1
|
Romanet P, Charnay T, Sahakian N, Cuny T, Castinetti F, Barlier A. Challenges in molecular diagnosis of multiple endocrine neoplasia. Front Endocrinol (Lausanne) 2024; 15:1445633. [PMID: 39398337 PMCID: PMC11466760 DOI: 10.3389/fendo.2024.1445633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 10/15/2024] Open
Abstract
Multiple endocrine neoplasia (MEN) is a group of rare genetic diseases characterized by the occurrence of multiple tumors of the endocrine system in the same patient. The first MEN described was MEN1, followed by MEN2A, and MEN2B. The identification of the genes responsible for these syndromes led to the introduction of family genetic screening programs. More than twenty years later, not all cases of MENs have been resolved from a genetic point of view, and new clinicogenetic entities have been described. In this review, we will discuss the strategies and difficulties of genetic screening for classic and newly described MENs in a clinical setting, from limitations in sequencing, to problems in classifying variants, to the identification of new candidate genes. In the era of genomic medicine, characterization of new candidate genes and their specific tumor risk is essential for inclusion of patients in personalized medicine programs as well as to permit accurate genetic counseling to be proposed for families.
Collapse
Affiliation(s)
- Pauline Romanet
- Aix Marseille Univ, APHM, INSERM, MMG, La Timone University Hospital, Laboratory of Molecular Biology GEnOPé, BIOGENOPOLE, Marseille, France
| | - Théo Charnay
- Aix Marseille Univ, APHM, INSERM, MMG, La Timone University Hospital, Laboratory of Molecular Biology GEnOPé, BIOGENOPOLE, Marseille, France
| | - Nicolas Sahakian
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Department of Endocrinology, Marseille, France
| | - Thomas Cuny
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Department of Endocrinology, Marseille, France
| | - Frédéric Castinetti
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Department of Endocrinology, Marseille, France
| | - Anne Barlier
- Aix Marseille Univ, APHM, INSERM, MMG, La Timone University Hospital, Laboratory of Molecular Biology GEnOPé, BIOGENOPOLE, Marseille, France
| |
Collapse
|
2
|
Boudreault J, Canaff L, Ghozlan M, Wang N, Guarnieri V, Salcuni AS, Scillitani A, Goltzman D, Ali S, Lebrun JJ. Multiple Endocrine Neoplasia Type 1 Regulates TGFβ-Mediated Suppression of Tumor Formation and Metastasis in Melanoma. Cells 2024; 13:973. [PMID: 38891107 PMCID: PMC11172218 DOI: 10.3390/cells13110973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Over the past few decades, the worldwide incidence of cutaneous melanoma, a malignant neoplasm arising from melanocytes, has been increasing markedly, leading to the highest rate of skin cancer-related deaths. While localized tumors are easily removed by excision surgery, late-stage metastatic melanomas are refractory to treatment and exhibit a poor prognosis. Consequently, unraveling the molecular mechanisms underlying melanoma tumorigenesis and metastasis is crucial for developing novel targeted therapies. We found that the multiple endocrine neoplasia type 1 (MEN1) gene product Menin is required for the transforming growth factor beta (TGFβ) signaling pathway to induce cell growth arrest and apoptosis in vitro and prevent tumorigenesis in vivo in preclinical xenograft models of melanoma. We further identified point mutations in two MEN1 family members affected by melanoma that led to proteasomal degradation of the MEN1 gene product and to a loss of TGFβ signaling. Interestingly, blocking the proteasome degradation pathway using an FDA-approved drug and RNAi targeting could efficiently restore MEN1 expression and TGFβ transcriptional responses. Together, these results provide new potential therapeutic strategies and patient stratification for the treatment of cutaneous melanoma.
Collapse
Affiliation(s)
- Julien Boudreault
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Lucie Canaff
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Mostafa Ghozlan
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Ni Wang
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Vito Guarnieri
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Antonio Stefano Salcuni
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia, 33100 Udine, Italy;
| | - Alfredo Scillitani
- Endocrinology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - David Goltzman
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Suhad Ali
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| | - Jean-Jacques Lebrun
- Cancer Research Program, Department of Medicine, Research Institute of McGill University Health Center, Montreal, QC H4A 3J1, Canada; (J.B.); (L.C.); (M.G.); (N.W.); (D.G.); (S.A.)
| |
Collapse
|
3
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa-SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [DOI: https:/doi.org/10.3390/biomedicines12020395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3−3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
4
|
Salgado I, Prado Montes de Oca E, Chairez I, Figueroa-Yáñez L, Pereira-Santana A, Rivera Chávez A, Velázquez-Fernandez JB, Alvarado Parra T, Vallejo A. Deep Learning Techniques to Characterize the RPS28P7 Pseudogene and the Metazoa- SRP Gene as Drug Potential Targets in Pancreatic Cancer Patients. Biomedicines 2024; 12:395. [PMID: 38397997 PMCID: PMC11154313 DOI: 10.3390/biomedicines12020395] [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: 10/20/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 02/25/2024] Open
Abstract
The molecular explanation about why some pancreatic cancer (PaCa) patients die early and others die later is poorly understood. This study aimed to discover potential novel markers and drug targets that could be useful to stratify and extend expected survival in prospective early-death patients. We deployed a deep learning algorithm and analyzed the gene copy number, gene expression, and protein expression data of death versus alive PaCa patients from the GDC cohort. The genes with higher relative amplification (copy number >4 times in the dead compared with the alive group) were EWSR1, FLT3, GPC3, HIF1A, HLF, and MEN1. The most highly up-regulated genes (>8.5-fold change) in the death group were RPL30, RPL37, RPS28P7, RPS11, Metazoa_SRP, CAPNS1, FN1, H3-3B, LCN2, and OAZ1. None of their corresponding proteins were up or down-regulated in the death group. The mRNA of the RPS28P7 pseudogene could act as ceRNA sponging the miRNA that was originally directed to the parental gene RPS28. We propose RPS28P7 mRNA as the most druggable target that can be modulated with small molecules or the RNA technology approach. These markers could be added as criteria to patient stratification in future PaCa drug trials, but further validation in the target populations is encouraged.
