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
|
Xie L, Miao X, Luo Q, Zhao H, Qin X. Impact of FecB Mutation on Ovarian DNA Methylome in Small-Tail Han Sheep. Genes (Basel) 2023; 14:203. [PMID: 36672944 PMCID: PMC9859159 DOI: 10.3390/genes14010203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/31/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Booroola fecundity (FecB) gene, a mutant of bone morphogenetic protein 1B (BMPR-1B) that was discovered in Booroola Merino, was the first prolificacy gene identified in sheep related to increased ovulation rate and litter size. The mechanism of FecB impact on reproduction is unclear. METHODS In this study, adult Han ewes with homozygous FecB(B)/FecB(B) mutations (Han BB group) and ewes with FecB(+)/FecB(+) wildtype (Han ++ group) were selected. Methylated DNA immunoprecipitation and high-throughput sequencing (MeDIP-seq) was used to identify differences in methylated genes in ovary tissue. RESULTS We examined differences in DNA methylation patterns between HanBB and Han ++ sheep. In both sheep, methylated reads were mainly distributed at the gene body regions, CpG islands and introns. The differentially methylated genes were enriched in neurotrophy in signaling pathway, Gonadotropin Releasing Hormone (GnRH) signaling pathway, Wnt signaling pathway, oocyte meiosis, vascular endothelial growth factor (VEGF) signaling pathway, etc. Differentially-methylated genes were co-analyzed with differentially-expressed mRNAs. Several genes which could be associated with female reproduction were identified, such as FOXP3 (forkhead box P3), TMEFF2 (Transmembrane Protein with EGF Like and Two Follistatin Like Domains 2) and ADAT2 (Adenosine Deaminase TRNA Specific 2). CONCLUSIONS We constructed a MeDIP-seq based methylomic study to investigate the ovarian DNA methylation differences between Small-Tail Han sheep with homozygous FecB mutant and wildtype, and successfully identified FecB gene-associated differentially-methylated genes. This study has provided information with which to understand the mechanisms of FecB gene-induced hyperprolificacy in sheep.
Collapse
Affiliation(s)
| | - Xiangyang Miao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | | | | | | |
Collapse
|
3
|
Hale MD, Robinson CD, Cox CL, Cox RM. Ontogenetic Change in Male Expression of Testosterone-Responsive Genes Contributes to the Emergence of Sex-Biased Gene Expression in Anolis sagrei. Front Physiol 2022; 13:886973. [PMID: 35721538 PMCID: PMC9203151 DOI: 10.3389/fphys.2022.886973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Sex differences in gene expression tend to increase with age across a variety of species, often coincident with the development of sexual dimorphism and maturational changes in hormone levels. However, because most transcriptome-wide characterizations of sexual divergence are framed as comparisons of sex-biased gene expression across ages, it can be difficult to determine the extent to which age-biased gene expression within each sex contributes to the emergence of sex-biased gene expression. Using RNAseq in the liver of the sexually dimorphic brown anole lizard (Anolis sagrei), we found that a pronounced increase in sex-biased gene expression with age was associated with a much greater degree of age-biased gene expression in males than in females. This pattern suggests that developmental changes in males, such as maturational increases in circulating testosterone, contribute disproportionately to the ontogenetic emergence of sex-biased gene expression. To test this hypothesis, we used four different experimental contrasts to independently characterize sets of genes whose expression differed as a function of castration and/or treatment with exogenous testosterone. We found that genes that were significantly male-biased in expression or upregulated as males matured tended to be upregulated by testosterone, whereas genes that were female-biased or downregulated as males matured tended to be downregulated by testosterone. Moreover, the first two principal components describing multivariate gene expression indicated that exogenous testosterone reversed many of the feminizing effects of castration on the liver transcriptome of maturing males. Collectively, our results suggest that developmental changes that occur in males contribute disproportionately to the emergence of sex-biased gene expression in the Anolis liver, and that many of these changes are orchestrated by androgens such as testosterone.
