1
|
Xu HY, Wang YT, Yang HQ, Cao YY, Fan ZP. EZH2, via an association with KDM2B, modulates osteogenic differentiation of root apical papillary stem cells. World J Stem Cells 2025; 17:103482. [PMID: 40308879 PMCID: PMC12038457 DOI: 10.4252/wjsc.v17.i4.103482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/23/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Stem cells from apical papilla (SCAPs) represent promising candidates for bone regenerative therapies due to their osteogenic potential. However, enhancing their differentiation capacity remains a critical challenge. Enhancer of zeste homolog 2 (EZH2), a histone H3 lysine 27 methyltransferase, regulates osteogenesis through epigenetic mechanisms, but its role in SCAPs remains unclear. We hypothesized that EZH2 modulates SCAP osteogenic differentiation via interaction with lysine demethylase 2B (KDM2B), offering a target for therapeutic intervention. AIM To investigate the functional role and molecular mechanism of EZH2 in SCAP osteogenic differentiation. METHODS SCAPs were isolated from healthy human third molars (n = 6 donors). Osteogenic differentiation was assessed via Alizarin red staining and alkaline phosphatase assays. EZH2 overexpression/knockdown models were established using lentiviral vectors. Protein interactions were analyzed by co-immunoprecipitation, transcriptomic changes via microarray (Affymetrix platform), and chromatin binding by chromatin immunoprecipitation-quantitative polymerase chain reaction. In vivo bone formation was evaluated in immunodeficient mice (n = 8/group) transplanted with SCAPs-hydroxyapatite scaffolds. Data were analyzed using Student's t-test and ANOVA. RESULTS EZH2 overexpression increased osteogenic markers and mineralized nodule formation. In vivo, EZH2-overexpressing SCAPs generated 10% more bone/dentin-like tissue. Co-immunoprecipitation confirmed EZH2-KDM2B interaction, and peptide-mediated disruption of this binding enhanced osteogenesis. Transcriptome analysis identified 1648 differentially expressed genes (971 upregulated; 677 downregulated), with pathway enrichment in Wnt/β-catenin signaling. CONCLUSION EZH2 promotes SCAP osteogenesis via antagonistic interaction with KDM2B, and targeted disruption of this axis offers a translatable strategy for bone regeneration.
Collapse
Affiliation(s)
- Hui-Yue Xu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100050, China
| | - Yan-Tong Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100050, China
| | - Hao-Qing Yang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100050, China
| | - Yang-Yang Cao
- School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Zhi-Peng Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
2
|
Garg B, Khan S, Courelli AS, Panneerpandian P, Sheik Pran Babu D, Mose ES, Gulay KCM, Sharma S, Sood D, Wenzel AT, Martsinkovskiy A, Rajbhandari N, Patel J, Jaquish D, Esparza E, Jaque K, Aggarwal N, Lambies G, D’Ippolito A, Austgen K, Johnston B, Orlando DA, Jang GH, Gallinger S, Goodfellow E, Brodt P, Commisso C, Tamayo P, Mesirov JP, Tiriac H, Lowy AM. MICAL2 Promotes Pancreatic Cancer Growth and Metastasis. Cancer Res 2025; 85:1049-1063. [PMID: 39745352 PMCID: PMC11907191 DOI: 10.1158/0008-5472.can-24-0744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/11/2024] [Accepted: 12/18/2024] [Indexed: 02/23/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest solid cancers; thus, identifying more effective therapies is a major unmet need. In this study, we characterized the super-enhancer (SE) landscape of human PDAC to identify drivers of the disease that might be targetable. This analysis revealed MICAL2 as an SE-associated gene in human PDAC, which encodes the flavin monooxygenase enzyme that induces actin depolymerization and indirectly promotes serum response factor transcription by modulating the availability of serum response factor coactivators such as myocardin-related transcription factors (MRTF-A and MRTF-B). MICAL2 was overexpressed in PDAC, and high-MICAL2 expression correlated with poor patient prognosis. Transcriptional analysis revealed that MICAL2 upregulates KRAS and epithelial-mesenchymal transition signaling pathways, contributing to tumor growth and metastasis. In loss- and gain-of-function experiments in human and mouse PDAC cells, MICAL2 promoted both ERK1/2 and AKT activation. Consistent with its role in actin depolymerization and KRAS signaling, loss of MICAL2 also inhibited macropinocytosis. MICAL2, MRTF-A, and MRTF-B influenced PDAC cell proliferation and migration and promoted cell-cycle progression in vitro. Importantly, MICAL2 supported in vivo tumor growth and metastasis. Interestingly, MRTF-B, but not MRTF-A, phenocopied MICAL2-driven phenotypes in vivo. This study highlights the multiple ways in which MICAL2 affects PDAC biology and provides a foundation for future investigations into the potential of targeting MICAL2 for therapeutic intervention. Significance: Characterization of the epigenomic landscape of pancreatic cancer to identify early drivers of tumorigenesis uncovered MICAL2 as a super-enhancer-associated gene critical for tumor progression that represents a potential pharmacologic target.
