1
|
Fernández-Rhodes M, Suárez M, Molet I, Suárez-García S, Arola L, Sze SK, Gallart-Palau X, Serra A. Molecular Imprints of Clinical Comorbidities in Hypothalamic Extracellular Vesicles at the Onset of Obesity. ACS OMEGA 2025; 10:23563-23581. [PMID: 40521556 PMCID: PMC12163634 DOI: 10.1021/acsomega.5c02274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025]
Abstract
The onset of obesity is characterized by early physiological and molecular changes, including leptin resistance and hypothalamic dysfunction, preceding significant weight gain and metabolic complications. Extracellular vesicles (EVs) are key mediators of intercellular communication, reflecting early pathological shifts in metabolic disorders. This study investigates the role of hypothalamic EVs (hEVs) in early obesogenic insult and their potential implications for obesity-related comorbidities. Using a hamster model fed a high fat diet for 30 days, next-generation proteomics revealed altered hEV protein compositions linked to cellular metabolism, neuroinflammation, and metabolic dysfunction, mirroring early obesity-related dysfunction. These findings highlight the adaptive molecular profiles of hEVs during early obesogenic insult and their potential as biomarkers and molecular mediators in obesity progression and its comorbidities. In conclusion, this study provides new insights into the molecular mechanisms underlying the onset of obesity and highlights hEVs as promising targets for early detection and intervention.
Collapse
Affiliation(s)
- María Fernández-Rhodes
- Biomedical
Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA)+Pec
Proteomics Research Group (+PPRG)Neuroscience Area, University Hospital Arnau de Vilanova (HUAV), 80 Avenue Rovira Roure, 25198Lleida, Catalonia, Spain
- Department
of Medical Basic Sciences (UdL)Biomedical Research Institute
of Lleida Dr. Pifarré Foundation (IRBLLEIDA), +Pec Proteomics
Research Group (+PPRG), University of Lleida, 25198Lleida, Catalonia, Spain
| | - Manuel Suárez
- Departament
de Bioquímica i Biotecnologia, Nutrigenomics Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Institut
d’Investigació Sanitària Pere Virgili, Nutrigenomics
Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Center
of Environmental, Food and Toxicological Technology (TecnATox), Nutrigenomics
Research Group, University Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
| | - Itziar Molet
- Biomedical
Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA)+Pec
Proteomics Research Group (+PPRG)Neuroscience Area, University Hospital Arnau de Vilanova (HUAV), 80 Avenue Rovira Roure, 25198Lleida, Catalonia, Spain
- Department
of Medical Basic Sciences (UdL)Biomedical Research Institute
of Lleida Dr. Pifarré Foundation (IRBLLEIDA), +Pec Proteomics
Research Group (+PPRG), University of Lleida, 25198Lleida, Catalonia, Spain
| | - Susana Suárez-García
- Departament
de Bioquímica i Biotecnologia, Nutrigenomics Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Institut
d’Investigació Sanitària Pere Virgili, Nutrigenomics
Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Center
of Environmental, Food and Toxicological Technology (TecnATox), Nutrigenomics
Research Group, University Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
| | - Lluís Arola
- Departament
de Bioquímica i Biotecnologia, Nutrigenomics Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Institut
d’Investigació Sanitària Pere Virgili, Nutrigenomics
Research Group, Universitat Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
- Center
of Environmental, Food and Toxicological Technology (TecnATox), Nutrigenomics
Research Group, University Rovira i Virgili, C/Marcel·lí Domingo
s/n, 43007Tarragona, Spain
| | - Siu Kwan Sze
- Department
of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, OntarioL2S 3A1, Canada
| | - Xavier Gallart-Palau
- Biomedical
Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA)+Pec
Proteomics Research Group (+PPRG)Neuroscience Area, University Hospital Arnau de Vilanova (HUAV), 80 Avenue Rovira Roure, 25198Lleida, Catalonia, Spain
- Department
of Medical Basic Sciences (UdL)Biomedical Research Institute
of Lleida Dr. Pifarré Foundation (IRBLLEIDA), +Pec Proteomics
Research Group (+PPRG), University of Lleida, 25198Lleida, Catalonia, Spain
| | - Aida Serra
- Biomedical
Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA)+Pec
Proteomics Research Group (+PPRG)Neuroscience Area, University Hospital Arnau de Vilanova (HUAV), 80 Avenue Rovira Roure, 25198Lleida, Catalonia, Spain
- Department
of Medical Basic Sciences (UdL)Biomedical Research Institute
of Lleida Dr. Pifarré Foundation (IRBLLEIDA), +Pec Proteomics
Research Group (+PPRG), University of Lleida, 25198Lleida, Catalonia, Spain
| |
Collapse
|
2
|
Bai YF, Shi XH, Zhang ML, Gu JH, Bai TL, Bai YB. Advances in the study of CCT3 in malignant tumors: A review. Medicine (Baltimore) 2025; 104:e41069. [PMID: 39928781 PMCID: PMC11813047 DOI: 10.1097/md.0000000000041069] [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] [Received: 04/16/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 02/12/2025] Open
Abstract
Malignant tumors are among the leading causes of death worldwide, with their underlying mechanisms remaining largely unclear. Tumorigenesis is a complex process involving multiple factors, genes, and pathways. Tumor cells are characterized by abnormal proliferation, infiltration, invasion, and metastasis. Improving tumor diagnosis rates and identifying novel molecular therapeutic targets are of great significance for the advancement of modern medicine. Chaperonin containing TCP-1 subunit 3 (CCT3) is one of the subunits of the chaperonin containing TCP-1 complex, a molecular chaperone involved in protein folding and remodeling. CCT3 plays a crucial role in maintaining protein homeostasis, with key substrates including tubulin and actin. In recent years, CCT3 has been reported to be abnormally expressed in various cancers, correlating with prognosis and therapeutic outcomes. In this review, we summarize the basic structure and function of chaperonin containing TCP-1 complex and CCT3, and discuss the role of CCT3 in tumor development. Additionally, we explore its potential applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yun-Feng Bai
- Baotou Medical College, Baotou, Inner Mongolia, China
| | - Xiao-Hui Shi
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | | | - Jia-hui Gu
- Baotou Medical College, Baotou, Inner Mongolia, China
| | - Ta-La Bai
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yin-Bao Bai
- Department of Thyroid Tumor Surgery, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Du Y, Wu S, Xi S, Xu W, Sun L, Yan J, Gao H, Wang Y, Zheng J, Wang F, Yang H, Xie D, Chen X, Ou X, Guan X, Li Y. ASH1L in Hepatoma Cells and Hepatic Stellate Cells Promotes Fibrosis-Associated Hepatocellular Carcinoma by Modulating Tumor-Associated Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404756. [PMID: 39377228 PMCID: PMC11615825 DOI: 10.1002/advs.202404756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/06/2024] [Indexed: 10/09/2024]
Abstract
Hepatocellular carcinoma (HCC) often occurs in the context of fibrosis or cirrhosis. Methylation of histone is an important epigenetic mechanism, but it is unclear whether histone methyltransferases are potent targets for fibrosis-associated HCC therapy. ASH1L, an H3K4 methyltransferase, is found at higher levels in activated hepatic stellate cells (HSCs) and hepatoma cells. To determine the role of ASH1L in vivo, transgenic mice with conditional Ash1l depletion in the hepatocyte cell lineage (Ash1lflox/floxAlbcre) or HSCs (Ash1lflox/floxGFAPcreERT2) are generated, and these mice are challenged in a diethylnitrosamine (DEN)/carbon tetrachloride (CCl4)-induced model of liver fibrosis and HCC. Depleting Ash1l in both hepatocytes and HSCs mitigates hepatic fibrosis and HCC development. Multicolor flow cytometry, bulk, and single-cell transcriptomic sequencing reveal that ASH1L creates an immunosuppressive microenvironment. Mechanically, ASH1L-mediated H3K4me3 modification increases the expression of CCL2 and CSF1, which recruites and polarizes M2-like pro-tumorigenic macrophages. The M2-like macrophages further enhance tumor cell proliferation and suppress CD8+ T cell activation. AS-99, a small molecule inhibitor of ASH1L, demonstrates similar anti-fibrosis and tumor-suppressive effects. Of pathophysiological significance, the increased expression levels of mesenchymal ASH1L and M2 marker CD68 are associated with poor prognosis of HCC. The findings reveal ASH1L as a potential small-molecule therapeutic target against fibrosis-related HCC.
Collapse
Affiliation(s)
- Yuyang Du
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Shasha Wu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Shaoyan Xi
- Department of PathologySun Yat‐Sen University Cancer CenterGuangzhou510275China
| | - Wei Xu
- GMU‐GIBH Joint School of Life Sciences, The Guangdong‐Hong Kong‐Macau Joint Laboratory for Cell Fate Regulation and DiseasesGuangzhou Medical UniversityGuangzhou511436China
| | - Liangzhan Sun
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
- Department of Clinical OncologyThe University of Hong KongHong Kong999077China
- Institute of Cancer ResearchShenzhen Bay LaboratoryShenzhen518067China
| | - Jingsong Yan
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Han Gao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Yanchen Wang
- Shenzhen HospitalSouthern Medical UniversityShenzhen518000China
| | - Jingyi Zheng
- Shenzhen HospitalSouthern Medical UniversityShenzhen518000China
| | - Fenfen Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Hui Yang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Dan Xie
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510080China
| | - Xi Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and TechnologyShenzhen518055China
| | - Xijun Ou
- School of Life SciencesSouthern University of Science and TechnologyShenzhen518055China
| | - Xin‐Yuan Guan
- Department of Clinical OncologyThe University of Hong KongHong Kong999077China
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510080China
- The University of Hong Kong‐Shenzhen HospitalShenzhen518053China
| | - Yan Li
- Shenzhen HospitalSouthern Medical UniversityShenzhen518000China
| |
Collapse
|
4
|
Kodera K, Hishida R, Sakai A, Nyuzuki H, Matsui N, Yamanaka T, Saitoh A, Matsui H. GPATCH4 contributes to nucleolus morphology and its dysfunction impairs cell viability. Biochem Biophys Res Commun 2024; 693:149384. [PMID: 38113722 DOI: 10.1016/j.bbrc.2023.149384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
The nucleolus serves a multifaceted role encompassing not only rRNA transcription and ribosome synthesis, but also the intricate orchestration of cell cycle regulation and the modulation of cellular senescence. G-patch domain containing 4 (GPATCH4) stands as one among the nucleolar proteins; however, its functional significances remain still unclear. In order to elucidate the functions of GPATCH4, we examined the effects of its dysfunction on cellular proliferation, alterations in nucleolar architecture, apoptotic events, and cellular senescence. Through experimentation conducted on cultured neuroblastoma SH-SY5Y cells, the reduction of GPATCH4 caused inhibition of cellular proliferation, concurrently fostering escalated apoptotic susceptibilities upon exposure to high-dose etoposide. In the realm of nucleolar morphology comparisons, a discernible decline was noted in the count of nucleoli per nucleus, concomitant with a significant expansion in the area occupied by individual nucleoli. Upon induction of senescence prompted by low-dose etoposide, GPATCH4 knockdown resulted in decreased cell viability and increased expression of senescence-associated markers, namely senescence-associated β-galactosidase (SA-β-GAL) and p16. Furthermore, GPATCH4 dysfunction elicited alterations in the gene expression profile of the ribosomal system. In sum, our findings showed that GPATCH4 is a pivotal nucleolar protein that regulates nucleolar morphology and is correlated with cell viability.
Collapse
Affiliation(s)
- Kazuki Kodera
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan; Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Ryuichi Hishida
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Akiko Sakai
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Hiromi Nyuzuki
- Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Noriko Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Tomoyuki Yamanaka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Akihiko Saitoh
- Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan.
| |
Collapse
|
5
|
Kothapalli KSD, Park HG, Kothapalli NSL, Brenna JT. FADS2 function at the major cancer hotspot 11q13 locus alters fatty acid metabolism in cancer. Prog Lipid Res 2023; 92:101242. [PMID: 37597812 DOI: 10.1016/j.plipres.2023.101242] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Dysregulation of fatty acid metabolism and de novo lipogenesis is a key driver of several cancer types through highly unsaturated fatty acid (HUFA) signaling precursors such as arachidonic acid. The human chromosome 11q13 locus has long been established as the most frequently amplified in a variety of human cancers. The fatty acid desaturase genes (FADS1, FADS2 and FADS3) responsible for HUFA biosynthesis localize to the 11q12-13.1 region. FADS2 activity is promiscuous, catalyzing biosynthesis of several unsaturated fatty acids by Δ6, Δ8, and Δ4 desaturation. Our main aim here is to review known and putative consequences of FADS2 dysregulation due to effects on the 11q13 locus potentially driving various cancer types. FADS2 silencing causes synthesis of sciadonic acid (5Z,11Z,14Z-20:3) in MCF7 cells and breast cancer in vivo. 5Z,11Z,14Z-20:3 is structurally identical to arachidonic acid (5Z,8Z,11Z,14Z-20:4) except it lacks the internal Δ8 double bond required for prostaglandin and leukotriene synthesis, among other eicosanoids. Palmitic acid has substrate specificity for both SCD and FADS2. Melanoma, prostate, liver and lung cancer cells insensitive to SCD inhibition show increased FADS2 activity and sapienic acid biosynthesis. Elevated serum mead acid levels found in hepatocellular carcinoma patients suggest an unsatisfied demand for arachidonic acid. FADS2 circular RNAs are at high levels in colorectal and lung cancer tissues. FADS2 circular RNAs are associated with shorter overall survival in colorectal cancer patients. The evidence thusfar supports an effort for future research on the role of FADS2 as a tumor suppressor in a range of neoplastic disorders.
