1
|
Xing B, Lei Z, Wang Z, Wang Q, Jiang Q, Zhang Z, Liu X, Qi Y, Li S, Guo X, Liu Y, Li X, Shu K, Zhang H, Bartsch JW, Nimsky C, Huang Y, Lei T. A disintegrin and metalloproteinase 22 activates integrin β1 through its disintegrin domain to promote the progression of pituitary adenoma. Neuro Oncol 2024; 26:137-152. [PMID: 37555799 PMCID: PMC10768997 DOI: 10.1093/neuonc/noad148] [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: 03/03/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Approximately 35% of pituitary adenoma (PA) display an aggressive profile, resulting in low surgical total resection rates, high recurrence rates, and worse prognosis. However, the molecular mechanism of PA invasion remains poorly understood. Although "a disintegrin and metalloproteinases" (ADAMs) are associated with the progression of many tumors, there are no reports on ADAM22 in PA. METHODS PA transcriptomics databases and clinical specimens were used to analyze the expression of ADAM22. PA cell lines overexpressing wild-type ADAM22, the point mutation ADAM22, the mutated ADAM22 without disintegrin domain, and knocking down ADAM22 were generated. Cell proliferation/invasion assays, flow cytometry, immunohistochemistry, immunofluorescence, co-immunoprecipitation, mass spectrometry, Reverse transcription-quantitative real-time PCR, phos-tag SDS-PAGE, and Western blot were performed for function and mechanism research. Nude mice xenograft models and rat prolactinoma orthotopic models were used to validate in vitro findings. RESULTS ADAM22 was significantly overexpressed in PA and could promote the proliferation, migration, and invasion of PA cells. ADAM22 interacted with integrin β1 (ITGB1) and activated FAK/PI3K and FAK/ERK signaling pathways through its disintegrin domain to promote PA progression. ADAM22 was phosphorylated by PKA and recruited 14-3-3, thereby delaying its degradation. ITGB1-targeted inhibitor (anti-itgb1) exerted antitumor effects and synergistic effects in combination with somatostatin analogs or dopamine agonists in treating PA. CONCLUSIONS ADAM22 was upregulated in PA and was able to promote PA proliferation, migration, and invasion by activating ITGB1 signaling. PKA may regulate the degradation of ADAM22 through post-transcriptional modification levels. ITGB1 may be a potential therapeutic target for PA.
Collapse
Affiliation(s)
- Biao Xing
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Zhuowei Lei
- Department of Orthopedics, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Zihan Wang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Quanji Wang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Qian Jiang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Zhang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojin Liu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Yiwei Qi
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Sihan Li
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Guo
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Liu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Xingbo Li
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Huaqiu Zhang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Jörg Walter Bartsch
- Department of Neurosurgery, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps-University Marburg, University Hospital Marburg (UKGM), Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Marburg, Germany
| | - Yimin Huang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital of Tongji medical college of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Rai A, Yelamanchi SD, Radotra BD, Gupta SK, Mukherjee KK, Tripathi M, Chhabra R, Ahuja CK, Kumar N, Pandey A, Korbonits M, Dutta P, Gaston-Massuet C. Phosphorylation of β-catenin at Serine552 correlates with invasion and recurrence of non-functioning pituitary neuroendocrine tumours. Acta Neuropathol Commun 2022; 10:138. [PMID: 36114575 PMCID: PMC9482208 DOI: 10.1186/s40478-022-01441-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Non-functioning pituitary tumours (NF-PitNETs) are common intracranial benign neoplasms that can exhibit aggressive behaviour by invading neighbouring structures and, in some cases, have multiple recurrences. Despite resulting in severe co-morbidities, no predictive biomarkers of recurrence have been identified for NF-PitNETs. In this study we have used high-throughput mass spectrometry-based analysis to examine the phosphorylation pattern of different subsets of NF-PitNETs. Based on histopathological, radiological, surgical and clinical features, we have grouped NF-PitNETs into non-invasive, invasive, and recurrent disease groups. Tumour recurrence was determined based on regular clinical and radiological data of patients for a mean follow-up of 10 years (SD ± 5.4 years). Phosphoproteomic analyses identified a unique phosphopeptide enrichment pattern which correlates with disease recurrence. Candidate phosphorylated proteins were validated in a large cohort of NF-PitNET patients by western blot and immunohistochemistry. We identified a cluster of 22 phosphopeptides upregulated in recurrent NF-PitNETs compared to non-invasive and invasive subgroups. We reveal significant phosphorylation of the β-catenin at Ser552 in recurrent and invasive NF-PitNETs, compared to non-invasive/non-recurrent NF-PitNET subgroup. Moreover, β-catenin pSer552 correlates with the recurrence free survival among 200 patients with NF-PitNET. Together, our results suggest that the phosphorylation status of β-catenin at Ser552 could act as potential biomarker of tumour recurrence in NF-PitNETs.
