1
|
Møller MW, Nortvig MJ, Andersen MS, Poulsen FR. DNA Methylation in Pituitary Adenomas: A Scoping Review. Int J Mol Sci 2025; 26:531. [PMID: 39859246 PMCID: PMC11765255 DOI: 10.3390/ijms26020531] [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: 12/06/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Pituitary adenomas are a diverse group of neoplasms with variable clinical behavior. Despite advances in genetic analysis, understanding the role of epigenetic modifications, particularly DNA methylation, remains an area under investigation. This scoping review aimed to update and synthesize the current body of literature on DNA methylation in pituitary adenomas, focusing on methodological advancements and clinical correlations. A systematic search conducted across multiple databases, including Embase, Scopus, MEDLINE, and CENTRAL, identified 107 eligible studies. Early methods, such as methylation-restricted digestion and methylation-specific PCR (MSP), have evolved into more comprehensive approaches, such as chip-based DNA methylation analysis. Key findings suggest that genes like POMC, SOCS-1, and RASSF1A show a significant association between methylation and clinical behavior. However, methylation patterns alone are insufficient to fully explain tumorigenesis. Emerging data suggest that DNA methylation might serve as a prognostic marker for invasive growth and recurrence, but further longitudinal studies are needed. This review highlights the need for future research to explore the methylome more thoroughly and to better define the clinical impact of epigenetic modifications in pituitary adenomas.
Collapse
Affiliation(s)
- Morten Winkler Møller
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Mathias Just Nortvig
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Mikkel Schou Andersen
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital, DK-5000 Odense, Denmark; (M.J.N.); (M.S.A.); (F.R.P.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense, Denmark
- BRIDGE (Brain Research-Inter Disciplinary Guided Excellence), University of Southern Denmark, DK-5000 Odense, Denmark
| |
Collapse
|
2
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [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] [Indexed: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
3
|
Torres-Morán M, Franco-Álvarez AL, Rebollar-Vega RG, Hernández-Ramírez LC. Hotspots of Somatic Genetic Variation in Pituitary Neuroendocrine Tumors. Cancers (Basel) 2023; 15:5685. [PMID: 38067388 PMCID: PMC10705109 DOI: 10.3390/cancers15235685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 02/13/2025] Open
Abstract
The most common genetic drivers of pituitary neuroendocrine tumors (PitNETs) lie within mutational hotspots, which are genomic regions where variants tend to cluster. Some of these hotspot defects are unique to PitNETs, while others are associated with additional neoplasms. Hotspot variants in GNAS and USP8 are the most common genetic causes of acromegaly and Cushing's disease, respectively. Although it has been proposed that these genetic defects could define specific clinical phenotypes, results are highly variable among studies. In contrast, DICER1 hotspot variants are associated with a familial syndrome of cancer predisposition, and only exceptionally occur as somatic changes. A small number of non-USP8-driven corticotropinomas are due to somatic hotspot variants in USP48 or BRAF; the latter is a well-known mutational hotspot in cancer. Finally, somatic variants affecting a hotspot in SF3B1 have been associated with multiple cancers and, more recently, with prolactinomas. Since the associations of BRAF, USP48, and SF3B1 hotspot variants with PitNETs are very recent, their effects on clinical phenotypes are still unknown. Further research is required to fully define the role of these genetic defects as disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Laura C. Hernández-Ramírez
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
4
|
Peverelli E, Treppiedi D, Mangili F, Catalano R, Spada A, Mantovani G. Drug resistance in pituitary tumours: from cell membrane to intracellular signalling. Nat Rev Endocrinol 2021; 17:560-571. [PMID: 34194011 DOI: 10.1038/s41574-021-00514-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
The pharmacological treatment of pituitary tumours is based on the use of stable analogues of somatostatin and dopamine. The analogues bind to somatostatin receptor types 2 and 5 (SST2 and SST5) and dopamine receptor type 2 (DRD2), respectively, and generate signal transduction cascades in cancerous pituitary cells that culminate in the inhibition of hormone secretion, cell growth and invasion. Drug resistance occurs in a subset of patients and can involve different steps at different stages, such as following receptor activation by the agonist or during the final biological responses. Although the expression of somatostatin and dopamine receptors in cancer cells is a prerequisite for these drugs to reach a biological effect, their presence does not guarantee the success of the therapy. Successful therapy also requires the proper functioning of the machinery of signal transduction and the finely tuned regulation of receptor desensitization, internalization and intracellular trafficking. The present Review provides an updated overview of the molecular factors underlying the pharmacological resistance of pituitary tumours. The Review discusses the experimental evidence that supports a role for receptors and intracellular proteins in the function of SSTs and DRD2 and their clinical importance.
Collapse
Affiliation(s)
- Erika Peverelli
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy.
| | - Donatella Treppiedi
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Federica Mangili
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Rosa Catalano
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
- PhD Program in Endocrinological Sciences, Sapienza University of Rome, Rome, Italy
| | - Anna Spada
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Giovanna Mantovani
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Milan, Italy
| |
Collapse
|
5
|
Romanet P, Galluso J, Kamenicky P, Hage M, Theodoropoulou M, Roche C, Graillon T, Etchevers HC, De Murat D, Mougel G, Figarella-Branger D, Dufour H, Cuny T, Assié G, Barlier A. Somatotroph Tumors and the Epigenetic Status of the GNAS Locus. Int J Mol Sci 2021; 22:ijms22147570. [PMID: 34299200 PMCID: PMC8306130 DOI: 10.3390/ijms22147570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/16/2022] Open
Abstract
Forty percent of somatotroph tumors harbor recurrent activating GNAS mutations, historically called the gsp oncogene. In gsp-negative somatotroph tumors, GNAS expression itself is highly variable; those with GNAS overexpression most resemble phenotypically those carrying the gsp oncogene. GNAS is monoallelically expressed in the normal pituitary due to methylation-based imprinting. We hypothesize that changes in GNAS imprinting of gsp-negative tumors affect GNAS expression levels and tumorigenesis. We characterized the GNAS locus in two independent somatotroph tumor cohorts: one of 23 tumors previously published (PMID: 31883967) and classified by pan-genomic analysis, and a second with 82 tumors. Multi-omics analysis of the first cohort identified a significant difference between gsp-negative and gsp-positive tumors in the methylation index at the known differentially methylated region (DMR) of the GNAS A/B transcript promoter, which was confirmed in the larger series of 82 tumors. GNAS allelic expression was analyzed using a polymorphic Fok1 cleavage site in 32 heterozygous gsp-negative tumors. GNAS expression was significantly reduced in the 14 tumors with relaxed GNAS imprinting and biallelic expression, compared to 18 tumors with monoallelic expression. Tumors with relaxed GNAS imprinting showed significantly lower SSTR2 and AIP expression levels. Altered A/B DMR methylation was found exclusively in gsp-negative somatotroph tumors. 43% of gsp-negative tumors showed GNAS imprinting relaxation, which correlated with lower GNAS, SSTR2 and AIP expression, indicating lower sensitivity to somatostatin analogues and potentially aggressive behavior.
