1
|
Ren MJ, Zhang ZL, Tian C, Liu GQ, Zhang CS, Yu HB, Xin Q. Importance of early detection in multiple endocrine neoplasia type 1: Clinical insights and future directions. World J Gastrointest Oncol 2025; 17:100013. [PMID: 40235881 PMCID: PMC11995327 DOI: 10.4251/wjgo.v17.i4.100013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/11/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is an autosomal-inherited syndrome involving multiple endocrine tumors. It is characterized by multiple mutations in the tumor suppressor gene MEN1, which is located on chromosome 11q13. As main etiology of MEN1 is genetic mutations, clinical symptoms may vary. In this editorial, we comment on the article by Yuan et al. This article describes a case of (MEN1) characterized by low incidence and diagnostic complexity. MEN1 commonly presents as parathyroid, pancreatic, and pituitary tumors. Diagnosis requires a combination of serologic tests, magnetic resonance imaging, computed tomography, endoscopic ultrasonography, immunologic and pathology. The diagnosis is unique depending on the site of disease. Surgical resection is the treatment of choice for MEN1. The prognosis depends on the site of origin, but early detection and intervention is the most effective.
Collapse
Affiliation(s)
- Mei-Jing Ren
- Department of Pathology, Tianjin Third Center Hospital, Tianjin 300170, China
| | - Zi-Li Zhang
- Department of Gastrointestinal Surgery, Tianjin Third Center Hospital, Tianjin 300170, China
| | - Can Tian
- Department of Pathology, Tianjin Third Center Hospital, Tianjin 300170, China
| | - Gui-Qiu Liu
- Department of Pathology, Tianjin Third Center Hospital, Tianjin 300170, China
| | - Chuan-Shan Zhang
- Department of Pathology, Tianjin Third Center Hospital, Tianjin 300170, China
| | - Hai-Bo Yu
- Department of Laboratory, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qi Xin
- Department of Pathology, Tianjin Third Center Hospital, Tianjin 300170, China
| |
Collapse
|
2
|
Peng J, Yuan L, Kang P, Jin S, Ma S, Zhou W, Jia G, Zhang C, Jia W. Comprehensive transcriptomic analysis identifies three distinct subtypes of pituitary adenomas: insights into tumor behavior, prognosis, and stem cell characteristics. J Transl Med 2024; 22:892. [PMID: 39363281 PMCID: PMC11448088 DOI: 10.1186/s12967-024-05702-w] [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: 06/16/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Pituitary adenomas (PAs) are the second most common intracranial tumor. While current diagnostic practices rely primarily on histological testing, they often fail to capture the molecular complexities of pituitary adenomas, underscoring the need for a molecular-based classification to refine therapeutic strategies and prognostic assessments. This study aims to provide a molecularly unbiased classification of pituitary adenomas and explore their unique gene expression patterns and clinical features. METHODS We performed unsupervised hierarchical clustering of the gene expression profiles of 117 PA samples to identify three distinct molecular subtypes. Subsequently, we analyzed the compiled transcriptomic profiles of each individual subtype for pathway enrichment. We also validated the new classification with a validation set containing 158 PAs and 24 pituitary adenoma stem cells (PASCs). RESULTS Consensus clustering of transcriptomic data from 117 pituitary adenoma (PA) samples identified three distinct molecular subtypes, each showing unique gene expression patterns and associated biological processes: Group I is enriched in signaling pathways, such as the cAMP signaling pathway and the calcium signaling pathway. Group II is primarily related to metabolic processes, including nitrogen metabolism and arginine biosynthesis in cancer. Group III predominantly shows enrichment in immune responses and potential malignant transformation of the disease, especially through cancer-related pathways such as the JAK-STAT signaling pathway and the PI3K-Akt signaling pathway. The immune profiling revealed distinct patterns for each subtype: Group I had higher dendritic cells and fewer CD8+ T cells, Group II had more monocytes and macrophages, and Group III had elevated levels of T cells. Additionally, there were differences in clinical characteristics and prognosis among the subtypes, with Group III having a worse prognosis, despite the smaller tumor size compared to other groups. Notably, differences in PASCs correlated with the molecular subtypes, with Group III stem cells being enriched in tumorigenesis pathways, PI3K-Akt signaling pathway and Ras signaling pathway. CONCLUSION Our study introduces a novel molecular classification for pituitary adenomas, independent of traditional histological methods. Each subtype features distinct genetic, molecular, and immunological profiles. We have isolated pituitary adenoma stem-like cells (PASCs), pairing them with tumor tissues for detailed transcriptomic analysis. These PASCs exhibit diverse molecular traits consistent with the new classification.