Collapse
Affiliation(s)
- Kumar S D Kothapalli
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA.
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA
| | | | - J Thomas Brenna
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA.
| |
Collapse
|
6
|
Batista MDP, Roffé M, Romero I, López-Guerrero JA, Illueca C, Lopez R, Balieiro Anastácio da Costa AA, De Brot L, Molina JP, Barboza L, Peria FM, Chaud F, Gouvêa Yamada AS, Poveda A, Rego EM. Genomic landscapes of ovarian clear cell carcinoma from latin countries reveal aberrations linked to survival and progression. BMC Cancer 2023; 23:613. [PMID: 37400764 DOI: 10.1186/s12885-023-11095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 06/21/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Ovarian clear cell carcinomas (OCCCs) are rare, aggressive and chemoresistant tumors. Geographical and ethnic differences in the incidence of OCCC have been reported with a higher incidence in Asiatic countries. There is a paucity of information regarding OCCC in Latin America (LA) and other countries. METHODS Here, we characterized two cohorts of 33 patients with OCCC from LA (24 from Brazil and 9 from Costa Rica) and a cohort of 27 patients from Spain. Genomic analysis was performed for 26 OCCC using the OncoScan platform. Tumors were classified according to their genomic landscapes into subgroups. Clinical parameters were related to the frequency of genomic aberrations. RESULTS The median overall survival (OS) was not significantly different between the cohorts. Genomic landscapes were characterized by different homologous recombination deficiency (HRD) levels. No difference in the distribution of genomic landscapes profiles was detected between patients from the different cohorts. OCCCs with MYC-amplified tumors harboring a concomitant loss of a region in chromosome 13q12-q13 that includes the BRCA2 gene had the longest OS. In contrast, patients carrying a high number (> 30) of total copy number (CN) aberrations with no concomitant alterations in MYC and BRCA2 genes presented the shortest OS. Furthermore, amplification of the ASH1L gene was also associated with a shorter OS. Initial-stage OCCCs with early progression were characterized by gains in the JNK1 and MKL1 genes. CONCLUSIONS Our results provide new data from understudied OCCC populations and reveal new potential markers for OCCCs.
Collapse
Affiliation(s)
- Mariana de Paiva Batista
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil.
| | - Martín Roffé
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Ignacio Romero
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | | | - Carmen Illueca
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | - Raquel Lopez
- Gynecological Oncology Area, Valencian Institute of Oncology Foundation, Valencia, Spain
| | | | - Louise De Brot
- Department of Pathology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Juan Pablo Molina
- Medical Oncology Service, México Hospital, CCSS, San José, Costa Rica
| | - Laura Barboza
- Pathological Anatomy Service, San Juan de Dios Hospital, San José, Costa Rica
| | - Fernanda Maris Peria
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Fernando Chaud
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Ana Silvia Gouvêa Yamada
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| | - Andres Poveda
- Oncogynecologic Department, Quironsalud Hospital, Valencia, Spain
| | - Eduardo Magalhães Rego
- Department of Internal Medicine, Medical School of Ribeirão Preto and Center for Cell Based Therapy, University of São Paulo, 14051-140, Tenente Catao Roxo, 2501, Ribeirão Preto, Brazil
| |
Collapse
|
7
|
Song F, Dai Q, Grimm MO, Steinbach D. The Antithetic Roles of IQGAP2 and IQGAP3 in Cancers. Cancers (Basel) 2023; 15:cancers15041115. [PMID: 36831467 PMCID: PMC9953781 DOI: 10.3390/cancers15041115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
The scaffold protein family of IQ motif-containing GTPase-activating proteins (IQGAP1, 2, and 3) share a high degree of homology and comprise six functional domains. IQGAPs bind and regulate the cytoskeleton, interact with MAP kinases and calmodulin, and have GTPase-related activity, as well as a RasGAP domain. Thus, IQGAPs regulate multiple cellular processes and pathways, affecting cell division, growth, cell-cell interactions, migration, and invasion. In the past decade, significant evidence on the function of IQGAPs in signal transduction during carcinogenesis has emerged. Compared with IQGAP1, IQGAP2 and IQGAP3 were less analyzed. In this review, we summarize the different signaling pathways affected by IQGAP2 and IQGAP3, and the antithetic roles of IQGAP2 and IQGAP3 in different types of cancer. IQGAP2 expression is reduced and plays a tumor suppressor role in most solid cancer types, while IQGAP3 is overexpressed and acts as an oncogene. In lymphoma, for example, IQGAPs have partially opposite functions. There is considerable evidence that IQGAPs regulate a multitude of pathways to modulate cancer processes and chemoresistance, but some questions, such as how they trigger this signaling, through which domains, and why they play opposite roles on the same pathways, are still unanswered.
Collapse
Affiliation(s)
- Fei Song
- Department of Urology, Jena University Hospital, 07740 Jena, Germany
| | - Qingqing Dai
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07740 Jena, Germany
| | - Marc-Oliver Grimm
- Department of Urology, Jena University Hospital, 07740 Jena, Germany
| | - Daniel Steinbach
- Department of Urology, Jena University Hospital, 07740 Jena, Germany
- Correspondence:
| |
Collapse
|
8
|
Wu L, Huang J, Trivedi P, Sun X, Yu H, He Z, Zhang X. Zinc finger myeloid Nervy DEAF-1 type (ZMYND) domain containing proteins exert molecular interactions to implicate in carcinogenesis. Discov Oncol 2022; 13:139. [PMID: 36520265 PMCID: PMC9755447 DOI: 10.1007/s12672-022-00597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Morphogenesis and organogenesis in the low organisms have been found to be modulated by a number of proteins, and one of such factor, deformed epidermal auto-regulatory factor-1 (DEAF-1) has been initially identified in Drosophila. The mammalian homologue of DEAF-1 and structurally related proteins have been identified, and they formed a family with over 20 members. The factors regulate gene expression through association with co-repressors, recognition of genomic marker, to exert histone modification by catalyze addition of some chemical groups to certain amino acid residues on histone and non-histone proteins, and degradation host proteins, so as to regulate cell cycle progression and execution of cell death. The formation of fused genes during chromosomal translocation, exemplified with myeloid transforming gene on chromosome 8 (MTG8)/eight-to-twenty one translocation (ETO) /ZMYND2, MTG receptor 1 (MTGR1)/ZMYND3, MTG on chromosome 16/MTGR2/ZMYND4 and BS69/ZMYND11 contributes to malignant transformation. Other anomaly like copy number variation (CNV) of BS69/ZMYND11 and promoter hyper methylation of BLU/ZMYND10 has been noted in malignancies. It has been reported that when fusing with Runt-related transcription factor 1 (RUNX1), the binding of MTG8/ZMYND2 with co-repressors is disturbed, and silencing of BLU/ZMYND10 abrogates its ability to inhibition of cell cycle and promotion of apoptotic death. Further characterization of the implication of ZMYND proteins in carcinogenesis would enhance understanding of the mechanisms of occurrence and early diagnosis of tumors, and effective antitumor efficacy.
Collapse
Affiliation(s)
- Longji Wu
- Department of Pathophysiology, School of Basic Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Songshan Lake Scientific and Industrial Park, Dongguan, 523808, Guangdong, People's Republic of China
- Chinese-American Tumor Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
- Institute of Modern Biology, Nanjing University, Nanjing, Jiangsu, China
| | - Jing Huang
- Department of Pathophysiology, School of Basic Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Songshan Lake Scientific and Industrial Park, Dongguan, 523808, Guangdong, People's Republic of China
- Chinese-American Tumor Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Pankaj Trivedi
- Department of Experimental Medicine, La Sapienza University, Rome, Italy
| | - Xuerong Sun
- Institute of Aging, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Hongbing Yu
- Chinese-American Tumor Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China.
| | - Zhiwei He
- Department of Pathophysiology, School of Basic Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Songshan Lake Scientific and Industrial Park, Dongguan, 523808, Guangdong, People's Republic of China
- Chinese-American Tumor Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Xiangning Zhang
- Department of Pathophysiology, School of Basic Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Songshan Lake Scientific and Industrial Park, Dongguan, 523808, Guangdong, People's Republic of China.
- Chinese-American Tumor Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, People's Republic of China.
| |
Collapse
|
9
|
Mohamed EE, Ahmed OM, Abdel-Moneim A, Zoheir KMA, Elesawy BH, Al Askary A, Hassaballa A, El-Shahawy AAG. Protective Effects of Naringin-Dextrin Nanoformula against Chemically Induced Hepatocellular Carcinoma in Wistar Rats: Roles of Oxidative Stress, Inflammation, Cell Apoptosis, and Proliferation. Pharmaceuticals (Basel) 2022; 15:1558. [PMID: 36559011 PMCID: PMC9786090 DOI: 10.3390/ph15121558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Nanotechnology holds great promise for the development of treatments for deadly human diseases, such as hepatocellular carcinoma (HCC). In the current study, we compared the hepatoprotective effects of naringin-dextrin nanoparticles (NDNPs) against HCC in male Wistar rats with those of pure naringin and investigated the underlying cellular and molecular mechanisms. HCC was induced by intraperitoneal injection of diethylnitrosamine (DEN, 150 mg/kg body weight (b.w.) per week) for two weeks, followed by oral administration of 2-acetylaminofluorene (2AAF, 20 mg/kg b.w.) four times per week for three weeks. DEN/2AAF-administered rats were divided into three groups that respectively received 1% carboxymethyl cellulose (as vehicle), 10 mg/kg b.w. naringin, or 10 mg/kg b.w. NDNP every other day by oral gavage for 24 weeks. Both naringin and NDNP significantly attenuated the harmful effects of DEN on liver function. Both compounds also suppressed tumorigenesis as indicated by the reduced serum concentrations of liver tumor markers, and this antitumor effect was confirmed by histopathological evaluation. Additionally, naringin and NDNP prevented DEN-induced changes in hepatic oxidative stress and antioxidant activities. In addition, naringin and NDNP suppressed inflammation induced by DEN. Moreover, naringin and NDNP significantly reduced the hepatic expression of Bcl-2 and increased Bax, p53, and PDCD5 expressions. Naringin and NDNP also reduced expression of IQGAP1, IQGAP3, Ras signaling, and Ki-67 while increasing expression of IQGAP2. Notably, NDNP more effectively mitigated oxidative stress and inflammatory signaling than free naringin and demonstrated improved antitumor efficacy, suggesting that this nanoformulation improves bioavailability within nascent tumor sites.
Collapse
Affiliation(s)
- Eman E. Mohamed
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Osama M. Ahmed
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 2722165, Egypt
| | - Khairy M. A. Zoheir
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Basem H. Elesawy
- Department of Pathology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmad Al Askary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed Hassaballa
- Nutrition and Food Science, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI 48202, USA
- ZeroHarm L.C., Farmington Hills, Farmington, MI 48333, USA
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 2722165, Egypt
| |
Collapse
|
10
|
Hymel E, Fisher KW, Farazi PA. Differential methylation patterns in lean and obese non-alcoholic steatohepatitis-associated hepatocellular carcinoma. BMC Cancer 2022; 22:1276. [PMID: 36474183 PMCID: PMC9727966 DOI: 10.1186/s12885-022-10389-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease affects about 24% of the world's population and may progress to nonalcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). While more common in those that are obese, NASH-HCC can develop in lean individuals. The mechanisms by which HCC develops and the role of epigenetic changes in the context of obesity and normal weight are not well understood. METHODS In this study, we used previously generated mouse models of lean and obese HCC using a choline deficient/high trans-fat/fructose/cholesterol diet and a choline supplemented/high trans-fat/fructose/cholesterol diet, respectively, to evaluate methylation differences in HCC progression in lean versus obese mice. Differentially methylated regions were determined using reduced representation bisulfite sequencing. RESULTS A larger number of differentially methylated regions (DMRs) were seen in NASH-HCC progression in the obese mice compared to the non-obese mice. No overlap existed in the DMRs with the largest methylation differences between the two models. In lean NASH-HCC, methylation differences were seen in genes involved with cancer progression and prognosis (including HCC), such as CHCHD2, FSCN1, and ZDHHC12, and lipid metabolism, including PNPLA6 and LDLRAP1. In obese NASH- HCC, methylation differences were seen in genes known to be associated with HCC, including RNF217, GJA8, PTPRE, PSAPL1, and LRRC8D. Genes involved in Wnt-signaling pathways were enriched in hypomethylated DMRs in the obese NASH-HCC. CONCLUSIONS These data suggest that differential methylation may play a role in hepatocarcinogenesis in lean versus obese NASH. Hypomethylation of Wnt signaling pathway-related genes in obese mice may drive progression of HCC, while progression of HCC in lean mice may be driven through other signaling pathways, including lipid metabolism.
