1
|
Vamvoukaki R, Chrysoulaki M, Betsi G, Xekouki P. Pituitary Tumorigenesis-Implications for Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040812. [PMID: 37109772 PMCID: PMC10145673 DOI: 10.3390/medicina59040812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs), the third most common intracranial tumor, are mostly benign. However, some of them may display a more aggressive behavior, invading into the surrounding structures. While they may rarely metastasize, they may resist different treatment modalities. Several major advances in molecular biology in the past few years led to the discovery of the possible mechanisms involved in pituitary tumorigenesis with a possible therapeutic implication. The mutations in the different proteins involved in the Gsa/protein kinase A/c AMP signaling pathway are well-known and are responsible for many PitNETS, such as somatotropinomas and, in the context of syndromes, as the McCune-Albright syndrome, Carney complex, familiar isolated pituitary adenoma (FIPA), and X-linked acrogigantism (XLAG). The other pathways involved are the MAPK/ERK, PI3K/Akt, Wnt, and the most recently studied HIPPO pathways. Moreover, the mutations in several other tumor suppressor genes, such as menin and CDKN1B, are responsible for the MEN1 and MEN4 syndromes and succinate dehydrogenase (SDHx) in the context of the 3PAs syndrome. Furthermore, the pituitary stem cells and miRNAs hold an essential role in pituitary tumorigenesis and may represent new molecular targets for their diagnosis and treatment. This review aims to summarize the different cell signaling pathways and genes involved in pituitary tumorigenesis in an attempt to clarify their implications for diagnosis and management.
Collapse
Affiliation(s)
- Rodanthi Vamvoukaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Maria Chrysoulaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Grigoria Betsi
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| |
Collapse
|
2
|
Helderman N, Lucas M, Blank C. Autoantibodies involved in primary and secondary adrenal insufficiency following treatment with immune checkpoint inhibitors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 17:100374. [PMID: 36937704 PMCID: PMC10014276 DOI: 10.1016/j.iotech.2023.100374] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Primary and secondary adrenal insufficiency (AI) are commonly known immune-related adverse events following treatment with immune checkpoint inhibitors (ICIs), and are clinically relevant due to their morbidity and potential mortality. For this reason, upfront identification of patients susceptible for ICI-induced AI could be a step in improving patient's safety. Multiple studies have focused on the identification of novel biomarkers for ICI-induced AI, including autoantibodies, which may be involved in ICI-induced AI as a result of the T-cell-mediated activation of autoreactive B cells. This review highlights the currently described autoantibodies that may be involved in either primary [e.g. anti-21-hydroxylase, anti-17α-hydroxylase, anti-P450scc, anti-aromatic L-amino acid decarboxylase (AADC), anti-interferon (IFN)α and anti-IFNΩ] or secondary AI [e.g. anti-guanine nucleotide-binding protein G(olf) subunit alpha (GNAL), anti-integral membrane protein 2B (ITM2B), anti-zinc finger CCHC-type containing 8 (ZCCHC8), anti-pro-opiomelanocortin (POMC), anti-TPIT (corticotroph-specific transcription factor), anti-pituitary-specific transcriptional factor-1 (PIT-1) and others], and discusses the current evidence concerning their role as biomarker for ICI-induced AI. Standardized autoantibody measurements in patients (to be) treated with ICIs would be a clinically accessible and patient-friendly screening method to identify the patients at risk, and could change the management of ICI-induced AI.
Collapse
Affiliation(s)
- N.C. Helderman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - M.W. Lucas
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam
| | - C.U. Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam
- Correspondence to: Prof. Dr Christian U. Blank, Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121 A, 1066 CX, Amsterdam, The Netherlands. Tel: +31-(0)20-512-9111
| |
Collapse
|
3
|
Taylor J, Gandhi A, Gray E, Zaenker P. Checkpoint inhibitor immune-related adverse events: A focused review on autoantibodies and B cells as biomarkers, advancements and future possibilities. Front Immunol 2023; 13:991433. [PMID: 36713389 PMCID: PMC9874109 DOI: 10.3389/fimmu.2022.991433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/07/2022] [Indexed: 01/13/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has evolved rapidly with unprecedented treatment benefits being obtained for cancer patients, including improved patient survival. However, over half of the patients experience immune related adverse events (irAEs) or toxicities, which can be fatal, affect the quality of life of patients and potentially cause treatment interruption or cessation. Complications from these toxicities can also cause long term irreversible organ damage and other chronic health conditions. Toxicities can occur in various organ systems, with common observations in the skin, rheumatologic, gastrointestinal, hepatic, endocrine system and the lungs. These are not only challenging to manage but also difficult to detect during the early stages of treatment. Currently, no biomarker exists to predict which patients are likely to develop toxicities from ICI therapy and efforts to identify robust biomarkers are ongoing. B cells and antibodies against autologous antigens (autoantibodies) have shown promise and are emerging as markers to predict the development of irAEs in cancer patients. In this review, we discuss the interplay between ICIs and toxicities in cancer patients, insights into the underlying mechanisms of irAEs, and the involvement of the humoral immune response, particularly by B cells and autoantibodies in irAE development. We also provide an appraisal of the progress, key empirical results and advances in B cell and autoantibody research as biomarkers for predicting irAEs. We conclude the review by outlining the challenges and steps required for their potential clinical application in the future.