Collapse
Affiliation(s)
- Bharti Garg
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Sohini Khan
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Asimina S. Courelli
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Ponmathi Panneerpandian
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Deepa Sheik Pran Babu
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Evangeline S. Mose
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Kevin Christian Montecillo Gulay
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Shweta Sharma
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Divya Sood
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Alexander T. Wenzel
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Alexei Martsinkovskiy
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Nirakar Rajbhandari
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jay Patel
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Dawn Jaquish
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Edgar Esparza
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Katelin Jaque
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Neetu Aggarwal
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Guillem Lambies
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | | | | | | | | | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Elliot Goodfellow
- Department of Surgery, McGill University, Montreal, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Pnina Brodt
- Department of Surgery, McGill University, Montreal, Canada
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
- Department of Medicine, McGill University, Montreal, Canada
| | - Cosimo Commisso
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Pablo Tamayo
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jill P. Mesirov
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Hervé Tiriac
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Andrew M. Lowy
- Division of Surgical Oncology, Department of Surgery, Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
3
|
Liu Y, Tan X, Wang L, Jin W, He Y, Yan Y, Hu K, Wang H, Xiang C, Hou M, Lai Y. Identification of the CeRNA axis of circ_0000006/miR-483-5p/KDM2B in the progression of aortic aneurysm to aorta dissection. BMC Cardiovasc Disord 2025; 25:141. [PMID: 40021969 PMCID: PMC11869725 DOI: 10.1186/s12872-025-04598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Aortic aneurysm (AA) and aortic dissection (AD) are serious cardiovascular disorders with a high risk of mortality. The molecular mechanisms underlying the progression from AA to AD are not well understood. This study aimed to identify the key circular RNA (circRNA)-microRNA (miRNA)-messenger RNA (mRNA) regulatory axis involved in this disease progression. METHODS CircRNA microarray, miRNA microarray, and mRNA sequencing were performed on plasma samples from healthy controls, AA patients, and AD patients. Bioinformatics analysis integrated the expression profiles to identify dysregulated circRNA-miRNA-mRNA networks. Key molecules were validated in vascular smooth muscle cells (VSMCs) and an AD mouse model. Cell proliferation, migration, and phenotypic transition assays were conducted after modulating the identified circRNA. The impact on AD progression was evaluated in mice upon circRNA knockdown. RESULTS A total of 12 circRNAs were found upregulated in AD compared to AA samples. miR-483-5p was downregulated while its targets KDM2B and circ_0000006 were upregulated in AD. Silencing circ_0000006 in VSMCs inhibited PDGF-induced phenotypic switching, proliferation, and migration by increasing miR-483-5p and decreasing KDM2B levels. In the AD mouse model, knockdown of circ_0000006 alleviated disease progression with similar molecular changes. CONCLUSION The study identified a novel circ_0000006/miR-483-5p/KDM2B axis dysregulated during AD progression. Targeting this axis, especially circ_0000006, could be a potential strategy to mitigate the transition from AA to AD by modulating VSMC phenotype and function.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Animals
- Humans
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/metabolism
- Disease Progression
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Cell Movement
- Disease Models, Animal
- Cell Proliferation
- Male
- Cells, Cultured
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/metabolism
- Case-Control Studies
- Female
- Middle Aged
- Mice, Inbred C57BL
- Signal Transduction
- Gene Regulatory Networks
- Gene Expression Regulation
- Mice
- Phenotype
- Aged
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Yong Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China.