Collapse
Affiliation(s)
- Emma Hymel
- grid.266813.80000 0001 0666 4105Department of Epidemiology, University of Nebraska Medical Center, 984395 Nebraska Medical Center, Omaha, NE 68198-4395 USA
| | - Kurt W. Fisher
- grid.266813.80000 0001 0666 4105Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Paraskevi A. Farazi
- grid.266813.80000 0001 0666 4105Department of Epidemiology, University of Nebraska Medical Center, 984395 Nebraska Medical Center, Omaha, NE 68198-4395 USA
| |
Collapse
|
11
|
Mohamed EE, Abdel-Moneim A, Ahmed OM, Zoheir KM, Eldin ZE, El-Shahawy AA. Anticancer activity of a novel naringin‒dextrin nanoformula: Preparation, characterization, and in vitro induction of apoptosis in human hepatocellular carcinoma cells by inducing ROS generation, DNA fragmentation, and cell cycle arrest. J Drug Deliv Sci Technol 2022; 75:103677. [DOI: 10.1016/j.jddst.2022.103677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Oleocanthal Attenuates Metastatic Castration-Resistant Prostate Cancer Progression and Recurrence by Targeting SMYD2. Cancers (Basel) 2022; 14:cancers14143542. [PMID: 35884603 PMCID: PMC9317016 DOI: 10.3390/cancers14143542] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The Mediterranean, extra-virgin-olive-oil-rich diet ingredient S-(-)-oleocanthal (OC) has emerged as a potential inhibitor for the growth and relapse of the most aggressive prostate cancer type. This effect is mediated through suppression of important enzyme, SMYD2, that drives the activation of several downstream protein effectors. OC treatments reduced SMYD2 downstream substrates, which are critical for prostate cancer growth and relapse. OC is more advantageous than other reported SMYD2 inhibitors because it has shown potent anticancer activity in animal models. OC’s anti-prostate-cancer effect was prominent compared with some standard drugs currently used to control prostate cancer. OC is a potential, novel natural compound appropriate for immediate use by prostate cancer patients and survivors as a nutraceutical or dietary supplement product. Abstract Metastatic castration-resistant prostate cancer (mCRPC) is the most aggressive prostate cancer (PC) phenotype. Cellular lysine methylation is driven by protein lysine methyltransferases (PKMTs), such as those in the SET- and MYND-containing protein (SMYD) family, including SMYD2 methylate, and several histone and non-histone proteins. SMYD2 is dysregulated in metastatic PC patients with high Gleason score and shorter survival. The Mediterranean, extra-virgin-olive-oil-rich diet ingredient S-(-)-oleocanthal (OC) inhibited SMYD2 in biochemical assays and suppressed viability, migration, invasion, and colony formation of PC-3, CWR-R1ca, PC-3M, and DU-145 PC cell lines with IC50 range from high nM to low µM. OC’s in vitro antiproliferative effect was comparable to standard anti-PC chemotherapies or hormone therapies. A daily, oral 10 mg/kg dose of OC for 11 days effectively suppressed the progression of the mCRPC CWR-R1ca cells engrafted into male nude mice. Daily, oral OC treatment for 30 days suppressed tumor locoregional and distant recurrences after the primary tumors’ surgical excision. Collected OC-treated animal tumors showed marked SMYD2 reduction. OC-treated mice showed significant serum PSA reduction. For the first time, this study showed SMYD2 as novel molecular target in mCRPC, and OC emerged as a specific SMYD2 lead inhibitor. OC prevailed over previously reported SMYD2 inhibitors, with validated in vivo potency and high safety profile, and, therefore, is proposed as a novel nutraceutical for mCRPC progression and recurrence control.
Collapse
|
13
|
Dai Q, Ain Q, Rooney M, Song F, Zipprich A. Role of IQ Motif-Containing GTPase-Activating Proteins in Hepatocellular Carcinoma. Front Oncol 2022; 12:920652. [PMID: 35785216 PMCID: PMC9243542 DOI: 10.3389/fonc.2022.920652] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) are a class of scaffolding proteins, including IQGAP1, IQGAP2, and IQGAP3, which govern multiple cellular activities by facilitating cytoskeletal remodeling and cellular signal transduction. The role of IQGAPs in cancer initiation and progression has received increasing attention in recent years, especially in hepatocellular carcinoma (HCC), where the aberrant expression of IQGAPs is closely related to patient prognosis. IQGAP1 and 3 are upregulated and are considered oncogenes in HCC, while IQGAP2 is downregulated and functions as a tumor suppressor. This review details the three IQGAP isoforms and their respective structures. The expression and role of each protein in different liver diseases and mainly in HCC, as well as the underlying mechanisms, are also presented. This review also provides a reference for further studies on IQGAPs in HCC.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
- Else Kröner Graduate School for Medical Students “Jena School for Ageing Medicine (JSAM)”, Jena University Hospital, Jena, Germany
| | - Quratul Ain
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Michael Rooney
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Fei Song
- Department of Urology, Jena University Hospital, Jena, Germany
| | - Alexander Zipprich
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
- *Correspondence: Alexander Zipprich,
| |
Collapse
|
14
|
Ye T, Lin L, Cao L, Huang W, Wei S, Shan Y, Zhang Z. Novel Prognostic Signatures of Hepatocellular Carcinoma Based on Metabolic Pathway Phenotypes. Front Oncol 2022; 12:863266. [PMID: 35677150 PMCID: PMC9168273 DOI: 10.3389/fonc.2022.863266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Hepatocellular carcinoma is a disastrous cancer with an aberrant metabolism. In this study, we aimed to assess the role of metabolism in the prognosis of hepatocellular carcinoma. Ten metabolism-related pathways were identified to classify the hepatocellular carcinoma into two clusters: Metabolism_H and Metabolism_L. Compared with Metabolism_L, patients in Metabolism_H had lower survival rates with more mutated TP53 genes and more immune infiltration. Moreover, risk scores for predicting overall survival based on eleven differentially expressed metabolic genes were developed by the least absolute shrinkage and selection operator (LASSO)-Cox regression model in The Cancer Genome Atlas (TCGA) dataset, which was validated in the International Cancer Genome Consortium (ICGC) dataset. The immunohistochemistry staining of liver cancer patient specimens also identified that the 11 genes were associated with the prognosis of liver cancer patients. Multivariate Cox regression analyses indicated that the differentially expressed metabolic gene-based risk score was also an independent prognostic factor for overall survival. Furthermore, the risk score (AUC = 0.767) outperformed other clinical variables in predicting overall survival. Therefore, the metabolism-related survival-predictor model may predict overall survival excellently for HCC patients.
Collapse
Affiliation(s)
- Tingbo Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Leilei Lin
- Department of Ultrasound, Wenzhou People's Hospital, Wenzhou, China
| | - Lulu Cao
- Department of Pathology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Weiguo Huang
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shengzhe Wei
- Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunfeng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongjing Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Suzuki H, Iwamoto H, Yamamoto K, Tsukaguchi M, Nakamura T, Masuda A, Sakaue T, Tanaka T, Niizeki T, Okamura S, Shimose S, Shirono T, Noda Y, Kamachi N, Kuromatsu R, Hisaka T, Yano H, Koga H, Torimura T. DNA Methylation in Noncancerous Liver Tissues as Biomarker for Multicentric Occurrence of Hepatitis C Virus-Related Hepatocellular Carcinoma. GASTRO HEP ADVANCES 2022; 1:555-562. [PMID: 39132059 PMCID: PMC11307517 DOI: 10.1016/j.gastha.2022.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/16/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Hepatitis C virus (HCV)-related hepatocellular carcinoma (HCC) progresses with a highly multicentric occurrence (MO) even after radical hepatectomy. Despite several efforts to clarify the pathogenesis of MO, the underlying molecular mechanism remains elusive. The aim of this study was to evaluate alterations in DNA methylation in noncancerous liver tissues in the MO of HCC. Methods A total of 203 patients with HCV-related HCC who underwent radical hepatectomy at our hospital between January 2008 and January 2012 were recruited. We defined a group of nonearly recurrence of HCC (NR) for ≥3 years after radical hepatectomy and a group of early recurrence of HCC (ER) with MO within 2 years after radical hepatectomy. Results Three patients each were selected in the NR and ER groups in the first set, and 13 patients in the NR group and 17 patients in the ER group were selected in the second set. Genome-wide DNA methylation profiles were obtained from noncancerous liver tissues using a Human Methylation 450 BeadChip, and the differences between the groups were analyzed for each set. After excluding single nucleotide polymorphism-associated methylation sites and low-call sites, 401,282 sites were assessed using a generalized linear model without any adjustments. Nine gene regions, APBB1P, CLSTN3, DLG5, IRX5, OAS1, SOX12, SNX19, TENM2, and TRIM54, exhibiting a significant difference (P < .001) in DNA methylation levels were identified in the common direction between the 2 analysis sets. Conclusion Alterations in DNA methylation of 9 genes in noncancerous liver tissues appear to be involved in MO after radical hepatectomy for HCV-related HCC.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Mai Tsukaguchi
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toshimitsu Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Niizeki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shusuke Okamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomotake Shirono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yu Noda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Naoki Kamachi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Ryoko Kuromatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toru Hisaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
16
|
Feoli A, Viviano M, Cipriano A, Milite C, Castellano S, Sbardella G. Lysine methyltransferase inhibitors: where we are now. RSC Chem Biol 2022; 3:359-406. [PMID: 35441141 PMCID: PMC8985178 DOI: 10.1039/d1cb00196e] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Protein lysine methyltransferases constitute a large family of epigenetic writers that catalyse the transfer of a methyl group from the cofactor S-adenosyl-l-methionine to histone- and non-histone-specific substrates. Alterations in the expression and activity of these proteins have been linked to the genesis and progress of several diseases, including cancer, neurological disorders, and growing defects, hence they represent interesting targets for new therapeutic approaches. Over the past two decades, the identification of modulators of lysine methyltransferases has increased tremendously, clarifying the role of these proteins in different physio-pathological states. The aim of this review is to furnish an updated outlook about the protein lysine methyltransferases disclosed modulators, reporting their potency, their mechanism of action and their eventual use in clinical and preclinical studies.
Collapse
Affiliation(s)
- Alessandra Feoli
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| | - Monica Viviano
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| | - Alessandra Cipriano
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| | - Ciro Milite
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| | - Sabrina Castellano
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| | - Gianluca Sbardella
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno via Giovanni Paolo II 132 I-84084 Fisciano SA Italy +39-089-96-9602 +39-089-96-9770
| |
Collapse
|
17
|
Li X, Cheng L, Li C, Hu X, Hu X, Tan L, Li Q, Liu C, Wang J. Associating Preoperative MRI Features and Gene Expression Signatures of Early-stage Hepatocellular Carcinoma Patients using Machine Learning. J Clin Transl Hepatol 2022; 10:63-71. [PMID: 35233374 PMCID: PMC8845145 DOI: 10.14218/jcth.2021.00023] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS The relationship between quantitative magnetic resonance imaging (MRI) imaging features and gene-expression signatures associated with the recurrence of hepatocellular carcinoma (HCC) is not well studied. METHODS In this study, we generated multivariable regression models to explore the correlation between the preoperative MRI features and Golgi membrane protein 1 (GOLM1), SET domain containing 7 (SETD7), and Rho family GTPase 1 (RND1) gene expression levels in a cohort study including 92 early-stage HCC patients. A total of 307 imaging features of tumor texture and shape were computed from T2-weighted MRI. The key MRI features were identified by performing a multi-step feature selection procedure including the correlation analysis and the application of RELIEFF algorithm. Afterward, regression models were generated using kernel-based support vector machines with 5-fold cross-validation. RESULTS The features computed from higher specificity MRI better described GOLM1 and RND1 gene-expression levels, while imaging features computed from lower specificity MRI data were more descriptive for the SETD7 gene. The GOLM1 regression model generated with three features demonstrated a moderate positive correlation (p<0.001), and the RND1 model developed with five variables was positively associated (p<0.001) with gene expression levels. Moreover, RND1 regression model integrating four features was moderately correlated with expressed RND1 levels (p<0.001). CONCLUSIONS The results demonstrated that MRI radiomics features could help quantify GOLM1, SETD7, and RND1 expression levels noninvasively and predict the recurrence risk for early-stage HCC patients.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Lin Cheng
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Chuanming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xianling Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xiaofei Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
- Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau, China
| | - Qing Li
- MR Collaborations, Siemens Healthcare Ltd., Shanghai, China
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| |
Collapse
|
18
|
Htun MW, Shibata Y, Soe K, Koji T. Nuclear Expression of Pygo2 Correlates with Poorly Differentiated State Involving c-Myc, PCNA and Bcl9 in Myanmar Hepatocellular Carcinoma. Acta Histochem Cytochem 2021; 54:195-206. [PMID: 35023882 PMCID: PMC8727843 DOI: 10.1267/ahc.21-00090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/03/2021] [Indexed: 01/10/2023] Open
Abstract
In Myanmar, hepatocellular carcinoma (HCC) is commonly seen in young adult and associated with poor prognosis, while the molecular mechanisms that characterize HCC in Myanmar are unknown. As co-activation of Wnt/β-catenin signaling and c-Myc (Myc) are reported to associate with malignancy of HCC, we immunohistochemically investigated the expression of Pygo2 and Bcl9, the co-activators of the Wnt/β-catenin signaling, Myc and PCNA in 60 cases of Myanmar HCC. Pygo2 expression was confirmed by in situ hybridization. The signal intensity was measured by image analyzer and then statistically analyzed. As a result, the expression of Pygo2 was significantly higher in HCC compared to normal liver tissue and the nuclear signal was the most intense in poorly differentiated HCC. Cytoplasmic Bcl9 was expressed in the normal liver tissue but decreased in HCC with the progression of histopathological grade. Myc was significantly higher in poorly differentiated HCC, whereas PCNA labeling index increased with the progression of histopathological grade. Nuclear Pygo2 showed strong correlation with nuclear Myc (P < 0.01) and PCNA (P < 0.001), and inversely correlated with cytoplasmic Bcl9 (P < 0.01). Our results suggested Wnt/β-catenin and Myc signaling is commonly activated in Myanmar HCC and that the correlative upregulation of nuclear Pygo2 and Myc characterizes the malignant features of HCC in Myanmar.