Collapse
Affiliation(s)
- Matthew D. Hale
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | | | - Christian L. Cox
- College of Arts, Sciences, and Education, Florida International University, Miami, FL, United States
| | - Robert M. Cox
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Oliveira HA, Bueno AC, Pugliesi RS, da Silva Júnior RMP, de Castro M, Martins CS. PI3K inhibition by BKM120 results in anti-proliferative effects on corticotroph tumor cells. J Endocrinol Invest 2022; 45:999-1009. [PMID: 34988938 DOI: 10.1007/s40618-021-01735-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Cushing's disease is associated with significant morbidity; thus, additional tumor-directed drugs with the potential to exert antineoplastic effects on corticotroph adenoma cells are desired. The phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) pathway, which plays regulatory role in cell survival and proliferation, is activated in pituitary adenomas. The present study evaluated the effects of BKM120 (Buparlisib), an oral PI3K inhibitor, on cell viability, apoptosis, cell cycle phase distribution, and ACTH production in mouse corticotroph tumor cells. METHODS AtT-20/D16v-F2 mouse pituitary corticotroph tumor cells were treated with increasing concentrations of BKM120 or vehicle. Cell viability was measured using an MTS-based assay. Apoptosis was evaluated by Annexin V staining. Cell cycle analysis was performed by propidium iodide DNA staining and flow cytometry. Gene expression of cell cycle regulators (Cdkn1b, Ccnd1, Ccne1, Cdk2, Cdk4, Myc, and Rb1) was assessed by qPCR. Protein expression of p27, total and phosphorylated Akt was assessed by Western blot. ACTH levels were measured in the culture supernatants by chemiluminescent immunometric assay. RESULTS Treatment with BKM120 decreased AtT-20/D16v-F2 cell viability, induced a G0/G1 cell cycle arrest, reduced the phosphorylation of Akt at Serine 473, and increased p27 expression. Furthermore, BKM120 treatment diminished ACTH levels in the cell culture supernatants. CONCLUSION In vitro inhibition of PI3K/AKT pathway by BKM120 resulted in anti-proliferative effects on corticotroph tumor cells, decreasing cell viability and ACTH production. These encouraging findings shape the path for further experiments with the inhibition of PI3K/AKT pathway in Cushing's disease.
Collapse
Affiliation(s)
- H A Oliveira
- Molecular Biology Laboratory, Endocrinology Division, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo. Av Bandeirantes, 3900 Bloco G, Ribeirão Preto, SP, 14049-900, Brazil
| | - A C Bueno
- Departments of Pediatrics of Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R S Pugliesi
- Molecular Biology Laboratory, Endocrinology Division, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo. Av Bandeirantes, 3900 Bloco G, Ribeirão Preto, SP, 14049-900, Brazil
| | - R M P da Silva Júnior
- Molecular Biology Laboratory, Endocrinology Division, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo. Av Bandeirantes, 3900 Bloco G, Ribeirão Preto, SP, 14049-900, Brazil
| | - M de Castro
- Molecular Biology Laboratory, Endocrinology Division, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo. Av Bandeirantes, 3900 Bloco G, Ribeirão Preto, SP, 14049-900, Brazil
| | - C S Martins
- Molecular Biology Laboratory, Endocrinology Division, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo. Av Bandeirantes, 3900 Bloco G, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
5
|
Masood M, Grimm S, El-Bahrawy M, Yagüe E. TMEFF2: A Transmembrane Proteoglycan with Multifaceted Actions in Cancer and Disease. Cancers (Basel) 2020; 12:cancers12123862. [PMID: 33371267 PMCID: PMC7766544 DOI: 10.3390/cancers12123862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 11/25/2022] Open
Abstract
Simple Summary We recently came across an intriguing protein while screening for tumour-specific apoptosis inducers. It is known as the transmembrane protein with an EGF-like and two Follistatin-like domains 2 (TMEFF2). The gene was identified and characterized by five different groups almost simultaneously around 2000. Physiological function of TMEFF2 is elusive; however, the protein is reported to be involved in wide-ranging physiological and pathological functions including neuroprotection in Alzheimer’s diseases, interferon induction and one-carbon metabolism. Moreover, the TMEFF2 promoter and 5′-upstream regions harbour a CpG island which is progressively methylated upon progression in a wide variety of cancers. Numerous primary publications suggest the methylation of TMEFF2 as a prognostic and even diagnostic marker in different cancers. The primary literature regarding TMEFF2 is distributed far and wide, and despite having more than 150 primary publications mentioning TMEFF2 (or its aliases) in the title or abstract on PubMed, a comprehensive literature review is not available. We believe the reason behind this is firstly the sheer diversity of subjects of these publications and secondly the numerous primary publications reporting