a neoplasia-associated translocation. Furthermore, in contrast to the prevailing assumption that benign tumors harbor relatively few genetic aberrations, the present analyses demonstrate that a considerable number of chromosome breaks are involved in the pathogenesis of hibernoma.
Collapse
|
48
|
Varier RA, Timmers HTM. Histone lysine methylation and demethylation pathways in cancer. Biochim Biophys Acta Rev Cancer 2010; 1815:75-89. [PMID: 20951770 DOI: 10.1016/j.bbcan.2010.10.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/06/2010] [Accepted: 10/07/2010] [Indexed: 12/21/2022]
Abstract
The genetic changes leading to the development of human cancer are accompanied by alterations in the structure and modification status of chromatin, which represent powerful regulatory mechanisms for gene expression and genome stability. These epigenetic alterations have sparked interest into deciphering the regulatory pathways and function of post-translational modifications of histones during the initiation and progression of cancer. In this review we describe and summarize the current knowledge of several histone lysine methyltransferase and demethylase pathways relevant to cancer. Mechanistic insight into histone modifications will pave the way for the development and therapeutic application of "epidrugs" in cancer.
Collapse
Affiliation(s)
- Radhika A Varier
- Department of Physiological Chemistry, University Medical Centre, Utrecht, The Netherlands
| | | |
Collapse
|
49
|
Molecular Mechanisms and Genome-Wide Aspects of PPAR Subtype Specific Transactivation. PPAR Res 2010; 2010. [PMID: 20862367 PMCID: PMC2938449 DOI: 10.1155/2010/169506] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 06/27/2010] [Indexed: 12/13/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are central regulators of fat metabolism, energy homeostasis, proliferation, and inflammation. The three PPAR subtypes, PPARα, β/δ, and γ activate overlapping but also very different target gene programs. This review summarizes the insights into PPAR subtype-specific transactivation provided by genome-wide studies and discusses the recent advances in the understanding of the molecular mechanisms underlying PPAR subtype specificity with special focus on the regulatory role of AF-1.
Collapse
|
50
|
Seigne C, Fontanière S, Carreira C, Lu J, Tong WM, Fontanière B, Wang ZQ, Zhang CX, Frappart L. Characterisation of prostate cancer lesions in heterozygous Men1 mutant mice. BMC Cancer 2010; 10:395. [PMID: 20663219 PMCID: PMC2920881 DOI: 10.1186/1471-2407-10-395] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 07/27/2010] [Indexed: 01/14/2023] Open
Abstract
Background Mutations of the MEN1 gene predispose to multiple endocrine neoplasia type 1 (MEN1) syndrome. Our group and others have shown that Men1 disruption in mice recapitulates MEN1 pathology. Intriguingly, rare lesions in hormone-dependent tissues, such as prostate and mammary glands, were also observed in the Men1 mutant mice. Methods To study the occurrence of prostate lesions, we followed a male mouse cohort of 47 Men1+/- mice and 23 age-matched control littermates, starting at 18 months of age, and analysed the prostate glands from the cohort. Results Six Men1+/- mice (12.8%) developed prostate cancer, including two adenocarcinomas and four in situ carcinomas, while none of the control mice developed cancerous lesions. The expression of menin encoded by the Men1 gene was found to be drastically reduced in all carcinomas, and partial LOH of the wild-type Men1 allele was detected in three of the five analysed lesions. Using immunostaining for the androgen receptor and p63, a basal epithelial cell marker, we demonstrated that the menin-negative prostate cancer cells did not display p63 expression and that the androgen receptor was expressed but more heterogeneous in these lesions. Furthermore, our data showed that the expression of the cyclin-dependent kinase inhibitor CDKN1B (p27), a Men1 target gene known to be inactivated during prostate cell tumorigenesis, was notably decreased in the prostate cancers that developed in the mutant mice. Conclusion Our work suggests the possible involvement of Men1 inactivation in the tumorigenesis of the prostate gland.
Collapse
Affiliation(s)
- Christelle Seigne
- CNRS UMR5201, Laboratoire de Génétique Moléculaire, Signalisation et Cancer, Centre Léon Bérard, Lyon F-69008, France
| | | | | | | | | | | | | | | | | |
Collapse
|