Collapse
Affiliation(s)
- Myo Win Htun
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences
| | - Yasuaki Shibata
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences
| | | | - Takehiko Koji
- Office for Research Initiative and Development, Nagasaki University
| |
Collapse
|
19
|
Zhao D, Qian L, Zhuang D, Wang L, Cao Y, Zhou F, Zhang S, Liu Y, Liang Y, Zhang W, Kang W, Zhang M, Wang Y, Zhang F, Zhang W, Xiao J, Xu G, Lv Y, Zou X, Zhuge Y, Zhang B. Inhibition of ribosomal RNA processing 15 Homolog (RRP15), which is overexpressed in hepatocellular carcinoma, suppresses tumour growth via induction of senescence and apoptosis. Cancer Lett 2021; 519:315-327. [PMID: 34343634 DOI: 10.1016/j.canlet.2021.07.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023]
Abstract
Recent studies suggest that RRP15 (Ribosomal RNA Processing 15 Homolog) might be a potential target for cancer therapy. However, the role of RRP15 in hepatocarcinogenesis remains poorly delineated. In this study, we aimed to evaluate the expression and biological function of RRP15 in human hepatocellular carcinoma (HCC). We show that RRP15 was up regulated in HCC cell lines and tumours. Up-regulation of RRP15 in HCC tumours was also correlated with unfavorable prognosis. We further show that the frequent up-regulation of RRP15 in HCCs is at least partly driven by recurrent gene copy gain at chromosome 1q41. Functional studies indicated that RRP15 knockdown suppresses HCC proliferation and growth both in vitro and in vivo. Mechanistically, RRP15 depletion in p53-wild-type HepG2 cells induced senescence via activation of the p53-p21 signalling pathway through enhanced interaction of RPL11 with MDM2, as well as inhibition of SIRT1-mediated p53 deacetylation. Moreover, RRP15 depletion in p53-mutant PLC5 and p53-deleted Hep3B cells induced metabolic shift from the glycolytic pentose-phosphate to mitochondrial oxidative phosphorylation via regulating a series of key genes such as HK2 and TIGAR, and thus, promoted the generation of ROS and apoptosis. Taken together, our findings provide evidence for an important role of the RRP15 gene in hepatocarcinogenesis through regulation of HCC proliferation and growth, raising the possibility that targeting RRP15 may represent a potential therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Dian Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Liping Qian
- Centre for Experimental Animal, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Duanming Zhuang
- Department of Gastroenterology, Gaochun People's Hospital, Nanjing, Jiangsu, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yu Cao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University/Naval Medical University, Shanghai, China
| | - Fan Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shu Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ying Liu
- Department of Ultrasound Diagnostics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ying Liang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenjie Zhang
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yi Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Feng Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jiangqiang Xiao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ying Lv
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Yuzheng Zhuge
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Bin Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China; Department of Gastroenterology, Gaochun People's Hospital, Nanjing, Jiangsu, China; Department of Gastroenterology, Yining People's Hospital, Yining, China.
| |
Collapse
|
20
|
Temiz E, Koyuncu İ, Sahin E. CCT3 suppression prompts apoptotic machinery through oxidative stress and energy deprivation in breast and prostate cancers. Free Radic Biol Med 2021; 165:88-99. [PMID: 33508424 DOI: 10.1016/j.freeradbiomed.2021.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
Mediated by chaperon proteins, protein misfolding plays a crucial role in cancer pathogenesis. Chaperonin Containing TCP1 Subunit 3 (CCT3) is one of eight subunits forming eukaryotic chaperons that catalyzes correct folding of the proteins employed in cell division, proliferation, and apoptosis pathway. Moreover, CCT3 expression increases responsively with carcinogenesis. However, how CCT3 drives the cancerous process has not been documented. Here we probed the mechanistic and functional interactions between CCT3 and apoptotic pathways and cell stressors. First, we profiled CCT3 expression levels of different 16 cell lines and found that CCT3 expression levels of CRL-2329 and PC3 were significantly increased. Then, we suppressed CCT3 levels in CRL-2329 and PC3 lines by miR-24-3p, miR-128-3p, and miR-149-5p mimics, and measured apoptotic response of the cell lines to the knockdown of CCT3 by acridine orange/ethidium bromide and Annexin V/PI staining, cell-cycle and mitochondria membrane potential (MMP) analyses, intracellular reactive oxygen species (ROS) measurement and analysis of expression levels of the apoptotic genes. After having suppressed CCT3, the cell cycle was arrested in the G0/G1 phase, MMP was impaired, and the intracellular ROS level was increased. These signs of apoptotic flux were corroborated by morphological images, statistically enhanced expression levels of the apoptotic pathway modulators and intracellular free amino acids profile. The free amino acid profile, which is heavily implicated in energy metabolism and cell division, is fluctuated in the progress of canceration. Strikingly, suppressed CCT3 shifted intracellular levels of glutamine, beta-alanine, glycine, serin, asparagine and sarcosine, which are employed in energy metabolism. Consequently, miRNA-mediated CCT3 suppression spur apoptosis by unbalancing the homeostasis in intracellular ROS and the profile of free amino acids in energy metabolism. Taken together, we anticipate that miRNA-mediated CCT3 suppression might provide a "dual therapeutic strategy" through conventional cellular toxicity as well as energy withdrawal.
Collapse
Affiliation(s)
- Ebru Temiz
- Program of Medical Promotion and Marketing, Health Services Vocational School, Harran University, Sanliurfa, Turkey; Department of Medical Biology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - İsmail Koyuncu
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Emel Sahin
- Department of Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey.
| |
Collapse
|
21
|
Na K, Kim M, Kim CY, Lim JS, Cho JY, Shin H, Lee HJ, Kang BJ, Han DH, Kim H, Baik JH, Swiatek-de Lange M, Karl J, Paik YK. Potential Regulatory Role of Human-Carboxylesterase-1 Glycosylation in Liver Cancer Cell Growth. J Proteome Res 2020; 19:4867-4883. [PMID: 33206527 DOI: 10.1021/acs.jproteome.0c00787] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We previously reported that human carboxylesterase 1 (CES1), a serine esterase containing a unique N-linked glycosyl group at Asn79 (N79 CES1), is a candidate serological marker of hepatocellular carcinoma (HCC). CES1 is normally present at low-to-undetectable levels in normal human plasma, HCC tumors, and major liver cancer cell lines. To investigate the potential mechanism underlying the suppression of CES1 expression in liver cancer cells, we took advantage of the low detectability of this marker in tumors by overexpressing CES1 in multiple HCC cell lines, including stable Hep3B cells. We found that the population of CES1-overexpressing (OE) cells decreased and that their doubling time was longer compared with mock control liver cancer cells. Using interactive transcriptome, proteome, and subsequent Gene Ontology enrichment analysis of CES1-OE cells, we found substantial decreases in the expression levels of genes involved in cell cycle regulation and proliferation. This antiproliferative function of the N79 glycan of CES1 was further supported by quantitative real-time polymerase chain reaction, flow cytometry, and an apoptosis protein array assay. An analysis of the levels of key signaling target proteins via Western blotting suggested that CES1 overexpression exerted an antiproliferative effect via the PKD1/PKCμ signaling pathway. Similar results were also seen in another HCC cell line (PLC/RFP/5) after transient transfection with CES1 but not in similarly treated non-HCC cell lines (e.g., HeLa and Tera-1 cells), suggesting that CES1 likely exerts a liver cell-type-specific suppressive effect. Given that the N-linked glycosyl group at Asn79 (N79 glycan) of CES1 is known to influence CES1 enzyme activity, we hypothesized that the post-translational modification of CES1 at N79 may be linked to its antiproliferative activity. To investigate the regulatory effect of the N79 glycan on cellular growth, we mutated the single N-glycosylation site in CES1 from Asn to Gln (CES1-N79Q) via site-directed mutagenesis. Fluorescence 2-D difference gel electrophoresis protein expression analysis of cell lysates revealed an increase in cell growth and a decrease in doubling time in cells carrying the N79Q mutation. Thus our results suggest that CES1 exerts an antiproliferative effect in liver cancer cells and that the single N-linked glycosylation at Asn79 plays a potential regulatory role. These functions may underlie the undetectability of CES1 in human HCC tumors and liver cancer cell lines. Mass spectrometry data are available via ProteomeXchange under the identifier PXD021573.
Collapse
Affiliation(s)
- Keun Na
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Minjoo Kim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Chae-Yeon Kim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Jong-Sun Lim
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Heon Shin
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| | - Hyo Jin Lee
- Department of Life Sciences, Korea University, 145 Anamro, Seongbuk-ku, Seoul 02841, South Korea
| | - Byeong Jun Kang
- Department of Life Sciences, Korea University, 145 Anamro, Seongbuk-ku, Seoul 02841, South Korea
| | | | | | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, 145 Anamro, Seongbuk-ku, Seoul 02841, South Korea
| | | | - Johann Karl
- Roche Diagnostics, GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemoon-ku, Seoul 03722, South Korea
| |
Collapse
|
22
|
Zhou A, Liu H, Tang B. Comprehensive Evaluation of Endocytosis-Associated Protein SCAMP3 in Hepatocellular Carcinoma. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:415-426. [PMID: 33116758 PMCID: PMC7548866 DOI: 10.2147/pgpm.s270062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
Background Secretory carrier membrane proteins 3 (SCAMP3) is an endocytosis-associated protein involved in regulating endosomal pathways and the trafficking of vital signaling receptors. This study aimed to comprehensively assess the role of SCAMP3 in hepatocellular carcinoma (HCC) by integrated bioinformatics analysis. Methods In this study, bioinformatics databases were used to explore the differential expression status and prognostic value of SCAMP3 gene in HCC, and bioinformatics analyses of survival data and interactors of SCAMP3 were conducted to predict the prognostic value of SCAMP3 in HCC. Results Using the TCGA data, our data shows that SCAMP3 mRNA expression is most significantly different between liver and hepatocellular carcinoma tissues and higher expression of SCAMP3 has unfavorable prognostic significance in HCC. Tumor grade, stage, and gender also showed a significant relevance with SCAMP3 expression. High SCAMP3 expression of males revealed significantly poorer survival and progression compared with low SCAMP3 expression of males. BioGRID statistics explores 79 unique interactions with SCAMP3 and multiple post translational modifications. Further analysis finds that SOCS2 may negatively correlate with SCAMP3, while GBA, MX1, and DDOST positively correlate with SCAMP3. Moreover, ncRNA analysis shows that SCAMP3 gene expression is positively associated with lncRNA SBF2-AS1 and negatively related with Has-miR-145. The expressions of SBF2-AS1 and Has-miR-145 are also significantly related with survival in HCC. Discussion SCAMP3 expression can be affected by multiple genes or ncRNAs expression that are associated with survival, thus suggesting that SCAMP3 can be used as a clinical diagnosis and prognostic biomarker in HCC.
Collapse
Affiliation(s)
- Ao Zhou
- Basic Medical College, Southwest Medical University, Luzhou 646100, People's Republic of China
| | - Hongjing Liu
- The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646100, People's Republic of China
| | - Bin Tang
- Basic Medical College, Southwest Medical University, Luzhou 646100, People's Republic of China.,Key Laboratory of Process Analysis and Control of Sichuan Universities, Yibin University, Yibin 644000, People's Republic of China
| |
Collapse
|
23
|
Yan J, Shu M, Li X, Yu H, Chen S, Xie S. Prognostic Score-based Clinical Factors and Metabolism-related Biomarkers for Predicting the Progression of Hepatocellular Carcinoma. Evol Bioinform Online 2020; 16:1176934320951571. [PMID: 33013158 PMCID: PMC7518001 DOI: 10.1177/1176934320951571] [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: 05/07/2020] [Accepted: 07/24/2020] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor representing more than 90% of primary liver cancer. This study aimed to identify metabolism-related biomarkers with prognostic value by developing the novel prognostic score (PS) model. Transcriptomic profiles derived from TCGA and EBIArray databases were analyzed to identify differentially expressed genes (DEGs) in HCC tumor samples compared with normal samples. The overlapped genes between DEGs and metabolism-related genes (crucial genes) were screened and functionally analyzed. A novel PS model was constructed to identify optimal signature genes. Cox regression analysis was performed to identify independent clinical factors related to prognosis. Nomogram model was constructed to estimate the predictability of clinical factors. Finally, protein expression of crucial genes was explored in different cancer tissues and cell types from the Human Protein Atlas (HPA). We screened a total of 305 overlapped genes (differentially expressed metabolism-related genes). These genes were mainly involved in "oxidation reduction," "steroid hormone biosynthesis," "fatty acid metabolic process," and "linoleic acid metabolism." Furthermore, we screened ten optimal DEGs (CYP2C9, CYP3A4, and TKT, among others) by using the PS model. Two clinical factors of pathologic stage (P < .001, HR: 1.512 [1.219-1.875]) and PS status (P <.001, HR: 2.259 [1.522-3.354]) were independent prognostic predictors by cox regression analysis. Nomogram model showed a high predicted probability of overall survival time, and the AUC value was 0.837. The expression status of 7 proteins was frequently altered in normal or differential tumor tissues, such as liver cancer and stomach cancer samples.We have identified several metabolism-related biomarkers for prognosis prediction of HCC based on the PS model. Two clinical factors were independent prognostic predictors of pathologic stage and PS status (high/low risk). The prognosis prediction model described in this study is a useful and stable method for novel biomarker identification.