contradictory information about TMEFF2, especially when it comes to its oncogenic versus the onco-suppressive roles. The interest in TMEFF2 is growing again; PubMed returning at least 60 publications mentioning TMEFF2 (or its aliases) within the last year. We have made a laborious effort and written a comprehensive review article on TMEFF2 where we have not only compiled and contextualized the information regarding it but also critically analysed the information in the major primary publications. In addition, we have proposed some answers to the apparent TMEFF2 disagreements on its function. This information could serve as a valuable tool for readers not only about TMEFF2 but also on the dual role of type-I transmembrane proteoglycans (harbouring Follistatin-like domains) in oncogenesis and onco-suppression. Abstract Transmembrane protein with an EGF-like and two Follistatin-like domains 2 (TMEFF2) is a 374-residue long type-I transmembrane proteoglycan which is proteolytically shed from the cell surface. The protein is involved in a range of functions including metabolism, neuroprotection, apoptosis, embryonic development, onco-suppression and endocrine function. TMEFF2 is methylated in numerous cancers, and an inverse correlation with the stage, response to therapy and survival outcome has been observed. Moreover, TMEFF2 methylation increases with breast, colon and gastric cancer progression. TMEFF2 is methylated early during oncogenesis in breast and colorectal cancer, and the detection of methylated free-circulating TMEFF2 DNA has been suggested as a potential diagnostic tool. The TMEFF2 downregulation signature equals and sometimes outperforms the Gleason and pathological scores in prostate cancer. TMEFF2 is downregulated in glioma and cotricotropinomas, and it impairs the production of adrenocorticotropic hormone in glioma cells. Interestingly, through binding the amyloid β protein, its precursor and derivatives, TMEFF2 provides neuroprotection in Alzheimer’s disease. Despite undergoing extensive investigation over the last two decades, the primary literature regarding TMEFF2 is incoherent and offers conflicting information, in particular, the oncogenic vs. onco-suppressive role of TMEFF2 in prostate cancer. For the first time, we have compiled, contextualised and critically analysed the vast body of TMEFF2-related literature and answered the apparent discrepancies regarding its function, tissue expression, intracellular localization and oncogenic vs. onco-suppressive role.
Collapse
Affiliation(s)
- Motasim Masood
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK;
| | - Stefan Grimm
- Department of Medicine, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK;
| | - Mona El-Bahrawy
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
- Correspondence: (M.E.-B.); (E.Y.); Tel.: +44-(0)77-7157-4968 (M.E.B.); +44-(0)20-7594-2802 (E.Y.)
| | - Ernesto Yagüe
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK;
- Correspondence: (M.E.-B.); (E.Y.); Tel.: +44-(0)77-7157-4968 (M.E.B.); +44-(0)20-7594-2802 (E.Y.)
| |
Collapse
|
6
|
van den Akker GGH, Eijssen LMT, Richardson SM, Rhijn LWV, Hoyland JA, Welting TJM, Voncken JW. A Membranome-Centered Approach Defines Novel Biomarkers for Cellular Subtypes in the Intervertebral Disc. Cartilage 2020; 11:203-220. [PMID: 29629573 PMCID: PMC7097986 DOI: 10.1177/1947603518764260] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Lack of specific marker-sets prohibits definition and functional distinction of cellular subtypes in the intervertebral disc (IVD), such as those from the annulus fibrosus (AF) and the nucleus pulposus (NP). DESIGN We recently generated immortalized cell lines from human NP and AF tissues; these comprise a set of functionally distinct clonal subtypes. Whole transcriptome analyses were performed of 12 phenotypically distinct clonal cell lines (4× NP-Responder, 4× NP-nonResponder, 2× AF-Sheet forming, and 2× AF-nonSheet forming). Data sets were filtered for membrane-associated marker genes and compared to literature. RESULTS Comparison of our immortal cell lines to published primary NP, AF, and articular chondrocytes (AC) transcriptome datasets revealed preservation of AF and NP phenotypes. NP-specific membrane-associated genes were defined by comparison to AF cells in both the primary dataset (46 genes) and immortal cell-lines (161 genes). Definition of AF-specific membrane-associated genes yielded 125 primary AF cell and 92 immortal cell-line markers. Overlap between primary and immortal NP cells yielded high-confidence NP-specific marker genes for NP-R (CLDN11, TMEFF2, CA12, ANXA2, CD44) and NP-nR (EFNA1, NETO2, SLC2A1). Overlap between AF and immortal AF subtypes yielded specific markers for AF-S (COLEC12, LPAR1) and AF-nS (CHIC1). CONCLUSIONS The current study provides a reference platform for preclinical evaluation of novel membrane-associated cell type-specific markers in the IVD. Future research will focus on their biological relevance for IVD function in development, homeostasis, and degenerate conditions.