Collapse
|
3
|
Hu J, Yang J, Chen L, Meng X, Zhang X, Li W, Li Z, Huang G. Alterations of the Gut Microbiome in Patients With Pituitary Adenoma. Pathol Oncol Res 2022; 28:1610402. [PMID: 35991836 PMCID: PMC9385953 DOI: 10.3389/pore.2022.1610402] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022]
Abstract
Pituitary adenoma (PA) includes invasive pituitary adenoma (IPA) and noninvasive pituitary adenoma (NIPA), which are associated with the endocrine system. The gut microbiome plays an important role in human metabolism, but the association between the gut microbiome and pituitary adenoma remains unclear. A total of 44 subjects were enrolled in this study. Of these, 29 PA patients were further divided into IPA patients (n = 13) and NIPA patients (n = 16), while 15 healthy age-matched subjects were defined as control subjects. We collected faecal samples and characterized the gut microbial profiles by metagenomic sequencing using the Illumina X-ten platform. PLS-DA showed different microbial clusters among the three groups, and slightly different microbial ecological networks were observed. LEfSe analysis revealed significant alterations in the microbial community among PA patients. In particular, the enrichment of Clostridium innocuum, along with the reduced abundance of Oscillibacter sp. 57_20 and Fusobacterium mortiferum, were observed both in the IPA and NIPA groups compared to the control group. Moreover, PA patients could be effectively classified based on these bacteria using a support vector machine algorithm. In summary, this study demonstrated significant differences in the gut microbiome between PA patients and healthy controls. Future mechanistic experiments are needed to determine whether such alterations are a cause or consequence of pituitary adenoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zongyang Li
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Guodong Huang
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
4
|
Raverot G, Ilie MD, Lasolle H, Amodru V, Trouillas J, Castinetti F, Brue T. Aggressive pituitary tumours and pituitary carcinomas. Nat Rev Endocrinol 2021; 17:671-684. [PMID: 34493834 DOI: 10.1038/s41574-021-00550-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2021] [Indexed: 02/07/2023]
Abstract
Although usually benign, anterior pituitary tumours occasionally exhibit aggressive behaviour, with invasion of surrounding tissues, rapid growth, resistance to conventional treatments and multiple recurrences. In very rare cases, they metastasize and are termed pituitary carcinomas. The time between a 'classical' pituitary tumour and a pituitary carcinoma can be years, which means that monitoring should be performed regularly in patients with clinical (invasion and/or tumour growth) or pathological (Ki67 index, mitotic count and/or p53 detection) markers suggesting aggressiveness. However, although both invasion and proliferation have prognostic value, such parameters cannot predict outcome or malignancy without metastasis. Future research should focus on the biology of both tumour cells and their microenvironment, hopefully with improved therapeutic outcomes. Currently, the initial therapeutic approach for aggressive pituitary tumours is generally to repeat surgery or radiotherapy in expert centres. Standard medical treatments usually have no effect on tumour progression but they can be maintained on a long-term basis to, at least partly, control hypersecretion. In cases where standard treatments prove ineffective, temozolomide, the sole formally recommended treatment, is effective in only one-third of patients. Personalized use of emerging therapies, including peptide receptor radionuclide therapy, angiogenesis-targeted therapy and immunotherapy, will hopefully improve the outcomes of patients with this severe condition.
Collapse
Affiliation(s)
- Gérald Raverot
- Endocrinology Department, Reference Centre for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
- Lyon 1 University, Villeurbanne, France
- INSERM U1052, CNRS UMR5286, Cancer Research Centre of Lyon (CRLC), Lyon, France
| | - Mirela Diana Ilie
- Lyon 1 University, Villeurbanne, France
- INSERM U1052, CNRS UMR5286, Cancer Research Centre of Lyon (CRLC), Lyon, France
- Endocrinology Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Hélène Lasolle
- Endocrinology Department, Reference Centre for Rare Pituitary Diseases HYPO, "Groupement Hospitalier Est" Hospices Civils de Lyon, Bron, France
- Lyon 1 University, Villeurbanne, France
- INSERM U1052, CNRS UMR5286, Cancer Research Centre of Lyon (CRLC), Lyon, France
| | - Vincent Amodru
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Endocrinology Department, Hôpital de la Conception, Reference Centre for Rare Pituitary Diseases HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | | | - Frédéric Castinetti
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Endocrinology Department, Hôpital de la Conception, Reference Centre for Rare Pituitary Diseases HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - Thierry Brue
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Endocrinology Department, Hôpital de la Conception, Reference Centre for Rare Pituitary Diseases HYPO, Marseille, France.
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Marseille, France.
| |
Collapse
|
5
|
Large Scale Molecular Studies of Pituitary Neuroendocrine Tumors: Novel Markers, Mechanisms and Translational Perspectives. Cancers (Basel) 2021; 13:cancers13061395. [PMID: 33808624 PMCID: PMC8003417 DOI: 10.3390/cancers13061395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/28/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pituitary neuroendocrine tumors are non-cancerous tumors of the pituitary gland, that may overproduce hormones leading to serious health conditions or due to tumor size cause chronic headache, vertigo or visual impairment. In recent years pituitary neuroendocrine tumors are studied with the latest molecular biology methods that simultaneously investigate a large number of factors to understand the mechanisms of how these tumors develop and how they could be diagnosed or treated. In this review article, we have studied literature reports, compiled information and described molecular factors that could affect the development and clinical characteristics of pituitary neuroendocrine tumors, discovered factors that overlap between several studies using large scale molecular analysis and interpreted the potential involvement of these factors in pituitary tumor development. Overall, this study provides a valuable resource for understanding the biology of pituitary neuroendocrine tumors. Abstract Pituitary neuroendocrine tumors (PitNETs) are non-metastatic neoplasms of the pituitary, which overproduce hormones leading to systemic disorders, or tumor mass effects causing headaches, vertigo or visual impairment. Recently, PitNETs have been investigated in large scale (exome and genome) molecular analyses (transcriptome microarrays and sequencing), to uncover novel markers. We performed a literature analysis on these studies to summarize the research data and extrapolate overlapping gene candidates, biomarkers, and molecular mechanisms. We observed a tendency in samples with driver mutations (GNAS, USP8) to have a smaller overall mutational rate, suggesting driver-promoted tumorigenesis, potentially changing transcriptome profiles in tumors. However, direct links from drivers to signaling pathways altered in PitNETs (Notch, Wnt, TGF-β, and cell cycle regulators) require further investigation. Modern technologies have also identified circulating nucleic acids, and pinpointed these as novel PitNET markers, i.e., miR-143-3p, miR-16-5p, miR-145-5p, and let-7g-5p, therefore these molecules must be investigated in the future translational studies. Overall, large-scale molecular studies have provided key insight into the molecular mechanisms behind PitNET pathogenesis, highlighting previously reported molecular markers, bringing new candidates into the research field, and reapplying traditional perspectives to newly discovered molecular mechanisms.