Collapse
Affiliation(s)
- John Taylor
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia,*Correspondence: John Taylor,
| | - Aesha Gandhi
- Sir Charles Gairdner Hospital, Department of Medical Oncology, Nedlands, WA, Australia
| | - Elin Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Pauline Zaenker
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
4
|
cAMP Signalling Pathway in Biocontrol Fungi. Curr Issues Mol Biol 2022; 44:2622-2634. [PMID: 35735620 PMCID: PMC9221721 DOI: 10.3390/cimb44060179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 01/07/2023] Open
Abstract
Biocontrol is a complex process, in which a variety of physiological and biochemical characteristics are altered. The cAMP signalling pathway is an important signal transduction pathway in biocontrol fungi and consists of several key components. The G-protein system contains G-protein coupled receptors (GPCRs), heterotrimeric G-proteins, adenylate cyclase (AC), cAMP-dependent protein kinase (PKA), and downstream transcription factors (TFs). The cAMP signalling pathway can regulate fungal growth, development, differentiation, sporulation, morphology, secondary metabolite production, environmental stress tolerance, and the biocontrol of pathogens. However, few reviews of the cAMP signalling pathway in comprehensive biocontrol processes have been reported. This work reviews and discusses the functions and applications of genes encoding each component in the cAMP signalling pathway from biocontrol fungi, including the G-protein system components, AC, PKA, and TFs, in biocontrol behaviour. Finally, future suggestions are provided for constructing a complete cAMP signalling pathway in biocontrol fungi containing all the components and downstream effectors involved in biocontrol behavior. This review provides useful information for the understanding the biocontrol mechanism of biocontrol fungi by utilising the cAMP signalling pathway.
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
|
Bogner EM, Daly AF, Gulde S, Karhu A, Irmler M, Beckers J, Mohr H, Beckers A, Pellegata NS. miR-34a is upregulated in AIP-mutated somatotropinomas and promotes octreotide resistance. Int J Cancer 2020; 147:3523-3538. [PMID: 32856736 DOI: 10.1002/ijc.33268] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 07/15/2020] [Accepted: 08/12/2020] [Indexed: 12/17/2022]
Abstract
Pituitary adenomas (PAs) are intracranial tumors associated with significant morbidity due to hormonal dysregulation, mass effects and have a heavy treatment burden. Growth hormone (GH)-secreting PAs (somatotropinomas) cause acromegaly-gigantism. Genetic forms of somatotropinomas due to germline AIP mutations (AIPmut+) have an early onset and are aggressive and resistant to treatment with somatostatin analogs (SSAs), including octreotide. The molecular underpinnings of these clinical features remain unclear. We investigated the role of miRNA dysregulation in AIPmut+ vs AIPmut- PA samples by array analysis. miR-34a and miR-145 were highly expressed in AIPmut+ vs AIPmut- somatotropinomas. Ectopic expression of AIPmut (p.R271W) in Aip-/- mouse embryonic fibroblasts (MEFs) upregulated miR-34a and miR-145, establishing a causal link between AIPmut and miRNA expression. In PA cells (GH3), miR-34a overexpression promoted proliferation, clonogenicity, migration and suppressed apoptosis, whereas miR-145 moderately affected proliferation and apoptosis. Moreover, high miR-34a expression increased intracellular cAMP, a critical mitogenic factor in PAs. Crucially, high miR-34a expression significantly blunted octreotide-mediated GH inhibition and antiproliferative effects. miR-34a directly targets Gnai2 encoding Gαi2, a G protein subunit inhibiting cAMP production. Accordingly, Gαi2 levels were significantly lower in AIPmut+ vs AIPmut- PA. Taken together, somatotropinomas with AIP mutations overexpress miR-34a, which in turn downregulates Gαi2 expression, increases cAMP concentration and ultimately promotes cell growth. Upregulation of miR-34a also impairs the hormonal and antiproliferative response of PA cells to octreotide. Thus, miR-34a is a novel downstream target of mutant AIP that promotes a cellular phenotype mirroring the aggressive clinical features of AIPmut+ acromegaly.