Collapse
Affiliation(s)
- Jiayi Peng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Linhao Yuan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peng Kang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shucheng Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shunchang Ma
- China National Clinical Research Center for Neurological Diseases, Fengtai, Beijing, China
| | - Wenjianlong Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guijun Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Fengtai, Beijing, China
| | - Chuanbao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Fengtai, Beijing, China.
- Beijing Neurosurgical Institute, Beijing, China.
| |
Collapse
|
3
|
Kazzaz SA, Tawil J, Harhaj EW. The aryl hydrocarbon receptor-interacting protein in cancer and immunity: Beyond a chaperone protein for the dioxin receptor. J Biol Chem 2024; 300:107157. [PMID: 38479600 PMCID: PMC11002312 DOI: 10.1016/j.jbc.2024.107157] [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: 01/09/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR)-interacting protein (AIP) is a ubiquitously expressed, immunophilin-like protein best known for its role as a co-chaperone in the AhR-AIP-Hsp90 cytoplasmic complex. In addition to regulating AhR and the xenobiotic response, AIP has been linked to various aspects of cancer and immunity that will be the focus of this review article. Loss-of-function AIP mutations are associated with pituitary adenomas, suggesting that AIP acts as a tumor suppressor in the pituitary gland. However, the tumor suppressor mechanisms of AIP remain unclear, and AIP can exert oncogenic functions in other tissues. While global deletion of AIP in mice yields embryonically lethal cardiac malformations, heterozygote, and tissue-specific conditional AIP knockout mice have revealed various physiological roles of AIP. Emerging studies have established the regulatory roles of AIP in both innate and adaptive immunity. AIP interacts with and inhibits the nuclear translocation of the transcription factor IRF7 to inhibit type I interferon production. AIP also interacts with the CARMA1-BCL10-MALT1 complex in T cells to enhance IKK/NF-κB signaling and T cell activation. Taken together, AIP has diverse functions that vary considerably depending on the client protein, the tissue, and the species.
Collapse
Affiliation(s)
- Sarah A Kazzaz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - John Tawil
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Edward W Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
4
|
Tapoi DA, Popa ML, Tanase C, Derewicz D, Gheorghișan-Gălățeanu AA. Role of Tumor Microenvironment in Pituitary Neuroendocrine Tumors: New Approaches in Classification, Diagnosis and Therapy. Cancers (Basel) 2023; 15:5301. [PMID: 37958474 PMCID: PMC10649263 DOI: 10.3390/cancers15215301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/15/2023] Open
Abstract
Adenohypophysal pituitary tumors account for 10-15% of all intracranial tumors, and 25-55% display signs of invasiveness. Nevertheless, oncology still relies on histopathological examination to establish the diagnosis. Considering that the classification of pituitary tumors has changed significantly in recent years, we discuss the definition of aggressive and invasive tumors and the latest molecular criteria used for classifying these entities. The pituitary tumor microenvironment (TME) is essential for neoplastic development and progression. This review aims to reveal the impact of TME characteristics on stratifying these tumors in view of finding appropriate therapeutic approaches. The role of the pituitary tumor microenvironment and its main components, non-tumoral cells and soluble factors, has been addressed. The variable display of different immune cell types, tumor-associated fibroblasts, and folliculostellate cells is discussed in relation to tumor development and aggressiveness. The molecules secreted by both tumoral and non-tumoral cells, such as VEGF, FGF, EGF, IL6, TNFα, and immune checkpoint molecules, contribute to the crosstalk between the tumor and its microenvironment. They could be considered potential biomarkers for diagnosis and the invasiveness of these tumors, together with emerging non-coding RNA molecules. Therefore, assessing this complex network associated with pituitary neuroendocrine tumors could bring a new era in diagnosing and treating this pathology.
Collapse
Affiliation(s)
- Dana Antonia Tapoi
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pathology, University Emergency Hospital, 050098 Bucharest, Romania
| | - Maria-Linda Popa
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Cell Biology and Clinical Biochemistry, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Diana Derewicz
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pediatric Hematology and Oncology, Marie Sklodowska Curie Clinical Emergency Hospital, 041447 Bucharest, Romania
| | - Ancuța-Augustina Gheorghișan-Gălățeanu
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
| |
Collapse
|
5
|
Лапшина АМ. [Molecular genetic abnormalities in ACTH-secreting pituitary tumors (corticotropinomas): fundamental research and prospects for use in clinical practice]. PROBLEMY ENDOKRINOLOGII 2023; 70:23-30. [PMID: 39069770 PMCID: PMC11334230 DOI: 10.14341/probl13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/16/2023] [Accepted: 11/02/2023] [Indexed: 07/30/2024]
Abstract
In recent years, a large number of studies have been carried out to research molecular genetic abnormalities in ACTH--secreting pituitary tumors. This review presents a comprehensive analysis of exome studies results (germline and somatic mutations, chromosomal abnormalities in corticotropinomas which developed as part of hereditary syndromes MEN 1, 2, 4, DICER1, Carney complex etc., and isolated tumors, respectively) and transcriptome (specific genes expression profiles in hormonally active and inactive corticotropinomas, regulation of cell cycles and signal pathways). Modern technologies (next-generation sequencing - NGS) allow us to study the state of the microRNAome, DNA methylome and inactive chromatin sites, in particular using RNA sequencing. Thus, a wide range of fundamental studies is shown, the results of which allow us to identify and comprehend the key previously known and new pathogenesis mechanisms and biomarkers of corticotropinomas. The characteristics of the most promising molecular genetic factors that can be used in clinical practice for screening and earlier diagnosis of hereditary syndromes and isolated corticotropinomas, differential diagnosis of various forms of endogenous hypercorticism, sensitivity to existing and potential therapies and personalized outcome determination of Cushing`s disease.