Collapse
Affiliation(s)
- Guus G. H. van den Akker
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Lars M. T. Eijssen
- Department of Bioinformatics, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Stephen M. Richardson
- Centre for Regenerative Medicine, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Lodewijk W. van Rhijn
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Judith A. Hoyland
- Centre for Regenerative Medicine, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Tim J. M. Welting
- Department of Orthopedic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jan Willem Voncken
- Department of Molecular Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
7
|
Han H, Zhan Z, Xu J, Song Z. TMEFF2 inhibits pancreatic cancer cells proliferation, migration, and invasion by suppressing phosphorylation of the MAPK signaling pathway. Onco Targets Ther 2019; 12:11371-11382. [PMID: 31920328 PMCID: PMC6939404 DOI: 10.2147/ott.s210619] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/30/2019] [Indexed: 12/27/2022] Open
Abstract
PURPOSE This paper studied the effect of TMEFF2 expression on pancreatic cancer and its mechanism. METHODS A total of 72 pancreatic cancer patients were enrolled. AsPC1 and Panc1 cells were transfected. SB203580 was used to treat AsPC1 cells. CCK8 assay, colony formation analysis, Transwell experiment and Tunel test were performed. In vivo studies in nude mice were conducted. Immunohistochemistry, qRT-PCR and Western blot were used to detect genes expression. RESULTS TMEFF2 was downregulated in pancreatic cancer tissues and cells (P<0.001). Low TMEFF2 expression was associated with larger tumor size and advanced stage and poor differentiation (P<0.01). Compared with the NC group, AsPC1 and Panc1 cells of the TMEFF2 group exhibited much lower OD450 values, colony number, tumor volume and weight, migration and invasion cell numbers, obviously higher E-cadherin protein expression, lower Snail, Vimentin, MMP-2 and MMP-9 proteins expression, lower phosphorylation level of MAPK signaling pathway, and more apoptotic cells. AsPC1 cells of the SB203580 group showed much lower OD450 value when compared with the siTMEFF2 group. Significantly decreased colony number, migration and invasion number, higher E-cadherin protein expression and lower Snail, Vimentin, MMP-2 and MMP-9 proteins expression were found in AsPC1 cells of the siTMEFF2+ SB203580 group when compared with the siTMEFF2+ DMSO group. CONCLUSION TMEFF2 inhibits pancreatic cancer cells proliferation, migration, and invasion by suppressing the phosphorylation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Hongchao Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Shanghai No. 10 People’s Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of General Surgery, Yancheng Third People’s Hospital, Yancheng, People’s Republic of China
| | - Zhilin Zhan
- Department of Hepatobiliary Surgery, Chizhou People’s Hospital, Chizhou, People’s Republic of China
| | - Jie Xu
- Department of General Surgery, Yancheng Third People’s Hospital, Yancheng, People’s Republic of China
| | - Zhenshun Song
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Shanghai No. 10 People’s Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
8
|
Lu M, Wang Y, Zhan X. The MAPK Pathway-Based Drug Therapeutic Targets in Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:330. [PMID: 31231308 PMCID: PMC6558377 DOI: 10.3389/fendo.2019.00330] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) include ERK, p38, and JNK MAPK subfamilies, which are crucial regulators of cellular physiology, cell pathology, and many diseases including cancers. For the MAPK signaling system in pituitary adenomas (PAs), the activation of ERK signaling is generally thought to promote cell proliferation and growth; whereas the activations of p38 and JNK signaling are generally thought to promote cell apoptosis. The role of MAPK in treatment of PAs is demonstrated through the effects of currently used medications such as somatostatin analogs such as SOM230 and OCT, dopamine agonists such as cabergoline and bromocriptine, and retinoic acid which inhibit the MAPK pathway. Further, there are potential novel therapies based on putative molecular targets of the MAPK pathway, including 18beta-glycyrrhetinic acid (GA), dopamine-somatostatin chimeric compound (BIM-23A760), ursolic acid (UA), fulvestrant, Raf kinase inhibitory protein (RKIP), epidermal growth factor pathway substrate number 8 (Eps8), transmembrane protein with EGF-like and two follistatin-like domains (TMEFF2), cold inducible RNA-binding protein (CIRP), miR-16, and mammaliansterile-20-like kinase (MST4). The combined use of ERK inhibitor (e.g., SOM230, OCT, or dopamine) plus p38 activator (e.g., cabergoline, bromocriptine, and fulvestrant) and/or JNK activator (e.g., UA), or the development of single drug (e.g., BIM-23A760) to target both ERK and p38 or JNK pathways, might produce better anti-tumor effects on PAs. This article reviews the advances in understanding the role of MAPK signaling in pituitary tumorigenesis, and the MAPK pathway-based potential therapeutic drugs for PAs.