Collapse
|
6
|
Zhao P, Cheng J, Li B, Nie D, Li C, Gui S, Wang H, Zhang Y. Up-regulation of the expressions of MiR-149-5p and MiR-99a-3p in exosome inhibits the progress of pituitary adenomas. Cell Biol Toxicol 2021; 37:633-651. [PMID: 33400021 DOI: 10.1007/s10565-020-09570-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 11/04/2020] [Indexed: 10/22/2022]
Abstract
This study explored the function of microRNAs (miRNAs) in invasive pituitary adenomas (IPA), and developed a microRNA-exosome strategy for the disease treatment. Differentially expressed miRNAs and tumor-associated markers in IPA, non-invasive pituitary adenoma (NIPA), and rat pituitary adenoma cells were identified by bioinformatics analysis and qRT-PCR. Then, the cells were treated by miR-149-5p and miR-99a-3p mimics or inhibitors, or incubated with modified exosome with overexpressed or silenced miRNAs. The cell behaviors were analyzed by molecular experiments. Xenograft assays were constructed by injection of pituitary adenoma cells and exosome into NU/NU nude mice. Tumor size, weight, and expressions of markers related to miRNAs and angiogenesis were determined. Target genes for miR-99a-3p and miR-149 were predicted and verified by bioinformatics analysis and molecular experiments. Twenty differentially expressed miRNAs were identified, among which miR-99a-3p and miR-149 were inhibited in both pituitary adenoma cells and tissues significantly. Expressions of E-cadherin and p53 were down-regulated, while those of MMP-2, MMP-9, N-cadherin, Vimentin, and VEGF were up-regulated in pituitary adenoma cells and tissues, especially in IPA. Further experiments revealed that overexpressed miR-149 and miR-99a-3p inhibited the growth and metastasis of pituitary adenoma cells and tube formation of endothelial cells. MiR-149 and miR-99a-3p overexpressed by exosome showed similar suppressive effects on cell viability, metastasis, tube formation ability, in vivo tumor growth, and expressions of angiogenesis-related markers. Further analysis showed that NOVA1, DTL, and RAB27B were targeted by miR-99a-3p. This study found that overexpressed miR-149-5p and miR-99a-3p induced by exosome could suppress the progression of IPA. 1. MiR-149-5p and miR-99a-3p affect the expression of EMT- and ECM-related markers and tumor-related genes in rat pituitary adenoma cells treated with exosomes. 2. Exosome inhibited the tumor growth. 3. Overexpressed miR-149-5p and miR-99a-3p induced by exosome.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| | - Jianhua Cheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Bin Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Ding Nie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Chuzhong Li
- Department of Cell Biology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Hongyun Wang
- Department of Cell Biology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Department of Cell Biology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Srirangam Nadhamuni V, Korbonits M. Novel Insights into Pituitary Tumorigenesis: Genetic and Epigenetic Mechanisms. Endocr Rev 2020; 41:bnaa006. [PMID: 32201880 PMCID: PMC7441741 DOI: 10.1210/endrev/bnaa006] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/19/2020] [Indexed: 02/08/2023]
Abstract
Substantial advances have been made recently in the pathobiology of pituitary tumors. Similar to many other endocrine tumors, over the last few years we have recognized the role of germline and somatic mutations in a number of syndromic or nonsyndromic conditions with pituitary tumor predisposition. These include the identification of novel germline variants in patients with familial or simplex pituitary tumors and establishment of novel somatic variants identified through next generation sequencing. Advanced techniques have allowed the exploration of epigenetic mechanisms mediated through DNA methylation, histone modifications and noncoding RNAs, such as microRNA, long noncoding RNAs and circular RNAs. These mechanisms can influence tumor formation, growth, and invasion. While genetic and epigenetic mechanisms often disrupt similar pathways, such as cell cycle regulation, in pituitary tumors there is little overlap between genes altered by germline, somatic, and epigenetic mechanisms. The interplay between these complex mechanisms driving tumorigenesis are best studied in the emerging multiomics studies. Here, we summarize insights from the recent developments in the regulation of pituitary tumorigenesis.