Collapse
Affiliation(s)
- Pauline Romanet
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Justine Galluso
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Peter Kamenicky
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, 94270 Le Kremlin-Bicêtre, Île-de-France, France; (P.K.); (M.H.)
| | - Mirella Hage
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, 94270 Le Kremlin-Bicêtre, Île-de-France, France; (P.K.); (M.H.)
| | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig Maximilian University Munich, 80336 Munich, Germany;
| | - Catherine Roche
- APHM, La Conception Hospital, Laboratory of Molecular Biology, 13385 Marseille, France;
| | - Thomas Graillon
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Timone Hospital Department of Neurosurgery, 13385 Marseille, France; (T.G.); (H.D.)
| | - Heather C. Etchevers
- Aix Marseille Univ, INSERM, MMG, UMR1251, Marmara Institute, 13385 Marseille, France;
| | - Daniel De Murat
- Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR8104, F-75014 Paris, France; (D.D.M.); (G.A.)
| | - Grégory Mougel
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Dominique Figarella-Branger
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13385 Marseille, France;
| | - Henry Dufour
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Timone Hospital Department of Neurosurgery, 13385 Marseille, France; (T.G.); (H.D.)
| | - Thomas Cuny
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, Department of Endocrinology, Hospital La Conception, 13385 Marseille, France;
| | - Guillaume Assié
- Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR8104, F-75014 Paris, France; (D.D.M.); (G.A.)
- Department of Endocrinology, Center for Rare Adrenal Diseases, Assistance Publique—Hôpitaux de Paris, Hôpital Cochin, 75014 Paris, France
| | - Anne Barlier
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
- Correspondence:
| |
Collapse
|
6
|
Vasilev V, Daly AF, Zacharieva S, Beckers A. Clinical and Molecular Update on Genetic Causes of Pituitary Adenomas. Horm Metab Res 2020; 52:553-561. [PMID: 32299111 DOI: 10.1055/a-1143-5930] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pituitary adenomas are benign tumors with variable functional characteristics that can have a significant impact on patients. The majority arise sporadically, but an inherited genetic susceptibility is increasingly being recognized. Recent advances in genetics have widened the scope of our understanding of pituitary tumorigenesis. The clinical and genetic characteristics of pituitary adenomas that develop in the setting of germline-mosaic and somatic GNAS mutations (McCune-Albright syndrome and sporadic acromegaly), germline MEN1 mutations (multiple endocrine neoplasia type 1), and germline PRKAR1A mutations (Carney complex) have been well described. Non-syndromic familial cases of isolated pituitary tumors can occur as familial isolated pituitary adenomas (FIPA); mutations/deletions of the AIP gene have been found in a minority of these. Genetic alterations in GPR101 have been identified recently as causing X-linked acro-gigantism (X-LAG) leading to very early-onset pediatric gigantism. Associations of pituitary adenomas with other tumors have been described in syndromes like multiple endocrine neoplasia type 4, pheochromocytoma-paraganglioma with pituitary adenoma association (3PAs) syndrome and some of their genetic causes have been elucidated. The genetic etiologies of a significant proportions of sporadic corticotropinomas have recently been identified with the discovery of USP8 and USP48 mutations. The elucidation of genetic and molecular pathophysiology in pituitary adenomas is a key factor for better patient management and effective follow-up.
Collapse
Affiliation(s)
- Vladimir Vasilev
- Department of Endocrinology, CHU de Liège, Liège Université, Liège, Belgium
- Department of Endocrinology, Medical University, Sofia, Bulgaria
| | - Adrian F Daly
- Department of Endocrinology, CHU de Liège, Liège Université, Liège, Belgium
| | | | - Albert Beckers
- Department of Endocrinology, CHU de Liège, Liège Université, Liège, Belgium
| |
Collapse
|
7
|
Li J, Dong W, Li Z, Wang H, Gao H, Zhang Y. Impact of SLC20A1 on the Wnt/β‑catenin signaling pathway in somatotroph adenomas. Mol Med Rep 2019; 20:3276-3284. [PMID: 31432167 PMCID: PMC6755178 DOI: 10.3892/mmr.2019.10555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Studies have revealed that genetic and functional aberrations of oncogenes, tumor‑suppressor genes, signaling pathways and receptors are among the most prominent events in pituitary tumorigenesis, and a potent biomarker would be helpful for early diagnosis, subsequent treatment and disease control. The present study investigated the expression signatures of solute carrier family 20 member 1, also known as phosphate transporter 1 (SLC20A1) and the Wnt/β‑catenin signaling pathway in 52 patients with somatotroph adenomas. According to immunohistochemistry analysis, the H‑score of SLC20A1 was 222.6±15.2 in invasive tumor samples and 144.5±30.4 in non‑invasive tumor samples (P<0.01), while the H‑scores of β‑catenin were 210.1±21.4 and 134.9±32.7, respectively (P<0.05). The H‑scores of Wnt inhibitory factor 1 (Wif1) exhibited the opposite trend, with scores of 134.5±22.7 and 253.6±14.8, respectively (P<0.01). The H‑scores of SLC20A1 were negatively associated with those of Wif1 in somatotroph adenomas (correlation coefficient r=‑0.367). The mean progression‑free survival in the low SLC20A1 group was longer than that in the group with high SLC20A1 H‑scores (P=0.024). Reverse transcription‑quantitative PCR (RT‑qPCR) and western blotting confirmed the interference efficiency of the segments short hairpin (Sh)‑B‑SLC20A1 and Sh‑C‑SLC20A1. Cell proliferation experiments revealed that the cell viability of the Sh‑B‑SLC20A1 group was 76.3±4.5, 65.7±3.7 and 53.1±3.2% of that of control GH3 cells after 24, 48 and 72 h of transfection, respectively, while the cell viability of the Sh‑C‑SLC20A1 group was 86.4±5.7, 75.6±4.4 and 67.5±3.8%, respectively (P<0.05). ELISA analysis demonstrated the growth hormone (GH) levels in the Sh‑B‑SLC20A1 and Sh‑C‑SLC20A1 groups to be 34.7±10.4 and 54.6±14.4%, respectively, of that of control GH3 cells (P<0.05). The transmembrane invasion assay revealed that knocking down SLC20A1 significantly suppressed cell invasion in the Sh‑B‑SLC20A1 and Sh‑C‑SLC20A1 groups. RT‑qPCR and western blotting demonstrated that Sh‑B‑SLC20A1 and Sh‑C‑SLC20A1 evidently increased the levels of Wif1 and secreted frizzled‑related protein 4. The present data suggested that SLC20A1 levels are positively associated with tumor size, invasive behavior and tumor recurrence in somatotroph adenomas. Furthermore, SLC20A1 may be associated with the activation of the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Jianhua Li
- Key Laboratory of Central Nervous System Injury Research, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China
| | - Wei Dong
- Department of Neurosurgery, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Zhenye Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Hongyun Wang
- Key Laboratory of Central Nervous System Injury Research, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China