Collapse
Affiliation(s)
- А. М. Лапшина
- Национальный медицинский исследовательский центр эндокринологии
| |
Collapse
|
6
|
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
|
7
|
Batchu S, Diaz MJ, Lin K, Arya N, Patel K, Lucke-Wold B. Single Cell Metabolic Landscape of Pituitary Neuroendocrine Tumor Subgroups and Lineages. OBM NEUROBIOLOGY 2023; 7:10. [PMID: 37007673 PMCID: PMC10062196 DOI: 10.21926/obm.neurobiol.2301157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs) are common intracranial tumors comprising numerous subtypes whose metabolic profiles have yet to be fully examined. The present in silico study analyzed single-cell expression profiles from 2311 PitNET cells from various lineages and subtypes to elucidate differences in metabolic activities. Gonadotroph tumors exhibited high activities with histidine metabolism, whose activity is low in lactotroph tumors. Somatotroph tumors enriched for sulfur and tyrosine metabolism, while lactotroph tumors were enriched metabolism of nitrogen, ascorbate, and aldarate. PIT-1 lineage tumors exhibited high sulfur and thiamine metabolism. These results set precedence for further translational studies for subgroup/lineage specific targeted therapies.
Collapse
Affiliation(s)
| | | | - Keldon Lin
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, United States
| | - Namrata Arya
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, United States
| | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, United States
| |
Collapse
|
8
|
Saksis R, Rogoza O, Niedra H, Megnis K, Mandrika I, Balcere I, Steina L, Stukens J, Breiksa A, Nazarovs J, Sokolovska J, Konrade I, Peculis R, Rovite V. Transcriptome of GH-producing pituitary neuroendocrine tumours and models are significantly affected by somatostatin analogues. Cancer Cell Int 2023; 23:25. [PMID: 36774501 PMCID: PMC9922463 DOI: 10.1186/s12935-023-02863-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/01/2023] [Indexed: 02/13/2023] Open
Abstract
Pituitary neuroendocrine tumours (PitNETs) are neoplasms of the pituitary that overproduce hormones or cause unspecific symptoms due to mass effect. Growth hormone overproducing GH-producing PitNETs cause acromegaly leading to connective tissue, metabolic or oncologic disorders. The medical treatment of acromegaly is somatostatin analogues (SSA) in specific cases combined with dopamine agonists (DA), but almost half of patients display partial or full SSA resistance and potential causes of this are unknown. In this study we investigated transcriptomic landscape of GH-producing PitNETs on several levels and functional models-tumour tissue of patients with and without SSA preoperative treatment, tumour derived pituispheres and GH3 cell line incubated with SSA to study effect of medication on gene expression. MGI sequencing platform was used to sequence total RNA from PitNET tissue, pituispheres, mesenchymal stromal stem-like cells (MSC), and GH3 cell cultures, and data were analysed with Salmon-DeSeq2 pipeline. We observed that the GH-producing PitNETs have distinct changes in growth hormone related pathways related to its functional status alongside inner cell signalling, ion transport, cell adhesion and extracellular matrix characteristic patterns. In pituispheres model, treatment regimens (octreotide and cabergoline) affect specific cell proliferation (MKI67) and core functionality pathways (RYR2, COL8A2, HLA-G, ARFGAP1, TGFBR2). In GH3 cells we observed that medication did not have transcriptomic effects similar to preoperative treatment in PitNET tissue or pituisphere model. This study highlights the importance of correct model system selection for cell transcriptomic profiling and data interpretation that could be achieved in future by incorporating NGS methods and detailed cell omics profiling in PitNET model research.