Collapse
Affiliation(s)
- Vinaya Srirangam Nadhamuni
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| |
Collapse
|
8
|
Structural and functional annotation of PR/SET Domain (PRDM) protein family: In-silico study elaborating role of PRDM12 mutation in congenital insensitivity to pain. Comput Biol Chem 2020; 89:107382. [PMID: 33010785 DOI: 10.1016/j.compbiolchem.2020.107382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/11/2020] [Accepted: 09/18/2020] [Indexed: 11/22/2022]
Abstract
Congenital insensitivity to pain (CIP), classified as a type of hereditary sensory and autonomic neuropathies, is a rare disease in which the affected individuals fail to perceive sensation of pain. One of the PR/SET Domain Proteins, PRDM12, has been identified in recent past as a candidate gene for congenital insensitivity to pain. In the present study, we performed whole exome sequencing in a Pakistani family with CIP phenotype to ascertain the causative mutation. We identified a previously described alanine repeat duplication in PRDM12 (Ala353_Ala359dup) in this family. After this, we performed structural annotations for PR/SET Domain (PRDM) containing protein family to prognosticate the potential hypothetical structure of PRDM proteins with physical and chemical parameters. Out of nineteen members of this family, four members (PRDM5, PRDM8, PRDM12 and PRDM13) were specially focused because of their role in neurological disorders. Predictions about structure and interactions of these proteins revealed novel interacting molecules and pathways. Detailed in silico analysis of PRDM12 was performed to elaborate importance of its domain structure in interaction with other proteins and its role in pain insensitivity phenotype. These results have substantially enhanced our understanding regarding the etiology of congenital pain insensitivity and would stimulate further research on therapy and prevention.
Collapse
|
9
|
Casamassimi A, Rienzo M, Di Zazzo E, Sorrentino A, Fiore D, Proto MC, Moncharmont B, Gazzerro P, Bifulco M, Abbondanza C. Multifaceted Role of PRDM Proteins in Human Cancer. Int J Mol Sci 2020; 21:ijms21072648. [PMID: 32290321 PMCID: PMC7177584 DOI: 10.3390/ijms21072648] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
The PR/SET domain family (PRDM) comprise a family of genes whose protein products share a conserved N-terminal PR [PRDI-BF1 (positive regulatory domain I-binding factor 1) and RIZ1 (retinoblastoma protein-interacting zinc finger gene 1)] homologous domain structurally and functionally similar to the catalytic SET [Su(var)3-9, enhancer-of-zeste and trithorax] domain of histone methyltransferases (HMTs). These genes are involved in epigenetic regulation of gene expression through their intrinsic HMTase activity or via interactions with other chromatin modifying enzymes. In this way they control a broad spectrum of biological processes, including proliferation and differentiation control, cell cycle progression, and maintenance of immune cell homeostasis. In cancer, tumor-specific dysfunctions of PRDM genes alter their expression by genetic and/or epigenetic modifications. A common characteristic of most PRDM genes is to encode for two main molecular variants with or without the PR domain. They are generated by either alternative splicing or alternative use of different promoters and play opposite roles, particularly in cancer where their imbalance can be often observed. In this scenario, PRDM proteins are involved in cancer onset, invasion, and metastasis and their altered expression is related to poor prognosis and clinical outcome. These functions strongly suggest their potential use in cancer management as diagnostic or prognostic tools and as new targets of therapeutic intervention.
Collapse
Affiliation(s)
- Amelia Casamassimi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (E.D.Z.); (A.S.)
- Correspondence: (A.C.); (C.A.); Tel.: +39-081-566-7579 (A.C.); +39-081-566-7568 (C.A.)
| | - Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (E.D.Z.); (A.S.)
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Anna Sorrentino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (E.D.Z.); (A.S.)
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (D.F.); (M.C.P.); (P.G.)
| | - Maria Chiara Proto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (D.F.); (M.C.P.); (P.G.)
| | - Bruno Moncharmont
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (D.F.); (M.C.P.); (P.G.)
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (E.D.Z.); (A.S.)
- Correspondence: (A.C.); (C.A.); Tel.: +39-081-566-7579 (A.C.); +39-081-566-7568 (C.A.)
| |
Collapse
|
10
|
Wang H, Sang W. Association of NM23 polymorphisms and clinicopathological features and recurrence of invasive pituitary adenomas. Pituitary 2020; 23:113-119. [PMID: 31734851 DOI: 10.1007/s11102-019-01006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Pituitary adenomas (PAs) are intracranial tumors, deriving from anterior pituitary cells. Previously, expression of non-metastasis-23 (NM23) gene has been shown to correlate with the progression of PAs. In this study, we aim to determine whether there is association between specific NM23 polymorphisms and invasive pituitary adenoma (IPA). METHODS Genotypes of rs2302254 and rs16949649 of NM23 were identified in the peripheral venous blood of patients by PCR-RLFP. Next, the correlation between specific genotypes of rs2302254 and rs16949649 and risk of IPA was investigated. Finally, the correlations between NM23 polymorphisms and tumor size, Ki67 LI and recurrence of IPA were analyzed with 3 to 24 months follow-up for the enrolled patients. RESULTS We observed that the TT genotype at rs16949649 correlated closely with a high risk of IPA, while CC and CT genotypes reduced the risk of IPA. CC genotype at rs2302254 increased the risk of IPA, while CT and TT genotypes reduced the risk of IPA. Trs16949649Crs2302254 haplotype of NM23 was found to be a high-risk haplotype for IPA. TT genotype at rs16949649 and CC genotype at rs2302254 were associated with higher rates of tumors larger than 20 mm, Ki67 LI and tumor recurrence. CONCLUSION Taken together, the present study provides evidence that NM23 polymorphisms are closely associated with the incidence and recurrence of IPA. Specifically, TT genotype at rs16949649 and CC genotype at rs2302254 are risk factors of IPA. NM23 polymorphisms could therefore be used as a reference for clinical diagnosis and prognosis of IPA.