| | - Hua Gao
- Key Laboratory of Central Nervous System Injury Research, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China
| | - Yazhuo Zhang
- Key Laboratory of Central Nervous System Injury Research, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
8
|
Foltran RK, Amorim PVGH, Duarte FH, Grande IPP, Freire ACTB, Frassetto FP, Dettoni JB, Alves VA, Castro I, Trarbach EB, Bronstein MD, Jallad RS. Study of major genetic factors involved in pituitary tumorigenesis and their impact on clinical and biological characteristics of sporadic somatotropinomas and non-functioning pituitary adenomas. ACTA ACUST UNITED AC 2018; 51:e7427. [PMID: 29947650 PMCID: PMC6040863 DOI: 10.1590/1414-431x20187427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/02/2018] [Indexed: 11/25/2022]
Abstract
Genetic and functional aberrations of guanine nucleotide-binding protein, alpha stimulating (GNAS), aryl hydrocarbon receptor interacting protein (AIP), and pituitary tumor transforming gene (PTTG) are among the most prominent events in pituitary tumorigenesis. A cohort of Brazilian patients with somatotropinomas (n=41) and non-functioning pituitary adenomas (NFPA, n=21) from a single tertiary-referral center were evaluated for GNAS and AIP mutations and gene expression of AIP and PTTG. Results were compared to the clinical and biological (Ki67 and p53 expression) characteristics of tumors and their response to therapy, if applicable. Genetic analysis revealed that 27% of somatotropinomas and 4.8% of NFPA harbored GNAS mutations (P=0.05). However, no differences were observed in clinical characteristics, tumor extension, response to somatostatin analog therapy, hormonal/surgical remission rates, Ki67 index, and p53 expression between mutated and non-mutated somatotropinomas patients. PTTG overexpression (RQ mean=10.6, min=4.39, max=11.9) and AIP underexpression (RQ mean=0.56, min=0.46-max=0.92) were found in virtually all cases without a statistically significant relationship with clinical and biological tumor features. No patients exhibited somatic or germline pathogenic AIP mutations. In conclusion, mutations in GNAS and abnormal PTTG and AIP expression had no impact on tumor features and treatment outcomes in this cohort. Our data support some previous studies and point to the need for further investigations, probably involving epigenetic and transcriptome analysis, to improve our understanding of pituitary tumor behavior.
Collapse
Affiliation(s)
- R K Foltran
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - P V G H Amorim
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F H Duarte
- Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Serviço de Endocrinologia, A.C. Camargo Center, São Paulo, SP, Brasil
| | - I P P Grande
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A C T B Freire
- Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F P Frassetto
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - J B Dettoni
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V A Alves
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - I Castro
- Divisao de Medicina Molecular, Departamento de Medicina, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E B Trarbach
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M D Bronstein
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - R S Jallad
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
9
|
Innamorati G, Wilkie TM, Kantheti HS, Valenti MT, Dalle Carbonare L, Giacomello L, Parenti M, Melisi D, Bassi C. The curious case of Gαs gain-of-function in neoplasia. BMC Cancer 2018; 18:293. [PMID: 29544460 PMCID: PMC5856294 DOI: 10.1186/s12885-018-4133-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 02/15/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mutations activating the α subunit of heterotrimeric Gs protein are associated with a number of highly specific pathological molecular phenotypes. One of the best characterized is the McCune Albright syndrome. The disease presents with an increased incidence of neoplasias in specific tissues. MAIN BODY A similar repertoire of neoplasms can develop whether mutations occur spontaneously in somatic tissues during fetal development or after birth. Glands are the most "permissive" tissues, recently found to include the entire gastrointestinal tract. High frequency of activating Gαs mutations is associated with precise diagnoses (e.g., IPMN, Pyloric gland adenoma, pituitary toxic adenoma). Typically, most neoplastic lesions, from thyroid to pancreas, remain well differentiated but may be a precursor to aggressive cancer. CONCLUSIONS Here we propose the possibility that gain-of-function mutations of Gαs interfere with signals in the microenvironment of permissive tissues and lead to a transversal neoplastic phenotype.
Collapse
Affiliation(s)
- Giulio Innamorati
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Thomas M. Wilkie
- Pharmacology Department, UT Southwestern Medical Center, Dallas, TX USA
| | | | - Maria Teresa Valenti
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Luca Dalle Carbonare
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Luca Giacomello
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Marco Parenti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Davide Melisi
- Laboratory of Oncology and Molecular Therapy, Department of Medicine, University of Verona, Verona, Italy
| | - Claudio Bassi
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Hernández-Ramírez LC, Trivellin G, Stratakis CA. Cyclic 3',5'-adenosine monophosphate (cAMP) signaling in the anterior pituitary gland in health and disease. Mol Cell Endocrinol 2018; 463:72-86. [PMID: 28822849 DOI: 10.1016/j.mce.2017.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 11/28/2022]
Abstract
The cyclic 3',5'-adenosine monophosphate (cAMP) was the first among the so-called "second messengers" to be described. It is conserved in most organisms and functions as a signal transducer by mediating the intracellular effects of multiple hormones and neurotransmitters. In this review, we first delineate how different members of the cAMP pathway ensure its correct compartmentalization and activity, mediate the terminal intracellular effects, and allow the crosstalk with other signaling pathways. We then focus on the pituitary gland, where cAMP exerts a crucial function by controlling the responsiveness of the cells to hypothalamic hormones, neurotransmitters and peripheral factors. We discuss the most relevant physiological functions mediated by cAMP in the different pituitary cell types, and summarize the defects affecting this pathway that have been reported in the literature. We finally discuss how a deregulated cAMP pathway is involved in the pathogenesis of pituitary disorders and how it affects the response to therapy.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 10 Center Drive, CRC, Room 1E-3216, Bethesda, MD 20892-1862, USA
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 10 Center Drive, CRC, Room 1E-3216, Bethesda, MD 20892-1862, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), 10 Center Drive, CRC, Room 1E-3216, Bethesda, MD 20892-1862, USA.