Collapse
Affiliation(s)
- Eva-Maria Bogner
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Adrian F Daly
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, Liège Université, Liège, Belgium
| | - Sebastian Gulde
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Auli Karhu
- Department of Medical and Clinical Genetics & Genome-Scale Biology Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Technische Universität München, Chair of Experimental Genetics, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, Liège Université, Liège, Belgium
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
7
|
Pinto EM, Faucz FR, Paza LZ, Wu G, Fernandes ES, Bertherat J, Stratakis CA, Lalli E, Ribeiro RC, Rodriguez-Galindo C, Figueiredo BC, Zambetti GP. Germline Variants in Phosphodiesterase Genes and Genetic Predisposition to Pediatric Adrenocortical Tumors. Cancers (Basel) 2020; 12:506. [PMID: 32098292 PMCID: PMC7072638 DOI: 10.3390/cancers12020506] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Phosphodiesterases (PDEs) form a superfamily of enzymes that catalyze the hydrolysis of cyclic nucleotides adenosine 3'5'-cyclic monophosphate (cAMP) and guanosine 3'5'-cyclic monophosphate (cGMP) to their inactive 5' monophosphates. cAMP plays a critical role as a second messenger in endocrine tissues, and activation of cAMP signaling has been reported in endocrine tumors. Germline variants in PDEs have been associated with benign cortisol-secreting adrenocortical adenomas and testicular germ cell cancer but not adrenocortical carcinoma. We performed whole genome sequencing (WGS) and whole exome sequencing (WES) of paired blood and tumor samples from 37 pediatric adrenocortical tumors (ACTs). Germline inactivating variants in PDEs were observed in 9 of 37 (24%) patients. Tumor DNA analysis revealed loss of heterozygosity, with maintenance of the mutated allele in all cases. Our results suggest that germline variants in PDEs and other regulators of the cAMP-signaling pathway may contribute to pediatric adrenocortical tumorigenesis, perhaps by cooperating with germline hypomorphic mutant TP53 alleles and uniparental disomy of chromosome 11p15 (Beckwith-Wiedemann syndrome).
Collapse
Affiliation(s)
- Emilia Modolo Pinto
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105-2794, USA
| | - Fabio R. Faucz
- Section on Genetics and Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD 20892-1862, USA; (F.R.F.); (C.A.S.)
| | - Luana Z. Paza
- Pele Pequeno Principe Research Institute and Faculdades Pequeno Principe, Curitiba PR 80250-200, Brazil;
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38105-2794, USA;
| | - Elizabeth S. Fernandes
- Pele Pequeno Principe Research Institute, Faculdades Pequeno Principe, Curitiba PR 80250-200, Brazil;
| | - Jerome Bertherat
- Institut Cochin, Université Paris Descartes, Paris. Service d’Endocrinologie, Centre de référence des maladies rares de la surrénale, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, F-75014 Paris, France;
| | - Constantine A. Stratakis
- Section on Genetics and Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD 20892-1862, USA; (F.R.F.); (C.A.S.)
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, 06560 Valbonne, France;
| | - Raul C. Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105-2794, USA;
| | - Carlos Rodriguez-Galindo
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105-2794, USA;
| | - Bonald C. Figueiredo
- Pele Pequeno Principe Research Institute, Faculdades Pequeno Principe, Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC) and Departamento de Saúde Coletiva, Federal University of Paraná, Curitiba PR 80250-200, Brazil;
| | - Gerard P. Zambetti
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105-2794, USA
| |
Collapse
|
8
|
Wang X, Fang Y, Zhou Y, Guo X, Xu K, Li C, Zhang J, Hong Y. SDF-1α/MicroRNA-134 Axis Regulates Nonfunctioning Pituitary Neuroendocrine Tumor Growth via Targeting VEGFA. Front Endocrinol (Lausanne) 2020; 11:566761. [PMID: 33362712 PMCID: PMC7756115 DOI: 10.3389/fendo.2020.566761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Nonfunctioning pituitary neuroendocrine tumor (NF-PitNET) is difficult to resect. Except for surgery, there is no effective treatment for NF-PitNET. MicroRNA-134 (miR-134) has been reported to inhibit proliferation and invasion ability of tumor cells. Herein, the mechanism underlying the effect of miR-134 on alleviating NF-PitNET tumor cells growth is explored. METHODS Mouse pituitary αT3-1 cells were transfected with miR-134 mimics and inhibitor, followed by treatment with stromal cell-derived factor-1α (SDF-1α) in vitro. MiR-134 expression level: we used quantitative real-time PCR (qRT-PCR) to detect the expression of miR-134. Cell behavior level: cell viability and invasion ability were assessed using a cell counting kit-8 (CCK8) assay and Transwell invasion assay respectively. Cytomolecular level: tumor cell proliferation was evaluated by Ki-67 staining; propidium iodide (PI) staining analyzed the effect of miR-134 on cell cycle arrest; western blot analysis and immunofluorescence staining evaluated tumor migration and invasive ability. Additionally, we collected 27 NF-PitNET tumor specimens and related clinical data. The specimens were subjected to qRT-PCR to obtain the relative miR-134 expression level of each specimen; linear regression analysis was used to analyze the miR-134 expression level in tumor specimens and the age of the NF-PitNET population, gender, tumor invasion, prognosis, and other indicators. RESULTS In vitro experiment, miR-134 was observed to significantly inhibit αT3-1 cells proliferation characterized by inhibited cell viability and expressions of vascular endothelial growth factor A (VEGFA) and cell cycle transition from G1 to S phase (P < 0.01). VEGFA was verified as a target of miR-134. Additionally, miR-134-induced inhibition of αT3-1 cell proliferation and invasion was attenuated by SDF-1α and VEGFA overexpression (P < 0.01). In primary NF-PitNET tumor analysis, miR-134 expression level was negatively correlated with tumor invasion (P = 0.003). CONCLUSION The regulation of the SDF-1α/miR-134/VEGFA axis represents a novel mechanism in the pathogenesis of NF-PitNETs and may serve as a potential therapeutic target for the treatment of NF-PitNETs.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoming Guo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ke Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chenguang Li