Collapse
Affiliation(s)
- Hua Wang
- 1st Ward of Department of Neurosurgery, Chifeng Municipal Hospital, No. 1, Zhaowuda Road, Chifeng, 024000, Inner Mongolia Autonomous Region, People's Republic of China
| | - Wenyuan Sang
- 1st Ward of Department of Neurosurgery, Chifeng Municipal Hospital, No. 1, Zhaowuda Road, Chifeng, 024000, Inner Mongolia Autonomous Region, People's Republic of China.
| |
Collapse
|
11
|
Genetics of Pituitary Tumours. EXPERIENTIA. SUPPLEMENTUM 2019. [PMID: 31588533 DOI: 10.1007/978-3-030-25905-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Pituitary tumours are relatively common in the general population. Most often they occur sporadically, with somatic mutations accounting for a significant minority of somatotroph and corticotroph adenomas. Pituitary tumours can also develop secondary to germline mutations as part of a complex syndrome or as familial isolated pituitary adenomas. Tumours occurring in a familial setting may present at a younger age and can behave more aggressively with resistance to treatment. This chapter will focus on the genetics and molecular pathogenesis of pituitary tumours.
Collapse
|
12
|
Välimäki N, Schalin-Jäntti C, Karppinen A, Paetau A, Kivipelto L, Aaltonen LA, Karhu A. Genetic and Epigenetic Characterization of Growth Hormone-Secreting Pituitary Tumors. Mol Cancer Res 2019; 17:2432-2443. [PMID: 31578227 DOI: 10.1158/1541-7786.mcr-19-0434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Somatic driver mechanisms of pituitary adenoma pathogenesis have remained incompletely characterized; apart from mutations in the stimulatory Gα protein (Gαs encoded by GNAS) causing activated cAMP synthesis, pathogenic variants are rarely found in growth hormone-secreting pituitary tumors (somatotropinomas). The purpose of the current work was to clarify how genetic and epigenetic alterations contribute to the development of somatotropinomas by conducting an integrated copy number alteration, whole-genome and bisulfite sequencing, and transcriptome analysis of 21 tumors. Somatic mutation burden was low, but somatotropinomas formed two subtypes associated with distinct aneuploidy rates and unique transcription profiles. Tumors with recurrent chromosome aneuploidy (CA) were GNAS mutation negative (Gsp- ). The chromosome stable (CS) -group contained Gsp+ somatotropinomas and two totally aneuploidy-free Gsp- tumors. Genes related to the mitotic G1-S-checkpoint transition were differentially expressed in CA- and CS-tumors, indicating difference in mitotic progression. Also, pituitary tumor transforming gene 1 (PTTG1), a regulator of sister chromatid segregation, showed abundant expression in CA-tumors. Moreover, somatotropinomas displayed distinct Gsp genotype-specific methylation profiles and expression quantitative methylation (eQTM) analysis revealed that inhibitory Gα (Gαi) signaling is activated in Gsp+ tumors. These findings suggest that aneuploidy through modulated driver pathways may be a causative mechanism for tumorigenesis in Gsp- somatotropinomas, whereas Gsp+ tumors with constitutively activated cAMP synthesis seem to be characterized by DNA methylation activated Gαi signaling. IMPLICATIONS: These findings provide valuable new information about subtype-specific pituitary tumorigenesis and may help to elucidate the mechanisms of aneuploidy also in other tumor types.
Collapse
Affiliation(s)
- Niko Välimäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Atte Karppinen
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, HUSLAB, University of Helsinki, Helsinki, Finland
| | - Leena Kivipelto
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland. .,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| |
Collapse
|
13
|
Németh K, Darvasi O, Likó I, Szücs N, Czirják S, Reiniger L, Szabó B, Kurucz PA, Krokker L, Igaz P, Patócs A, Butz H. Next-generation sequencing identifies novel mitochondrial variants in pituitary adenomas. J Endocrinol Invest 2019; 42:931-940. [PMID: 30684245 PMCID: PMC6647476 DOI: 10.1007/s40618-019-1005-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/08/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Disrupted mitochondrial functions and genetic variants of mitochondrial DNA (mtDNA) have been observed in different human neoplasms. Next-generation sequencing (NGS) can be used to detect even low heteroplasmy-level mtDNA variants. We aimed to investigate the mitochondrial genome in pituitary adenomas by NGS. METHODS We analysed 11 growth hormone producing and 33 non-functioning [22 gonadotroph and 11 hormone immunonegative] pituitary adenomas using VariantPro™ Mitochondrion Panel on Illumina MiSeq instrument. Revised Cambridge Reference Sequence (rCRS) of the mtDNA was used as reference. Heteroplasmy was determined using a 3% cutoff. RESULTS 496 variants were identified in pituitary adenomas with overall low level of heteroplasmy (7.22%). On average, 35 variants were detected per sample. Samples harbouring the highest number of variants had the highest Ki-67 indices independently of histological subtypes. We identified eight variants (A11251G, T4216C, T16126C, C15452A, T14798C, A188G, G185A, and T16093C) with different prevalences among different histological groups. T16189C was found in 40% of non-recurrent adenomas, while it was not present in the recurrent ones. T14798C and T4216C were confirmed by Sanger sequencing in all 44 samples. 100% concordance was found between NGS and Sanger method. CONCLUSIONS NGS is a reliable method for investigating mitochondrial genome and heteroplasmy in pituitary adenomas. Out of the 496 detected variants, 414 have not been previously reported in pituitary adenoma. The high number of mtDNA variants may contribute to adenoma genesis, and some variants (i.e., T16189C) might associate with benign behaviour.