| |
Collapse
|
11
|
Ronchi CL, Peverelli E, Herterich S, Weigand I, Mantovani G, Schwarzmayr T, Sbiera S, Allolio B, Honegger J, Appenzeller S, Lania AG, Reincke M, Calebiro D, Spada A, Buchfelder M, Flitsch J, Strom TM, Fassnacht M. Landscape of somatic mutations in sporadic GH-secreting pituitary adenomas. Eur J Endocrinol 2016; 174:363-72. [PMID: 26701869 DOI: 10.1530/eje-15-1064] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
CONTEXT Alterations in the cAMP signaling pathway are common in hormonally active endocrine tumors. Somatic mutations at GNAS are causative in 30-40% of GH-secreting adenomas. Recently, mutations affecting the USP8 and PRKACA gene have been reported in ACTH-secreting pituitary adenomas and cortisol-secreting adrenocortical adenomas respectively. However, the pathogenesis of many GH-secreting adenomas remains unclear. AIM Comprehensive genetic characterization of sporadic GH-secreting adenomas and identification of new driver mutations. DESIGN Screening for somatic mutations was performed in 67 GH-secreting adenomas by targeted sequencing for GNAS, PRKACA, and USP8 mutations (n=31) and next-generation exome sequencing (n=36). RESULTS By targeted sequencing, known activating mutations in GNAS were detected in five cases (16.1%), while no somatic mutations were observed in both PRKACA and USP8. Whole-exome sequencing identified 132 protein-altering somatic mutations in 31/36 tumors with a median of three mutations per sample (range: 1-13). The only recurrent mutations have been observed in GNAS (31.4% of cases). However, seven genes involved in cAMP signaling pathway were affected in 14 of 36 samples and eight samples harbored variants in genes involved in the calcium signaling or metabolism. At the enrichment analysis, several altered genes resulted to be associated with developmental processes. No significant correlation between genetic alterations and the clinical data was observed. CONCLUSION This study provides a comprehensive analysis of somatic mutations in a large series of GH-secreting adenomas. No novel recurrent genetic alterations have been observed, but the data suggest that beside cAMP pathway, calcium signaling might be involved in the pathogenesis of these tumors.
Collapse
Affiliation(s)
- Cristina L Ronchi
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Erika Peverelli
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Sabine Herterich
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Isabel Weigand
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Giovanna Mantovani
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Thomas Schwarzmayr
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Silviu Sbiera
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Bruno Allolio
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Jürgen Honegger
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Silke Appenzeller
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, W
| | - Andrea G Lania
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Martin Reincke
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Davide Calebiro
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Anna Spada
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Michael Buchfelder
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Joerg Flitsch
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany
| | - Tim M Strom
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, W
| | - Martin Fassnacht
- Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, Wuerzburg, GermanyDepartment of NeurosurgeryUniversity Hospital of Erlangen, Erlangen, GermanyNeurosurgeryUniversity Hospital of Hamburg-Eppendorf, Hamburg, GermanyInstitute of Human GeneticsTechnische Universitaet Muenchen, Munich, Germany Department of Internal Medicine IEndocrine and Diabetes Unit, University Hospital, University of Wuerzburg, Oberduerrbacherstrasse 6, 97080 Wuerzburg, GermanyEndocrinology and Diabetology UnitDepartment of Clinical Sciences and Community Health, University of Milan, Milan, ItalyCentral LaboratoryUniversity Hospital, University of Wuerzburg, Wuerzburg, GermanyInstitute of Human GeneticsHelmholtz Zentrum Munich, Neuherberg, GermanyComprehensive Cancer Center MainfrankenUniversity of Wuerzburg, Wuerzburg, GermanyMedizinische Klinik and Poliklinik IVLudwig-Maximilians University, Munich, GermanyCore Unit Systems MedicineUniversity of Wuerzburg, Wuerzburg, GermanyEndocrinology UnitDepartment of Biomedical Sciences, Humanitas Research Hospital, Humanitas University, Rozzano, Milan, ItalyInstitute of Pharmacology and Toxicology and Bioimaging CenterUniversity of Wuerzburg, W
| |
Collapse
|
12
|
Potorac I, Petrossians P, Daly AF, Schillo F, Ben Slama C, Nagi S, Sahnoun M, Brue T, Girard N, Chanson P, Nasser G, Caron P, Bonneville F, Raverot G, Lapras V, Cotton F, Delemer B, Higel B, Boulin A, Gaillard S, Luca F, Goichot B, Dietemann JL, Beckers A, Bonneville JF. Pituitary MRI characteristics in 297 acromegaly patients based on T2-weighted sequences. Endocr Relat Cancer 2015; 22:169-77. [PMID: 25556181 DOI: 10.1530/erc-14-0305] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Responses of GH-secreting adenomas to multimodal management of acromegaly vary widely between patients. Understanding the behavioral patterns of GH-secreting adenomas by identifying factors predictive of their evolution is a research priority. The aim of this study was to clarify the relationship between the T2-weighted adenoma signal on diagnostic magnetic resonance imaging (MRI) in acromegaly and clinical and biological features at diagnosis. An international, multicenter, retrospective analysis was performed using a large population of 297 acromegalic patients recently diagnosed with available diagnostic MRI evaluations. The study was conducted at ten endocrine tertiary referral centers. Clinical and biochemical characteristics, and MRI signal findings were evaluated. T2-hypointense adenomas represented 52.9% of the series, were smaller than their T2-hyperintense and isointense counterparts (P<0.0001), were associated with higher IGF1 levels (P=0.0001), invaded the cavernous sinus less frequently (P=0.0002), and rarely caused optic chiasm compression (P<0.0001). Acromegalic men tended to be younger at diagnosis than women (P=0.067) and presented higher IGF1 values (P=0.01). Although in total, adenomas had a predominantly inferior extension in 45.8% of cases, in men this was more frequent (P<0.0001), whereas in women optic chiasm compression of macroadenomas occurred more often (P=0.0067). Most adenomas (45.1%) measured between 11 and 20 mm in maximal diameter and bigger adenomas were diagnosed at younger ages (P=0.0001). The T2-weighted signal differentiates GH-secreting adenomas into subgroups with particular behaviors. This raises the question of whether the T2-weighted signal could represent a factor in the classification of acromegalic patients in future studies.
Collapse
Affiliation(s)
- Iulia Potorac
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Patrick Petrossians
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Adrian F Daly
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Franck Schillo
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Claude Ben Slama
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Sonia Nagi
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Mouna Sahnoun
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Thierry Brue
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Nadine Girard
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Philippe Chanson
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Ghaidaa Nasser
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Philippe Caron
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Fabrice Bonneville
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Gérald Raverot
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Véronique Lapras
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - François Cotton
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Brigitte Delemer
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Brigitte Higel
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Anne Boulin
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Stéphan Gaillard
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Florina Luca
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Bernard Goichot
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Jean-Louis Dietemann
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Albert Beckers
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| | - Jean-François Bonneville
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart Tilman, 4000 Liège, BelgiumDepartment of EndocrinologyCHU Besançon, Besançon, FranceDepartments of EndocrinologyNeuroradiologySchool of Medicine, Tunis University, Tunis, TunisiaDepartments of EndocrinologyNeuroradiologyCHU Marseille, Marseille, FranceDepartments of EndocrinologyNeuroradiologyCHU Bicêtre, Le Kremlin-Bicêtre, FranceDepartments of EndocrinologyNeuroradiologyCHU Toulouse, Toulouse, FranceDepartments of EndocrinologyRadiologyHospices Civils de Lyon, Lyon, FranceDepartments of EndocrinologyNeuroradiologyCHU Reims, Reims, FranceDepartments of NeuroradiologyNeurosurgeryCH Foch, Suresnes, FranceDepartments of EndocrinologyNeuroradiologyCHU Strasbourg, Strasbourg, France
| |
Collapse
|
13
|
Abstract
Pituitary adenomas are a heterogeneous group of tumors that may occur as part of a complex syndrome or as an isolated endocrinopathy and both forms can be familial or non-familial. Studies of syndromic and non-syndromic pituitary adenomas have yielded important insights about the molecular mechanisms underlying tumorigenesis. Thus, syndromic forms, including multiple endocrine neoplasia type 1 (MEN1), MEN4, Carney Complex and McCune Albright syndrome, have been shown to be due to mutations of the tumor-suppressor protein menin, a cyclin-dependent kinase inhibitor (p27Kip1), the protein kinase A regulatory subunit 1-α, and the G-protein α-stimulatory subunit (Gsα), respectively. Non-syndromic forms, which include familial isolated pituitary adenoma (FIPA) and sporadic tumors, have been shown to be due to abnormalities of: the aryl hydrocarbon receptor-interacting protein; Gsα; signal transducers; cell cycle regulators; transcriptional modulators and miRNAs. The roles of these molecular abnormalities and epigenetic mechanisms in pituitary tumorigenesis, and their therapeutic implications are reviewed.