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
- *Correspondence: Jianmin Zhang, ; Yuan Hong,
| | - Yuan Hong
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- *Correspondence: Jianmin Zhang, ; Yuan Hong,
| |
Collapse
|
9
|
Autoimmune antibodies correlate with immune checkpoint therapy-induced toxicities. Proc Natl Acad Sci U S A 2019; 116:22246-22251. [PMID: 31611368 DOI: 10.1073/pnas.1908079116] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint (IC) therapy provides substantial benefits to cancer patients but can also cause distinctive toxicities termed immune-related adverse events (irAEs). Biomarkers to predict toxicities will be necessary to improve management of patients receiving IC therapy. We relied on serological analysis of recombinant cDNA expression libraries to evaluate plasma samples from patients treated with IC therapy and identified autoantibodies, both in pretreatment and on-treatment samples prior to the development of irAEs, which correlate with the development of immune-related hypophysitis (anti-GNAL and anti-ITM2B autoantibodies) and pneumonitis (anti-CD74 autoantibody). We developed an enzyme-linked immunosorbent assay and tested additional patient samples to confirm our initial findings. Collectively, our data suggest that autoantibodies may correlate with irAEs related to IC therapy, and specific autoantibodies may be detected early for the management of irAEs.
Collapse
|
10
|
Tang C, Zhong C, Cong Z, Yang J, Wen G, Zhu J, Ma C. MEG3 is associated with gsp oncogene regulation of growth hormone hypersecretion, proliferation and invasiveness of human GH-secreting adenomas. Oncol Lett 2019; 17:3495-3502. [PMID: 30867789 DOI: 10.3892/ol.2019.10006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 11/07/2018] [Indexed: 11/05/2022] Open
Abstract
Overactivation of the Gs-mediated pathway by mutations of the G-protein α subunit (Gsα), a gsp oncogene, results in increased growth hormone (GH) hypersecretion and reduced tumor volume in patients with GH-secreting pituitary tumors. However, the mechanism underlying the clinical characteristics of gsp oncogene requires further investigation. Cyclic adenosine monophosphate-responsive element binding (CREB), as a downstream target gene of gsp oncogene, is implicated in activating maternally expressed gene 3 (MEG3). The present study proposes that gsp oncogene mediates MEG3-regulating GH hypersecretion, resulting in the small tumor size of GH-secreting tumors. Therefore, the present study detected Gsα mutations by polymerase chain reaction in GH-secreting tumors, and revealed that Gsα mutations were observed in 7/25 (28%) GH-secreting tumors. Gsp-positive tumors indicated significantly increased levels of phosphorylated p-CREB (P<0.0001) and MEG3 (P=0.039), compared with gsp-negative tumors. The results indicated that MEG3 levels were positively correlated with GH and IGF-1 levels, and negatively correlated with the tumor volume of GH-secreting tumors. The group with gsp-positive or with high MEG3 expression indicated a significantly reduced proportion of invasiveness and lower Ki-67 index, compared with the gsp-negative or low MEG3 expression group. In conclusion, gsp oncogene may mediate MEG3 by promoting GH hypersecretion, resulting in smaller tumors, as well as suppressing proliferation and invasiveness of GH-secreting pituitary tumors.
Collapse
Affiliation(s)
- Chao Tang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Chunyu Zhong
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Zixiang Cong
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jin Yang
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Guodao Wen
- Department of Neurosurgery, Dongguan Donghua Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Junhao Zhu
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Chiyuan Ma
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
11
|
Bizzi MF, Bolger GB, Korbonits M, Ribeiro-Oliveira Jr. A. Phosphodiesterases and cAMP Pathway in Pituitary Diseases. Front Endocrinol (Lausanne) 2019; 10:141. [PMID: 30941100 PMCID: PMC6433792 DOI: 10.3389/fendo.2019.00141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
Human phosphodiesterases (PDEs) comprise a complex superfamily of enzymes derived from 24 genes separated into 11 PDE gene families (PDEs 1-11), expressed in different tissues and cells, including heart and brain. The isoforms PDE4, PDE7, and PDE8 are specific for the second messenger cAMP, which is responsible for mediating diverse physiological actions involving different hormones and neurotransmitters. The cAMP pathway plays an important role in the development and function of endocrine tissues while phosphodiesterases are responsible for ensuring the appropriate intensity of the actions of this pathway by hydrolyzing cAMP to its inactive form 5'-AMP. PDE1, PDE2, PDE4, and PDE11A are highly expressed in the pituitary, and overexpression of some PDE4 isoforms have been demonstrated in different pituitary adenoma subtypes. This observed over-expression in pituitary adenomas, although of unknown etiology, has been considered a compensatory response to tumorigenesis. PDE4A4/5 has a unique interaction with the co-chaperone aryl hydrocarbon receptor-interacting protein (AIP), a protein implicated in somatotroph tumorigenesis via germline loss-of-function mutations. Based on the association of low PDE4A4 expression with germline AIP-mutation-positive samples, the available data suggest that lack of AIP hinders the upregulation of PDE4A4 protein seen in sporadic somatotrophinomas. This unique disturbance of the cAMP-PDE pathway observed in the majority of AIP-mutation positive adenomas could contribute to their well-described poor response to somatostatin analogs and may support a role in tumorigenesis.