- , No. 63, Culture Road, Nanchong, Sichuan, 637000, China.
| | - Xiong Tan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Liang Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Weitao Jin
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yangchen He
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yu Yan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Kai Hu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Hao Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Chaohu Xiang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Ming Hou
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yinglong Lai
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
4
|
Garg B, Khan S, Babu DS, Mose E, Gulay K, Sharma S, Sood D, Wenzel AT, Martsinkovskiy A, Patel J, Jaquish D, Lambies G, D'Ippolito A, Austgen K, Johnston B, Orlando D, Jang GH, Gallinger S, Goodfellow E, Brodt P, Commisso C, Tamayo P, Mesirov JP, Tiriac H, Lowy AM. MICAL2 Is a Super Enhancer Associated Gene that Promotes Pancreatic Cancer Growth and Metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600548. [PMID: 38979336 PMCID: PMC11230455 DOI: 10.1101/2024.06.26.600548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest solid cancers and thus identifying more effective therapies is a major unmet need. In this study we characterized the super enhancer (SE) landscape of human PDAC to identify novel, potentially targetable, drivers of the disease. Our analysis revealed that MICAL2 is a super enhancer-associated gene in human PDAC. MICAL2 is a flavin monooxygenase that induces actin depolymerization and indirectly promotes SRF transcription by modulating the availability of serum response factor coactivators myocardin related transcription factors (MRTF-A and MRTF-B). We found that MICAL2 is overexpressed in PDAC and correlates with poor patient prognosis. Transcriptional analysis revealed that MICAL2 upregulates KRAS and EMT signaling pathways, contributing to tumor growth and metastasis. In loss and gain of function experiments in human and mouse PDAC cells, we observed that MICAL2 promotes both ERK1/2 and AKT activation. Consistent with its role in actin depolymerization and KRAS signaling, loss of MICAL2 expression also inhibited macropinocytosis. Through in vitro phenotypic analyses, we show that MICAL2, MRTF-A and MRTF-B influence PDAC cell proliferation, migration and promote cell cycle progression. Importantly, we demonstrate that MICAL2 is essential for in vivo tumor growth and metastasis. Interestingly, we find that MRTF-B, but not MRTF-A, phenocopies MICAL2-driven phenotypes in vivo . This study highlights the multiple ways in which MICAL2 impacts PDAC biology and suggests that its inhibition may impede PDAC progression. Our results provide a foundation for future investigations into the role of MICAL2 in PDAC and its potential as a target for therapeutic intervention.
Collapse
|
5
|
Duan N, Hua Y, Yan X, He Y, Zeng T, Gong J, Fu Z, Li W, Yin Y. An Imbalance in Histone Modifiers Induces tRNA-Cys-GCA Overexpression and tRF-27 Accumulation by Attenuating Promoter H3K27me3 in Primary Trastuzumab-Resistant Breast Cancer. Cancers (Basel) 2024; 16:1118. [PMID: 38539453 PMCID: PMC10968641 DOI: 10.3390/cancers16061118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 11/11/2024] Open
Abstract
tRNA-derived fragments (tRFs) play crucial roles in cancer progression. Among them, tRF-27 has been identified as a key factor in promoting naïve trastuzumab resistance in HER2-positive breast cancer. However, the origin of tRF-27 remains uncertain. In this study, we propose that the upregulated expression of specific cysteine tRNAs may lead to the increased accumulation of tRF-27 in trastuzumab-resistant JIMT1 cells. Mechanistically, the reduced inhibitory H3K27me3 modification at the promoter regions of tRF-27-related tRNA genes in JIMT1 cells, potentially resulting from decreased EZH2 and increased KDM6A activity, may be a critical factor stimulating the transcriptional activity of these tRNA genes. Our research offers fresh insights into the mechanisms underlying elevated tRF-27 levels in trastuzumab-resistant breast cancer cells and suggests potential strategies to mitigate trastuzumab resistance in clinical treatments.