Collapse
Affiliation(s)
- Jia Yan
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Ming Shu
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Xiang Li
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Hua Yu
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Shuhuai Chen
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Shujie Xie
- Department of Hepatobiliary Pancreatic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| |
Collapse
|
24
|
Wang T, Zhang KH. New Blood Biomarkers for the Diagnosis of AFP-Negative Hepatocellular Carcinoma. Front Oncol 2020; 10:1316. [PMID: 32923383 PMCID: PMC7456927 DOI: 10.3389/fonc.2020.01316] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022] Open
Abstract
An early diagnosis of hepatocellular carcinoma (HCC) followed by effective treatment is currently critical for improving the prognosis and reducing the associated economic burden. Alpha-fetoprotein (AFP) is the most widely used biomarker for HCC diagnosis. Based on elevated serum AFP levels as well as typical imaging features, AFP-positive HCC (APHC) can be easily diagnosed, but AFP-negative HCC (ANHC) is not easily detected due to lack of ideal biomarkers and thus mainly reliance on imaging. Imaging for the diagnosis of ANHC is probably insufficient in sensitivity and/or specificity because most ANHC tumors are small and early-stage HCC, and it is involved in sophisticated techniques and high costs. Moreover, ANHC accounts for nearly half of HCC and exhibits a better prognosis compared with APHC. Therefore, the diagnosis of ANHC in clinical practice has been a critical issue for the early treatment and prognosis improvement of HCC. In recent years, tremendous efforts have been made to discover new biomarkers complementary to AFP for HCC diagnosis. In this review, we systematically review and discuss the recent advances of blood biomarkers for HCC diagnosis, including DNA biomarkers, RNA biomarkers, protein biomarkers, and conventional laboratory metrics, focusing on their diagnostic evaluation alone and in combination, in particular on their diagnostic performance for ANHC.
Collapse
Affiliation(s)
- Ting Wang
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Xu B, Qin T, Yu J, Giordano TJ, Sartor MA, Koenig RJ. Novel role of ASH1L histone methyltransferase in anaplastic thyroid carcinoma. J Biol Chem 2020; 295:8834-8845. [PMID: 32398261 DOI: 10.1074/jbc.ra120.013530] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most aggressive human malignancies, with an average life expectancy of ∼6 months from the time of diagnosis. The genetic and epigenetic changes that underlie this malignancy are incompletely understood. We found that ASH1-like histone lysine methyltransferase (ASH1L) is overexpressed in ATC relative to the much less aggressive and more common differentiated thyroid cancer. This increased expression was due at least in part to reduced levels of microRNA-200b-3p (miR-200b-3p), which represses ASH1L expression, in ATC. Genetic knockout of ASH1L protein expression in ATC cell lines decreased cell growth both in culture and in mouse xenografts. RNA-Seq analysis of ASH1L knockout versus WT ATC cell lines revealed that ASH1L is involved in the regulation of numerous cancer-related genes and gene sets. The pro-oncogenic long noncoding RNA colon cancer-associated transcript 1 (CCAT1) was one of the most highly (approximately 68-fold) down-regulated transcripts in ASH1L knockout cells. Therefore, we investigated CCAT1 as a potential mediator of the growth-inducing activity of ASH1L. Supporting this hypothesis, CCAT1 knockdown in ATC cells decreased their growth rate, and ChIP-Seq data indicated that CCAT1 is likely a direct target of ASH1L's histone methyltransferase activity. These results indicate that ASH1L contributes to the aggressiveness of ATC and suggest that ASH1L, along with its upstream regulator miR-200b-3p and its downstream mediator CCAT1, represents a potential therapeutic target in ATC.
Collapse
Affiliation(s)
- Bin Xu
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jingcheng Yu
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Thomas J Giordano
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ronald J Koenig
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
26
|
Li C, Zhang Z, Lv P, Zhan Y, Zhong Q. SCAMP3 Promotes Glioma Proliferation and Indicates Unfavorable Prognosis via Multiple Pathways. Onco Targets Ther 2020; 13:3677-3687. [PMID: 32431518 PMCID: PMC7200257 DOI: 10.2147/ott.s242462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction The secretory carrier-associated membrane protein 3 (SCAMP3) is a component of post-Golgi membranes, functions as a protein carrier and is critical for subcellular protein transportation. Limited studies revealed an elevated expression of SCAMP3 in breast cancer and hepatocellular carcinoma; however, its role in glioma remains unknown. The aim of our study is to investigate the expression pattern and functional mechanisms of SCAMP3 in glioma. Methods mRNA and protein levels of SCAMP3 were examined in glioma tissues together with nontumorous brain tissues by using quantitative real-time-PCR and immunohistochemistry staining. The prognostic role of SCAMP3 in glioma was evaluated through univariate and multivariate analyses. In vitro and in vivo assays were conducted to explore the underlying mechanisms of SCAMP3-induced glioma progression. Results The expression level of SCAMP3 was higher in glioma tissues than that in normal brain tissues. High protein level of SCAMP3 was correlated with larger tumor size and advanced WHO grade. Glioma patients with high-SCAMP3 level had worse overall survival. In addition, SCAMP3 was defined as an independent risk factor of glioma prognosis. Cellular and xenograft studies revealed that SCAMP3 promotes glioma proliferation possibly through enhancing EGFR and mTORC1 signaling. Discussion Our studies revealed that high-SCAMP3 expression level was closely related to the unfavorable clinical features and poor prognosis of glioma patients. SCAMP3 may serve as an invaluable prognostic indicator and novel therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Chunliu Li
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, People's Republic of China
| | - Zhen Zhang
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, People's Republic of China
| | - Peng Lv
- Department of Oncology, Yantai Yuhuangding Hospital Affiliated to College of Qingdao University, Yantai, Shandong 264100, People's Republic of China
| | - Yan Zhan
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, People's Republic of China
| | - Qianwei Zhong
- Department of Neurology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, People's Republic of China
| |
Collapse
|
27
|
Han SS, Feng ZQ, Liu R, Ye J, Cheng WW, Bao JB. Bioinformatics Analysis and RNA-Sequencing of SCAMP3 Expression and Correlated Gene Regulation in Hepatocellular Carcinoma. Onco Targets Ther 2020; 13:1047-1057. [PMID: 32099407 PMCID: PMC7007781 DOI: 10.2147/ott.s221785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Background Secretory Carrier Membrane Proteins 3 (SCAMP3) is a transmembrane protein that affects intracellular trafficking, protein sorting and vesicle formation. Overexpression of SCAMP3 correlates with poorly differentiated hepatocellular carcinoma (HCC). However, the expression and corresponding gene regulation of SCAMP3 in HCC remain unclear. Methods Bioinformatics analyses of clinical parameters and survival data were conducted to predict the prognostic value of SCAMP3 in HCC. RNA sequencing and real-time PCR were conducted to confirm the SCAMP3 expression in HCC tissue. Expression was analyzed using OncomineTM and UALCAN, while SCAMP3 alterations and survival analysis were identified by cBioPortal. Differential gene expression with SCAMP3 was analyzed by LinkedOmics and GEPIA. The target networks of enzymes and co-transcriptional factors were identified using Gene enrichment analysis. Expression of SCAMP3 in HCC tissue was detected by RNA-sequencing and Western-blotting. Results Based on bioinformatics analysis and detection of mRNA expression, SCAMP3 was over-expressed in numerous tumors, especially in HCC. SCAMP3 level was positively correlated with disease stages and tumor grades and negatively correlated with patient survival. Furthermore, functional network analysis indicated that SCAMP3 regulated metabolic process and DNA replication through oxidative phosphorylation and chromatin remodeling or Ribosome. SCAMP3 regulated a number of gene expressions including PPAP2B, SNRK, ARID4A, PRCC, VPS72 via protein binding and proteasome, which may affect cell adhesion, proliferation, transcription, cell cycle and metabolism. Further, Real-time PCR and Western-blotting showed that the SCAMP3 level was increased in HCC tissue. Conclusion The present data analysis efficiently reveals information about SCAMP3 expression and correlated function in HCC, laying a foundation for further study of SCAMP3 in the tumor.
Collapse
Affiliation(s)
- Shan-Shan Han
- Beijing Chaoyang Emergency Medical Center, Department of General Surgery, Chaoyang, Beijing 100020, People's Republic of China
| | - Zhi-Qiang Feng
- Beijing Chaoyang Emergency Medical Center, Department of General Surgery, Chaoyang, Beijing 100020, People's Republic of China
| | - Rui Liu
- Medical University of Anhui Air Force Clinical School, Department of Hepatobiliary Surgery, Beijing 100142, People's Republic of China
| | - Jun Ye
- Medical University of Anhui Air Force Clinical School, Department of Hepatobiliary Surgery, Beijing 100142, People's Republic of China
| | - Wei-Wei Cheng
- Medical University of Anhui Air Force Clinical School, Department of Hepatobiliary Surgery, Beijing 100142, People's Republic of China
| | - Jun-Bo Bao
- First People's Hospital of Suqian, Department of Medicine, Suqian, Jiangsu 223800, People's Republic of China
| |
Collapse
|
28
|
Loss of alanine-glyoxylate and serine-pyruvate aminotransferase expression accelerated the progression of hepatocellular carcinoma and predicted poor prognosis. J Transl Med 2019; 17:390. [PMID: 31771612 PMCID: PMC6880547 DOI: 10.1186/s12967-019-02138-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Accumulated studies reported abnormal gene expression profiles of hepatocellular carcinoma (HCC) by cDNA microarray. We tried to merge cDNA microarray data from different studies to search for stably changed genes, and to find out better diagnostic and prognostic markers for HCC. METHODS A systematic review was performed by searching publications indexed in Pubmed from March 1, 2001 to July 1, 2016. Studies that reporting cDNA microarray profiles in HCC, containing both tumor and nontumor data and published in English-language were retrieved. The differentially expressed genes from eligible studies were summarized and ranked according to the frequency. High frequency genes were subjected to survival analyses. The expression and prognostic value of alanine-glyoxylate and serine-pyruvate aminotransferase (AGXT) was further evaluated in HCC datasets in Oncomine and an independent HCC tissue array cohort. The role of AGXT in HCC progression was evaluated by proliferation and migration assays in a human HCC cell line. RESULTS A total of 43 eligible studies that containing 1917 HCC patients were included, a list of 2022 non redundant abnormally expressed genes in HCC were extracted. The frequencies of reported genes were ranked. We finally obtained a list of only five genes (AGXT; ALDOB; CYP2E1; IGFBP3; TOP2A) that were differentially expressed in tumor and nontumor tissues across studies and were significantly correlated to HCC prognosis. Only AGXT had not been reported in HCC. Reduced expression of AGXT reflected poor differentiation of HCC and predicts poor survival. Knocking down of AGXT enhanced cell proliferation and migration of HCC cell line. CONCLUSIONS The present study supported the feasibility and necessity of systematic review on discovering new and reliable biomarkers for HCC. We also identified a list of high frequency prognostic genes and emphasized a critical role of AGXT deletion during HCC progression.
Collapse
|
29
|
Small molecule inhibitors and CRISPR/Cas9 mutagenesis demonstrate that SMYD2 and SMYD3 activity are dispensable for autonomous cancer cell proliferation. PLoS One 2018; 13:e0197372. [PMID: 29856759 PMCID: PMC5983452 DOI: 10.1371/journal.pone.0197372] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/01/2018] [Indexed: 12/13/2022] Open
Abstract
A key challenge in the development of precision medicine is defining the phenotypic consequences of pharmacological modulation of specific target macromolecules. To address this issue, a variety of genetic, molecular and chemical tools can be used. All of these approaches can produce misleading results if the specificity of the tools is not well understood and the proper controls are not performed. In this paper we illustrate these general themes by providing detailed studies of small molecule inhibitors of the enzymatic activity of two members of the SMYD branch of the protein lysine methyltransferases, SMYD2 and SMYD3. We show that tool compounds as well as CRISPR/Cas9 fail to reproduce many of the cell proliferation findings associated with SMYD2 and SMYD3 inhibition previously obtained with RNAi based approaches and with early stage chemical probes.
Collapse
|
30
|
Li LX, Zhou JX, Calvet JP, Godwin AK, Jensen RA, Li X. Lysine methyltransferase SMYD2 promotes triple negative breast cancer progression. Cell Death Dis 2018; 9:326. [PMID: 29487338 PMCID: PMC5832424 DOI: 10.1038/s41419-018-0347-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/06/2018] [Accepted: 01/22/2018] [Indexed: 02/06/2023]
Abstract
We identified SMYD2, a SMYD (SET and MYND domain) family protein with lysine methyltransferase activity, as a novel breast cancer oncogene. SMYD2 was expressed at significantly higher levels in breast cancer cell lines and in breast tumor tissues. Silencing of SMYD2 by RNAi in triple-negative breast cancer (TNBC) cell lines or inhibition of SMYD2 with its specific inhibitor, AZ505, significantly reduced tumor growth in vivo. SMYD2 executes this activity via methylation and activation of its novel non-histone substrates, including STAT3 and the p65 subunit of NF-κB, leading to increased TNBC cell proliferation and survival. There are cross-talk and synergistic effects among SMYD2, STAT3, and NF-κB in TNBC cells, in that STAT3 can contribute to the modification of NF-κB p65 subunit post-translationally by recruitment of SMYD2, whereas the p65 subunit of NF-κB can also contribute to the modification of STAT3 post-translationally by recruitment of SMYD2, leading to methylation and activation of STAT3 and p65 in these cells. The expression of SMYD2 can be upregulated by IL-6-STAT3 and TNFα-NF-κB signaling, which integrates epigenetic regulation to inflammation in TNBC development. In addition, we have identified a novel SMYD2 transcriptional target gene, PTPN13, which links SMYD2 to other known breast cancer associated signaling pathways, including ERK, mTOR, and Akt signaling via PTPN13 mediated phosphorylation.