Collapse
Affiliation(s)
- Mariana Ferreira Bizzi
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Graeme B. Bolger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Márta Korbonits
- Center for Endocrinology, Barts and The London School of Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Ribeiro-Oliveira Jr.
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Antonio Ribeiro-Oliveira Jr.
| |
Collapse
|
12
|
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
|
13
|
Geng L, Zhou W, Liu B, Wang X, Chen B. DHA induces apoptosis of human malignant breast cancer tissues by the TLR-4/PPAR-α pathways. Oncol Lett 2017; 15:2967-2977. [PMID: 29435026 PMCID: PMC5778790 DOI: 10.3892/ol.2017.7702] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 09/15/2017] [Indexed: 01/16/2023] Open
Abstract
Docosahexaenoic acid (DHA) oil is an important polyunsaturated fatty acid for the human body. Evidence has demonstrated that DHA is beneficial for inhibiting mammary carcinogenesis. However, the mechanisms of DHA mediating apoptosis induction have not been fully elucidated. Thus, in the present study, the activity levels of total-superoxide dismutase (t-SOD), catalase (CAT), glutathione-peroxidase (GSH-PX) and the concentration of malondialdehyde (MDA) were determined in DHA oil-treated human malignant breast tissues. The results revealed that compared with control, DHA significantly increased the main antioxidant enzymes levels, including t-SOD, CAT, and GSH-PX, but decreased the MDA concentration in the DHA oil treated breast cancer tissues. Furthermore, DHA significantly increased the ratio of cyclic (c)AMP/cGMP levels and promoted the expression of Toll-like receptor 4 (TLR-4) and peroxisome proliferator activated receptor (PPAR)-α, thus DHA induced breast cancer cell apoptosis. We hypothesized that the levels of TLR-4 and PPAR-α are involved in the antitumorigenesis properties of DHA in breast cancer. The results of the present study hold significance for the further clinical development of DHA oil in breast cancer treatment.
Collapse
Affiliation(s)
- Lijing Geng
- Key Laboratory of Molecular Cell Biology and New Drug Development of The Educational Department of Jinzhou Province, Food Science and Engineer College, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Wei Zhou
- Key Laboratory of Molecular Cell Biology and New Drug Development of The Educational Department of Jinzhou Province, Food Science and Engineer College, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Bing Liu
- Key Laboratory of Molecular Cell Biology and New Drug Development of The Educational Department of Jinzhou Province, Food Science and Engineer College, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xinyun Wang
- Key Laboratory of Molecular Cell Biology and New Drug Development of The Educational Department of Jinzhou Province, Food Science and Engineer College, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Bo Chen
- Key Laboratory of Molecular Cell Biology and New Drug Development of The Educational Department of Jinzhou Province, Food Science and Engineer College, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
14
|
Formosa R, Vassallo J. Aryl Hydrocarbon Receptor-Interacting Protein (AIP) N-Terminus Gene Mutations Identified in Pituitary Adenoma Patients Alter Protein Stability and Function. Discov Oncol 2017; 8:174-184. [PMID: 28255869 DOI: 10.1007/s12672-017-0288-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 02/10/2017] [Indexed: 01/23/2023] Open
Abstract
Mutations spanning the entire aryl hydrocarbon receptor-interacting protein (AIP) gene have been found in isolated familial cases of pituitary adenomas (PA). Missense mutations located in the N-terminus of the gene have been identified in several patients. However, the functional significance of these mutations remains a matter of controversy. In most studies, the N-terminus of AIP has been shown to regulate protein stability and subcellular localization of the AIP-AHR-HSP90 complex but not to be involved in protein-protein interactions. Other studies found that the N-terminal domain interacts directly with other proteins. The aim of this study was to analyze whether specific N-terminus AIP mutations identified in PA patients would be functionally different from wild-type (WT) AIP. In vitro analyses were used to assess the role of known N-terminus variants, a locally identified mutant, R9Q, and three other commonly genotyped N-terminus mutations R16H, V49M and K103R are found in PA patients. Given the functional effect of WT AIP on cAMP signalling alterations caused by N-terminus mutants on this pathway were also analyzed in GH3 cells. Results indicate that N-terminus mutations lead to de-regulation of the effect of WT AIP on cAMP signalling and increased cAMP thresholds in GH3 cells resulting in increased growth hormone (GH) secretion. Cycloheximide chase analysis identified a variation in protein degradation patterns between WT and N-terminus variants. Therefore, both functional and structural studies reveal that N-terminus mutations in the AIP gene alter protein behaviour significantly and hence can truly be pathogenic in nature.