Collapse
Affiliation(s)
- Miaolong Lu
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Ya Wang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Abstract
INTRODUCTION Cushing's disease is a rare systemic and disabling disease due to oversecretion of adrenocorticotrophic hormone (ACTH) resulting in excess cortisol levels. Diagnosis and treatment are difficult; despite the availability of various pharmaceutical treatment options, there is an ongoing, unmet need for even more effective treatment. AREAS COVERED The present review aims at providing an overview of available drugs and presenting new developments. Focusing on the pituitary as a target, the review covers compounds targeting pituitary cell signaling or cell cycle control such as heat shock protein inhibitors (e.g. silibinin), histone deacetylase inhibitors (trichostatin A, vorinostat), kinase inhibitors (gefitinib, seliciclib), and others (such as triptolide, AT-101). Levoketoconazole and osilodrostat are in clinical testing and inhibit steroidogenesis. Blockade of ACTH receptor binding at the adrenal level is explained as a theoretical drug target. Inhibition of binding of the glucocorticoid receptor in the peripheral tissue plays a minor role due to its lack of biomonitoring options. EXPERT OPINION In our opinion, further research and drug development of pituitary-directed targets are necessary. Combination therapies may exert synergistic effects and allow for smaller and better tolerated doses, but more experience and data are needed to guide such treatment schemes.
Collapse
Affiliation(s)
- Sylvère Störmann
- a Medizinische Klinik und Poliklinik IV , Klinikum der Universität München , München , Germany
| | - Jochen Schopohl
- a Medizinische Klinik und Poliklinik IV , Klinikum der Universität München , München , Germany
| |
Collapse
|
10
|
Roof AK, Gutierrez-Hartmann A. Consider the context: Ras/ERK and PI3K/AKT/mTOR signaling outcomes are pituitary cell type-specific. Mol Cell Endocrinol 2018; 463:87-96. [PMID: 28445712 DOI: 10.1016/j.mce.2017.04.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022]
Abstract
Conserved signaling pathways are critical regulators of pituitary homeostasis and, when dysregulated, contribute to adenoma formation. Pituitary adenomas are typically benign and rarely progress to malignant cancer. Pituitary and other neuroendocrine cell types often display non-proliferative responses to ERK and PI3K, in contrast to non-endocrine cell types which typically proliferate in response to ERK and PI3K activation. These differences likely contribute to the infrequent progression to malignancy in many endocrine tumors. In this review, we highlight the Ras/ERK and PI3K/AKT/mTOR signaling pathways in each pituitary cell type, as well as in other endocrine tissues. Furthermore, we provide evidence that a balance of ERK and PI3K signaling is required to maintain pituitary homeostasis. It is unlikely that one sole oncogene will be identified as being responsible for sporadic pituitary adenoma formation. This review emphasizes the necessity to consider endocrine cell-specific contexts and the interplay of signaling pathways to define the mechanisms underlying pituitary tumorigenesis.
