1
|
Xu D, Wang L, Zheng M. Advancements in Molecular Diagnosis and Pharmacotherapeutic Strategies for Invasive Pituitary Adenomas. Immun Inflamm Dis 2024; 12:e70098. [PMID: 39688352 PMCID: PMC11650491 DOI: 10.1002/iid3.70098] [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: 08/14/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The overwhelming majority of pituitary tumors consist of pituitary adenomas (PAs), which have recently also been termed pituitary neuroendocrine tumors (PitNETs). Clinically significant PAs occur in approximately one in every 1000 individuals, while other types of pituitary tumors, such as craniopharyngiomas and pituicytomas, are significantly less common. Although PAs are generally benign, a subset of them exhibits malignant-like biological traits. They tend to infiltrate and grow aggressively into adjacent tissues and organs, including the dura mater, cavernous sinus, and sphenoid sinus. This invasive behavior often results in the destruction of the normal anatomical architecture of the sella turcica and skull base. Clinically, such tumors are classified as invasive PAs (IPAs), emphasizing their aggressive and destructive nature. OBJECTIVE AND SIGNIFICANCE Currently, the diagnostic indicators for IPAs frequently suffer from suboptimal sensitivity and specificity. The invasiveness assessment of PAs lacks a definitive gold standard and instead serves as a predictive tool, with a greater number of indicators met suggesting a higher likelihood of invasiveness. Consequently, a comprehensive approach that integrates imaging, pathological, molecular biological, and other disciplinary metrics is crucial for accurate evaluation. Despite surgery being the primary treatment modality for IPAs, their malignant-like behavior complicates complete resection, resulting in lower resection rates and heightened postoperative recurrence, necessitating multiple surgeries. Therefore, adjunctive drug therapy is often necessary for IPA patients. Preoperative drug therapy can shrink tumor size, facilitating resection and postoperative recovery, mitigating hormone imbalances, delaying recurrence, and enhancing patients' quality of life. CONCLUSIONS This article comprehensively reviews the diagnostic criteria for assessing the invasiveness of PAs in the domains of imaging, pathology, and molecular biology, provides an overview of the current research status of drug therapy for these conditions, and deepens our insight into the biological and therapeutic aspects of the tumor microenvironment in PAs.
Collapse
Affiliation(s)
- Dingkai Xu
- Department of NeurosurgeryThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Ling Wang
- Department of EndocrinologyLiangzhou HospitalWuweiGansuChina
| | - Maohua Zheng
- Department of NeurosurgeryThe First Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
2
|
Potthoff TE, Walter C, Jeising D, Münter D, Verma A, Suero Molina E, Stummer W, Dugas M, Hartmann W, Dottermusch M, Altendorf L, Schüller U, Scheuermann S, Seitz C, Albert TK, Kerl K. Single-cell transcriptomics link gene expression signatures to clinicopathological features of gonadotroph and lactotroph PitNET. J Transl Med 2024; 22:1027. [PMID: 39548496 PMCID: PMC11566263 DOI: 10.1186/s12967-024-05821-4] [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: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNET) are among the most common intracranial tumors. Despite a frequent benign course, aggressive behavior can occur. Tumor behavior is known to be under the influence of the tumor microenvironment (TME). However, the relationship between TME cells and aggressive tumor behavior has not been adequately explored in PitNET. METHODS We performed differential expression analysis as well as gene expression program identification based on single-cell RNA sequencing to comparatively characterize the transcriptome of seven gonadotroph and three lactotroph PitNET and correlate it with clinical features using bulk RNA-seq data from an independent cohort of 134 PitNET. Tumor immune infiltration was quantified via immunostaining on tissue sections of gonadotroph and lactotroph PitNET. RESULTS In lactotroph PitNET, we detect a highly proliferative gene profile with significantly increased expression levels in aggressively growing tumors within bulk RNA-seq data of an independent cohort of 134 PitNET samples. We also report high intratumoral heterogeneity in gonadotroph PitNET (GoPN) and lactotroph PitNET (LaPN) and identify signatures of epithelial, endocrine, and immunological gene networks in both subtypes. A comparison of their TME composition shows enrichment of SPP1+ macrophages and CD4+ T cells in GoPN, as well as enrichment of CD4/CD8 double-negative T cells (DN) and natural killer cells (NK) in LaPN. Also notable is the presence of proliferative lymphocytes, the occurrence of which positively correlates with more aggressive tumor behavior in the bulk RNA-seq cohort. However, increased CD8+ T and NK cell abundances correlate significantly with reduced aggressiveness indicating potential anti-tumoral effects. CONCLUSIONS Our study expands the knowledge of the differences in cellular composition of gonadotroph and lactotroph PitNET subtypes. It lays the foundation for further studies on the influence of lymphoid cells on the variable aggressive behavior of PitNET. Regarding the treatment of drug-resistant lactotroph PitNET, proliferative lymphocytes, CD8+ T, and NK cells could represent potentially valuable targets for developing new cancer immunotherapies.