Collapse
Affiliation(s)
- Iván Salgado
- Medical Robotics and Biosignals Laboratory, Centro de Innovación y Desarrollo Tecnológico en Cómputo, Instituto Politécnico Nacional (IPN), Mexico City 07700, Mexico;
| | - Ernesto Prado Montes de Oca
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Isaac Chairez
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Jalisco, Mexico;
| | - Luis Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Alejandro Pereira-Santana
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico; (L.F.-Y.); (A.P.-S.)
| | - Andrés Rivera Chávez
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | | | - Teresa Alvarado Parra
- Regulatory SNPs Laboratory, Personalized Medicine National Laboratory (LAMPER), Guadalajara Unit, Medical and Pharmaceutical Biotechnology Department, Research Center in Technology and Design Assistance of Jalisco State (CIATEJ), National Council of Science and Technology (CONACYT), Guadalajara 44270, Jalisco, Mexico; (A.R.C.); (T.A.P.)
| | - Adriana Vallejo
- Unidad de Biotecnología Médica y Farmacéutica, CONACYT-Centro de Investigación y Asistencia en Tecnologia y Diseño del Estado de Jalisco AC, Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico
| |
Collapse
|
5
|
Ramachandran A, Le Quesne Stabej P, Boyle V, Elston MS, Pattison S, Lawrence B, Print C. Alternative splicing generates isoform diversity in MEN1. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2024; 4:e240014. [PMID: 39649118 PMCID: PMC11623284 DOI: 10.1530/eo-24-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/24/2024] [Indexed: 12/10/2024]
Abstract
Although the gene MEN1 has a long-standing association with cancer, its mechanisms of action remain incompletely understood, acting both as a tumour suppressor in neuroendocrine tumours and as an oncogene in leukaemia. The best-characterised isoform of the encoded protein, MENIN, is the 610-amino acid MENIN isoform 2. We hypothesise that some of the complexity of MEN1 biology can be attributed to a currently unappreciated contribution of different MENIN isoforms. Through in silico data mining, we show alternative splicing along the entire length of MEN1. Splice junction data suggest that the transcript encoding MENIN isoform 2 is the most abundant in all tissues examined, making a strong argument for this to be the reference transcript/protein isoform of MEN1. We also report novel splicing events, including a novel exon from within intron 7 that is relatively highly expressed in many tissues. These splicing events are predicted to contribute to MENIN diversity by generating isoforms with in-frame insertions, deletions or unique amino termini that, in turn, could have altered interactions with partner proteins. Finally, we have compiled 2574 unique genomic variants reported in MEN1 within somatic and germline databases and have identified several variants that could impact individual MENIN isoforms. We have also collated studies pertinent to MENIN function in the literature and summarised the impact of MEN1 variants on 74 biological variables. We propose a set of four MEN1 variants, MENINL22R, MENINH139D, MENINA242V and MENINW436R, that represent a cohort with different biological properties, which should be investigated concurrently to better dissect MENIN function.
Collapse
Affiliation(s)
- Anassuya Ramachandran
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Polona Le Quesne Stabej
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Veronica Boyle
- Endocrinology Unit, Te Whatu Ora Waikato, Hamilton, New Zealand
- School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Marianne S Elston
- School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sharon Pattison
- Te Whatu Ora Capital, Coast, and Hutt Valley, New Zealand
- Department of Pathology, Otago Medical School – Dunedin Campus, University of Otago, Dunedin, New Zealand
- Department of Oncology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ben Lawrence
- Department of Oncology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Medical Oncology, Te Toka Tumai, Health New Zealand, Auckland, New Zealand
| | - Cris Print
- Department of Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Miranda ISDM, Valadares LP, Barra GB, Mesquita PG, de Santana LB, de Castro LF, Rita THS, Naves LA. Clinical and molecular features of four Brazilian families with multiple endocrine neoplasia type 1. Front Endocrinol (Lausanne) 2023; 14:1117873. [PMID: 36967793 PMCID: PMC10036827 DOI: 10.3389/fendo.2023.1117873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Objective Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant syndrome characterized by its clinical variability and complexity in diagnosis and treatment. We performed both clinical and molecular descriptions of four families with MEN1 in a follow-up at a tertiary center in Brasília. Methods From a preliminary review of approximately 500 medical records of patients with pituitary neuroendocrine tumor (PitNET) from the database of the Neuroendocrinology Outpatient Clinic of the University Hospital of Brasília, a total of 135 patients met the criteria of at least two affected family members. From this cohort, we have identified 34 families: only four with a phenotype of MEN1 and the other 30 families with the phenotype of familial isolated pituitary adenoma (FIPA). Eleven patients with a clinical diagnosis of MEN1 from these four families were selected. Results Variants in MEN1 gene were identified in all families. One individual from each family underwent genetic testing using targeted high-throughput sequencing (HTS). All patients had primary hyperparathyroidism (PHPT), and the second most common manifestation was PitNET. One individual had well-differentiated liposarcoma, which has been previously reported in a single case of MEN1. Three variants previously described in the database and a novel variant in exon 2 have been found. Conclusions The study allowed the genotypic and phenotypic characterization of families with MEN1 in a follow-up at a tertiary center in Brasília.