Collapse
Affiliation(s)
- Christopher J Yates
- a 1 Academic Endocrine Unit, Radcliffe Department of Clinical Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, Oxfordshire, OX3 7LJ, UK
- b 2 Department of Diabetes and Endocrinology, Melbourne Health, The Royal Melbourne Hospital, Grattan Street, Parkville, Vic 3050, Australia
| | - Kate E Lines
- a 1 Academic Endocrine Unit, Radcliffe Department of Clinical Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, Oxfordshire, OX3 7LJ, UK
| | - Rajesh V Thakker
- a 1 Academic Endocrine Unit, Radcliffe Department of Clinical Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, Oxfordshire, OX3 7LJ, UK
| |
Collapse
|
14
|
MacConaill LE, Garcia E, Shivdasani P, Ducar M, Adusumilli R, Breneiser M, Byrne M, Chung L, Conneely J, Crosby L, Garraway LA, Gong X, Hahn WC, Hatton C, Kantoff PW, Kluk M, Kuo F, Jia Y, Joshi R, Longtine J, Manning A, Palescandolo E, Sharaf N, Sholl L, van Hummelen P, Wade J, Wollinson BM, Zepf D, Rollins BJ, Lindeman NI. Prospective enterprise-level molecular genotyping of a cohort of cancer patients. J Mol Diagn 2014; 16:660-72. [PMID: 25157968 DOI: 10.1016/j.jmoldx.2014.06.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/31/2014] [Accepted: 06/25/2014] [Indexed: 12/22/2022] Open
Abstract
Ongoing cancer genome characterization studies continue to elucidate the spectrum of genomic abnormalities that drive many cancers, and in the clinical arena assessment of the driver genetic alterations in patients is playing an increasingly important diagnostic and/or prognostic role for many cancer types. However, the landscape of genomic abnormalities is still unknown for less common cancers, and the influence of specific genotypes on clinical behavior is often still unclear. To address some of these deficiencies, we developed Profile, a prospective cohort study to obtain genomic information on all patients at a large tertiary care medical center for cancer-related care. We enrolled patients with any cancer diagnosis, and, for each patient (unselected for cancer site or type) we applied mass spectrometric genotyping (OncoMap) of 471 common recurrent mutations in 41 cancer-related genes. We report the results of the first 5000 patients, of which 26% exhibited potentially actionable somatic mutations. These observations indicate the utility of genotyping in advancing the field of precision oncology.
Collapse
Affiliation(s)
- Laura E MacConaill
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts; Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts.
| | - Elizabeth Garcia
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Priyanka Shivdasani
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Matthew Ducar
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ravali Adusumilli
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marc Breneiser
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark Byrne
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lawrence Chung
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jodie Conneely
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lauren Crosby
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Levi A Garraway
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Xin Gong
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - William C Hahn
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Charlie Hatton
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Philip W Kantoff
- Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Michael Kluk
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Frank Kuo
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Yonghui Jia
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ruchi Joshi
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Janina Longtine
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Allison Manning
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Emanuele Palescandolo
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nematullah Sharaf
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Paul van Hummelen
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacqueline Wade
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bruce M Wollinson
- Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Dimity Zepf
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Barrett J Rollins
- Department of Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
15
|
Goto Y, Kinoshita M, Oshino S, Arita H, Kitamura T, Otsuki M, Shimomura I, Yoshimine T, Saitoh Y. Gsp mutation in acromegaly and its influence on TRH-induced paradoxical GH response. Clin Endocrinol (Oxf) 2014; 80:714-9. [PMID: 24111551 DOI: 10.1111/cen.12336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/04/2013] [Accepted: 09/13/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVE We recently reported that paradoxical GH response to TRH administration reflects biological characteristics in patients with acromegaly. The aim of this study is to elucidate the relationship between gsp mutations and the paradoxical GH response to TRH. PATIENTS Sixty-seven patients with acromegaly were included for analysis. Paradoxical increase in serum GH level to TRH, GH suppression by octreotide and bromocriptine, radiological profiles and histopathological findings were analysed with respect to tumour gsp-mutation status. RESULTS Twenty-six (38·8%) gsp mutations were detected, and the number of paradoxical GH responders to TRH, defined as an increase of 100% or more in GH after TRH, was 49 (73·1%). Among the paradoxical GH responders to TRH, 21 patients (42·9%) had a gsp mutation and 28 patients (57·1%) did not. The percentage of paradoxical GH responders to TRH in gsp-positive and gsp-negative patients was not significantly different (80·8% and 68·3%, respectively). The gsp-positive group showed a significantly higher paradoxical increase in serum GH level by TRH administration (1830% vs 650% GH increase, P = 0·045) and greater GH suppression by octreotide (88·7% vs 75·4% GH decrease, P = 0·003) than the gsp-negative group. CONCLUSION Paradoxical GH response to TRH was observed regardless of gsp mutation, although the rate of increase was significantly higher in gsp-positive patients. These results suggest that gsp mutation is not sufficient to cause the paradoxical GH response to TRH, while other unidentified factors have a strong influence on paradoxical GH response to TRH in patients with acromegaly.
Collapse
Affiliation(s)
- Yuko Goto
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Salpea P, Stratakis CA. Carney complex and McCune Albright syndrome: an overview of clinical manifestations and human molecular genetics. Mol Cell Endocrinol 2014; 386:85-91. [PMID: 24012779 PMCID: PMC3943598 DOI: 10.1016/j.mce.2013.08.022] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/25/2022]
Abstract
Endocrine neoplasia syndromes feature a wide spectrum of benign and malignant tumors of endocrine and non-endocrine organs associated with other clinical manifestations. This study outlines the main clinical features, genetic basis, and molecular mechanisms behind two multiple endocrine neoplasia syndromes that share quite a bit of similarities, but one can be inherited whereas the other is always sporadic, Carney complex (CNC) and McCune-Albright (MAS), respectively. Spotty skin pigmentation, cardiac and other myxomas, and different types of endocrine tumors and other characterize Carney complex, which is caused largely by inactivating Protein kinase A, regulatory subunit, type I, Alpha (PRKAR1A) gene mutations. The main features of McCune-Albright are fibrous dysplasia of bone (FD), café-au-lait macules and precocious puberty; the disease is caused by activating mutations in the Guanine Nucleotide-binding protein, Alpha-stimulating activity polypeptide (GNAS) gene which are always somatic. We review the clinical manifestations of the two syndromes and provide an update on their molecular genetics.