Collapse
Affiliation(s)
- T Elise Potthoff
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Daniela Jeising
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Daniel Münter
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Archana Verma
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149, Münster, Germany
| | - Matthias Dottermusch
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Lea Altendorf
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Sophia Scheuermann
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Christian Seitz
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Thomas K Albert
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Kornelius Kerl
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
3
|
Retraction: Correlations of Pituitary Tumor Transforming Gene Expression with Human Pituitary Adenomas: A Meta-Analysis. PLoS One 2022; 17:e0279013. [PMID: 36473002 PMCID: PMC9725118 DOI: 10.1371/journal.pone.0279013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
4
|
Hosseinkhan N, Honardoost M, Emami Z, Cheraghi S, Hashemi-Madani N, Khamseh ME. A systematic review of molecular alterations in invasive non-functioning pituitary adenoma. Endocrine 2022; 77:500-509. [PMID: 35711030 DOI: 10.1007/s12020-022-03105-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/03/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Invasive non-functional pituitary adenomas (NFPAs) constitute 35% of NFPAs. Despite a relatively large body of molecular investigations on the invasiveness of NFPA, the underlying molecular mechanisms of invasiveness are yet to be determined. Herein, we aimed to provide an overview of gene/microRNA(miRNAs) expression alterations in invasive NFPA. METHODS This article describes a systematic literature review of articles published up to March 23, 2021, on the transcriptional alterations of invasive NFPA. Five digital libraries were searched, and 42 articles in total fulfilled the eligibility criteria. Pathway enrichment was conducted, and protein interactions among the identified deregulated genes were inferred. RESULTS In total 133 gene/protein transcriptional alterations, comprising 87 increased and 46 decreased expressions, were detected in a collective number of 1001 invasive compared with 1007 non-invasive patients with NFPA. Deregulation of CDH1, PTTG1, CCNB1, SNAI1, SLUG, EZR, and PRKACB, which are associated with epidermal-mesenchymal transition (EMT), was identified. Moreover, six members of the angiogenesis pathway, i.e., VEGFA, FLT1, CCND1, CTNNB1, MYC(c-MYC), and PTTG1, were detected. SLC2A1, FLT1, and VEGFA were also recognized in the hypoxia pathway. Physical interactions of CTNNB1 with FLT1, CCND1, and EZR as well as its indirect interactions with VEGFA, MYC, CCNB1, and PCNA indicate the tight interplay between EMT, angiogenesis, and hypoxia pathways in invasive NFPAs. In addition, Hippo, JAK-STAT, MAPK, Wnt, PI3K-Akt, Ras, TGF-b, VEGF, and ErbB were identified as interwoven signaling pathways. CONCLUSION In conclusion, invasive NFPA shares very common deregulated signaling pathways with invasive cancers. A large amount of heterogeneity in the reported deregulations in different studies necessitates the validation of the expressional changes of the suggested biomarkers in a large number of patients with invasive NFPA.