Collapse
Affiliation(s)
- Ningjun Duan
- Department of Oncology, First Affiliation Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.H.); (X.Y.); (Y.H.); (T.Z.); (J.G.); (Z.F.); (W.L.)
| | | | | | | | | | | | | | | | - Yongmei Yin
- Department of Oncology, First Affiliation Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.H.); (X.Y.); (Y.H.); (T.Z.); (J.G.); (Z.F.); (W.L.)
| |
Collapse
|
6
|
Suzuki T, Henshaw MJ, Yanagi T, Aoshima K. Current understanding of comparative pathology and prospective research approaches for canine hemangiosarcoma. Res Vet Sci 2024; 167:105120. [PMID: 38150941 DOI: 10.1016/j.rvsc.2023.105120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Hemangiosarcoma (HSA) is a malignant tumor originating from endothelial cells. HSA typically develops in dogs, but is rare in other animals, including humans. Although surgery and chemotherapy are conventional treatments for HSA, neither treatment can significantly improve patient prognosis. To develop novel and effective therapeutics, a deeper understanding of HSA pathogenesis must be acquired. However, the limited research tools for HSA have been unable to make a breakthrough; therefore, it is crucial to widely utilize or establish novel research tools such as patient-derived xenograft models, organoids, and chicken embryo xenograft models. The pathogenesis of the human counterpart of HSA, angiosarcoma (AS), also remains incompletely understood, preventing the extrapolation of findings from humans to dogs, unlike other diseases. In this review, we summarize the clinicopathological and morphological features of HSA, and then we discuss the current understanding of the molecular pathology of HSA. Finally, we highlight promising research tools that may accelerate HSA basic research toward developing novel therapeutics. We also briefly summarize AS to help researchers comprehend HSA from the perspective of comparative pathology.
Collapse
Affiliation(s)
- Tamami Suzuki
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Michael James Henshaw
- English Education Section, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan; Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan.
| |
Collapse
|
7
|
Asari Y, Yamazaki J, Thandar O, Suzuki T, Aoshima K, Takeuchi K, Kinoshita R, Kim S, Hosoya K, Ishizaki T, Kagawa Y, Jelinek J, Yokoyama S, Sasaki N, Ohta H, Nakamura K, Takiguchi M. Diverse genome-wide DNA methylation alterations in canine hepatocellular tumours. Vet Med Sci 2023; 9:2006-2014. [PMID: 37483163 PMCID: PMC10508506 DOI: 10.1002/vms3.1204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Canine hepatocellular tumours (HCTs) are common primary liver tumours. However, the exact mechanisms of tumourigenesis remain unclear. Although some genetic mutations have been reported, DNA methylation alterations in canine HCT have not been well studied. OBJECTIVES In this study, we aimed to analyse the DNA methylation status of canine HCT. METHODS Tissues from 33 hepatocellular carcinomas, 3 hepatocellular adenomas, 1 nodular hyperplasia, 21 non-tumour livers from the patients and normal livers from 5 healthy dogs were used. We analysed the DNA methylation levels of 72,367 cytosine-guanine dinucleotides (CpG sites) in all 63 samples. RESULTS AND CONCLUSIONS Although a large fraction of CpG sites that were highly methylated in the normal liver became hypomethylated in tumours from most patients, we also found some patients with less remarkable change or no change in DNA methylation. Hierarchical clustering analysis revealed that 32 of 37 tumour samples differed from normal livers, although the remaining 5 tumour livers fell into the same cluster as normal livers. In addition, the number of hypermethylated genes in tumour livers varied among tumour cases, suggesting various DNA methylation patterns in different tumour groups. However, patient and clinical parameters, such as age, were not associated with DNA methylation status. In conclusion, we found that HCTs undergo aberrant and diverse patterns of genome-wide DNA methylation compared with normal liver tissue, suggesting a complex epigenetic mechanism in canine HCT.