Collapse
Affiliation(s)
- Paraskevi Salpea
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology & Genetics (PDEGEN) & Pediatric Endocrinology Inter-Institute Training Program, Eunice Kennedy Shriver, National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology & Genetics (PDEGEN) & Pediatric Endocrinology Inter-Institute Training Program, Eunice Kennedy Shriver, National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Abstract
The cyclic nucleotide cAMP is a universal regulator of a variety of cell functions in response to activated G-protein coupled receptors. In particular, cAMP exerts positive or negative effects on cell proliferation in different cell types. As demonstrated by several in vitro studies, in somatotrophs and in other endocrine cells, cAMP is a mitogenic factor. In agreement with this notion, it has been found that the mutations of genes coding for proteins that contribute to increases in the cAMP signaling cascade may cause endocrine tumor development. This review will discuss the central role of cAMP signaling in the pituitary, focusing on the cAMP pathway alterations involved in pituitary tumorigenesis, as well as on poorly investigated the aspects of cAMP cascade, such as crosstalk with the ERK signaling pathway and new cAMP effectors.
Collapse
Affiliation(s)
- Erika Peverelli
- Endocrine Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Endocrine Unit, IRCCS Humanitas Clinical Institute, Rozzano, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
18
|
Beckers A, Aaltonen LA, Daly AF, Karhu A. Familial isolated pituitary adenomas (FIPA) and the pituitary adenoma predisposition due to mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene. Endocr Rev 2013; 34:239-77. [PMID: 23371967 PMCID: PMC3610678 DOI: 10.1210/er.2012-1013] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pituitary adenomas are one of the most frequent intracranial tumors and occur with a prevalence of approximately 1:1000 in the developed world. Pituitary adenomas have a serious disease burden, and their management involves neurosurgery, biological therapies, and radiotherapy. Early diagnosis of pituitary tumors while they are smaller may help increase cure rates. Few genetic predictors of pituitary adenoma development exist. Recent years have seen two separate, complimentary advances in inherited pituitary tumor research. The clinical condition of familial isolated pituitary adenomas (FIPA) has been described, which encompasses the familial occurrence of isolated pituitary adenomas outside of the setting of syndromic conditions like multiple endocrine neoplasia type 1 and Carney complex. FIPA families comprise approximately 2% of pituitary adenomas and represent a clinical entity with homogeneous or heterogeneous pituitary adenoma types occurring within the same kindred. The aryl hydrocarbon receptor interacting protein (AIP) gene has been identified as causing a pituitary adenoma predisposition of variable penetrance that accounts for 20% of FIPA families. Germline AIP mutations have been shown to associate with the occurrence of large pituitary adenomas that occur at a young age, predominantly in children/adolescents and young adults. AIP mutations are usually associated with somatotropinomas, but prolactinomas, nonfunctioning pituitary adenomas, Cushing disease, and other infrequent clinical adenoma types can also occur. Gigantism is a particular feature of AIP mutations and occurs in more than one third of affected somatotropinoma patients. Study of pituitary adenoma patients with AIP mutations has demonstrated that these cases raise clinical challenges to successful treatment. Extensive research on the biology of AIP and new advances in mouse Aip knockout models demonstrate multiple pathways by which AIP may contribute to tumorigenesis. This review assesses the current clinical and therapeutic characteristics of more than 200 FIPA families and addresses research findings among AIP mutation-bearing patients in different populations with pituitary adenomas.
Collapse
Affiliation(s)
- Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium.
| | | | | | | |
Collapse
|
19
|
Cozzi R, Attanasio R. Octreotide long-acting repeatable for acromegaly. Expert Rev Clin Pharmacol 2012; 5:125-43. [PMID: 22390555 DOI: 10.1586/ecp.12.4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acromegaly remains a therapeutic challenge for the endocrinologist. Among the available therapeutic options, octreotide long-acting repeatable (Sandostatin(®) LAR(®), Novartis) plays a chief role, both as a primary therapy and as an adjuvant treatment after unsuccessful surgery. A plethora of papers and a meta-analysis have demonstrated its efficacy in: control of clinical picture; achievement of safe growth hormone and normal age-matched IGF-I levels (both factors associated with restoration of normal life expectancy) in 60-70% of patients; control of tumor volume (with real shrinkage in over half of cases); and halt or reversal of most acromegaly-associated comorbidities. Treatment is well tolerated in most patients and can be safely prolonged for many years if required.
Collapse
Affiliation(s)
- Renato Cozzi
- Division of Endocrinology, Ospedale Niguarda, Via Canonica 81, I-20154 Milan, Italy.
| | | |
Collapse
|
20
|
Szalontay L, Benveniste RJ, Schally AV, Vidaurre I, Nadji M, Zarandi M, Block NL, Kovacs M. Inhibitory effects of GHRH antagonists on human GH-secreting adenoma tissue. Neuroendocrinology 2012; 96:81-8. [PMID: 22377963 DOI: 10.1159/000335989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 12/06/2011] [Indexed: 11/19/2022]
Abstract
Experimental data indicate that antagonists of growth hormone-releasing hormone (GHRH) could be used clinically in disorders characterized by excessive GHRH/growth hormone (GH) secretion, but direct evidence for the effectiveness of GHRH antagonists on human pituitary tissue is still lacking. In this study, we investigated the inhibitory effect of our GHRH antagonists MZ-4-71 and JV-1-36 and the somatostatin (SST) analog RC-160 on superfused pituitary cells obtained from a human GH-secreting adenoma. Using Western blot analysis and immunohistochemistry, we demonstrated profuse expression of the GHRH receptor and its major splice variant SV1 and an increase in the expression of Gsa protein in the adenoma tissue. Exposure of the tumor cells to exogenous pulses of GHRH induced definite GH responses, causing a 3- to 5-fold elevation of the basal GH level. The antagonists MZ-4-71 and JV-1-36 did not alter basal GH secretion, indicating that the adenoma cells did not secrete GHRH in an autocrine manner. However, both antagonists prevented the stimulatory effect of exogenous GHRH. Similarly to the GHRH antagonists, neither SST-14 nor the SST analog RC-160 had an effect on the basal GH secretion of the tumor cells, but both peptides inhibited the stimulatory effect of exogenous GHRH, with RC-160 being more potent than SST. Our study provides direct evidence for the effectiveness of potent GHRH antagonists such as MZ-4-71 and JV-1-36 on human pituitary GH-secreting adenoma tissue and strongly suggests that these drugs could be used for therapy of GHRH-associated forms of acromegaly, particularly for those patients in whom surgery fails or is not an option.