Collapse
Affiliation(s)
- Nazanin Hosseinkhan
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Honardoost
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Emami
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Hashemi-Madani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad E Khamseh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Pituitary Apoplexy: Risk Factors and Underlying Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms23158721. [PMID: 35955859 PMCID: PMC9369054 DOI: 10.3390/ijms23158721] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 12/02/2022] Open
Abstract
Pituitary apoplexy is a rare syndrome, graded from asymptomatic subclinical apoplexy to a life-threatening condition due to pituitary ischemia or haemorrhage of an enlarged pituitary gland. The risk factors and the molecular underlying mechanisms are yet to be elucidated. We provide an overview of the general concepts, the potential factors associated with pituitary adenoma susceptibility for apoplectic events and the molecular mechanisms that could be involved such as HIF-1α/VEGF pathways and metalloproteinases activation, among others. The knowledge of the molecular mechanisms that could participate in the pathogenesis of pituitary apoplexy is crucial to advancement in the identification of future diagnostic tools and therapeutic targets in this rare but sometimes fatal condition.
Collapse
|
6
|
Kum SJ, Lee HW, Kim SG, Park H, Hwang I, Kim SP. Association of PTTG1 expression with invasiveness of non-functioning pituitary adenomas. J Pathol Transl Med 2021; 56:22-31. [PMID: 34645111 PMCID: PMC8743807 DOI: 10.4132/jptm.2021.08.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/31/2021] [Indexed: 11/17/2022] Open
Abstract
Background Pituitary tumor transforming gene 1 (PTTG1), paired-like homeodomain 2 (PITX2), and galectin-3 have been widely studied as predictive biomarkers for various tumors and are involved in tumorigenesis and tumor progression. We evaluated the usefulness of PTTG1, PITX2, and galectin-3 as predictive biomarkers for invasive non-functioning pituitary adenomas (NFPAs) by determining the relationship between the expressions of these three proteins and the invasiveness of the NFPAs. We also investigated whether PTTG1, E-cadherin, and Ki-67, which are known to be related to each other, show a correlation with NFPA features. Methods A retrospective study was conducted on 87 patients with NPFAs who underwent surgical removal. The NFPAs were classified into three groups based on magnetic resonance imaging findings of suprasellar extension and cavernous sinus invasion. Immunohistochemical staining for PTTG1, PITX2, galectin-3, E-cadherin, and Ki-67 was performed on tissue microarrays. Results PTTG1 expression showed a statistically significant correlation with the invasiveness of NFPAs, whereas PITX2 and galectin-3 did not have a relationship with the invasiveness of NFPAs. Moreover, there was no association among PTTG1, E-cadherin, and Ki-67 expression. Conclusion PTTG1 has the potential to serve as a predictive biomarker for invasive NFPA. Furthermore, this study may serve as a reference for the development of PTTG1-targeted therapeutic agents.
Collapse
Affiliation(s)
- Su Jung Kum
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Hye Won Lee
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Soon Gu Kim
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Hyungsik Park
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Ilseon Hwang
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Sang Pyo Kim
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea
| |
Collapse
|
7
|
Cui L, Xu L, Wang G, Wen J, Luo L, Zhao H, Chen S, Zheng M, Sun C, Jin X, Yang L. STAT3-PTTG11 abrogation inhibits proliferation and induces apoptosis in malignant glioma cells. Oncol Lett 2020; 20:6. [PMID: 32774480 DOI: 10.3892/ol.2020.11867] [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: 11/27/2019] [Accepted: 05/13/2020] [Indexed: 11/06/2022] Open
Abstract
Pituitary tumor transforming gene 1 (PTTG11) is abundantly expressed in glioma. Our previous study demonstrated that the downregulation of PTTG11 gene expression significantly inhibited the proliferation, migration and invasion ability, and increased the apoptosis of SHG44 glioma cells. However, the molecular mechanisms that regulate PTTG11 and its actions remain elusive. In the present study, CCK-8 and flow cytometry assays were used to assess the proliferation/viability and apoptosis, respectively, of the human glioma U251 cell line. STAT3-PTTG1 signals were further evaluated by western blotting. The findings of the present study revealed that STAT3 induced PTTG11 expression, which subsequently induced downstream c-Myc and Bcl-2 expression while inhibiting Bax expression, thereby promoting cell viability and inhibiting apoptosis. PTTG11 suppression via siRNA inhibited the viability and increased the apoptosis of glioma cells induced by the STAT3 activator S3I-201. c-Myc and Bcl-2 expression was suppressed by PTTG11 inhibition. The findings of the present study suggest that the STAT3-PTTG11 signaling pathway may play an important role in glioma progression by regulating cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Lishan Cui