Collapse
|
7
|
Zou J, Yu C, Zhang C, Guan Y, Zhang Y, Tolbert E, Zhang W, Zhao T, Bayliss G, Li X, Ye Z, Zhuang S. Inhibition of MLL1-menin interaction attenuates renal fibrosis in obstructive nephropathy. FASEB J 2023; 37:e22712. [PMID: 36527439 DOI: 10.1096/fj.202100634rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 11/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Mixed lineage leukemia 1 (MLL1), a histone H3 lysine 4 (H3K4) methyltransferase, exerts its enzymatic activity by interacting with menin and other proteins. It is unclear whether inhibition of the MLL1-menin interaction influences epithelial-mesenchymal transition (EMT), renal fibroblast activation, and renal fibrosis. In this study, we investigated the effect of disrupting MLL1-menin interaction on those events and mechanisms involved in a murine model of renal fibrosis induced by unilateral ureteral obstruction (UUO), in cultured mouse proximal tubular cells and renal interstitial fibroblasts. Injury to the kidney increased the expression of MLL1 and menin and H3K4 monomethylation (H3K4me1); MLL1 and menin were expressed in renal epithelial cells and renal interstitial fibroblasts. Inhibition of the MLL1-menin interaction by MI-503 administration or siRNA-mediated silencing of MLL1 attenuated UUO-induced renal fibrosis, and reduced expression of α-smooth muscle actin (α-SMA) and fibronectin. These treatments also inhibited UUO-induced expression of transcription factors Snail and Twist and transforming growth factor β1 (TGF-β1) while expression of E-cadherin was preserved. Moreover, treatment with MI-503 and transfection with either MLL siRNA or menin siRNA inhibited TGF-β1-induced upregulation of α-SMA, fibronectin and Snail, phosphorylation of Smad3 and AKT, and downregulation of E-cadherin in cultured renal epithelial cells. Finally, MI-503 was effective in abrogating serum or TGFβ1-induced transformation of renal interstitial fibroblasts to myofibroblasts in vitro. Taken together, these results suggest that targeting disruption of the MLL1-menin interaction attenuates renal fibrosis through inhibition of partial EMT and renal fibroblast activation.
Collapse
Affiliation(s)
- Jianan Zou
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Chao Yu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunyun Zhang
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Yingjie Guan
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Yunhe Zhang
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Evelyn Tolbert
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Wei Zhang
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Ting Zhao
- Department of Surgery, Rhode Island Hospital, Brown University, Providence, Rhode Island, USA
| | - George Bayliss
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Xiaogang Li
- Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Troka I, Griffanti G, Canaff L, Hendy GN, Goltzman D, Nazhat SN. Effect of Menin Deletion in Early Osteoblast Lineage on the Mineralization of an In Vitro 3D Osteoid-like Dense Collagen Gel Matrix. Biomimetics (Basel) 2022; 7:biomimetics7030101. [PMID: 35892371 PMCID: PMC9329857 DOI: 10.3390/biomimetics7030101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/24/2022] [Accepted: 07/16/2022] [Indexed: 02/05/2023] Open
Abstract
Bone has a complex microenvironment formed by an extracellular matrix (ECM) composed mainly of mineralized type I collagen fibres. Bone ECM regulates signaling pathways important in the differentiation of osteoblast-lineage cells, necessary for bone mineralization and in preserving tissue architecture. Compared to conventional 2D cell cultures, 3D in vitro models may better mimic bone ECM and provide an environment to support osteoblastic differentiation. In this study, a biomimetic 3D osteoid-like dense collagen gel model was used to investigate the role of the nuclear protein menin plays in osteoblastic differentiation and matrix mineralization. Previous in vitro and in vivo studies have shown that when expressed at later stages of osteoblastic differentiation, menin modulates osteoblastogenesis and regulates bone mass in adult mice. To investigate the role of menin when expressed at earlier stages of the osteoblastic lineage, conditional knockout mice in which the Men1 gene is specifically deleted early (i.e., at the level of the pluripotent mesenchymal stem cell lineage), where generated and primary calvarial osteoblasts were cultured in plastically compressed dense collagen gels for 21 days. The proliferation, morphology and differentiation of isolated seeded primary calvarial osteoblasts from knockout (Prx1-Cre; Men1f/f) mice were compared to those isolated from wild-type (Men1f/f) mice. Primary calvarial osteoblasts from knockout and wild-type mice did not show differences in terms of proliferation. However, in comparison to wild-type cells, primary osteoblast cells derived from knockout mice demonstrated deficient mineralization capabilities and an altered gene expression profile when cultured in 3D dense collagen gels. In summary, these findings indicate that when expressed at earlier stages of osteoblast differentiation, menin is important in maintaining matrix mineralization in 3D dense collagen gel matrices, in vitro.
Collapse
Affiliation(s)
- Ildi Troka
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada;
| | - Gabriele Griffanti
- Department of Mining and Materials Engineering, McGill University, Montreal, QC H3A 0C5, Canada;
| | - Lucie Canaff
- Department of Medicine, McGill University and McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (L.C.); (D.G.)
| | - Geoffrey N. Hendy
- Department of Medicine, McGill University and McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (L.C.); (D.G.)
| | - David Goltzman
- Department of Medicine, McGill University and McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (L.C.); (D.G.)