Collapse
Affiliation(s)
- Luca Szalontay
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Cuny T, Gerard C, Saveanu A, Barlier A, Enjalbert A. Physiopathology of somatolactotroph cells: from transduction mechanisms to cotargeting therapy. Ann N Y Acad Sci 2011; 1220:60-70. [PMID: 21388404 DOI: 10.1111/j.1749-6632.2010.05924.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In pituitary somatolactotroph cells, G protein-coupled receptors and receptor tyrosine kinases binding their specific ligands trigger an enzymatic cascade that converges to MAP kinase activation in the subcellular compartment. Different signaling pathways, such as AC/cAMP/PKA and PI3K/Akt pathways, interact with MAP kinase to regulate key physiological functions, such as hormonal secretion and cell proliferation. Abnormalities affecting these signaling pathways have been identified as preponderant factors of pituitary tumorigenesis. In addition to trans-sphenoidal surgery, somatostatin analogs are used to control hormonal hypersecretion in GH-secreting adenomas. However, a subset of these tumors remains uncontrolled with these treatFments, calling for new therapeutic approaches. In these cases, novel multivalent somatostatin analogs or new somatostatin-dopamine chimeric molecules could be of interest. Another attractive therapeutic approach may be to use one or several inhibitors acting downstream in the signaling pathway, such as mammalian target of rapamycin inhibitor. Cotargeting therapy and gene therapy are promising tools for these problematic pituitary tumors.
Collapse
Affiliation(s)
- Thomas Cuny
- Research Center of Neurobiology and Neurophysiology of Marseille, CRN2M, UMR 6231 CNRS, University of Mediterranée, Institut Fédératif Jean Roche, Marseille, France.
| | | | | | | | | |
Collapse
|
22
|
Pertuit M, Romano D, Zeiller C, Barlier A, Enjalbert A, Gerard C. The gsp oncogene disrupts Ras/ERK-dependent prolactin gene regulation in gsp inducible somatotroph cell line. Endocrinology 2011; 152:1234-43. [PMID: 21285319 DOI: 10.1210/en.2010-1077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The MAPK ERK1/2 cascade regulates all the critical cellular functions, and in many pathological situations, these regulatory processes are perturbed. It has been clearly established that this cascade is an integrative point in the control of the pituitary functions exerted by various extracellular signals. In particular, ERK1/2 cross talk with the cAMP pathway is determinant in the control of somatolactotroph hormonal secretion exerted via neuropeptide receptors. GH-secreting adenomas are characterized by frequent cAMP pathway alterations, such as constitutive activation of the α-subunit of the heterotrimeric Gs protein (the gsp oncogene), overexpression of Gsα, and changes in the protein kinase A regulatory subunits. However, it has not yet been established exactly how these alterations result in GH-secreting adenomas or how the ERK1/2 cascade contributes to the process of GH-secreting adenoma tumorigenesis. In this study on the conditional gsp-oncogene-expressing GH4C1 cell line, expression of the gsp oncogene, which was observed in up to 40% of GH-secreting adenomas, was found to induce sustained ERK1/2 activation, which required activation of the protein kinase A and the GTPases Ras and Rap1. All these signaling components contribute to the chronic activation of the human prolactin promoter. The data obtained here show that Ras plays a crucial role in these processes: in a physiopathological context, i.e. in the presence of the gsp oncogene, it switched from being a repressor of the cAMP/ protein kinase A ERK-sensitive prolactin gene control exerted by neuropeptides to an activator of the prolactin promoter.
Collapse
Affiliation(s)
- M Pertuit
- CRN2M, Unité Mixte de Recherche 6231, Department of Neuroendocrinology-Neuroimmunology, Institut Fédératif Jean-Roche, Faculté de Médecine Secteur Nord, Université de la Méditerranée CS80011, Boulevard Pierre Dramard, 13344 Marseille cedex 15, France
| | | | | | | | | | | |
Collapse
|
23
|
Izzi B, Decallonne B, Devriendt K, Bouillon R, Vanderschueren D, Levtchenko E, de Zegher F, Van den Bruel A, Lambrechts D, Van Geet C, Freson K. A new approach to imprinting mutation detection in GNAS by Sequenom EpiTYPER system. Clin Chim Acta 2010; 411:2033-9. [PMID: 20807523 DOI: 10.1016/j.cca.2010.08.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 08/19/2010] [Accepted: 08/24/2010] [Indexed: 11/28/2022]
Abstract
BACKGROUND Pseudohypoparathyroidism type Ib (PHPIb) results from abnormal imprinting of GNAS. Familial and sporadic forms of PHPIb have distinct GNAS imprinting patterns: familial PHPIb patients have an exon A/B-only imprinting defect and an intragenic STX16 deletion, whereas sporadic PHPIb cases have abnormal imprinting of the three differentially methylated regions (DMRs) in GNAS without the STX16 deletion. Overall GNAS methylation defects have recently been detected in some PHPIa patients. METHODS This study describes the first quantitative methylation analysis of multiple CpG sites for three different GNAS DMRs using the Sequenom EpiTYPER in 35 controls, 12 PHPIb patients, 2 PHPIa patients and 2 patients without parathormone (PTH) resistance but having only hypocalcemia and hyperphosphatemia. RESULTS All patients have GNAS methylation defects typically with NESP hypermethylation versus XL and exon A/B hypomethylation while the imprinting of SNURF/SNRPN was normal. PHPIa patients showed an abnormal methylation in the three DMRs of GNAS. For the first time, a marked abnormal GNAS methylation was also found in 2 patients without PTH resistance but having hypocalcemia and hyperphosphatemia. CONCLUSIONS The Sequenom EpiTYPER proves to be very sensitive in detecting DNA methylation changes. Our analysis also suggests that GNAS imprinting defects might be more frequent and diverse than previously thought.
Collapse
Affiliation(s)
- Benedetta Izzi
- Center for Molecular and Vascular Biology, University of Leuven, Leuven, 3000, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Xekouki P, Azevedo M, Stratakis CA. Anterior pituitary adenomas: inherited syndromes, novel genes and molecular pathways. Expert Rev Endocrinol Metab 2010; 5:697-709. [PMID: 21264206 PMCID: PMC3024595 DOI: 10.1586/eem.10.47] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pituitary adenomas are common tumors. Although rarely malignant, pituitary adenomas cause significant morbidity due to mass effects and/or hormonal hypo- and/or hyper-secretion. Molecular understanding of pituitary adenoma formation is essential for the development of medical therapies and the treatment of post-operative recurrences. In general, mutations in genes involved in genetic syndromes associated with pituitary tumors are not a common finding in sporadic lesions. By contrast, multiple endocrine neoplasia type 1 (MEN-1) and aryl hydrocarbon receptor-interacting protein (AIP) mutations may be more frequent among specific subgroups of patients, such as children and young adults, with growth hormone-producing adenomas. In this article, we present the most recent data on the molecular pathogenesis of pituitary adenomas and discuss some of the most recent findings from our laboratory. Guidelines for genetic screening and clinical counseling of patients with pituitary tumors are provided.