Collapse
Affiliation(s)
- K Németh
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - O Darvasi
- "Lendulet" Hereditary Endocrine Tumours Research Group, Hungarian Academy of Sciences and Semmelweis University, 46 Szentkiralyi Street, Budapest, H-1088, Hungary
| | - I Likó
- "Lendulet" Hereditary Endocrine Tumours Research Group, Hungarian Academy of Sciences and Semmelweis University, 46 Szentkiralyi Street, Budapest, H-1088, Hungary
| | - N Szücs
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - S Czirják
- National Institute of Clinical Neurosciences, Budapest, Hungary
| | - L Reiniger
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - B Szabó
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - P A Kurucz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - L Krokker
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - P Igaz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - A Patócs
- "Lendulet" Hereditary Endocrine Tumours Research Group, Hungarian Academy of Sciences and Semmelweis University, 46 Szentkiralyi Street, Budapest, H-1088, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| | - H Butz
- "Lendulet" Hereditary Endocrine Tumours Research Group, Hungarian Academy of Sciences and Semmelweis University, 46 Szentkiralyi Street, Budapest, H-1088, Hungary.
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
14
|
Wang K, Zhou W, Meng P, Wang P, Zhou C, Yao Y, Wu S, Wang Y, Zhao J, Zou D, Jin G. Immune-related somatic mutation genes are enriched in PDACs with diabetes. Transl Oncol 2019; 12:1147-1154. [PMID: 31203147 PMCID: PMC6581966 DOI: 10.1016/j.tranon.2019.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
The bidirectional interaction between pancreatic cancer (PanCa) and diabetes has been confirmed by epidemiological studies, but until now, the underlying molecular mechanisms for this connection is not fully understood yet. Here, we analyzed the clinical and genomic data of 26 pancreatic ductal adenocarcinoma (PDAC) patients without diabetes, and six diabetic PDAC patients, whose tumors underwent targeted next-generation sequencing (551 cancer-related genes included). Ingenuity Pathway Analysis (IPA) was performed to investigate genetic alterations and biological consequences associated with PDACs with or without diabetes. We identified 345 somatic mutations of 153 genes in test cohort and a positive association between diabetes duration and somatic mutation burden. KRAS, TP53, and SMAD4 were the top3 commonly mutated genes at a similar frequency compared to the Cancer Genome Atlas (TCGA) data. Several novel but infrequent mutations in other genes (MDC1, PRB2, and PRB4) were also found. Besides, 13 mutated genes (PIK3CD, SNCAIP, IRF4, HLA-A, NOTCH4, PIM1, ETV6, B2M, CD70, PRDM14, TGFBR1, FLT1, and PARP2) were uniquely found in the diabetic group, mainly involved in immune-related pathways. Further targeted sequencing of these genes in an independent validation cohort (n = 50) revealed significant enrichment in the diabetic group (n = 18, P = 2.6964E-08). Long-standing diabetes (≥3-year duration) may induce increasing somatic mutations with time, facilitating tumor initiation. Gene mutants associated with immune-related pathways could be used to distinguish the diabetic PDAC patients from the non-diabetic cases and allow more selective treatment.
Collapse
Affiliation(s)
- Kaixuan Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Wei Zhou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Peng Meng
- Biotecan Pharmaceuticals Co., Ltd, Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Peng Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chunhua Zhou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yao Yao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shouxin Wu
- Biotecan Pharmaceuticals Co., Ltd, Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Yu Wang
- Biotecan Pharmaceuticals Co., Ltd, Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Jiangman Zhao
- Biotecan Pharmaceuticals Co., Ltd, Shanghai Zhangjiang Institute of Medical Innovation, Shanghai, 201204, China
| | - Duowu Zou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China; Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Jin
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
15
|
Zhang Y, Yan L, Yao W, Chen K, Xu H, Ye Z. Integrated Analysis of Genetic Abnormalities of the Histone Lysine Methyltransferases in Prostate Cancer. Med Sci Monit 2019; 25:193-239. [PMID: 30616239 PMCID: PMC6330996 DOI: 10.12659/msm.912294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The histone methyltransferase (HMT) family includes histone lysine methyltransferases (HKMTs) and histone/protein arginine methyltransferases (PRMTs). The role of HMT gene variants in prostate cancer remains unknown. Therefore, this study aimed to evaluate HMT gene variants in the pathogenesis and prognosis of human prostate cancer, using in vitro cell studies and bioinformatics analysis. Material/Methods Integrative bioinformatics analysis of the expression of 51 HMT genes in human prostate cancer was based on datasets from the Cancer Genome Atlas (TCGA). Correlation and regression analysis were used to identify critical HMTs in prostate cancer. Kaplan-Meier and the area under the receiver operating characteristics curve (AUROC) were performed to evaluate the function of the HMTs on prognosis. Gene expression and function of 22Rv1 human prostate carcinoma cells were studied. Results The HMT genes identified to have a role in the pathogenesis of prostate cancer included the EZH2, SETD5, PRDM12, NSD1, SETD6, SMYD1, and the WHSC1L1 gene. The EZH2, SETD5, and SMYD1 genes were selected as a prognostic panel, with the SUV420H2 HMT gene. SETD2, NSD1, and ASH1L were identified as critical genes in the development of castration-resistant prostate cancer (CRPC), similar to mixed-lineage leukemia (MLL) complex family members. Knockdown of the SETD5 gene in 22Rv1 prostate carcinoma cells in vitro inhibited cancer cell growth and migration. Conclusions HMT gene variants may have a role in the pathogenesis of prostate cancer. Future studies may determine the role of HMT genes as prognostic biomarkers in patients with prostate cancer.