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering, McGill University, Montreal, QC H3A 0C5, Canada;
- Correspondence:
| |
Collapse
|
9
|
Huby T, Le Guillou E, Burin des Roziers C, Pacot L, Briand-Suleau A, Chansavang A, Toussaint A, Duchossoy V, Vaucouleur N, Benoit V, Lodé L, Molac C, North MO, Grotto S, Tsatsaris V, Jouinot A, Cochand-Priollet B, Paepegaey AC, Nectoux J, Groussin L, Pasmant E. Noninvasive Prenatal Diagnosis of a Paternally Inherited MEN1 Pathogenic Splicing Variant. J Clin Endocrinol Metab 2022; 107:e1367-e1373. [PMID: 34897474 DOI: 10.1210/clinem/dgab894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disease caused by mutations in the tumor suppressor gene MEN1. The uncertainty of pathogenicity of MEN1 variants complexifies the selection of the patients likely to benefit from specific care. OBJECTIVE MEN1-mutated patients should be offered tailored tumor screening and genetic counseling. We present a patient with hyperparathyroidism for whom genetic analysis identified a variant of uncertain significance in the MEN1 gene (NM_130799.2): c.654G > T p.(Arg218=). Additional functional genetic tests were performed to classify the variant as pathogenic and allowed prenatal testing. DESIGN Targeted next generation sequencing identified a synonymous variant in the MEN1 gene in a 26-year-old male with symptomatic primary hyperparathyroidism. In silico and in vitro genetic tests were performed to assess variant pathogenicity. RESULTS Genetic testing of the proband's unaffected parents showed the variant occurred de novo. Transcript study showed a splicing defect leading to an in-frame deletion. The classification of the MEN1 variant as pathogenic confirmed the diagnosis of MEN1 and recommended an adapted medical care and follow-up. Pathogenic classification also allowed to propose a genetic counseling to the proband and his wife. Noninvasive prenatal diagnosis was performed with a personalized medicine-based protocol by detection of the paternally inherited variant in maternal plasmatic cell free DNA, using digital PCR. CONCLUSION We showed that functional genetic analysis can help to assess the pathogenicity of a MEN1 variant with crucial consequences for medical care and genetic counseling decisions.
Collapse
Affiliation(s)
- Thomas Huby
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Edouard Le Guillou
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Cyril Burin des Roziers
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| | - Laurence Pacot
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| | - Audrey Briand-Suleau
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| | - Albain Chansavang
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| | - Aurélie Toussaint
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Véronique Duchossoy
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Nicolas Vaucouleur
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Virginie Benoit
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Laurence Lodé
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Clémence Molac
- Maternité Port-Royal, FHU PREMA, AP-HP, Hôpital Cochin, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | - Marie-Odile North
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Sarah Grotto
- Maternité Port-Royal, FHU PREMA, AP-HP, Hôpital Cochin, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | - Vassilis Tsatsaris
- Maternité Port-Royal, FHU PREMA, AP-HP, Hôpital Cochin, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | - Anne Jouinot
- Department of Endocrinology, Hôpital Cochin, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | - Béatrix Cochand-Priollet
- Department of Pathology, Cochin Hospital, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | | | - Juliette Nectoux
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
| | - Lionel Groussin
- Department of Endocrinology, Hôpital Cochin, AP-HP.Centre-Université de Paris, Université de Paris, Paris, France
| | - Eric Pasmant
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP.Centre-Université de Paris, Paris, France
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, CARPEM, Paris, France
| |
Collapse
|
10
|
Siavoshi A, Taghizadeh M, Dookhe E, Piran M. Gene expression profiles and pathway enrichment analysis to identification of differentially expressed gene and signaling pathways in epithelial ovarian cancer based on high-throughput RNA-seq data. Genomics 2021; 114:161-170. [PMID: 34839022 DOI: 10.1016/j.ygeno.2021.11.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 11/23/2021] [Indexed: 12/11/2022]
Abstract
Epithelial ovarian cancer (EOC) can be considered as a stressful and challenging disease among all women in the world, which has been associated with a poor prognosis and its molecular pathogenesis has remained unclear. In recent years, RNA Sequencing (RNA-seq) has become a functional and amazing technology for profiling gene expression. In the present study, RNA-seq raw data from Sequence Read Archive (SRA) of six tumor and normal ovarian sample was extracted, and then analysis and statistical interpretation was done with Linux and R Packages from the open-source Bioconductor. Gene Ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were applied for the identification of key genes and pathways involved in EOC. We identified 1091 Differential Expression Genes (DEGs) which have been reported in various studies of ovarian cancer as well as other types of cancer. Among them, 333 genes were up-regulated and 273 genes were down-regulated. In addition, Differentially Expressed Genes (DEGs) including RPL41, ALDH3A2, ERBB2, MIEN1, RBM25, ATF4, UPF2, DDIT3, HOXB8 and IL17D as well as Ribosome and Glycolysis/Gluconeogenesis pathway have had the potentiality to be used as targets for EOC diagnosis and treatment. In this study, unlike that of any other studies on various cancers, ALDH3A2 was most down-regulated gene in most KEGG pathways, and ATF4 was most up-regulated gene in leucine zipper domain binding term. In the other hand, RPL41 as a regulatory of cellular ATF4 level was up-regulated in many term and pathways and augmentation of ATF4 could justify the increase of RPL41 in the EOC. Pivotal pathways and significant genes, which were identified in the present study, can be used for adaptation of different EOC study. However, further molecular biological experiments and computational processes are required to confirm the function of the identified genes associated with EOC.
Collapse
Affiliation(s)
- A Siavoshi
- Department of Animal Sciences, Ramin University of Agriculture and Natural Resources, Ahvaz, Iran.