- Department of Neurosurgery, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005, P.R. China.,Department of Neurosurgery, Xiamen Fifth Hospital, Xiamen, Fujian 361005, P.R. China
| | - Lanxi Xu
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Guanling Wang
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Jing Wen
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Lili Luo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Haitao Zhao
- Department of Neurosurgery, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Shuide Chen
- Department of Neurosurgery, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Mingcheng Zheng
- Department of Neurosurgery, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Cuiling Sun
- School of Pharmacy, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Lichao Yang
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| |
Collapse
|
8
|
Xiong Z, Li X, Yang Q. PTTG has a Dual Role of Promotion-Inhibition in the Development of Pituitary Adenomas. Protein Pept Lett 2019; 26:800-818. [PMID: 37020362 DOI: 10.2174/0929866526666190722145449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/22/2022]
Abstract
Pituitary Tumor Transforming Gene (PTTG) of human is known as a checkpoint gene in the middle and late stages of mitosis, and is also a proto-oncogene that promotes cell cycle progression. In the nucleus, PTTG works as securin in controlling the mid-term segregation of sister chromatids. Overexpression of PTTG, entering the nucleus with the help of PBF in pituitary adenomas, participates in the regulation of cell cycle, interferes with DNA repair, induces genetic instability, transactivates FGF-2 and VEGF and promotes angiogenesis and tumor invasion. Simultaneously, overexpression of PTTG induces tumor cell senescence through the DNA damage pathway, making pituitary adenoma possessing the potential self-limiting ability. To elucidate the mechanism of PTTG in the regulation of pituitary adenomas, we focus on both the positive and negative function of PTTG and find out key factors interacted with PTTG in pituitary adenomas. Furthermore, we discuss other possible mechanisms correlate with PTTG in pituitary adenoma initiation and development and the potential value of PTTG in clinical treatment.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
9
|
Trott G, Ongaratti BR, de Oliveira Silva CB, Abech GD, Haag T, Rech CGSL, Ferreira NP, da Costa Oliveira M, Pereira-Lima JFS. PTTG overexpression in non-functioning pituitary adenomas: Correlation with invasiveness, female gender and younger age. Ann Diagn Pathol 2019; 41:83-89. [DOI: 10.1016/j.anndiagpath.2019.04.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/11/2019] [Accepted: 04/28/2019] [Indexed: 12/27/2022]
|
10
|
Yang Q, Li X. Molecular Network Basis of Invasive Pituitary Adenoma: A Review. Front Endocrinol (Lausanne) 2019; 10:7. [PMID: 30733705 PMCID: PMC6353782 DOI: 10.3389/fendo.2019.00007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cases with pituitary adenoma comprise 10-25% of intracranial neoplasm, being the third most common intracranial tumor, most of the adenomas are considered to be benign. About 35% of pituitary adenomas are invasive. This review summarized the known molecular basis of the invasiveness of pituitary adenomas. The study pointed out that hypoxia-inducible factor-1α, pituitary tumor transforming gene, vascular endothelial growth factor, fibroblast growth factor-2, and matrix metalloproteinases (MMPs, mainly MMP-2, and MMP-9) are core molecules responsible for the invasiveness of pituitary adenomas. The reason is that these molecules have the ability to directly or indirectly induce cell proliferation, epithelial-to-mesenchymal transition, angiogenesis, degradation, and remodeling of extracellular matrix. HIF-1α induced by hypoxia or apoplexy inside the adenoma might be the initiating factor of invasive transformation, followed with angiogenesis for overexpressed VEGF, EMT for overexpressed PTTG, degradation of ECM for overexpressed MMPs, creating a suitable microenvironment within the tumor. Together, they form a complex interactive network. More investigations are required to further elucidate the mechanisms underlying the invasiveness of pituitary adenomas.