Collapse
Affiliation(s)
- Yangjun Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Libin Yan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, AL, China (mainland).,Institute of Urology of Hubei Province, Wuhan, Hubei, China (mainland)
| |
Collapse
|
16
|
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: 42] [Impact Index Per Article: 8.4] [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
|
17
|
Fu YC, Chen N, Qiu ZL, Liu L, Shen J. Compound pathogenic mutation in the USH2A gene in Chinese RP families detected by whole‑exome sequencing. Mol Med Rep 2018; 18:5016-5022. [PMID: 30280194 PMCID: PMC6236299 DOI: 10.3892/mmr.2018.9530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Retinitis pigmentosa (RP) is a common form of inherited retinal degeneration that causes progressive loss of vision or adult blindness, characterized by the impairment of rod and cone photoreceptors. At present, mutations in >60 pathogenic genes have been confirmed to cause RP. The predominant modes of inheritance are autosomal dominant, autosomal recessive and X‑linked. In addition, other modes of inheritance, including digenic or mitochondrial inheritance, have been reported. In previous decades, with the development of sequencing techniques, significant advances in identifying novel RP pathogenic genes and screening mutations have been made. In the present study, whole‑exome sequencing was performed on samples from two Chinese pedigrees diagnosed with RP. A compound heterozygous mutation in the gene usherin 2A (USH2A; c.6,485+5G>A/c.11,156G>A) and a heterozygous X‑linked mutation in the gene retinitis pigmentosa 2 (RP2) ARL3 GTPase‑activating protein (RP2; c.358C>T) were identified by Sanger sequencing and co‑segregation analysis, of which the pathogenic mutation (c.6,485+5G>A) in USH2A has not been previously reported among Chinese patients. The findings of the present study may expand on current knowledge of RP among the Chinese population, providing essential assistance in the molecular diagnosis and screening of RP, and promoting further investigation of the pathogenesis of RP.
Collapse
Affiliation(s)
- Yue-Chuan Fu
- Department of Ophthalmology, The Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200120, P.R. China
| | - Na Chen
- Department of Ophthalmology, The Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200120, P.R. China
| | - Zi-Long Qiu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Lin Liu
- Department of Ophthalmology, The Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200120, P.R. China
| | - Jie Shen
- Department of Ophthalmology, The Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
18
|
Chen Z, Gao W, Pu L, Zhang L, Han G, Zuo X, Zhang Y, Li X, Shen H, Wu J, Wang X. PRDM8 exhibits antitumor activities toward hepatocellular carcinoma by targeting NAP1L1. Hepatology 2018; 68:994-1009. [PMID: 29572888 DOI: 10.1002/hep.29890] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is a major leading cause of cancer mortality worldwide. PRDI-BF1 and RIZ homology domain containing 8 (PRDM8) is a key regulator in neural development and testis steroidogenesis; however, its role in liver carcinogenesis remains to be investigated. In this study, PRDM8 was found to be down-regulated in HCC, which was linked with shorter recurrence-free survival. Lentiviral-based overexpression and knockdown approaches showed that PRDM8 inhibited HCC cell proliferation, migration, and invasion. PRDM8 caused G1/S cell cycle arrest and induced apoptosis. An in vivo tumor model confirmed the antitumor role of PRDM8 in HCC growth and metastasis. Mechanistic study showed that PRDM8 suppressed the PI3K/AKT/mTOR signaling cascade through the regulation of nucleosome assembly protein 1-like 1 (NAP1L1). Conclusion: PRDM8 as a functional tumor suppressor is frequently down-regulated in HCC. Through regulating NAP1L1, PRDM8 inhibits PI3K/AKT/mTOR signaling in HCC. PRDM8 is a potential target for therapies of HCC. (Hepatology 2018).
Collapse
Affiliation(s)
- Zhiqiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liyong Pu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Long Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Guoyong Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Xueliang Zuo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Yao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Xiangcheng Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jindao Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xuehao Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory on Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, China
| |
Collapse
|
19
|
Wang J, Wang D, Wan D, Ma Q, Liu Q, Li J, Li Z, Gao Y, Jiang G, Ma L, Liu J, Li C. Circular RNA In Invasive and Recurrent Clinical Nonfunctioning Pituitary Adenomas: Expression Profiles and Bioinformatic Analysis. World Neurosurg 2018; 117:e371-e386. [DOI: 10.1016/j.wneu.2018.06.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/28/2023]
|
20
|
Renner U, Ciato D, Stalla GK. Recent advances in understanding corticotroph pituitary tumor initiation and progression. F1000Res 2018; 7. [PMID: 30228864 PMCID: PMC6117851 DOI: 10.12688/f1000research.14789.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2018] [Indexed: 11/20/2022] Open
Abstract
Cushing’s disease is the most frequent form of hypercortisolism and is caused by hypophyseal corticotroph adenomas secreting excessive amounts of adrenocorticotropic hormone. Most of the tumors develop sporadically and only a limited number of corticotroph adenomas have been found to be associated with different neuroendocrine syndromes or with familial isolated pituitary adenomas. The pathogenic mechanisms of corticotroph adenomas are largely unknown, but the discovered aberrant chaperoning activity of heat shock protein 90 on the one hand and the presence of ubiquitin-specific protease 8 mutations on the other hand partially explained the causes of their development. Corticotroph tumors arise initially as benign microadenomas but with time form invasively growing aggressive macroadenomas which can switch to corticotroph carcinomas in extremely rare cases. The mechanisms through which corticotroph tumors escape from glucocorticoid negative feedback are still poorly understood, as are the processes that trigger the progression of benign corticotroph adenomas toward aggressive and malignant phenotypes. This review summarizes recent findings regarding initiation and progression of corticotroph pituitary tumors.