| | - M Taghizadeh
- Department of Medical Genetic, Tarbiat Modares University, Tehran, Iran
| | - E Dookhe
- Department of Biology, Research and Science Branch, Islamic Azad University, Tehran, Iran
| | - M Piran
- Department of Medical Biotechnology, Drug Design and Bioinformatics Unit, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Chen Cardenas SM, El-Kaissi S, Jarad O, Liaqat M, Korbonits M, Hamrahian AH. Unusual Combination of MEN-1 and the Contiguous Gene Deletion Syndrome of CAH and Ehlers-Danlos Syndrome (CAH-X). J Endocr Soc 2020; 4:bvaa077. [PMID: 32715272 PMCID: PMC7371387 DOI: 10.1210/jendso/bvaa077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Indexed: 11/19/2022] Open
Abstract
The contiguous gene deletion syndrome of congenital adrenal hyperplasia and Ehlers-Danlos syndrome, named CAH-X, is a rare entity that occurs because of a deletion of a chromosomal area containing 2 neighboring genes, TNXB and CYP21A. Here, we describe a patient from a consanguineous family in which coincidentally MEN-1 syndrome is associated with CAH-X, causing particular challenges explaining the phenotypic features of the patient. A 33-year-old man with salt-wasting congenital adrenal hyperplasia and classic-like Ehlers-Danlos syndrome presented with an adrenal crisis with a history of recurrent hypoglycemia, abdominal pain, and vomiting. He was found to have primary hyperparathyroidism, hyperprolactinemia, and pancreatic neuroendocrine tumors, as well as primary hypogonadism, large adrenal myelolipomas, and low bone mineral density. A bladder diverticulum was incidentally found. Genetic analysis revealed a heterozygous previously well-described MEN1 mutation (c.784-9G > A), a homozygous complete deletion of CYP21A2 (c.1-?_1488+? del), as well as a large deletion of the neighboring TNXB gene (c.11381-?_11524+?). The deletion includes the complete CYP21A2 gene and exons 35 through 44 of the TNXB gene. CGH array found 12% homozygosity over the whole genome. This rare case illustrates a complex clinical scenario with some initial diagnostic challenges.
Collapse
Affiliation(s)
- Stanley M Chen Cardenas
- Division of Endocrinology, Diabetes, and Metabolism. The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samer El-Kaissi
- Department of Endocrinology, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Ola Jarad
- Department of Endocrinology, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Muneezeh Liaqat
- Pathology and Laboratory Medicine Institute, Cleveland Clinic Abu Dhabi and National Reference Laboratory, Abu Dhabi, UAE
| | - Márta Korbonits
- Department of Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amir H Hamrahian
- Division of Endocrinology, Diabetes, and Metabolism. The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
The Study of Menin Expression as a Diagnostic Factor in HBV-Related Hepatocellular Carcinoma. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2020. [DOI: 10.5812/archcid.88188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Marx SJ, Goltzman D. Evolution of Our Understanding of the Hyperparathyroid Syndromes: A Historical Perspective. J Bone Miner Res 2019; 34:22-37. [PMID: 30536424 PMCID: PMC6396287 DOI: 10.1002/jbmr.3650] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022]
Abstract
We review advancing and overlapping stages for our understanding of the expressions of six hyperparathyroid (HPT) syndromes: multiple endocrine neoplasia type 1 (MEN1) or type 4, multiple endocrine neoplasia type 2A (MEN2A), hyperparathyroidism-jaw tumor syndrome, familial hypocalciuric hypercalcemia, neonatal severe primary hyperparathyroidism, and familial isolated hyperparathyroidism. During stage 1 (1903 to 1967), the introduction of robust measurement of serum calcium was a milestone that uncovered hypercalcemia as the first sign of dysfunction in many HPT subjects, and inheritability was reported in each syndrome. The earliest reports of HPT syndromes were biased toward severe or striking manifestations. During stage 2 (1959 to 1985), the early formulations of a syndrome were improved. Radioimmunoassays (parathyroid hormone [PTH], gastrin, insulin, prolactin, calcitonin) were breakthroughs. They could identify a syndrome carrier, indicate an emerging tumor, characterize a tumor, or monitor a tumor. During stage 3 (1981 to 2006), the assembly of many cases enabled recognition of further details. For example, hormone non-secreting skin lesions were discovered in MEN1 and MEN2A. During stage 4 (1985 to the present), new genomic tools were a revolution for gene identification. Four principal genes ("principal" implies mutated or deleted in 50% or more probands for its syndrome) (MEN1, RET, CASR, CDC73) were identified for five syndromes. During stage 5 (1993 to the present), seven syndromal genes other than a principal gene were identified (CDKN1B, CDKN2B, CDKN2C, CDKN1A, GNA11, AP2S1, GCM2). Identification of AP2S1 and GCM2 became possible because of whole-exome sequencing. During stages 4 and 5, the newly identified genes enabled many studies, including robust assignment of the carriers and non-carriers of a mutation. Furthermore, molecular pathways of RET and the calcium-sensing receptor were elaborated, thereby facilitating developments in pharmacotherapy. Current findings hold the promise that more genes for HPT syndromes will be identified and studied in the near future. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stephen J Marx
- Office of the Scientific Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - David Goltzman
- Calcium Research Laboratory, Metabolic Disorders and Complications Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
14
|
Cinque L, Pugliese F, Salcuni AS, Scillitani A, Guarnieri V. Molecular pathogenesis of parathyroid tumours. Best Pract Res Clin Endocrinol Metab 2018; 32:891-908. [PMID: 30477753 DOI: 10.1016/j.beem.2018.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Parathyroid tumors represent an elusive endocrine neoplasia, which lead to primary hyperparathyroidism, pHPT, a common endocrine calcium disorder characterized by hypercalcemia and normal-high parathormone secretion. Parathyroid tumours are benign adenomas or multiple glands hyperplasia in the vast majority (>99% of cases), while malignant neoplasms are rare (less than 1%). Despite pHPT is a common disorder, our knowledge about the genetic predisposition and molecular pathophysiology is limited to the familial syndromic forms of parathyroid tumour, that, however, represent not more than the 10% of all the cases; instead, the pathophysiology of sporadic forms remains an open field, although data about epigenetic mechanisms or private genes have been supposed. Here we present an overview of more recent acquisitions about the genetic causes along with their molecular mechanisms of benign, but also, malignant parathyroid tumours either in sporadic and familial presentation.