Collapse
Affiliation(s)
- Robert Formosa
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD2080, Malta.
| | - Josanne Vassallo
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD2080, Malta. .,Neuroendocrine Clinic, Mater Dei Hospital, Msida, Malta.
| |
Collapse
|
15
|
Sun W, Jiao W, Huang Y, Li R, Zhang Z, Wang J, Lei T. Exchange proteins directly activated by cAMP induce the proliferation of rat anterior pituitary GH3 cells via the activation of extracellular signal-regulated kinase. Biochem Biophys Res Commun 2017; 485:355-359. [PMID: 28216156 DOI: 10.1016/j.bbrc.2017.02.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 10/24/2022]
Abstract
Cyclic adenosine 3'-5'-monophosphate (cAMP) plays a crucial role in regulating pituitary cell proliferation and hormone synthesis. Recent evidence suggests that exchange proteins directly activated by cAMP (Epacs) may mediate the effects of cAMP. Here we used rat anterior pituitary GH3 cells as the experiment model to demonstrate that forskolin increased the proliferation of GH3 cells and the phosphorylation of ERK1/2, and these effects were inhibited by PKA or Epac inhibitors. Epac activator 8-pCPT-2'-O-Me-cAMP increased GH3 cell proliferation and this was blocked by ESI-09, an Epac inhibitor. In contrast, forskolin-induced phosphorylation of CREB was unaffected by Epac inhibition. Notably, increased phosphorylation of ERK1/2 was correlated with increased cyclin D3 expression in GH3 cells. Furthermore, knockdown of Epac as well as B-Raf and MEK inhibitors blocked 8-pCPT-2'-O-Me-cAMP induced proliferation of GH3 cells and the phosphorylation of ERK1/2. In conclusion, our study suggests that Epac mediates cAMP induced pituitary cell proliferation via B-Raf and MAPK dependent mechanism.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Wei Jiao
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Yimin Huang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ran Li
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Zhuo Zhang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Junwen Wang
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ting Lei
- Department of Neurosurgery, Sino-German Neuro-Oncology Molecular Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
16
|
Buslei R, Strissel PL, Henke C, Schey R, Lang N, Ruebner M, Stolt CC, Fabry B, Buchfelder M, Strick R. Activation and regulation of endogenous retroviral genes in the human pituitary gland and related endocrine tumours. Neuropathol Appl Neurobiol 2015; 41:180-200. [PMID: 24635849 DOI: 10.1111/nan.12136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 03/10/2014] [Indexed: 12/29/2022]
Abstract
AIMS Adenohypophysis (AH) hormone-producing cells represent the origin of diverse groups of pituitary adenomas (PA). Deregulation of hypothalamic hormone receptors, growth factors and cAMP signalling have been implicated in the aetiology of PA. Endogenous retroviruses (ERVs) are derived from past exogenous retroviral infections and represent more than 8% of the human genome. Some ERV genes encode open reading frames and produce functional proteins, for example, the ERVW-1 envelope gene Syncytin-1, essential for placentogenesis, but also deregulated in human tumours. Data concerning ERV expression in the AH and related endocrine tumours are missing. METHODS Syncytin-1 protein was analysed in normal AH (n = 15) and compared with five PA subtypes (n = 117) by immunohistochemistry. Absolute gene expression of 20 ERV functional envelope genes and ERVW-5 gag was measured. PA tissues were examined for Syncytin-1 and the cAMP signalling marker phospho-CREB-Ser133 using immunohistochemistry. Isolated primary human PA cells were treated with different hormones. Murine embryonic and adult pituitary gland ERV expressions were compared with human AH. RESULTS Syncytin-1 protein colocalized with corticotropic cells of AH. In contrast, all PA demonstrated significant Syncytin-1 protein overexpression, supporting deregulation. All other ERV genes showed significant up-regulations in different PA subtypes. Phospho-CREB-Ser133 and Syncytin-1 colocalized in PA cells. Cultivated primary PA cells with ACTH or CRH induced their respective receptors and ERV genes. Syncytin-A/-B, murine orthologues to human Syncytin-1/-2, localized to embryonic and adult pituitary glands demonstrating functional mammalian conservation. CONCLUSIONS Deregulated ERV genes may contribute to PA development via cAMP signalling.