Collapse
Affiliation(s)
- Yu Asari
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Jumpei Yamazaki
- Veterinary Teaching HospitalGraduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- Translational Research Unit, Veterinary Teaching Hospital, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
| | - Oo Thandar
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Tamami Suzuki
- Laboratory of Comparative Pathology, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Keisuke Aoshima
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
- Laboratory of Comparative Pathology, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Kyosuke Takeuchi
- Veterinary Teaching HospitalGraduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ryohei Kinoshita
- Veterinary Teaching HospitalGraduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
| | - Sangho Kim
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
- Laboratory of Veterinary Surgery, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Kenji Hosoya
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
- Laboratory of Veterinary Surgery, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Teita Ishizaki
- Veterinary Teaching HospitalGraduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- Laboratory of Comparative Pathology, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- North LabSapporoJapan
| | | | | | - Shoko Yokoyama
- Veterinary Teaching HospitalGraduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- Translational Research Unit, Veterinary Teaching Hospital, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
- One Health Research Center, Cancer Research UnitHokkaido UniversitySapporoJapan
| | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Hiroshi Ohta
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Kensuke Nakamura
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Graduate School of Veterinary MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
8
|
Kapturska KM, Pawlak A. New molecular targets in canine hemangiosarcoma-Comparative review and future of the precision medicine. Vet Comp Oncol 2023; 21:357-377. [PMID: 37308243 DOI: 10.1111/vco.12917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
Human angiosarcoma and canine hemangiosarcoma reveal similarities not only in their aggressive clinical behaviour, but especially in molecular landscape and genetic alterations involved in tumorigenesis and metastasis formation. Currently, no satisfying treatment that allows for achieving long overall survival or even prolonged time to progression does not exist. Due to the progress that has been made in targeted therapies and precision medicine the basis for a new treatment design is to uncover mutations and their functions as possible targets to provide tailored drugs for individual cases. Whole exome or genome sequencing studies and immunohistochemistry brought in the last few years important discoveries and identified the most common mutations with probably crucial role in this tumour development. Also, despite a lack of mutation in some of the culprit genes, the cancerogenesis cause may be buried in main cellular pathways connected with proteins encoded by those genes and involving, for example, pathological angiogenesis. The aim of this review is to highlight the most promising molecular targets for precision oncology treatment from the veterinary perspective aided by the principles of comparative science. Some of the drugs are only undergoing laboratory in vitro studies and others entered the clinic in the management of other cancer types in humans, but those used in dogs with promising responses have been mentioned as priorities.
Collapse
Affiliation(s)
- Karolina Małgorzata Kapturska
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
- Veterinary Clinic NEOVET s.c. Hildebrand, Jelonek, Michalek-Salt, Wroclaw, Poland
| | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
9
|
JMJD family proteins in cancer and inflammation. Signal Transduct Target Ther 2022; 7:304. [PMID: 36050314 PMCID: PMC9434538 DOI: 10.1038/s41392-022-01145-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
The occurrence of cancer entails a series of genetic mutations that favor uncontrollable tumor growth. It is believed that various factors collectively contribute to cancer, and there is no one single explanation for tumorigenesis. Epigenetic changes such as the dysregulation of enzymes modifying DNA or histones are actively involved in oncogenesis and inflammatory response. The methylation of lysine residues on histone proteins represents a class of post-translational modifications. The human Jumonji C domain-containing (JMJD) protein family consists of more than 30 members. The JMJD proteins have long been identified with histone lysine demethylases (KDM) and histone arginine demethylases activities and thus could function as epigenetic modulators in physiological processes and diseases. Importantly, growing evidence has demonstrated the aberrant expression of JMJD proteins in cancer and inflammatory diseases, which might serve as an underlying mechanism for the initiation and progression of such diseases. Here, we discuss the role of key JMJD proteins in cancer and inflammation, including the intensively studied histone lysine demethylases, as well as the understudied group of JMJD members. In particular, we focused on epigenetic changes induced by each JMJD member and summarized recent research progress evaluating their therapeutic potential for the treatment of cancer and inflammatory diseases.