Collapse
Affiliation(s)
- Luigia Cinque
- Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, Division of Medical Genetics, Italy.
| | - Flavia Pugliese
- Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, Unit of Endocrinology, San Giovanni Rotondo, FG, Italy.
| | | | - Alfredo Scillitani
- Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, Unit of Endocrinology, San Giovanni Rotondo, FG, Italy.
| | - Vito Guarnieri
- Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, Division of Medical Genetics, Italy.
| |
Collapse
|
15
|
Stevenson M, Lines KE, Thakker RV. Molecular Genetic Studies of Pancreatic Neuroendocrine Tumors: New Therapeutic Approaches. Endocrinol Metab Clin North Am 2018; 47:525-548. [PMID: 30098714 PMCID: PMC7614857 DOI: 10.1016/j.ecl.2018.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic neuroendocrine tumors (PNETs) arise sporadically or as part of familial syndromes. Genetic studies of hereditary syndromes and whole exome sequencing analysis of sporadic NETs have revealed the roles of some genes involved in PNET tumorigenesis. The multiple endocrine neoplasia type 1 (MEN1) gene is most commonly mutated. Its encoded protein, menin, has roles in transcriptional regulation, genome stability, DNA repair, protein degradation, cell motility and adhesion, microRNA biogenesis, cell division, cell cycle control, and epigenetic regulation. Therapies targeting epigenetic regulation and MEN1 gene replacement have been reported to be effective in preclinical models.
Collapse
Affiliation(s)
- Mark Stevenson
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - Kate E Lines
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - Rajesh V Thakker
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Headington, Oxford OX3 7LJ, UK.
| |
Collapse
|
16
|
Abstract
Pancreatic neuroendocrine tumours (PNETs) might occur as a non-familial isolated endocrinopathy or as part of a complex hereditary syndrome, such as multiple endocrine neoplasia type 1 (MEN1). MEN1 is an autosomal dominant disorder characterized by the combined occurrence of PNETs with tumours of the parathyroids and anterior pituitary. Treatments for primary PNETs include surgery. Treatments for non-resectable PNETs and metastases include biotherapy (for example, somatostatin analogues, inhibitors of receptors and monoclonal antibodies), chemotherapy and radiological therapy. All these treatments are effective for PNETs in patients without MEN1; however, there is a scarcity of clinical trials reporting the efficacy of the same treatments of PNETs in patients with MEN1. Treatment of PNETs in patients with MEN1 is challenging owing to the concomitant development of other tumours, which might have metastasized. In recent years, preclinical studies have identified potential new therapeutic targets for treating MEN1-associated neuroendocrine tumours (including PNETs), and these include epigenetic modification, the β-catenin-wingless (WNT) pathway, Hedgehog signalling, somatostatin receptors and MEN1 gene replacement therapy. This Review discusses these advances.
Collapse
Affiliation(s)
- Morten Frost
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
- Endocrine Research Unit, University of Southern Denmark, Odense, 5000, Denmark
| | - Kate E Lines
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
| |
Collapse
|
17
|
Feng Z, Ma J, Hua X. Epigenetic regulation by the menin pathway. Endocr Relat Cancer 2017; 24:T147-T159. [PMID: 28811300 PMCID: PMC5612327 DOI: 10.1530/erc-17-0298] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
There is a trend of increasing prevalence of neuroendocrine tumors (NETs), and the inherited multiple endocrine neoplasia type 1 (MEN1) syndrome serves as a genetic model to investigate how NETs develop and the underlying mechanisms. Menin, encoded by the MEN1 gene, at least partly acts as a scaffold protein by interacting with multiple partners to regulate cellular homeostasis of various endocrine organs. Menin has multiple functions including regulation of several important signaling pathways by controlling gene transcription. Here, we focus on reviewing the recent progress in elucidating the key biochemical role of menin in epigenetic regulation of gene transcription and cell signaling, as well as posttranslational regulation of menin itself. In particular, we will review the progress in studying structural and functional interactions of menin with various histone modifiers and transcription factors such as MLL, PRMT5, SUV39H1 and other transcription factors including c-Myb and JunD. Moreover, the role of menin in regulating cell signaling pathways such as TGF-beta, Wnt and Hedgehog, as well as miRNA biogenesis and processing will be described. Further, the regulation of the MEN1 gene transcription, posttranslational modifications and stability of menin protein will be reviewed. These various modes of regulation by menin as well as regulation of menin by various biological factors broaden the view regarding how menin controls various biological processes in neuroendocrine organ homeostasis.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Ma
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary BiotechnologyHarbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xianxin Hua
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Cinque L, Sparaneo A, Cetani F, Coco M, Clemente C, Chetta M, Balsamo T, Battista C, Sanpaolo E, Pardi E, D'Agruma L, Marcocci C, Maiello E, Hendy GN, Cole DEC, Scillitani A, Guarnieri V. Novel association of MEN1 gene mutations with parathyroid carcinoma. Oncol Lett 2017; 14:23-30. [PMID: 28693130 PMCID: PMC5494910 DOI: 10.3892/ol.2017.6162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/23/2016] [Indexed: 12/23/2022] Open
Abstract
Inactivating mutations of the multiple endocrine neoplasia 1 (MEN1) gene cause MEN1 syndrome, characterized by primary hyperparathyroidism (pHPT), and parathyroid and gastro-entero-pancreatic pituitary tumors. At present, only 14 cases of malignant parathyroid tumor have been associated with the syndrome, with 6 cases carrying an inactivating mutation of the MEN1 gene. The present study presents the case of a 48-year-old female who presented with multigland pHPT and multiple pancreatic lesions. The patient underwent surgery several times for the excision of parathyroid hyperplasia, carcinoma and adenoma. The MEN1 gene was screened, revealing three variants (in cis) at the intron/exon 3 boundary (IVS2-3G>C, c.497A>T and c.499G>T) detected on the DNA of the proband, not shared by her relatives. RNA sequencing revealed that the IVS2-3C>G variant caused the skipping of the exon 3. Therefore, the present study reports on a novel rare association of MEN1 syndrome and parathyroid carcinoma. The reported splicing mutation was previously identified in subjects who always developed malignant lesions; thus, a possible genotype-phenotype association may be considered.