Collapse
Affiliation(s)
- Linda Xiaoyan Li
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Julie Xia Zhou
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - James P Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Roy A Jensen
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Xiaogang Li
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA. .,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA. .,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
31
|
Hosseini A, Minucci S. Alterations of Histone Modifications in Cancer. EPIGENETICS IN HUMAN DISEASE 2018:141-217. [DOI: 10.1016/b978-0-12-812215-0.00006-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
32
|
Overexpression of SCAMP3 is an indicator of poor prognosis in hepatocellular carcinoma. Oncotarget 2017; 8:109247-109257. [PMID: 29312605 PMCID: PMC5752518 DOI: 10.18632/oncotarget.22665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/23/2017] [Indexed: 12/22/2022] Open
Abstract
SCAMP3, an isoform of the secretory carrier membrane proteins (SCAMPs) family, is a membrane-trafficking protein involved in endosome transport. Previous microarray data showed that SCAMP3 mRNA is highly expressed in hepatocellular carcinoma (HCC). In this study, the expression and clinical significance of SCAMP3 in 100 pairs of HCC and adjacent normal tissue were investigated. siRNA transfection was performed to silence SCAMP3 expression in HCC cells. The MTS assay and flow cytometry were used to detect the proliferation, cell cycle progression of HCC cells. Compared with adjacent normal tissues, SCAMP3 expression was dramatically increased in HCC tissues demonstrated by Western blotting (P < 0.05). In immunohistochemistry, compared with the adjacent normal tissues, SCAMP3 was detected in 96% of the HCC samples with a significant increase in intensity and number of stained cells (P < 0.05). Also, high SCAMP3 expression was found in 86% of the HCC samples (P < 0.05). The increased SCAMP3 expression was significantly correlated with vascular invasion (P = 0.004) and tumor stage (P = 0.001). Univariate and multivariate survival analyses showed that the expression of SCAMP3 was an independent prognostic factor of overall survival of HCC patients. Knockdown of SCAMP3 expression led to suppression of cell proliferation and blockage of cell cycle of HCC cells. In conclusion, our present study suggested that SCAMP3 may serve as a promising prognostic biomarker and molecular target of HCC and further investigation is warranted.
Collapse
|
33
|
Shi Y, Qin N, Zhou Q, Chen Y, Huang S, Chen B, Shen G, Jia H. Role of IQGAP3 in metastasis and epithelial-mesenchymal transition in human hepatocellular carcinoma. J Transl Med 2017; 15:176. [PMID: 28810875 PMCID: PMC5558666 DOI: 10.1186/s12967-017-1275-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/01/2017] [Indexed: 01/06/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide owing to its high rates of metastasis and recurrence. The oncogene IQ motif-containing GTPase activating protein 3 (IQGAP3) is ubiquitously overexpressed in several human cancers, including liver, ovary, lung, large intestine, gastric, bone marrow, and breast malignancies and is involved in the invasion and metastasis of cancer cells. Therefore, we aimed to determine the biological role and molecular mechanism of IQGAP3 in HCC. Methods We used 120 archived clinical HCC samples, 9 snap-frozen HCC tumor tissues, and 4 normal liver tissues. Expression of IQGAP3 mRNA and protein in HCC cell lines (Hep3B, SMMC-7721, HCCC-9810, HepG2, BEL-7404, HCCLM3, QGY-7701, Huh7, and MHCC97H) and normal liver epithelial cells LO2 was examined by western blot, quantitative polymerase chain reaction, and immunohistochemistry. In addition, wound-healing and transwell matrix penetration assays were used to assess the migratory and invasive abilities of HCC cells, respectively. Results Expression of the IQGAP3 was robustly upregulated in HCC cells and tissues. High expression of IQGAP3 in HCC correlated with aggressive clinicopathological features and was an independent poor prognostic factor for overall survival. Furthermore, ectopic expression of IQGAP3 markedly enhanced HCC cell migration, invasion, and epithelial-to-mesenchymal transition (EMT) in vitro and promoted metastasis of orthotopic hepatic tumors in nude mice. Conversely, silencing endogenous IQGAP3 showed an opposite effect. Mechanistically, IQGAP3 promoted EMT and metastasis by activating TGF-β signaling. Conclusions IQGAP3 functions as an important regulator of metastasis and EMT by constitutively activating the TGF-β signaling pathway in HCC. Our findings present new evidence of the role of IQGAP3 in EMT and metastasis, indicating its potential as a prognostic biomarker candidate and a therapeutic target against HCC.
Collapse
Affiliation(s)
- Yongjie Shi
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Nan Qin
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Qiang Zhou
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Yanqiu Chen
- Department of ENT, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9th Jinsui Road, Guangzhou, 510623, Guangdong, People's Republic of China
| | - Sicong Huang
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Bo Chen
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Gang Shen
- Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9th Jinsui Road, Guangzhou, 510623, Guangdong, People's Republic of China.
| | - Hongyun Jia
- Department of Clinical Examination, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
34
|
Elsemman IE, Mardinoglu A, Shoaie S, Soliman TH, Nielsen J. Systems biology analysis of hepatitis C virus infection reveals the role of copy number increases in regions of chromosome 1q in hepatocellular carcinoma metabolism. MOLECULAR BIOSYSTEMS 2017; 12:1496-506. [PMID: 27040643 DOI: 10.1039/c5mb00827a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hepatitis C virus (HCV) infection is a worldwide healthcare problem; however, traditional treatment methods have failed to cure all patients, and HCV has developed resistance to new drugs. Systems biology-based analyses could play an important role in the holistic analysis of the impact of HCV on hepatocellular metabolism. Here, we integrated HCV assembly reactions with a genome-scale hepatocyte metabolic model to identify metabolic targets for HCV assembly and metabolic alterations that occur between different HCV progression states (cirrhosis, dysplastic nodule, and early and advanced hepatocellular carcinoma (HCC)) and healthy liver tissue. We found that diacylglycerolipids were essential for HCV assembly. In addition, the metabolism of keratan sulfate and chondroitin sulfate was significantly changed in the cirrhosis stage, whereas the metabolism of acyl-carnitine was significantly changed in the dysplastic nodule and early HCC stages. Our results explained the role of the upregulated expression of BCAT1, PLOD3 and six other methyltransferase genes involved in carnitine biosynthesis and S-adenosylmethionine metabolism in the early and advanced HCC stages. Moreover, GNPAT and BCAP31 expression was upregulated in the early and advanced HCC stages and could lead to increased acyl-CoA consumption. By integrating our results with copy number variation analyses, we observed that GNPAT, PPOX and five of the methyltransferase genes (ASH1L, METTL13, SMYD2, TARBP1 and SMYD3), which are all located on chromosome 1q, had increased copy numbers in the cancer samples relative to the normal samples. Finally, we confirmed our predictions with the results of metabolomics studies and proposed that inhibiting the identified targets has the potential to provide an effective treatment strategy for HCV-associated liver disorders.
Collapse
Affiliation(s)
- Ibrahim E Elsemman
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. and Department of Mathematics, Faculty of Science, Assiut University, Assiut, Egypt and The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Hørsholm, Denmark
| | - Adil Mardinoglu
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. and Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Saeed Shoaie
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Taysir H Soliman
- Information Systems Department, Faculty of Computers and Information, Assiut University, Assiut, Egypt
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. and Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
35
|
Sandbothe M, Buurman R, Reich N, Greiwe L, Vajen B, Gürlevik E, Schäffer V, Eilers M, Kühnel F, Vaquero A, Longerich T, Roessler S, Schirmacher P, Manns MP, Illig T, Schlegelberger B, Skawran B. The microRNA-449 family inhibits TGF-β-mediated liver cancer cell migration by targeting SOX4. J Hepatol 2017; 66:1012-1021. [PMID: 28088579 DOI: 10.1016/j.jhep.2017.01.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Modulation of microRNA expression is a potential treatment for hepatocellular carcinoma (HCC). Therefore, the epigenetically regulated microRNA-449 family (miR-449a, miR-449b, miR-449c) was characterized with regards to its functional effects and target genes in HCC. METHODS After transfection of miR-449a, miR-449b, and/or miR-449c, tumor-relevant functional effects were analyzed using in vitro assays and a xenograft mouse model. Binding specificities, target genes, and regulated pathways of each miRNA were identified by microarray analyses. Target genes were validated by luciferase reporter assays and expression analyses in vitro. Furthermore, target gene expression was analyzed in 61 primary human HCCs compared to normal liver tissue. RESULTS Tumor suppressive effects, binding specificities, target genes, and regulated pathways of miR-449a and miR-449b differed from those of miR-449c. Transfection of miR-449a, miR-449b, and/or miR-449c inhibited cell proliferation and migration, induced apoptosis, and reduced tumor growth to different extents. Importantly, miR-449a, miR-449b, and, to a lesser degree, miR-449c directly targeted SOX4, which codes for a transcription factor involved in epithelial-mesenchymal transition and HCC metastasis, and thereby inhibited TGF-β-mediated cell migration. CONCLUSIONS This study provides detailed insights into the regulatory network of the epigenetically regulated miRNA-449 family and, for the first time, describes distinct tumor suppressive effects and target specificities of miR-449a, miR-449b, and miR-449c. Our results indicate that particularly miR-449a and miR-449b may be considered for miRNA replacement therapy to prevent HCC progression and metastasis. LAY SUMMARY In this study, we demonstrated that the microRNA-449 family acts as a tumor suppressor in liver cancer by causing cell death and inhibiting cell migration. These effects are caused by downregulation of the oncogene SOX4, which is frequently overexpressed in liver cancer. We conclude that the microRNA-449 family may be a target for liver cancer therapy.
Collapse
Affiliation(s)
- Maria Sandbothe
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Reena Buurman
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Nicole Reich
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Luisa Greiwe
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Beate Vajen
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Engin Gürlevik
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Vera Schäffer
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Marlies Eilers
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Thomas Longerich
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Institute of Pathology, University Hospital RWTH Aachen, Heidelberg, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael P Manns
- Clinic for Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Thomas Illig
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | - Britta Skawran
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
36
|
Naboulsi W, Bracht T, Megger DA, Reis H, Ahrens M, Turewicz M, Eisenacher M, Tautges S, Canbay AE, Meyer HE, Weber F, Baba HA, Sitek B. Quantitative proteome analysis reveals the correlation between endocytosis-associated proteins and hepatocellular carcinoma dedifferentiation. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1579-85. [PMID: 27519163 DOI: 10.1016/j.bbapap.2016.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 01/04/2023]
Abstract
The majority of poorly differentiated hepatocellular carcinomas (HCCs) develop from well-differentiated tumors. Endocytosis is a cellular function which is likely to take part in this development due to its important role in regulating the abundances of vital signaling receptors. Here, we aimed to investigate the abundance of endocytosis-associated proteins in HCCs with various differentiation grades. Therefore, we analyzed 36 tissue specimens from HCC patients via LC-MS/MS-based label-free quantitative proteomics including 19 HCC tissue samples with different degrees of histological grades and corresponding non-tumorous tissue controls. As a result, 277 proteins were differentially regulated between well-differentiated tumors and controls. In moderately and poorly differentiated tumors, 278 and 1181 proteins, respectively, were significantly differentially regulated compared to non-tumorous tissue. We explored the regulated proteins based on their functions and identified thirty endocytosis-associated proteins, mostly overexpressed in poorly differentiated tumors. These included proteins that have been shown to be up-regulated in HCC like clathrin heavy chain-1 (CLTC) as well as unknown proteins, such as secretory carrier-associated membrane protein 3 (SCAMP3). The abundances of SCAMP3 and CLTC were immunohistochemically examined in tissue sections of 84 HCC patients. We demonstrate the novel association of several endocytosis-associated proteins, in particular, SCAMP3 with HCC progression.
Collapse
Affiliation(s)
- Wael Naboulsi
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| | - Thilo Bracht
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Dominik A Megger
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Henning Reis
- Institute of Pathology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Maike Ahrens
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Michael Turewicz
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Stephanie Tautges
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Ali E Canbay
- Department of Gastroenterology and Hepatology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Helmut E Meyer
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Frank Weber
- Department of General, Visceral and Transplantation Surgery, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Hideo A Baba
- Institute of Pathology, University of Duisburg-Essen, University Hospital Essen, 45147 Essen, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany.
| |
Collapse
|
37
|
Qian EN, Han SY, Ding SZ, Lv X. Expression and diagnostic value of CCT3 and IQGAP3 in hepatocellular carcinoma. Cancer Cell Int 2016; 16:55. [PMID: 27390551 PMCID: PMC4936258 DOI: 10.1186/s12935-016-0332-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
Background To evaluate plasma chaperonin containing TCP1 complex subunit 3 (CCT3) and IQ-motif-containing GTPase-activating protein-3 (IQGAP3) as biomarker for hepatocellular carcinoma (HCC) screening and diagnosis. Methods Blood samples were collected from 126 HCC patients with HCC, 88 patients with cirrhosis and 50 healthy volunteers to detect plasma α-fetoprotein (AFP), CCT3 and IQGAP3 levels. Plasma AFP, CCT3 and IQGAP3 protein levels were measured by enzyme linked immunosorbent assay (ELISA). Results In the plasma of HCC patients, both CCT3 and IQGAP3 were significantly higher than in patients with cirrhosis and in healthy controls (P < 0.01). CCT3 and IQGAP3 protein level correlated well with HCC etiology, tumor size, TNM stage, and child-pugh classification. CCT3 protein had higher sensitivity in the diagnosis of HCC when compared with AFP (87.3 vs 69.8 %). In addition, CCT3 and IQGAP3 protein were able to complement AFP in detecting AFP-negative HCC patients with sensitivity and specificity of 92.1 and 70.5 % and 81.6 and 71.6 %, respectively. In the small HCC group, CCT3 and IQGAP3 protein had sensitivity of 76.6 and 74.5 %, respectively. The combination of AFP + CCT3 + IQGAP3 (0.954) had significantly superior discriminative ability than AFP alone (0.815; P < 0.01). Conclusions CCT3 and IQGAP3 are novel complementary biomarkers for HCC screening and diagnosis, especially for AFP-negative and small HCC patients.