Collapse
Affiliation(s)
- Rolf Buslei
- Institute of Neuropathology, University-Clinic Erlangen, Lab for Molecular Medicine, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Liu J, Tang X, Cheng J, Wang L, Yang X, Wang Y. Genetic analysis of a patient with coexisting acromegaly, thyroid papillary carcinoma and subcutaneous fibroma. Oncol Lett 2015; 9:1177-1180. [PMID: 25663877 PMCID: PMC4314992 DOI: 10.3892/ol.2014.2824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 11/24/2014] [Indexed: 12/03/2022] Open
Abstract
The aim of the present study was to analyze the MEN1 and gsα gene mutations in a Chinese patient with growth hormone-producing pituitary tumors causing acromegaly, papillary thyroid carcinoma and subcutaneous fibroma. Genomic DNA was isolated from the patient and 10 healthy controls, and prepared for polymerase chain reaction (PCR) analysis. Numerous pairs of primers were designed to amplify exons 1–10 of the MEN1 gene and exons 8 and 9 of the gsα gene, and the PCR products were sequenced to detect mutations. In the study patient, a heterozygous G→A mutation was detected at nucleotide 7848 within exon 10 of the MEN1 gene; the missense mutation caused the substitution of alanine with threonine at amino acid 541 (A541T) in the menin protein. In addition, a G→A mutation at nucleotide 7997 within exon 10 of the MEN1 gene was identified; the mutation was synonymous, therefore, the proline at amino acid 590 of the menin protein (P590P) did not change. No other mutations were observed in exons 8 and 9 of the gsα gene, therefore, the G7848A mutation within exon 10 of the MEN1 gene may represent the molecular pathology underlying pituitary somatotroph adenomas and papillary thyroid carcinoma. Furthermore, the pituitary adenomas, thyroid carcinoma and subcutaneous fibroma of the present patient may be considered as early manifestations of multiple endocrinologic neoplasia syndrome 1 as opposed to pure endocrine tumors, however, a long-term follow-up study is required to clarify this.
Collapse
Affiliation(s)
- Jingfang Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xulei Tang
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jianguo Cheng
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Liting Wang
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiaomei Yang
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yan Wang
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
18
|
The role of cyclic nucleotide signaling pathways in cancer: targets for prevention and treatment. Cancers (Basel) 2014; 6:436-58. [PMID: 24577242 PMCID: PMC3980602 DOI: 10.3390/cancers6010436] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/10/2014] [Accepted: 02/07/2014] [Indexed: 12/13/2022] Open
Abstract
For more than four decades, the cyclic nucleotides cyclic AMP (cAMP) and cyclic GMP (cGMP) have been recognized as important signaling molecules within cells. Under normal physiological conditions, cyclic nucleotides regulate a myriad of biological processes such as cell growth and adhesion, energy homeostasis, neuronal signaling, and muscle relaxation. In addition, altered cyclic nucleotide signaling has been observed in a number of pathophysiological conditions, including cancer. While the distinct molecular alterations responsible for these effects vary depending on the specific cancer type, several studies have demonstrated that activation of cyclic nucleotide signaling through one of three mechanisms-induction of cyclic nucleotide synthesis, inhibition of cyclic nucleotide degradation, or activation of cyclic nucleotide receptors-is sufficient to inhibit proliferation and activate apoptosis in many types of cancer cells. These findings suggest that targeting cyclic nucleotide signaling can provide a strategy for the discovery of novel agents for the prevention and/or treatment of selected cancers.
Collapse
|
19
|
Jaffrain-Rea ML, Rotondi S, Turchi A, Occhi G, Barlier A, Peverelli E, Rostomyan L, Defilles C, Angelini M, Oliva MA, Ceccato F, Maiorani O, Daly AF, Esposito V, Buttarelli F, Figarella-Branger D, Giangaspero F, Spada A, Scaroni C, Alesse E, Beckers A. Somatostatin analogues increase AIP expression in somatotropinomas, irrespective of Gsp mutations. Endocr Relat Cancer 2013; 20:753-66. [PMID: 23940012 DOI: 10.1530/erc-12-0322] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Germline aryl hydrocarbon receptor interacting protein (AIP) gene mutations confer a predisposition to pituitary adenoma (PA), predominantly GH-secreting (GH-PA). As recent data suggest a role for AIP in the pathogenesis of sporadic GH-PA and their response to somatostatin analogues (SSA), the expression of AIP and its partner, aryl hydrocarbon receptor (AHR), was determined by semiquantitative immunohistochemistry scoring in 62 sporadic GH-PA (37 treated with SSA preoperatively). The influence of Gsp status was studied in a subset of tumours (n=39, 14 Gsp(+)) and six GH-PA were available for primary cultures. AIP and AHR were detected in most cases, with a positive correlation between AIP and cytoplasmic AHR (P=0.012). Low AIP expression was significantly more frequent in untreated vs SSA-treated tumours (44.0 vs 20.5%, P=0.016). AHR expression or localisation did not differ between the two groups. Similarly, in vitro octreotide induced a median twofold increase in AIP expression (range 1.2-13.9, P=0.027) in GH-PA. In SSA-treated tumours, the AIP score was significantly higher in the presence of preoperative IGF1 decrease or tumour shrinkage (P=0.008 and P=0.014 respectively). In untreated tumours, low AIP expression was significantly associated with invasiveness (P=0.028) and suprasellar extension (P=0.019). The only effect of Gsp status was a significantly lower nuclear AHR score in Gsp(+) vs Gsp(-) tumours (P=0.025), irrespective of SSA. In conclusion, AIP is involved in the aggressiveness of sporadic GH-PA, regardless of Gsp status, and AIP up-regulation in SSA-treated tumours is associated with a better preoperative response, with no clear role for AHR.