Collapse
Affiliation(s)
- Luigia Cinque
- Department of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Angelo Sparaneo
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Filomena Cetani
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University Hospital of Pisa, I-56124 Pisa, Italy
| | - Michelina Coco
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Celeste Clemente
- Department of Pathology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Massimiliano Chetta
- Laboratory of Molecular Medicine and Genomics, University of Salerno, I-84081 Baronissi, Italy
| | - Teresa Balsamo
- Laboratory of Oncology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Claudia Battista
- Department of Endocrinology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Eliana Sanpaolo
- Department of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Elena Pardi
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University Hospital of Pisa, I-56124 Pisa, Italy
| | - Leonardo D'Agruma
- Department of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Claudio Marcocci
- Department of Clinical and Experimental Medicine, Endocrine Unit 2, University Hospital of Pisa, I-56124 Pisa, Italy
| | - Evaristo Maiello
- Department of Oncohematology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Geoffrey N Hendy
- Departments of Medicine, Physiology and Human Genetics, McGill University, Montreal, QC H4A 3J1, Canada.,Experimental Therapeutics and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| | - David E C Cole
- Departments of Laboratory Medicine and Pathobiology, Medicine and Genetics, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Alfredo Scillitani
- Department of Endocrinology, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| | - Vito Guarnieri
- Department of Medical Genetics, IRCCS Casa Sollievo della Sofferenza Hospital, I-71013 San Giovanni Rotondo, Italy
| |
Collapse
|
19
|
Nagy Z, Szabó PM, Grolmusz VK, Perge P, Igaz I, Patócs A, Igaz P. MEN1 and microRNAs: The link between sporadic pituitary, parathyroid and adrenocortical tumors? Med Hypotheses 2016; 99:40-44. [PMID: 28110695 DOI: 10.1016/j.mehy.2016.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 12/17/2016] [Indexed: 01/12/2023]
Abstract
Sporadic tumors of the pituitary, parathyroids and adrenal cortex are unique, as their benign forms are very common, but malignant forms are exceptionally rare. Hereditary forms of these tumors occur in multiple endocrine neoplasia syndrome type 1 (MEN1). We hypothesize that the pathogenic link among the sporadic tumors of these organs of different germ layers might be represented by common molecular pathways involving the MEN1 gene and microRNAs (miR). miR-24 might be a microRNA linking the three tumor entities, but other candidates such as miR-142-3p and microRNAs forming the DLK1-MEG3 miRNA cluster might also be of importance.
Collapse
Affiliation(s)
- Z Nagy
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary
| | - P M Szabó
- National Institutes of Health/NCI/DCTD/BRP, 9609 Medical Center Dr Bethesda MD, USA
| | - V K Grolmusz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary; "Lendület-2013" Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary
| | - P Perge
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary
| | - I Igaz
- Department of Gastroenterology, Szt Imre Teaching Hospital Budapest, Budapest, Hungary
| | - A Patócs
- "Lendület-2013" Research Group, Hungarian Academy of Sciences and Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary; Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary
| | - P Igaz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary.
| |
Collapse
|
20
|
HybridRanker: Integrating network topology and biomedical knowledge to prioritize cancer candidate genes. J Biomed Inform 2016; 64:139-146. [DOI: 10.1016/j.jbi.2016.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/13/2016] [Accepted: 10/06/2016] [Indexed: 11/20/2022]
|
21
|
Jyotsna VP, Malik E, Birla S, Sharma A. Novel MEN 1 gene findings in rare sporadic insulinoma--a case control study. BMC Endocr Disord 2015; 15:44. [PMID: 26307114 PMCID: PMC4549893 DOI: 10.1186/s12902-015-0041-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 08/21/2015] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Insulinomas, which are rare tumors causing hyperinsulinemic hypoglycemia are usually sporadic but may also occur in association with multiple endocrine neoplasia type 1 (MEN-1) syndrome an autosomal dominant disorder caused by MEN1 gene mutations. MEN1 encodes a nuclear protein Menin, a tumor suppressor which acts as an adapter and interacts with partner proteins involved in crucial activities like transcriptional regulation, cell division, proliferation and genome stability. This study reports on clinical findings and mutation screening in sporadic insulinoma patients. METHODS Seventeen patients diagnosed with insulinoma were recruited along with 30 healthy volunteers who acted as controls for the present study. The patients presented with symptoms of sweating, tremors, drowsiness, palpitations, loss of consciousness, abnormal behavior, seizures and weight gain. Detailed clinical and family history was collected from all the participants along with 5 ml of blood sample after taking informed consent. Genomic DNA isolated from blood was subjected to MEN1 gene amplification followed by direct sequencing. Nucleotide sequences obtained were compared with published MEN1 cDNA sequences. Prediction of functional effects of novel changes was done using various bioinformatics algorithms. RESULTS Molecular analysis revealed presence of three novel exonic mutations (M561K, Q192K and Q261Q), two novel intronic variations c.445-44G → A and c.913-42G → C in introns two and six respectively and three reported exon SNPs; H433H (rs540012), D418D (rs2071313), A541T (rs2959656) and one intronic SNP (rs669976). CONCLUSIONS The study identified presence of novel pathogenic MEN1 mutations in sporadic cases of insulinoma. The new mutations identified were in regions involved in defective binding of menin to proteins implicated in genetic and epigenetic mechanisms. The outcome of the study extends the growing list of MEN1 pathogenic mutations even in sporadic cases providing consequential insight into phenotypic heterogeneity and in the expression of individual mutations.