Collapse
|
11
|
Foltran RK, Amorim PVGH, Duarte FH, Grande IPP, Freire ACTB, Frassetto FP, Dettoni JB, Alves VA, Castro I, Trarbach EB, Bronstein MD, Jallad RS. Study of major genetic factors involved in pituitary tumorigenesis and their impact on clinical and biological characteristics of sporadic somatotropinomas and non-functioning pituitary adenomas. ACTA ACUST UNITED AC 2018; 51:e7427. [PMID: 29947650 PMCID: PMC6040863 DOI: 10.1590/1414-431x20187427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/02/2018] [Indexed: 11/25/2022]
Abstract
Genetic and functional aberrations of guanine nucleotide-binding protein, alpha stimulating (GNAS), aryl hydrocarbon receptor interacting protein (AIP), and pituitary tumor transforming gene (PTTG) are among the most prominent events in pituitary tumorigenesis. A cohort of Brazilian patients with somatotropinomas (n=41) and non-functioning pituitary adenomas (NFPA, n=21) from a single tertiary-referral center were evaluated for GNAS and AIP mutations and gene expression of AIP and PTTG. Results were compared to the clinical and biological (Ki67 and p53 expression) characteristics of tumors and their response to therapy, if applicable. Genetic analysis revealed that 27% of somatotropinomas and 4.8% of NFPA harbored GNAS mutations (P=0.05). However, no differences were observed in clinical characteristics, tumor extension, response to somatostatin analog therapy, hormonal/surgical remission rates, Ki67 index, and p53 expression between mutated and non-mutated somatotropinomas patients. PTTG overexpression (RQ mean=10.6, min=4.39, max=11.9) and AIP underexpression (RQ mean=0.56, min=0.46-max=0.92) were found in virtually all cases without a statistically significant relationship with clinical and biological tumor features. No patients exhibited somatic or germline pathogenic AIP mutations. In conclusion, mutations in GNAS and abnormal PTTG and AIP expression had no impact on tumor features and treatment outcomes in this cohort. Our data support some previous studies and point to the need for further investigations, probably involving epigenetic and transcriptome analysis, to improve our understanding of pituitary tumor behavior.
Collapse
Affiliation(s)
- R K Foltran
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - P V G H Amorim
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F H Duarte
- Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Serviço de Endocrinologia, A.C. Camargo Center, São Paulo, SP, Brasil
| | - I P P Grande
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A C T B Freire
- Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - F P Frassetto
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - J B Dettoni
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V A Alves
- Divisao de Anatomia Patológica, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - I Castro
- Divisao de Medicina Molecular, Departamento de Medicina, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E B Trarbach
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M D Bronstein
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - R S Jallad
- Laboratorio de Endocrinologia Celular e Molecular, LIM25, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.,Unidade de Neuroendocrinologia, Disciplina de Endocrinologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
12
|
PTTG regulates the metabolic switch of ovarian cancer cells via the c-myc pathway. Oncotarget 2016; 6:40959-69. [PMID: 26516926 PMCID: PMC4747382 DOI: 10.18632/oncotarget.5726] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 09/20/2015] [Indexed: 12/02/2022] Open
Abstract
Human pituitary tumor-transforming gene (PTTG) is a proto-oncogene involved in the development, invasion, and metastasis of many types of cancer, including ovarian cancer. However, little is known about the role of PTTG in the metabolic shift of ovarian cancer cells. In our study, we show that PTTG expression was positively correlated with the differentiation degree of ovarian cancer tissue. In addition, PTTG suppression by specific shRNA could inhibit the proliferation of ovarian cancer cells A2780 and SKOV-3. Furthermore, aerobic glycolysis was suppressed and oxidative phosphorylation was increased in ovarian cancer cells after PTTG suppression. We further found that the expression of c-myc and several crucial enzymes involved in aerobic glycolysis (e.g., PKM2, LDHA, and glucose transporter 1 (GLUT-1)) were downregulated by PTTG knockwown. Overexpression of c-myc could prevent the metabolic shift induced by PTTG knockwown. Together, our findings suggest that the oncogene PTTG promotes the progression of ovarian cancer cells, and its loss resists tumor development, in part, by regulating cellular metabolic reprogramming that supports cell growth and proliferation via c-myc pathway.