Collapse
Affiliation(s)
- Marie-Lise Jaffrain-Rea
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lania A, Mantovani G, Spada A. cAMP pathway and pituitary tumorigenesis. ANNALES D'ENDOCRINOLOGIE 2012; 73:73-5. [PMID: 22525824 DOI: 10.1016/j.ando.2012.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The pituitary is the target of different neurohormones that have a crucial role in the control of cell differentiation, cell proliferation and hormone secretion by recognizing specific receptors belonging to the G Protein-Coupled Receptor super-family (GPCR). Evidence from in vitro studies and naturally occurring human diseases indicate that several endocrine cells, and particularly somatotrophs, recognize cAMP as a growth factor. Accordingly, mutations of the alpha subunit of the stimulatory G protein gene (GNAS) leading to the constitutive activation of adenylyl cyclase (i.e. gsp oncogene) have been recognized in a significant proportion of GH-secreting pituitary adenomas. The role of cAMP in the control of cell proliferation in selected cell types and in particular in somatotroph cells has been further confirmed by identification of genetics defect affecting the regulatory subunit IA of PKA. The role of cAMP in the control of cell proliferation as well as the crosstalk with different intracellular signalling pathways will be discussed.
Collapse
Affiliation(s)
- Andrea Lania
- Department of Medical Sciences, University of Milan, Endocrine Unit, IRCCS Istituto Clinico Humanitas, Rozzano, Italy.
| | | | | |
Collapse
|
21
|
Silencing of the imprinted DLK1-MEG3 locus in human clinically nonfunctioning pituitary adenomas. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2120-30. [PMID: 21871428 DOI: 10.1016/j.ajpath.2011.07.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 06/13/2011] [Accepted: 07/01/2011] [Indexed: 01/04/2023]
Abstract
DLK1-MEG3 is an imprinted locus consisting of multiple maternally expressed noncoding RNA genes and paternally expressed protein-coding genes. The expression of maternally expressed gene 3 (MEG3) is selectively lost in clinically nonfunctioning adenomas (NFAs) of gonadotroph origin; however, expression status of other genes at this locus in human pituitary adenomas has not previously been reported. Using quantitative real-time RT-PCR, we evaluated expression of 24 genes from the DLK1-MEG3 locus in 44 human pituitary adenomas (25 NFAs, 7 ACTH-secreting, 7 GH-secreting, and 5 PRL-secreting adenomas) and 10 normal pituitaries. The effects on cell proliferation of five miRNAs whose expression was lost in NFAs were investigated by flow cytometry analysis. We found that 18 genes, including 13 miRNAs at the DLK1-MEG3 locus, were significantly down-regulated in human NFAs. In ACTH-secreting and PRL-secreting adenomas, 12 and 7 genes were significantly down-regulated, respectively; no genes were significantly down-regulated in GH-secreting tumors. One of the five miRNAs tested induced cell cycle arrest at the G2/M phase in PDFS cells derived from a human NFA. Our data indicate that the DLK1-MEG3 locus is silenced in NFAs. The growth suppression by miRNAs in PDFS cells is consistent with the hypothesis that the DLK1-MEG3 locus plays a tumor suppressor role in human NFAs.
Collapse
|
22
|
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
|
23
|
Abstract
Pituitary adenomas may hypersecrete hormones (including prolactin, growth hormone and adrenocorticotropic hormone, and rarely follicle-stimulating hormone, luteinizing hormone or TSH) or may be nonfunctional. Despite their high prevalence in the general population, these tumors are invariably benign and exhibit features of differentiated pituitary cell function as well as premature proliferative arrest. Pathogenesis of dysregulated pituitary cell proliferation and unrestrained hormone hypersecretion may be mediated by hypothalamic, intrapituitary and/or peripheral factors. Altered expression of pituitary cell cycle genes, activation of pituitary selective oncoproteins or loss of pituitary suppressor factors may be associated with aberrant growth factor signaling. Considerable information on the etiology of these tumors has been derived from transgenic animal models, which may not accurately and universally reflect human tumor pathophysiology. Understanding subcellular mechanisms that underlie pituitary tumorigenesis will enable development of tumor aggression markers as well as novel targeted therapies.
Collapse
Affiliation(s)
- Shlomo Melmed
- Cedars-Sinai Medical Center, Academic Affairs Room 2015, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:384-93. [PMID: 20588116 DOI: 10.1097/med.0b013e32833c4b2b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|