Collapse
Affiliation(s)
- Rihards Saksis
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Olesja Rogoza
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Helvijs Niedra
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Kaspars Megnis
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Ilona Mandrika
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Inga Balcere
- grid.488518.80000 0004 0375 2558Riga East Clinical University Hospital, Hipokrata Str 2, Riga, 1038 Latvia ,grid.17330.360000 0001 2173 9398Riga Stradins University, Dzirciema Str. 16, Riga, 1007 Latvia
| | - Liva Steina
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia ,grid.477807.b0000 0000 8673 8997Pauls Stradins Clinical University Hospital, Pilsonu Str 13, Riga, 1002 Latvia
| | - Janis Stukens
- grid.477807.b0000 0000 8673 8997Pauls Stradins Clinical University Hospital, Pilsonu Str 13, Riga, 1002 Latvia
| | - Austra Breiksa
- grid.477807.b0000 0000 8673 8997Pauls Stradins Clinical University Hospital, Pilsonu Str 13, Riga, 1002 Latvia
| | - Jurijs Nazarovs
- grid.477807.b0000 0000 8673 8997Pauls Stradins Clinical University Hospital, Pilsonu Str 13, Riga, 1002 Latvia
| | - Jelizaveta Sokolovska
- grid.9845.00000 0001 0775 3222Faculty of Medicine, University of Latvia, Raina Blvd 19, Riga, 1586 Latvia
| | - Ilze Konrade
- grid.488518.80000 0004 0375 2558Riga East Clinical University Hospital, Hipokrata Str 2, Riga, 1038 Latvia ,grid.17330.360000 0001 2173 9398Riga Stradins University, Dzirciema Str. 16, Riga, 1007 Latvia
| | - Raitis Peculis
- grid.419210.f0000 0004 4648 9892Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067 Latvia
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre, Ratsupites Str 1-k1, Riga, 1067, Latvia.
| |
Collapse
|
9
|
Jovanova T, Vichentijevikj I, Petrevska D, Ilievska E, Hiljadnikova-Bajro M. Current and future perspectives in laboratory analysis of Pituitary neuroendocrine tumors. MAKEDONSKO FARMACEVTSKI BILTEN 2022. [DOI: 10.33320/maced.pharm.bull.2022.68.03.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Teodora Jovanova
- Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, R.N. Macedonia
| | - Ivana Vichentijevikj
- Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, R.N. Macedonia
| | - Debora Petrevska
- Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, R.N. Macedonia
| | - Elena Ilievska
- Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, R.N. Macedonia
| | - Marija Hiljadnikova-Bajro
- Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, R.N. Macedonia
| |
Collapse
|
10
|
Banerjee A, Biswas D, Barpanda A, Halder A, Sibal S, Kattimani R, Shah A, Mahadevan A, Goel A, Srivastava S. The First Pituitary Proteome Landscape From Matched Anterior and Posterior Lobes for a Better Understanding of the Pituitary Gland. Mol Cell Proteomics 2022; 22:100478. [PMID: 36470533 PMCID: PMC9877467 DOI: 10.1016/j.mcpro.2022.100478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
To date, very few mass spectrometry (MS)-based proteomics studies are available on the anterior and posterior lobes of the pituitary. In the past, MS-based investigations have focused exclusively on the whole pituitary gland or anterior pituitary lobe. In this study, for the first time, we performed a deep MS-based analysis of five anterior and five posterior matched lobes to build the first lobe-specific pituitary proteome map, which documented 4090 proteins with isoforms, mostly mapped into chromosomes 1, 2, and 11. About 1446 differentially expressed significant proteins were identified, which were studied for lobe specificity, biological pathway enrichment, protein-protein interaction, regions specific to comparison of human brain and other neuroendocrine glands from Human Protein Atlas to identify pituitary-enriched proteins. Hormones specific to each lobe were also identified and validated with parallel reaction monitoring-based target verification. The study identified and validated hormones, growth hormone and thyroid-stimulating hormone subunit beta, exclusively to the anterior lobe whereas oxytocin-neurophysin 1 and arginine vasopressin to the posterior lobe. The study also identified proteins POU1F1 (pituitary-specific positive transcription factor 1), POMC (pro-opiomelanocortin), PCOLCE2 (procollagen C-endopeptidase enhancer 2), and NPTX2 (neuronal pentraxin-2) as pituitary-enriched proteins and was validated for their lobe specificity using parallel reaction monitoring. In addition, three uPE1 proteins, namely THEM6 (mesenchymal stem cell protein DSCD75), FSD1L (coiled-coil domain-containing protein 10), and METTL26 (methyltransferase-like 26), were identified using the NeXtProt database, and depicted tumor markers S100 proteins having high expression in the posterior lobe. In summary, the study documents the first matched anterior and posterior pituitary proteome map acting as a reference control for a better understanding of functional and nonfunctional pituitary adenomas and extrapolating the aim of the Human Proteome Project towards the investigation of the proteome of life.