Collapse
Affiliation(s)
- E-Na Qian
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Shuang-Yin Han
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Song-Ze Ding
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| | - Xun Lv
- Department of Gastroenterology and Hepatology, People's Hospital of Zhengzhou University, No. 7 Wei Wu Road, Zhengzhou, 450003 Henan China
| |
Collapse
|
38
|
Singh S, Colonna G, Di Bernardo G, Bergantino F, Cammarota M, Castello G, Costantini S. The gene expression profiling of hepatocellular carcinoma by a network analysis approach shows a dominance of intrinsically disordered proteins (IDPs) between hub nodes. MOLECULAR BIOSYSTEMS 2015; 11:2933-2945. [PMID: 26267014 DOI: 10.1039/c5mb00434a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have analyzed the transcriptomic data from patients with hepatocellular carcinoma (HCC) after viral HCV infection at the various stages of the disease by means of a networking analysis using the publicly available E-MTAB-950 dataset. The data was compared with those obtained in our group from HepG2 cells, a cancer cell line that lacks the viral infection. By sequential pruning of data, and also taking into account the data from cells of healthy patients as blanks, we were able to obtain a distribution of hub genes for the various stages that characterize the disease and finally, we isolated a metabolic sub-net specific to HCC alone. The general picture is that the basic organization to energetically and metabolically sustain the cells in both the normal and diseased conditions is the same, but a complex cluster of sub-networks controlled by hub genes drives the HCC progression with high metabolic flexibility and plasticity. In particular, we have extracted a sub-net of genes strictly correlated to other hub genes of the network from HepG2 cells, but specific for the HCC and mainly devoted to: (i) control at chromatin levels of cell division; (ii) control of ergastoplasmatic stress through protein degradation and misfolding; (iii) control of the immune response also through an increase of mature T-cells in the thymus. This sub-net is characterized by 26 hub genes coding for intrinsically disordered proteins with a high ability to interact with numerous molecular partners. Moreover, we have also noted that periphery molecules, that is, with one or very few interactions (e.g., cytokines or post-translational enzymes), which do not have a central role in the clusters that make up the global metabolic network, essentially have roles as information transporters. The results evidence a strong presence of intrinsically disordered proteins with key roles as hubs in the sub-networks that characterize the various stages of the disease, conferring a structural plasticity to the net nodes but an inherent functional versatility to the whole metabolic net. Thus, our present article provides a novel way of targeting the intrinsic disorder in HCC networks to dampen the cancer effects and provides new insight into the potential mechanisms of HCC. Taken together, the present findings suggest novel targets to design strategies for drug design and may support a rational intervention in the pharmacotherapy of HCC and other associated diseases.
Collapse
Affiliation(s)
- Sakshi Singh
- Dottorato in Biologia Computazionale, Dipartimento di Biochimica, Biofisica e Patologia generale, Seconda Università degli Studi di Napoli, Napoli, Italy
| | | | | | | | | | | | | |
Collapse
|
39
|
Xiong Y, Wu S, Du Q, Wang A, Wang Z. Integrated analysis of gene expression and genomic aberration data in osteosarcoma (OS). Cancer Gene Ther 2015; 22:524-9. [PMID: 26427513 DOI: 10.1038/cgt.2015.48] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/28/2015] [Accepted: 08/30/2015] [Indexed: 12/15/2022]
Abstract
Cytogenetic analyses have revealed that complex karyotypes with numerous and highly variable genomic aberrations including single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs), are observed in most of the conventional osteosarcomas (OSs). Several genome-wide studies have reported that the dysregulated expression of many genes is correlated with genomic aberrations in OS. We first compared OS gene expression in Gene Expression Omnibus (GEO) data sets and genomic aberrations in International Cancer Genome Consortium (ICGC) database to identify differentially expressed genes (DEGs) associated with SNPs or CNVs in OS. Then the function annotation of SNP- or CNV-associated DEGs was performed in terms of gene ontology analysis, pathway analysis and protein-protein interactions (PPIs). Finally, the expression of genes correlated with both SNPs and CNVs were confirmed by quantitative reverse-transcription PCR. Eight publicly available GEO data sets were obtained, and a set of 979 DEGs were identified (472 upregulated and 507 downregulated DEGs). Moreover, we obtained 1039 SNPs mapped in 938 genes, and 583 CNV sites mapped in 2915 genes. Comparing genomic aberrations and DGEs, we found 41 SNP-associated DEGs and 124 CNV-associated DEGs, in which 7 DGEs were associated with both SNPs and CNVs, including WWP1, EXT1, LDHB, C8orf59, PLEKHA5, CCT3 and VWF. The result of function annotation showed that ossification, bone development and skeletal system development were the significantly enriched terms of biological processes for DEGs. PPI network analysis showed that CCT3, COPS3 and WWP1 were the significant hub proteins. We conclude that these genes, including CCT3, COPS3 and WWP1 are candidate driver genes of importance in OS tumorigenesis.
Collapse
Affiliation(s)
- Y Xiong
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| | - S Wu
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Q Du
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| | - A Wang
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Z Wang
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
40
|
Fang X, Zhang B, Thisse B, Bloom GS, Thisse C. IQGAP3 is essential for cell proliferation and motility during zebrafish embryonic development. Cytoskeleton (Hoboken) 2015; 72:422-33. [PMID: 26286209 DOI: 10.1002/cm.21237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/07/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022]
Abstract
IQGAPs are scaffolding proteins that regulate actin assembly, exocyst function, cell motility, morphogenesis, adhesion and division. Vertebrates express 3 family members: IQGAP1, IQGAP2, and IQGAP3. IQGAP1 is known to stimulate nucleation of branched actin filaments through N-WASP and the Arp2/3 complex following direct binding to cytoplasmic tails of ligand-activated growth factor receptors, including EGFR, VEGFR2 and FGFR1. By contrast, little is known about functions of IQGAP2 or IQGAP3. Using in situ hybridization on whole mount zebrafish (Danio rerio) embryos, we show that IQGAP1 and IQGAP2 are associated with discrete tissues and organs, while IQGAP3 is mainly expressed in proliferative cells throughout embryonic and larval development. Morpholino knockdowns of IQGAP1 and IQGAP2 have little effect on embryo morphology while loss of function of IQGAP3 affects both cell proliferation and cell motility. IQGAP3 morphant phenotypes are similar to those resulting from overexpression of dominant negative forms of Ras or of Fibroblast Growth Factor Receptor 1 (FGFR1), suggesting that IQGAP3 plays a role in FGFR1-Ras-ERK signaling. In support of this hypothesis, dominant negative forms of FGFR1 or Ras could be rescued by co-injection of zebrafish IQGAP3 mRNA, strongly suggesting that IQGAP3 acts as a downstream regulator of the FGFR1-Ras signaling pathway.
Collapse
Affiliation(s)
- Xiaolan Fang
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Bianhong Zhang
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia.,Institute of Biomedical Science, School of Science, East China Normal University, Shanghai, China
| | - Bernard Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - George S Bloom
- Department of Biology, University of Virginia, Charlottesville, Virginia.,Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Christine Thisse
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
41
|
Ho DWH, Kai AKL, Ng IOL. TCGA whole-transcriptome sequencing data reveals significantly dysregulated genes and signaling pathways in hepatocellular carcinoma. Front Med 2015; 9:322-30. [DOI: 10.1007/s11684-015-0408-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/25/2015] [Indexed: 10/23/2022]
|
42
|
Mocellin S, Verdi D, Pooley KA, Nitti D. Genetic variation and gastric cancer risk: a field synopsis and meta-analysis. Gut 2015; 64:1209-19. [PMID: 25731870 DOI: 10.1136/gutjnl-2015-309168] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/06/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Data on genetic susceptibility to sporadic gastric carcinoma have been published at a growing pace, but to date no comprehensive overview and quantitative summary has been available. METHODS We conducted a systematic review and meta-analysis of the evidence on the association between DNA variation and risk of developing stomach cancer. To assess result credibility, summary evidence was graded according to the Venice criteria and false positive report probability (FPRP) was calculated to further validate result noteworthiness. Meta-analysis was also conducted for subgroups, which were defined by ethnicity (Asian vs Caucasian), tumour histology (intestinal vs diffuse), tumour site (cardia vs non-cardia) and Helicobacter pylori infection status (positive vs negative). RESULTS Literature search identified 824 eligible studies comprising 2 530 706 subjects (cases: 261 386 (10.3%)) and investigating 2841 polymorphisms involving 952 distinct genes. Overall, we performed 456 primary and subgroup meta-analyses on 156 variants involving 101 genes. We identified 11 variants significantly associated with disease risk and assessed to have a high level of summary evidence: MUC1 rs2070803 at 1q22 (diffuse carcinoma subgroup), MTX1 rs2075570 at 1q22 (diffuse), PSCA rs2294008 at 8q24.2 (non-cardia), PRKAA1 rs13361707 5p13 (non-cardia), PLCE1 rs2274223 10q23 (cardia), TGFBR2 rs3087465 3p22 (Asian), PKLR rs3762272 1q22 (diffuse), PSCA rs2976392 (intestinal), GSTP1 rs1695 11q13 (Asian), CASP8 rs3834129 2q33 (mixed) and TNF rs1799724 6p21.3 (mixed), with the first nine variants characterised by a low FPRP. We also identified polymorphisms with lower quality significant associations (n=110). CONCLUSIONS We have identified several high-quality biomarkers of gastric cancer susceptibility. These data will form the backbone of an annually updated online resource that will be integral to the study of gastric carcinoma genetics and may inform future screening programmes.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Daunia Verdi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Karen A Pooley
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Donato Nitti
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
43
|
Cui X, Hu ZP, Li Z, Gao PJ, Zhu JY. Overexpression of chaperonin containing TCP1, subunit 3 predicts poor prognosis in hepatocellular carcinoma. World J Gastroenterol 2015; 21:8588-8604. [PMID: 26229401 PMCID: PMC4515840 DOI: 10.3748/wjg.v21.i28.8588] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/03/2015] [Accepted: 07/03/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the value of chaperonin containing TCP1, subunit 3 (CCT3) to predict the prognosis of patients with hepatocellular carcinoma (HCC) and determine its function in HCC progression.
METHODS: CCT3 expression levels were examined in human non-cancerous liver tissues and a variety of HCC cell lines by quantitative real-time PCR and immunoblotting. CCT3 expression was suppressed by small interfering RNA. The effects of reducing CCT3 expression in HCC cells were tested. The 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) assay, cell counting experiment, cell cycle assay, apoptosis assay and invasion assay were employed to evaluate cell functions in vitro. Immunohistochemistry was performed on HCC specimens. In addition, CCT3 expression in HCC specimens was also assessed at the protein and mRNA level. Associations between clinicopathological characteristics and prognosis were analyzed, along with the possible mechanisms involved in CCT3’s function in HCC progression.
RESULTS: The expression levels of CCT3 mRNA and protein were upregulated in HCC cell lines in contrast to adjacent non-cancerous tissues. Reducing CCT3 expression not only suppressed cell proliferation in cell counts, MTT assay, cell cycle assay and induced cell apoptosis (P < 0.05 vs negative control), but also inhibited the tumor cell invasion capacity in vitro (P < 0.01 vs negative control). Overexpression of CCT3 in the nuclei of cancer cells in HCC specimens (58 of 104 patients, 55.8%) was associated with poor prognosis in HCC patients (3-year survival rate, 55.5% vs 84.2%, P = 0.020) after hepatectomy. Mechanistic analyses showed that signal transducer and activator of transcription 3 (STAT3) activation was decreased even when stimulated by interleukin-6 after knocking down CCT3 in the HepG2 cell line.
CONCLUSION: Overexpression of CCT3 in the nuclei of cancerous cells is associated with HCC progression. CCT3 may be a target that affects the activation of STAT3 in HCC.