Collapse
Affiliation(s)
- Allyson K Roof
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Arthur Gutierrez-Hartmann
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
11
|
Worm Ørntoft MB. Review of Blood-Based Colorectal Cancer Screening: How Far Are Circulating Cell-Free DNA Methylation Markers From Clinical Implementation? Clin Colorectal Cancer 2018; 17:e415-e433. [PMID: 29678513 DOI: 10.1016/j.clcc.2018.02.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer related deaths worldwide, and late stages (III-IV) in particular have low 5-year survival rates. Stage shifting by CRC screening programs has proven effective by decreasing morbidity and mortality and in many countries national CRC screening programs have been implemented. Currently, European, Asian, and American authorities recommend screening for CRC using fecal occult blood testing, sigmoidoscopy, or colonoscopy. Because these approaches all have weaknesses (eg, poor compliance, high costs, test invasiveness), much effort has been put into the development of alternative screening approaches, many of which are blood-based. Blood-based strategies especially present the advantages of minimally invasiveness compared to endoscopies and an expectantly higher compliance rate compared to stool-based tests. The last decades have seen many discovery studies identifying promising blood-based biomarkers of CRC; however, common to all of these markers is that their clinical usefulness remains evasive. At present only one blood-based CRC screening marker has been approved in the United States. The aim of this review is to discuss the development of blood-based cell-free DNA methylation marker candidates for CRC screening. On the basis of a methodical literature search, the past, present, and future of cell-free DNA screening markers for CRC are revised and discussed. Resource limitations and technical challenges related to sensitivity and specificity measurements keep many markers at bay. Possible solutions to these problems are offered to enable markers to benefit future screening participants.
Collapse
|
12
|
Gaweł‐Bęben K, Ali N, Ellis V, Velasco G, Poghosyan Z, Ager A, Knäuper V. TMEFF2 shedding is regulated by oxidative stress and mediated by ADAMs and transmembrane serine proteases implicated in prostate cancer. Cell Biol Int 2018; 42:273-280. [PMID: 28762604 PMCID: PMC5836882 DOI: 10.1002/cbin.10832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022]
Abstract
TMEFF2 is a type I transmembrane protein with two follistatin (FS) and one EGF-like domain over-expressed in prostate cancer; however its biological role in prostate cancer development and progression remains unclear, which may, at least in part, be explained by its proteolytic processing. The extracellular part of TMEFF2 (TMEFF2-ECD) is cleaved by ADAM17 and the membrane-retained fragment is further processed by the gamma-secretase complex. TMEFF2 shedding is increased with cell crowding, a condition associated with the tumour microenvironment, which was mediated by oxidative stress signalling, requiring jun-kinase (JNK) activation. Moreover, we have identified that TMEFF2 is also a novel substrate for other proteases implicated in prostate cancer, including two ADAMs (ADAM9 and ADAM12) and the type II transmembrane serine proteinases (TTSPs) matriptase-1 and hepsin. Whereas cleavage by ADAM9 and ADAM12 generates previously identified TMEFF2-ECD, proteolytic processing by matriptase-1 and hepsin produced TMEFF2 fragments, composed of TMEFF2-ECD or FS and/or EGF-like domains as well as novel membrane retained fragments. Differential TMEFF2 processing from a single transmembrane protein may be a general mechanism to modulate transmembrane protein levels and domains, dependent on the repertoire of ADAMs or TTSPs expressed by the target cell.
Collapse
Affiliation(s)
- Katarzyna Gaweł‐Bęben
- School of MedicineUniversity of Information Technology and Management in Rzeszow2 Sucharskiego Str.35‐225 RzeszowPoland
- School of DentistryCollege of Biomedical and Life SciencesCardiff UniversityCardiffCF14 4XYUnited Kingdom
| | - Nazim Ali
- School of DentistryCollege of Biomedical and Life SciencesCardiff UniversityCardiffCF14 4XYUnited Kingdom
- School of MedicineUniversity of KeeleKeeleST5 5BGUnited Kingdom
| | - Vincent Ellis
- School of Biological SciencesUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJUnited Kingdom
| | - Gloria Velasco
- Departamento de Bioquímica y Biología Molecular Facultad de MedicinaUniversidad de Oviedo33006 OviedoSpain
| | - Zaruhi Poghosyan
- School of MedicineCollege of Biomedical and Life SciencesCardiff UniversityCardiffCF14 4XYUnited Kingdom
| | - Ann Ager
- School of MedicineCollege of Biomedical and Life SciencesCardiff UniversityCardiffCF14 4XYUnited Kingdom
| | - Vera Knäuper
- School of DentistryCollege of Biomedical and Life SciencesCardiff UniversityCardiffCF14 4XYUnited Kingdom
| |
Collapse
|