Collapse
Affiliation(s)
- Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deepatarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Abhilash Barpanda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ankit Halder
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shamira Sibal
- Lokmanya Tilak Municipal Medical College, Mumbai, India
| | | | - Abhidha Shah
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Anita Mahadevan
- Human Brain Bank, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Atul Goel
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
11
|
Guaraldi F, Morandi L, Zoli M, Mazzatenta D, Righi A, Evangelisti S, Ambrosi F, Tonon C, Giannini C, Lloyd RV, Asioli S. Epigenomic and somatic mutations of pituitary tumors with clinical and pathological correlations in 111 patients. Clin Endocrinol (Oxf) 2022; 97:763-772. [PMID: 36161330 PMCID: PMC9828656 DOI: 10.1111/cen.14827] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/09/2022] [Accepted: 09/18/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To profile clinically non-aggressive and aggressive pituitary adenomas (PAs)/pituitary neuroendocrine tumours (PitNETs) and pituitary carcinomas for somatic mutations and epigenetic alterations of genes involved in cell proliferation/differentiation, microRNAs (miRNA)/long noncoding RNA (LncRNA)-post-transcriptional regulators and therapy targets. DESIGN Retrospective observational study. PATIENTS AND MEASUREMENTS A total of 64 non-aggressive and 41 aggressive PAs/PitNETs and 6 pituitary carcinomas treated by endoscopic surgery with ≥1-year follow-up were included. Somatic mutations of 17 genes and DNA methylation of 22 genes were assessed. Ten normal pituitaries were used as control. RESULTS We found at least one mutation in 17 tumours, including 6/64 non-aggressive, 10/41 aggressive PAs/PitNETs, and 1/6 pituitary carcinoma. AIP (N = 6) was the most frequently mutated gene, followed by NOTCH (4), and TP53 (3). Hypermethylation of PARP15, LINC00599, ZAP70 was more common in aggressive than non-aggressive PAs/PITNETs (p < .05). Lower levels of methylation of AIP, GNAS and PDCD1 were detected in aggressive PAs/PITNETs than non-aggressive ones (p < .05). For X-linked genes, males presented higher level of methylation of FLNA, UXT and MAGE family (MAGEA11, MAGEA1, MAGEC2) genes in aggressive vs. non-aggressive PAs/PITNETs (p < .05). In pituitary carcinomas, methylation of autosomal genes PARP15, LINC00599, MIR193 and ZAP70 was higher than in PAs/PITNETs, while X-linked genes methylation level was lower. CONCLUSIONS Somatic mutations and methylation levels of genes involved in cell proliferation/differentiation, miRNA/LncRNA-post-transcriptional regulators and targets of antineoplastic therapies are different in non-aggressive and in aggressive PAs/PitNETs. Methylation profile also varies according to gender. Combined genetic-epigenetic analysis, in association with clinico-radiological-pathological data, may be of help in predicting PA/PitNET behaviour.
Collapse
Affiliation(s)
| | - Luca Morandi
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Matteo Zoli
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Diego Mazzatenta
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Alberto Righi
- Department of PathologyIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefania Evangelisti
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Francesca Ambrosi
- Dipartimento Interaziendale Anatomia Patologica, Pathology Unit, Maggiore HospitalAUSL BolognaBolognaItaly
| | - Caterina Tonon
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Caterina Giannini
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
- Anatomic Pathology Unit, Department of Pathology and Laboratory MedicineMayo ClinicRochesterMinnesotaUSA
| | - Ricardo V. Lloyd
- Department of Pathology and Laboratory Medicine, School of Medicine and Public HealthUniversity of WisconsinMadisonMichiganUSA
| | - Sofia Asioli
- IRCCS Istituto delle Scienze Neurologiche di BolognaBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
- Dipartimento Interaziendale Anatomia Patologica, Unit of Anatomic PathologyAUSL BolognaBolognaItaly
| |
Collapse
|
12
|
Proteogenomic landscape and clinical characterization of GH-producing pituitary adenomas/somatotroph pituitary neuroendocrine tumors. Commun Biol 2022; 5:1304. [PMID: 36435867 PMCID: PMC9701206 DOI: 10.1038/s42003-022-04272-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/16/2022] [Indexed: 11/28/2022] Open
Abstract
The clinical characteristics of growth hormone (GH)-producing pituitary adenomas/somatotroph pituitary neuroendocrine tumors (GHomas/somatotroph PitNETs) vary across patients. In this study, we aimed to integrate the genetic alterations, protein expression profiles, transcriptomes, and clinical characteristics of GHomas/somatotroph PitNETs to identify molecules associated with acromegaly characteristics. Targeted capture sequencing and copy number analysis of 36 genes and nontargeted proteomics analysis were performed on fresh-frozen samples from 121 sporadic GHomas/somatotroph PitNETs. Targeted capture sequencing revealed GNAS as the only driver gene, as previously reported. Classification by consensus clustering using both RNA sequencing and proteomics revealed many similarities between the proteome and the transcriptome. Gene ontology analysis was performed for differentially expressed proteins between wild-type and mutant GNAS samples identified by nontargeted proteomics and involved in G protein-coupled receptor (GPCR) pathways. The results suggested that GNAS mutations impact endocrinological features in acromegaly through GPCR pathway induction. ATP2A2 and ARID5B correlated with the GH change rate in the octreotide loading test, and WWC3, SERINC1, and ZFAND3 correlated with the tumor volume change rate after somatostatin analog treatment. These results identified a biological connection between GNAS mutations and the clinical and biochemical characteristics of acromegaly, revealing molecules associated with acromegaly that may affect medical treatment efficacy.