Collapse
|
44
|
Jiang N, Chen WJ, Zhang JW, Xu C, Zeng XC, Zhang T, Li Y, Wang GY. Downregulation of miR-432 activates Wnt/β-catenin signaling and promotes human hepatocellular carcinoma proliferation. Oncotarget 2015; 6:7866-7879. [PMID: 25797263 PMCID: PMC4480722 DOI: 10.18632/oncotarget.3492] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/03/2015] [Indexed: 01/25/2023] Open
Abstract
Sustained cell growth and proliferation, one of the hallmarks of cancer, is considered to responsible for cancer-related deaths by disorganizing the balance of growth promotion and growth limitation. Aberrant activation of the Wnt/β-catenin signaling pathway leads to cell proliferation, growth and survival, and promotes the development of various human tumors, including hepatocellular carcinoma. Elucidating the molecular mechanism of this abnormality in hepatocellular carcinoma carcinogenesis may improve diagnostic and therapeutic strategies for this malignancy. Herein, we report that the expression of miR-432 was markedly downregulated in hepatocellular carcinoma cell lines and tissues, and upregulation of miR-432 inhibited, whereas downregulation of miR-432 enhanced the proliferation and tumorigenicity of hepatocellular carcinoma cells both in vitro and in vivo. Furthermore, miR-432 directly targeted and suppressed multiple regulators of the Wnt/β-catenin signaling cascade, including LRP6, TRIM29 and Pygo2, which subsequently deactivated Wnt/β-catenin signaling pathway. Finally, miR-432 expression was inversely correlated with three targets in clinical hepatocellular carcinoma samples. These results demonstrated that miR-432 functions as a tumor-suppressive miRNA by suppressing Wnt/β-catenin signaling activation and may represent a therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wen-Jie Chen
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Wen Zhang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chi Xu
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xian-Cheng Zeng
- Department of General Surgery and Clinical Laboratory, Zengcheng People's Hospital, (BoJi-Affiliated Hospital of Sun Yat-Sen University), Zengcheng, Guangdong, China
| | - Tong Zhang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yang Li
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Guo-Ying Wang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Nguyen H, Allali-Hassani A, Antonysamy S, Chang S, Chen LH, Curtis C, Emtage S, Fan L, Gheyi T, Li F, Liu S, Martin JR, Mendel D, Olsen JB, Pelletier L, Shatseva T, Wu S, Zhang FF, Arrowsmith CH, Brown PJ, Campbell RM, Garcia BA, Barsyte-Lovejoy D, Mader M, Vedadi M. LLY-507, a Cell-active, Potent, and Selective Inhibitor of Protein-lysine Methyltransferase SMYD2. J Biol Chem 2015; 290:13641-53. [PMID: 25825497 PMCID: PMC4447944 DOI: 10.1074/jbc.m114.626861] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Indexed: 12/13/2022] Open
Abstract
SMYD2 is a lysine methyltransferase that catalyzes the monomethylation of several protein substrates including p53. SMYD2 is overexpressed in a significant percentage of esophageal squamous primary carcinomas, and that overexpression correlates with poor patient survival. However, the mechanism(s) by which SMYD2 promotes oncogenesis is not understood. A small molecule probe for SMYD2 would allow for the pharmacological dissection of this biology. In this report, we disclose LLY-507, a cell-active, potent small molecule inhibitor of SMYD2. LLY-507 is >100-fold selective for SMYD2 over a broad range of methyltransferase and non-methyltransferase targets. A 1.63-Å resolution crystal structure of SMYD2 in complex with LLY-507 shows the inhibitor binding in the substrate peptide binding pocket. LLY-507 is active in cells as measured by reduction of SMYD2-induced monomethylation of p53 Lys370 at submicromolar concentrations. We used LLY-507 to further test other potential roles of SMYD2. Mass spectrometry-based proteomics showed that cellular global histone methylation levels were not significantly affected by SMYD2 inhibition with LLY-507, and subcellular fractionation studies indicate that SMYD2 is primarily cytoplasmic, suggesting that SMYD2 targets a very small subset of histones at specific chromatin loci and/or non-histone substrates. Breast and liver cancers were identified through in silico data mining as tumor types that display amplification and/or overexpression of SMYD2. LLY-507 inhibited the proliferation of several esophageal, liver, and breast cancer cell lines in a dose-dependent manner. These findings suggest that LLY-507 serves as a valuable chemical probe to aid in the dissection of SMYD2 function in cancer and other biological processes.
Collapse
Affiliation(s)
- Hannah Nguyen
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285,
| | - Abdellah Allali-Hassani
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| | - Stephen Antonysamy
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Shawn Chang
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Lisa Hong Chen
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Carmen Curtis
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Spencer Emtage
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Li Fan
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Tarun Gheyi
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Fengling Li
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| | - Shichong Liu
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Joseph R Martin
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - David Mendel
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Jonathan B Olsen
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Laura Pelletier
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Tatiana Shatseva
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| | - Song Wu
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Feiyu Fred Zhang
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada, Department of Medical Biophysics, University of Toronto and Princess Margaret Cancer Centre, 101 College Street, MaRS South Tower, Suite 707, Toronto, Ontario M5G 1L7, Canada, and
| | - Peter J Brown
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| | - Robert M Campbell
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Dalia Barsyte-Lovejoy
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| | - Mary Mader
- From the Departments of Oncology Discovery, Structural Biology, Tailored Therapeutics, and Discovery Chemistry Research and Technologies, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS South Tower, 7th floor, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
46
|
Zhang S, Li J, He F, Wang XM. Abnormal nuclear expression of Pygopus-2 in human primary hepatocellular carcinoma correlates with a poor prognosis. Histopathology 2015; 67:176-84. [PMID: 25545771 DOI: 10.1111/his.12637] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 12/21/2014] [Indexed: 12/13/2022]
Abstract
AIMS Pygopus-2 (Pygo2) is a critical element of the canonical Wnt/β-catenin transcriptional complex. The aim of the present study was to investigate the expression patterns and clinicopathological significance of Pygo2 in human primary hepatocellular carcinoma (HCC). METHODS AND RESULTS Real-time polymerase chain reaction (PCR) analysis of the mRNA levels of Pygo2 in 50 paired HCC cancer/adjacent non-cancerous tissues showed that Pygo2 mRNA expression was significantly higher in cancerous tissues (P = 0.009). Immunohistochemical analysis showed that abnormal Pygo2 protein expression in HCC patients was associated with age (P = 0.025), tumour size (P = 0.005), intra- or extra-hepatic metastasis (P = 0.029), vascular invasion (P = 0.026) and tumour differentiation (P = 0.004). Patients with normal Pygo2 protein expression showed a longer survival time (P = 0.031) and a higher 1-year survival rate (P = 0.032) than those with abnormal Pygo2 expression. Cox's proportional hazard regression model showed that abnormal Pygo2 protein expression was a risk factor associated with the prognosis of HCC patients (P = 0.043). CONCLUSION To the best of our knowledge, this is the first report investigating Pygo2 expression patterns and their clinicopathological significance in HCC. Our findings suggest that Pygo2 may be an important predictor of poor outcome in HCC patients, and could serve as a novel biomarker for HCC.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of General Surgery, Xiehe Hospital Fujian Medical University, Fujian, China
| | - Jie Li
- Department of Hepatobiliary Surgery, ZhongShan hospital Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Fujian, China
| | - Fei He
- Department of Hepatobiliary Surgery, ZhongShan hospital Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Fujian, China
| | - Xiao-Min Wang
- Department of General Surgery, Xiehe Hospital Fujian Medical University, Fujian, China.,Department of Hepatobiliary Surgery, ZhongShan hospital Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Fujian, China
| |
Collapse
|
47
|
Liu X, Wang SK, Zhang K, Zhang H, Pan Q, Liu Z, Pan H, Xue L, Yen Y, Chu PG. Expression of glypican 3 enriches hepatocellular carcinoma development-related genes and associates with carcinogenesis in cirrhotic livers. Carcinogenesis 2014; 36:232-42. [PMID: 25542894 DOI: 10.1093/carcin/bgu245] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Glypican-3 (GPC3) protein expression was determined by immunohistochemical analysis from 29 normal livers, 80 cirrhotic livers sample taken near hepatocellular carcinoma (HCC), and 87 cirrhotic livers without HCC. The levels for miR-657 and HCC-related gene mRNAs were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Also, a published microarray dataset was used for gene set enrichment analysis (GSEA) to investigate the relationship between GPC3- and HCC-related gene signatures. Kaplan-Meier analysis was used to evaluate the relationship between GPC3 and HCC recurrence. GPC3 protein expression was not detected in any of the 29 (0%) normal livers, but was detected in 32 of 87 (37%) cirrhotic livers without HCC, and 51 of 80 (64%) cirrhotic liver samples taken near HCC sites (P < 0.001). The GPC3-positive rate in cirrhotic livers of viral origin was 68% (27/40), which was significantly higher than for non-viral cirrhotic livers (11%, 5/47) (P < 0.001). Also, GPC3 expression positively correlated with mRNA expression of HCC-related genes in the qRT-PCR and GSEA evaluations. Furthermore, HCC recurrence in cirrhotic liver samples taken near HCC sites was significantly higher in the GPC3-positive group than the GPC3-negative group (Log-rank P = 0.02, HR = 3.26; 95% CI = 1.20-10.29). This study demonstrated that highly expression of GPC3 could enrich HCC-related genes' mRNA expression and positive associate with dysplasia in cirrhotic livers. Therefore, GPC3 may serve as a precancerous biomarker in cirrhotic livers.
Collapse
Affiliation(s)
- Xiyong Liu
- Department of Molecular Pharmacology, Beckman Research Institute
| | - Sean K Wang
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Keqiang Zhang
- Department of Molecular Pharmacology, Beckman Research Institute
| | - Hang Zhang
- Department of Molecular Pharmacology, Beckman Research Institute
| | - Qin Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China and
| | - Zhiwei Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China and
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China and
| | - Lijun Xue
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yun Yen
- Department of Molecular Pharmacology, Beckman Research Institute, Taipei Medical University, Taipei, Taiwan (R.O.C)
| | - Peiguo G Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA,
| |
Collapse
|
48
|
Wang L, Guo ZY, Zhang R, Xin B, Chen R, Zhao J, Wang T, Wen WH, Jia LT, Yao LB, Yang AG. Pseudogene OCT4-pg4 functions as a natural micro RNA sponge to regulate OCT4 expression by competing for miR-145 in hepatocellular carcinoma. Carcinogenesis 2013; 34:1773-1781. [PMID: 23615404 DOI: 10.1093/carcin/bgt139] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The POU transcription factor OCT4 is a pleiotropic regulator of gene expression in embryonic stem cells. Recent studies demonstrated that OCT4 is aberrantly expressed in multiple types of human cancer; however, the underlying molecular mechanism remains largely unknown. In this study, we report that OCT4-pg4, a pseudogene of OCT4, is abnormally activated in hepatocellular carcinoma (HCC). The expression level of OCT4-pg4 is positively correlated with that of OCT4, and both gene transcripts can be directly targeted by a tumor-suppressive micro RNA miR-145. We find that the non-coding RNA OCT4-pg4 is biologically active, as it can upregulate OCT4 protein level in HCC. Mechanistic analysis revealed that OCT4-pg4 functions as a natural micro RNA sponge to protect OCT4 transcript from being inhibited by miR-145. In addition, our study also showed that OCT4-pg4 can promote growth and tumorigenicity of HCC cells, thus exerting an oncogenic role in hepatocarcinogenesis. Furthermore, survival analysis suggests that high OCT4-pg4 level is significantly correlated with poor prognosis of HCC patients. Taken together, our finding adds a new layer of post-transcriptional regulation of OCT4 and sheds new light on the treatment of human HCC.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sarris M, Nikolaou K, Talianidis I. Context-specific regulation of cancer epigenomes by histone and transcription factor methylation. Oncogene 2013; 33:1207-17. [PMID: 23503463 DOI: 10.1038/onc.2013.87] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/01/2013] [Indexed: 12/18/2022]
Abstract
Altered expression or activity of histone lysine methylases and demethylases in cancer lead to aberrant chromatin modification patterns, which contribute to uncontrolled cell proliferation via cancer-specific deregulation of gene expression programs or the induction of genome instability. Several transcription factors that regulate growth-associated genes undergo lysine methylation, expanding the repertoire of regulatory targets modulated by histone-methylating enzymes during tumorigenesis. In certain specific tumor types or specific physiological conditions, these enzymes may trigger chromatin structure and/or transcription factor activity changes that result in opposite effects on cancer initiation or progression. The mechanisms of such context-specific dual functions and those involved in the crosstalk between factor and histone modifications are subject to extensive research, which is beginning to shed light into this novel level of complexity of cancer-related epigenetic pathways.
Collapse
Affiliation(s)
- M Sarris
- Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - K Nikolaou
- Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - I Talianidis
- Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| |
Collapse
|
50
|
Gonzalez-Perez A, Jene-Sanz A, Lopez-Bigas N. The mutational landscape of chromatin regulatory factors across 4,623 tumor samples. Genome Biol 2013; 14:r106. [PMID: 24063517 PMCID: PMC4054018 DOI: 10.1186/gb-2013-14-9-r106] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 09/24/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Chromatin regulatory factors are emerging as important genes in cancer development and are regarded as interesting candidates for novel targets for cancer treatment. However, we lack a comprehensive understanding of the role of this group of genes in different cancer types. RESULTS We have analyzed 4,623 tumor samples from thirteen anatomical sites to determine which chromatin regulatory factors are candidate drivers in these different sites. We identify 34 chromatin regulatory factors that are likely drivers in tumors from at least one site, all with relatively low mutational frequency. We also analyze the relative importance of mutations in this group of genes for the development of tumorigenesis in each site, and indifferent tumor types from the same site. CONCLUSIONS We find that, although tumors from all thirteen sites show mutations in likely driver chromatin regulatory factors, these are more prevalent in tumors arising from certain tissues. With the exception of hematopoietic, liver and kidney tumors, as a median, the mutated factors are less than one fifth of all mutated drivers across all sites analyzed. We also show that mutations in two of these genes, MLL and EP300, correlate with broad expression changes across cancer cell lines, thus presenting at least one mechanism through which these mutations could contribute to tumorigenesis in cells of the corresponding tissues.
Collapse
Affiliation(s)
- Abel Gonzalez-Perez
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona, Spain
| | - Alba Jene-Sanz
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona, Spain
| | - Nuria Lopez-Bigas
- Research Unit on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|