Collapse
Affiliation(s)
- Ulrich Renner
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Denis Ciato
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Günter K Stalla
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| |
Collapse
|
21
|
Mellby LD, Nyberg AP, Johansen JS, Wingren C, Nordestgaard BG, Bojesen SE, Mitchell BL, Sheppard BC, Sears RC, Borrebaeck CAK. Serum Biomarker Signature-Based Liquid Biopsy for Diagnosis of Early-Stage Pancreatic Cancer. J Clin Oncol 2018; 36:2887-2894. [PMID: 30106639 DOI: 10.1200/jco.2017.77.6658] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a 5-year survival of < 10% because of diffuse symptoms leading to late-stage diagnosis. That survival could increase significantly if localized tumors could be detected early. Therefore, we used multiparametric analysis of blood samples to obtain a novel biomarker signature of early-stage PDAC. The signature was derived from a large patient cohort, including patients with well-defined early-stage (I and II) PDAC. This biomarker signature was validated subsequently in an independent patient cohort. PATIENTS AND METHODS The biomarker signature was derived from a case-control study, using a Scandinavian cohort, consisting of 16 patients with stage I, 132 patients with stage II, 65 patients with stage III, and 230 patients with stage IV PDAC, and 888 controls. This signature was validated subsequently in an independent case-control cohort in the United States with 15 patients with stage I, 75 patients with stage II, 15 patients with stage III, and 38 patients with stage IV PDAC, and 219 controls. An antibody microarray platform was used to identify the serum biomarker signature associated with early-stage PDAC. RESULTS Using the Scandinavian case-control study, a biomarker signature was created, discriminating samples derived from patients with stage I and II from those from controls with a receiver operating characteristic area under the curve value of 0.96. This signature, consisting of 29 biomarkers, was then validated in an independent case-control study in the United States. The biomarker signature could discriminate patients with stage I and II PDAC from controls in this independent patient cohort with a receiver operating characteristic area under the curve value of 0.96. CONCLUSION This serum biomarker signature might represent a tenable approach to detecting early-stage, localized PDAC if these findings are supported by a prospective validation study.
Collapse
Affiliation(s)
- Linda D Mellby
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Andreas P Nyberg
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Julia S Johansen
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Christer Wingren
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Børge G Nordestgaard
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Stig E Bojesen
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Breeana L Mitchell
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Brett C Sheppard
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Rosalie C Sears
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| | - Carl A K Borrebaeck
- Linda D. Mellby and Andreas P. Nyberg, Immunovia AB; Christer Wingren and Carl A.K. Borrebaeck, Lund University, Lund, Sweden; Julia S. Johansen, University of Copenhagen, Copenhagen; Børge G. Nordestgaard and Stig E. Bojesen, Copenhagen University Hospital, Herlev, Denmark; and Breeana L. Mitchell, Brett C. Sheppard, and Rosalie C. Sears, Oregon Health and Science University, Portland, OR
| |
Collapse
|
22
|
Yan J, Chen G, Zhao X, Chen F, Wang T, Miao F. High expression of diffuse panbronchiolitis critical region 1 gene promotes cell proliferation, migration and invasion in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun 2017; 495:1908-1914. [PMID: 29242154 DOI: 10.1016/j.bbrc.2017.12.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
Diffuse panbronchiolitis critical region 1 (DPCR1) is located in the major histocompatibility complex (MHC) class I. It was reported to be downregulated in invasive pituitary adenoma compared with that in non-invasive tumors, but upregulated in the precursor of gastric carcinogenesis. However, the direct effect of DPCR1 on cancer cells has rarely been reported, and the role DPCR1 in pancreatic ductal adenocarcinoma (PDAC) remains unclear. The clinical sample validation and public data analysis of the present study demonstrated that DPCR1 was upregulated markedly in PDAC and this high expression was negatively correlated with the patient prognosis. Functionally, knocking down DPCR1 in PDAC cell lines inhibited cell proliferation, migration and invasion in vitro. Tumor xenograft experiments further showed that suppression of DPCR1 inhibited tumor growth in vivo. In addition, the results of RNA deep sequencing and qRT-PCR assay showed that DPCR1 participated in PADC progression by regulating nuclear factor-kappa B signaling pathway, suggesting that it might be a novel oncogene in tumor progression and a potential therapeutic target in PDAC as well.
Collapse
Affiliation(s)
- Jiayi Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Guanghui Chen
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Xuesong Zhao
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Fangying Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China
| | - Ting Wang
- Department of Bone Tumor Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Fei Miao
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai 200025, China.
| |
Collapse
|