Collapse
Affiliation(s)
- Paraskevi Xekouki
- SEGEN, PDEGEN & Pediatric Endocrinology Program, NICHD, NIH, Building 10, CRC (East Laboratories), Room 1-3330, 10 Center Drive, MSC1103, Bethesda, MD 20892, USA
| | - Monalisa Azevedo
- SEGEN, PDEGEN & Pediatric Endocrinology Program, NICHD, NIH, Building 10, CRC (East Laboratories), Room 1-3330, 10 Center Drive, MSC1103, Bethesda, MD 20892, USA
| | - Constantine A Stratakis
- SEGEN, PDEGEN & Pediatric Endocrinology Program, NICHD, NIH, Building 10, CRC (East Laboratories), Room 1-3330, 10 Center Drive, MSC1103, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Pertuit M, Barlier A, Enjalbert A, Gérard C. Signalling pathway alterations in pituitary adenomas: involvement of Gsalpha, cAMP and mitogen-activated protein kinases. J Neuroendocrinol 2009; 21:869-77. [PMID: 19732293 DOI: 10.1111/j.1365-2826.2009.01910.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite extensive research on sporadic pituitary adenomas, it is not yet possible to assign one protein alteration to one specific type of pituitary adenomas. Nevertheless, alterations of the cAMP pathway appear to be molecular hallmarks of most growth hormone (GH)-secreting adenomas. However, these alterations do not confer specific phenotypes to patients carrying these alterations. In this review, we summarise the literature regarding signalling alterations observed in GH-secreting adenomas. We focus on Gsalpha alterations and their possible cross-talk with the extracellular signal-related kinase (ERK)1/2 pathway. In the light of results obtained on human somatotroph adenoma cells in primary culture and on models of murine somatotroph cell lines, we postulate a crucial role for ERK1/2 in GH-secreting adenomas downstream of cAMP pathway alterations that might impact the tumoural phenotype.
Collapse
Affiliation(s)
- M Pertuit
- CRN2M, UMR 6231, CNRS, Department of Neuroendocrinology-Neuroimmunology, Institut Fédératif Jean-Roche, Faculté de Médecine Secteur Nord, Université de Méditerranée, Marseille, France
| | | | | | | |
Collapse
|
26
|
Theodoropoulou M, Tichomirowa MA, Sievers C, Yassouridis A, Arzberger T, Hougrand O, Deprez M, Daly AF, Petrossians P, Pagotto U, Beckers A, Stalla GK. Tumor ZAC1 expression is associated with the response to somatostatin analog therapy in patients with acromegaly. Int J Cancer 2009; 125:2122-6. [PMID: 19637311 DOI: 10.1002/ijc.24602] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Somatostatin analogs (SSA) with their potent antisecretory and antiproliferative effects are the main medical treatment option for patients with neuroendocrine tumors, such as gastroenteropancreatic and acromegaly-associated growth hormone secreting pituitary tumors. Although a good portion of acromegalic patients gets normalized after SSA treatment, strict hormonal control is not achieved in a sizeable proportion of these patients. The reasons for this incomplete response to SSA treatment are unclear. We have found that the tumor suppressor ZAC1 (LOT1/PLAGL1) is essential for the antiproliferative effect of SSA in pituitary tumor cells. The aim of the present retrospective cohort study was to determine whether ZAC1 immunoreactivity in archival somatotrophinoma tissue derived from 45 patients with acromegaly routinely pretreated with SSA before surgery, was associated with response to SSA (normalization of GH, IGF-I and presence of tumor shrinkage). All tumors displayed ZAC1 immunoreactivity [weak (+; n = 15), moderate (++; n = 16) and strong (+++; n = 14)]. A significant positive correlation was found between strong ZAC1 immunoreactivity and IGF-I normalization and presence of tumor shrinkage after SSA treatment, which was not affected by age at diagnosis, gender or duration of SSA treatment. These in vivo data combined with the antiproliferative properties of ZAC1/Zac1 provide evidence of a mechanistic role for this transcription factor on SSA induced tumor shrinkage and hormone normalization.
Collapse
Affiliation(s)
- Marily Theodoropoulou
- Department of Endocrinology, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Acunzo J, Saveanu A, Gérard C, Enjalbert A, Barlier A. Mécanismes de tumorigenèse hypophysaire. Presse Med 2009; 38:76-83. [DOI: 10.1016/j.lpm.2008.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 09/18/2008] [Indexed: 10/21/2022] Open
|
28
|
Bollerslev J, Fougner SL, Berg JP. New directions in pharmacological treatment of acromegaly. Expert Opin Investig Drugs 2008; 18:13-22. [DOI: 10.1517/13543780802554357] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
29
|
Faissolle P, Wagner-Mahler K, Mas JC, Moreigne M, Khalfi A, Léonetti V, Silvy M, Barlier A, Giudicelli H. Une cause de calcifications intracérébrales à connaître : la pseudohypoparathyroïdie de type Ib. Arch Pediatr 2008; 15:1433-6. [DOI: 10.1016/j.arcped.2008.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 04/24/2008] [Accepted: 06/19/2008] [Indexed: 12/22/2022]
|
30
|
Evans CO, Moreno CS, Zhan X, McCabe MT, Vertino PM, Desiderio DM, Oyesiku NM. Molecular pathogenesis of human prolactinomas identified by gene expression profiling, RT-qPCR, and proteomic analyses. Pituitary 2008; 11:231-45. [PMID: 18183490 DOI: 10.1007/s11102-007-0082-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The molecular pathogenesis of prolactinomas has resisted elucidation; with the exception of a RAS mutation in a single aggressive prolactinoma, no mutational changes have been identified. In prolactinomas, a further obstacle has been the paucity of surgical specimens suitable for molecular analysis since prolactionomas are infrequently removed due to the availability and effectiveness of medical therapy. In the absence of mutational events, gene expression changes have been sought and detected. Using high-throughput analysis from a large bank of human pituitary adenomas, we examined these tumors according to their molecular profiles rather than traditional immunohistochemistry. We examined six prolactinomas and eight normal pituitary glands using oligonucleotide GeneChip microarrays, reverse transcription-real time quantitative polymerase chain reaction using 10 prolactinomas, and proteomic analysis to examine protein expression in four prolactinomas. Microarray analyses identified 726 unique genes that were statistically significantly different between prolactinomas and normal glands, whereas proteomic analysis identified four differently up-regulated and 19 down-regulated proteins. Several components of the Notch pathway were altered in prolactinomas, and there was an increased expression of the Pit-1 transcription factor, and the survival factor BAG1 but decreased E-cadherin and N-cadherin expression. Taken together, expression profiling and proteomic analyses have identified molecular features unique to prolactinomas that may contribute to their pathogenesis. In the current era of molecular medicine, these findings greatly enhance our understanding and supercede immunohistochemical diagnosis.
Collapse
Affiliation(s)
- Chheng-Orn Evans
- Department of Neurosurgery and Laboratory of Molecular Neurosurgery and Biotechnology, Emory University School of Medicine, 1365 B Clifton Rd., NE, Suite. 6200, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | |
Collapse
|