Collapse
|
13
|
Melmed S, Kaiser UB, Lopes MB, Bertherat J, Syro LV, Raverot G, Reincke M, Johannsson G, Beckers A, Fleseriu M, Giustina A, Wass JAH, Ho KKY. Clinical Biology of the Pituitary Adenoma. Endocr Rev 2022; 43:1003-1037. [PMID: 35395078 PMCID: PMC9695123 DOI: 10.1210/endrev/bnac010] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 02/06/2023]
Abstract
All endocrine glands are susceptible to neoplastic growth, yet the health consequences of these neoplasms differ between endocrine tissues. Pituitary neoplasms are highly prevalent and overwhelmingly benign, exhibiting a spectrum of diverse behaviors and impact on health. To understand the clinical biology of these common yet often innocuous neoplasms, we review pituitary physiology and adenoma epidemiology, pathophysiology, behavior, and clinical consequences. The anterior pituitary develops in response to a range of complex brain signals integrating with intrinsic ectodermal cell transcriptional events that together determine gland growth, cell type differentiation, and hormonal production, in turn maintaining optimal endocrine health. Pituitary adenomas occur in 10% of the population; however, the overwhelming majority remain harmless during life. Triggered by somatic or germline mutations, disease-causing adenomas manifest pathogenic mechanisms that disrupt intrapituitary signaling to promote benign cell proliferation associated with chromosomal instability. Cellular senescence acts as a mechanistic buffer protecting against malignant transformation, an extremely rare event. It is estimated that fewer than one-thousandth of all pituitary adenomas cause clinically significant disease. Adenomas variably and adversely affect morbidity and mortality depending on cell type, hormone secretory activity, and growth behavior. For most clinically apparent adenomas, multimodal therapy controlling hormone secretion and adenoma growth lead to improved quality of life and normalized mortality. The clinical biology of pituitary adenomas, and particularly their benign nature, stands in marked contrast to other tumors of the endocrine system, such as thyroid and neuroendocrine tumors.
Collapse
Affiliation(s)
| | - Ursula B Kaiser
- Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - M Beatriz Lopes
- University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jerome Bertherat
- Université de Paris, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Luis V Syro
- Hospital Pablo Tobon Uribe and Clinica Medellin - Grupo Quirónsalud, Medellin, Colombia
| | - Gerald Raverot
- Hospices Civils de Lyon and Lyon 1 University, Lyon, France
| | - Martin Reincke
- University Hospital of LMU, Ludwig-Maximilians-Universität, Munich, Germany
| | - Gudmundur Johannsson
- Sahlgrenska University Hospital & Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Andrea Giustina
- San Raffaele Vita-Salute University and IRCCS Hospital, Milan, Italy
| | | | - Ken K Y Ho
- The Garvan Institute of Medical Research and St. Vincents Hospital, Sydney, Australia
| |
Collapse
|
14
|
Niedra H, Peculis R, Litvina HD, Megnis K, Mandrika I, Balcere I, Romanovs M, Steina L, Stukens J, Breiksa A, Nazarovs J, Sokolovska J, Liutkeviciene R, Vilkevicute A, Konrade I, Rovite V. Genome wide analysis of circulating miRNAs in growth hormone secreting pituitary neuroendocrine tumor patients’ plasma. Front Oncol 2022; 12:894317. [PMID: 36158656 PMCID: PMC9500360 DOI: 10.3389/fonc.2022.894317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/15/2022] [Indexed: 12/05/2022] Open
Abstract
Background Circulating plasma miRNAs have been increasingly studied in the field of pituitary neuroendocrine tumor (PitNET) research. Our aim was to discover circulating plasma miRNAs species associated with growth hormone (GH) secreting PitNETs versus assess how the plasma levels of discovered miRNA candidates are impacted by SSA therapy and whether there is a difference in their levels between GH secreting PitNETs versus other PitNET types and healthy individuals. Design We compared plasma miRNA content and levels before and after surgery focusing on GH secreting PitNET patients. Selected miRNA candidates from our data and literature were then tested in a longitudinal manner in somatostatin analogues (SSA) treatment group. Additionally, we validated selected targets in an independent GH secreting PitNET group. Methods miRNA candidates were discovered using the whole miRNA sequencing approach and differential expression