Collapse
|
13
|
Abstract
Pituitary carcinoma is a rare tumor originating from adenohypophyseal cells. Currently, diverse pathogenetic mechanisms, i.e. de novo versus malignant transformation from pituitary adenoma, remain obscure and require further investigation. During the last two decades, scientific research added new horizons not only in regards to general tumor concepts but also in next generation biomarker armamentarium that sheds light on alternate pathways in carcinogenesis. Areas covered: In this review, the impact of apoptotic and proliferative markers, angiogenesis, telomerase activity, H-ras, HIF-1, HER-2/neu, Rb gene, and microRNAs in pathogenetic mechanisms of pituitary carcinomas were revised. Expert commentary: It is becoming increasingly important for the need of standardization of new biomarkers but also for better comprehension of the diverse pathways in tumorigenesis. This can only be accomplished by tapping into the continuously expanding spectrum of new biomarkers.
Collapse
Affiliation(s)
- Aydin Sav
- a Division of Neuropathology, Nisantasi Pathology Group , Istanbul , Turkey
| | - Fabio Rotondo
- b Department of Laboratory Medicine, Division of Pathology, St Michael's Hospital , University of Toronto , Toronto , Canada
| | - Luis V Syro
- c Department of Neurosurgery , Hospital Pablo Tobon Uribe and Clinica Medellin , Medellin , Colombia
| | - Antonio Di Ieva
- d Neurosurgery Unit, Faculty of Medicine and Health Science , Macquarie University , Sydney , Australia
| | - Michael D Cusimano
- e Department of Surgery, Division of Neurosurgery, St. Michael's Hospital , University of Toronto , Toronto , Canada
| | - Kalman Kovacs
- b Department of Laboratory Medicine, Division of Pathology, St Michael's Hospital , University of Toronto , Toronto , Canada
| |
Collapse
|
14
|
Abstract
Acromegaly (ACM) is a chronic, progressive disorder caused by the persistent hypersecretion of GH, in the vast majority of cases secreted by a pituitary adenoma. The consequent increase in IGF1 (a GH-induced liver protein) is responsible for most clinical features and for the systemic complications associated with increased mortality. The clinical diagnosis, based on symptoms related to GH excess or the presence of a pituitary mass, is often delayed many years because of the slow progression of the disease. Initial testing relies on measuring the serum IGF1 concentration. The oral glucose tolerance test with concomitant GH measurement is the gold-standard diagnostic test. The therapeutic options for ACM are surgery, medical treatment, and radiotherapy (RT). The outcome of surgery is very good for microadenomas (80-90% cure rate), but at least half of the macroadenomas (most frequently encountered in ACM patients) are not cured surgically. Somatostatin analogs are mainly indicated after surgical failure. Currently their routine use as primary therapy is not recommended. Dopamine agonists are useful in a minority of cases. Pegvisomant is indicated for patients refractory to surgery and other medical treatments. RT is employed sparingly, in cases of persistent disease activity despite other treatments, due to its long-term side effects. With complex, combined treatment, at least three-quarters of the cases are controlled according to current criteria. With proper control of the disease, the specific complications are partially improved and the mortality rate is close to that of the background population.
Collapse
Affiliation(s)
- Cristina Capatina
- Department of EndocrinologyCarol Davila University of Medicine and Pharmacy, Bucharest, RomaniaCI Parhon National Institute of EndocrinologyBucharest, RomaniaDepartment of EndocrinologyOxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK Department of EndocrinologyCarol Davila University of Medicine and Pharmacy, Bucharest, RomaniaCI Parhon National Institute of EndocrinologyBucharest, RomaniaDepartment of EndocrinologyOxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - John A H Wass
- Department of EndocrinologyCarol Davila University of Medicine and Pharmacy, Bucharest, RomaniaCI Parhon National Institute of EndocrinologyBucharest, RomaniaDepartment of EndocrinologyOxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| |
Collapse
|