Collapse
Affiliation(s)
- Viveka P Jyotsna
- Department of Endocrinology and metabolism, All India Institute of Medical Sciences, Room No. 305, Third Floor, Biotechnology Building, New Delhi, India.
| | - Ekta Malik
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| | - Shweta Birla
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| | - Arundhati Sharma
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
22
|
Hall MJ, Innocent J, Rybak C, Veloski C, Scott WJ, Wu H, Ridge JA, Hoffman JP, Borghaei H, Turaka A, Daly MB. Bilateral granulosa cell tumors: a novel malignant manifestation of multiple endocrine neoplasia 1 syndrome found in a patient with a rare menin in-frame deletion. APPLICATION OF CLINICAL GENETICS 2015; 8:69-73. [PMID: 25733923 PMCID: PMC4337709 DOI: 10.2147/tacg.s72223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction Multiple endocrine neoplasia 1 (MEN1) is a cancer syndrome resulting from mutations of the MEN1 gene. The syndrome is characterized by neoplasia of the parathyroid and pituitary glands, and malignant tumors of the endocrine pancreas. Other manifestations include benign lipomas, angiofibromas, and carcinoid tumors commonly originating in the colon, thymus, and lung. This is the first report of MEN1 syndrome manifesting as bilateral granulosa cell ovarian tumors, and which is associated with a rare intronic mutation of the MEN1 gene. Case report A 41-year-old woman presented with abdominal pain, increasing abdominal girth, and dysmenorrhea. Ultrasound demonstrated enlarged ovaries and uterine fibroids. After an exploratory laparotomy, she subsequently underwent bilateral salpingo–oophorectomy with hysterectomy where the pathology revealed bilateral cystic granulosa cell tumors of the ovaries. Additional workup including computed tomography imaging discovered a thymic mass, which the pathology showed was malignant, along with a pancreatic mass suspicious for a neuroendocrine tumor. Hyperparathyroidism was also discovered and was found to be secondary to a parathyroid adenoma. Genetic testing revealed an exceedingly rare mutation in the MEN1 gene (c.654 + 1 G>A). Discussion Mutations of the menin gene leading to MEN1 syndrome are classically nonsense or missense mutations producing a dysfunctional protein product. Recently, researchers described a novel mutation of MEN1 (c.654 + 1 G>A) in a male proband meeting the criteria for clinical MEN1 syndrome. Functional analysis performed on the stable mutant protein showed selective disruption of the transforming growth factor beta signaling pathway, yet it maintained its wild-type ability to inhibit nuclear factor kappa B and to suppress JunD transcriptional activity. Conclusion To our knowledge, this is the first report of MEN1 syndrome associated with bilateral granulosa cell malignancy. We postulate that this presentation may be due to the novel menin gene mutation recently described.
Collapse
Affiliation(s)
- Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Julie Innocent
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Christina Rybak
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Colleen Veloski
- Department of Internal Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Walter J Scott
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hong Wu
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John A Ridge
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John P Hoffman
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hossein Borghaei
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aruna Turaka
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
23
|
Zhou Y, Zhang X, Klibanski A. Genetic and epigenetic mutations of tumor suppressive genes in sporadic pituitary adenoma. Mol Cell Endocrinol 2014; 386:16-33. [PMID: 24035864 PMCID: PMC3943596 DOI: 10.1016/j.mce.2013.09.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/03/2013] [Indexed: 12/28/2022]
Abstract
Human pituitary adenomas are the most common intracranial neoplasms. Approximately 5% of them are familial adenomas. Patients with familial tumors carry germline mutations in predisposition genes, including AIP, MEN1 and PRKAR1A. These mutations are extremely rare in sporadic pituitary adenomas, which therefore are caused by different mechanisms. Multiple tumor suppressive genes linked to sporadic tumors have been identified. Their inactivation is caused by epigenetic mechanisms, mainly promoter hypermethylation, and can be placed into two groups based on their functional interaction with tumor suppressors RB or p53. The RB group includes CDKN2A, CDKN2B, CDKN2C, RB1, BMP4, CDH1, CDH13, GADD45B and GADD45G; AIP and MEN1 genes also belong to this group. The p53 group includes MEG3, MGMT, PLAGL1, RASSF1, RASSF3 and SOCS1. We propose that the tumor suppression function of these genes is mainly mediated by the RB and p53 pathways. We also discuss possible tumor suppression mechanisms for individual genes.
Collapse
Affiliation(s)
- Yunli Zhou
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Xun Zhang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Anne Klibanski
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW To describe the recent efforts to understand the molecular and genetic mechanisms involved in the tumorigenesis of pituitary adenomas. RECENT FINDINGS There is rapidly accumulating evidence for the roles of molecular abnormalities in pituitary adenoma tumorigenesis, including dysregulation of the cell cycle, signal transduction pathways, oncogenes and tumor suppressor genes. MicroRNAs have been identified as important participants in gene regulation and may have a role in therapy. Stem cells have also provided novel concepts for tumorigenesis and potentially treatment. SUMMARY Pituitary adenomas are relatively common neoplasms, whose pathogenesis is still poorly understood. Although considered by many as benign monoclonal proliferations, their clinical spectrum is diverse including hormone hypersecretion, and various degrees of invasiveness, suggesting multiple steps and mechanisms. This review summarizes recent development in the molecular tumorigenesis of pituitary adenomas emphasizing the dysregulation of the cell cycle components, tumor suppressor genes, oncogenes, stem cells and microRNAs.
Collapse
Affiliation(s)
- Steve Rostad
- CellNetix Pathology and Laboratories, Seattle, Washington, USA
| |
Collapse
|