analysis. Selected miRNAs were then analyzed using real-time polymerase chain reaction (qPCR). Results Whole miRNA sequencing discovered a total of 16 differentially expressed miRNAs (DEMs) in GH secreting PitNET patients’ plasma 24 hours after surgery and 19 DEMs between GH secreting PitNET patients’ plasma and non-functioning (NF) PitNET patients’ plasma. Seven miRNAs were selected for further testing of which miR-625-5p, miR-503-5p miR-181a-2-3p and miR-130b-3p showed a significant downregulation in plasma after 1 month of SSA treatment. mir-625-5p was found to be significantly downregulated in plasma of GH secreting PitNET patients vs. NF PitNET patients. miR-625-5p alongside miR-130b-3p were also found to be downregulated in GH PitNETs compared to healthy individuals. Conclusions Our study suggests that expression of plasma miRNAs miR-625-5p, miR-503-5p miR-181a-2-3p and miR-130b-3p in GH secreting PitNETs is affected by SSA treatment. Additionally, miR-625-5p can distinguish GH secreting PitNETs from other PitNET types and healthy controls warranting further research on these miRNAs for treatment efficacy.
Collapse
Affiliation(s)
- Helvijs Niedra
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Raitis Peculis
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Helena Daiga Litvina
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Kaspars Megnis
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ilona Mandrika
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Inga Balcere
- Department of Endocrinology, Riga East Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Mihails Romanovs
- Department of Endocrinology, Riga East Clinical University Hospital, Riga, Latvia
| | - Liva Steina
- Department of Neurosurgery, Faculty of Medicine Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Janis Stukens
- Department of Neurosurgery, Faculty of Medicine Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Austra Breiksa
- Department of Neurosurgery, Faculty of Medicine Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Jurijs Nazarovs
- Department of Neurosurgery, Faculty of Medicine Pauls Stradins Clinical University Hospital, Riga, Latvia
| | | | - Rasa Liutkeviciene
- Institute of Neuroscience, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alvita Vilkevicute
- Institute of Neuroscience, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ilze Konrade
- Department of Endocrinology, Riga East Clinical University Hospital, Riga, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Vita Rovite
- Department of molecular and functional genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
- *Correspondence: Vita Rovite,
| |
Collapse
|
15
|
Yamamoto M, Takahashi Y. Genetic and Epigenetic Pathogenesis of Acromegaly. Cancers (Basel) 2022; 14:cancers14163861. [PMID: 36010855 PMCID: PMC9405703 DOI: 10.3390/cancers14163861] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Various genetic and epigenetic factors are involved in the pathogenesis of somatotroph tumors. Although GNAS mutations are the most prevalent cause of somatotroph tumors, the cause of half of all pathogenesis occurrences remains unclarified. However, recent findings including the pangenomic analysis, such as genome, transcriptome, and methylome approaches, and histological characteristics of pituitary tumors, the involvement of AIP and GPR101, the mechanisms of genomic instability, and possible involvement of miRNAs have gradually unveiled the whole landscape of underlying mechanisms of somatotroph tumors. In this review, we will focus on the recent advances in the pathogenesis of somatotroph tumors. Abstract Acromegaly is caused by excessive secretion of GH and IGF-I mostly from somatotroph tumors. Various genetic and epigenetic factors are involved in the pathogenesis of somatotroph tumors. While somatic mutations of GNAS are the most prevalent cause of somatotroph tumors, germline mutations in various genes (AIP, PRKAR1A, GPR101, GNAS, MEN1, CDKN1B, SDHx, MAX) are also known as the cause of somatotroph tumors. Moreover, recent findings based on multiple perspectives of the pangenomic approach including genome, transcriptome, and methylome analyses, histological characterization, genomic instability, and possible involvement of miRNAs have gradually unveiled the whole landscape of the underlying mechanisms of somatotroph tumors. In this review, we will focus on the recent advances in genetic and epigenetic pathogenesis of somatotroph tumors.
Collapse
Affiliation(s)
- Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Correspondence: ; Tel.: +81-78-382-5861
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
- Department of Diabetes and Endocrinology, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
16
|
Molecular Markers of Telomerase Complex for Patients with Pituitary Adenoma. Brain Sci 2022; 12:brainsci12080980. [PMID: 35892421 PMCID: PMC9331889 DOI: 10.3390/brainsci12080980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022] Open
Abstract
Pituitary adenoma (PA) is the most common benign tumor of the pituitary gland. The pathogenesis of most PA is considered as a multifactorial process, that involves genetic mutations, alterations in gene transcription, and epigenetic factors. Their interaction promotes tumorigenesis. The processes are increasingly focused on changes in telomere length. Our study enrolled 126 patients with PA and 368 healthy subjects. DNA samples from peripheral blood leukocytes were purified by the DNA salting-out method. The RT-PCR carried out SNPs and relative leukocyte telomere lengths (RLTL). ELISA determined the level of TEP1 in blood serum. Binary logistic regression revealed that TERC rs35073794 is likely associated with increased odds of PA development and macro-PA development. It is also associated with decreased odds of active PA, non-invasive PA, and PA without relapse development. Also, we discovered that PA patients with at least one G allele of the TEP1 gene polymorphism rs1713418 have lower serum TEP1 levels than healthy individuals (p = 0.035). To conclude, the study revealed that TERC rs35073794 might be a potential biomarker for PA development.
Collapse
|
17
|
Pastorino L, Grillo F, Albertelli M, Ghiorzo P, Bruno W. Insights into Mechanisms of Tumorigenesis in Neuroendocrine Neoplasms. Int J Mol Sci 2021; 22:ijms221910328. [PMID: 34638668 PMCID: PMC8508699 DOI: 10.3390/ijms221910328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Genomic studies have identified some of the most relevant genetic players in Neuroendocrine Neoplasm (NEN) tumorigenesis. However, we are still far from being able to draw a model that encompasses their heterogeneity, elucidates the different biological effects consequent to the identified molecular events, or incorporates extensive knowledge of molecular biomarkers and therapeutic targets. Here, we reviewed recent insights in NEN tumorigenesis from selected basic research studies on animal models, highlighting novel players in the intergenic cooperation and peculiar mechanisms including splicing dysregulation, chromatin stability, or cell dedifferentiation. Furthermore, models of tumorigenesis based on composite interactions other than a linear progression of events are proposed, exemplified by the involvement in NEN tumorigenesis of genes regulating complex functions, such as MEN1 or DAXX. Although limited by interspecies differences, animal models have proved helpful for the more in-depth study of every facet of tumorigenesis, showing that the identification of driver mutations is only one of the many necessary steps and that other mechanisms are worth investigating.
Collapse
Affiliation(s)
- Lorenza Pastorino
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (L.P.); (P.G.)
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, V.le Benedetto XV 6, 16132 Genoa, Italy;
| | - Federica Grillo
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy;
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 1632 Genoa, Italy
| | - Manuela Albertelli
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, V.le Benedetto XV 6, 16132 Genoa, Italy;
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Paola Ghiorzo
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (L.P.); (P.G.)
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, V.le Benedetto XV 6, 16132 Genoa, Italy;
| | - William Bruno
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy; (L.P.); (P.G.)
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, V.le Benedetto XV 6, 16132 Genoa, Italy;
- Correspondence: ; Tel.: +39-(01)-0555-7254
| |
Collapse
|
18
|
4-Nitrophenol-Loaded Magnetic Mesoporous Silica Hybrid Materials for Spectrometric Aptasensing of Carcinoembryonic Antigen. MICROMACHINES 2021; 12:mi12101138. [PMID: 34683189 PMCID: PMC8537709 DOI: 10.3390/mi12101138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/20/2022]
Abstract
Aptamer- or antibody-based sensing protocols have been reported for detecting carcinoembryonic antigen (CEA), but most exhibit complicated procedures or multiple reactions. In this work, we developed a one-step aptasensing protocol for the spectrometric determination of CEA based on 4-nitrophenol (4-NP)-loaded magnetic mesoporous silica nanohybrids (MMSNs) for bioresponsive controlled-release applications. To fabricate such a responsive–controlled sensing system, single-stranded complementary oligonucleotides relative to the CEA-specific aptamer were first modified on the aminated MMSN. Thereafter, 4-NP molecules blocked the pores with the assistance of the aptamers via a hybridization reaction. The introduced target CEA specifically reacted with the hybridized aptamer, thus detaching from the MMSN to open the gate. The loaded 4-NP molecules were released from the pores, as determined using ultraviolet–visible (UV–vis) absorption spectroscopy after magnetic separation. Under optimum conditions, the absorbance increased with an increase in the target CEA in the sample and exhibited a good linear relationship within the dynamic range of 0.1–100 ng mL−1, with a detection limit of 46 pg mL−1. Moreover, this system also displayed high specificity, good reproducibility, and acceptable accuracy for analyzing human serum specimens, in comparison with a commercialized human CEA-enzyme-linked immunosorbent assay (ELISA) kit.
Collapse
|