Collapse
|
10
|
Suzuki T, Aoshima K, Yamazaki J, Kobayashi A, Kimura T. Manipulating Histone Acetylation Leads to Antitumor Effects in Hemangiosarcoma Cells. Vet Comp Oncol 2022; 20:805-816. [PMID: 35568976 DOI: 10.1111/vco.12840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
Abstract
Canine hemangiosarcoma (HSA) is a malignant tumor derived from endothelial cells. No effective treatment has yet been developed because of the lack of understanding of its pathogenesis. Histone acetylation, an epigenetic modification, is highly associated with cancer pathogenesis. Manipulating histone acetylation by histone deacetylase inhibitors (HDACi) or bromodomain and extraterminal domain inhibitors (BETi) is one approach to treat various cancers. However, the role of histone acetylation in HSA remains unknown. This study aimed to investigate how histone acetylation functions in HSA pathogenesis using two HDACi, suberanilohydroxamic acid (SAHA) and valproic acid (VPA), and one BETi, JQ1, in vitro and in vivo. Histone acetylation levels were high in cell lines and heterogeneous in clinical cases. SAHA and JQ1 induced apoptosis in HSA cell lines. HSA cell lines treated with SAHA and VPA upregulated inflammatory-related genes and attracted macrophage cell line RAW264 cells, which suggests that SAHA and VPA can affect immune responses. JQ1 stimulated autophagy and inhibited the cell cycle in HSA cell lines. Finally, we demonstrated that JQ1 suppressed HSA tumor cell proliferation in vivo although SAHA and VPA did not affect tumor growth. These results suggest that BETi can be alternative drugs for HSA treatment. Although further research is required, our study indicated that dysregulation of histone acetylation is likely to be involved in HSA malignancy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tamami Suzuki
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Jumpei Yamazaki
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
11
|
Chen NP, Aretz J, Fässler R. CDK1-cyclin-B1-induced kindlin degradation drives focal adhesion disassembly at mitotic entry. Nat Cell Biol 2022; 24:723-736. [PMID: 35469017 PMCID: PMC9106588 DOI: 10.1038/s41556-022-00886-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/03/2022] [Indexed: 12/25/2022]
Abstract
The disassembly of integrin-containing focal adhesions (FAs) at mitotic entry is essential for cell rounding, mitotic retraction fibre formation, bipolar spindle positioning and chromosome segregation. The mechanism that drives FA disassembly at mitotic entry is unknown. Here, we show that the CDK1–cyclin B1 complex phosphorylates the integrin activator kindlin, which results in the recruitment of the cullin 9–FBXL10 ubiquitin ligase complex that mediates kindlin ubiquitination and degradation. This molecular pathway is essential for FA disassembly and cell rounding, as phospho-inhibitory mutations of the CDK1 motif prevent kindlin degradation, FA disassembly and mitotic cell rounding. Conversely, phospho-mimetic mutations promote kindlin degradation in interphase, accelerate mitotic cell rounding and impair mitotic retraction fibre formation. Despite the opposing effects on kindlin stability, both types of mutations cause severe mitotic spindle defects, apoptosis and aneuploidy. Thus, the exquisite regulation of kindlin levels at mitotic entry is essential for cells to progress accurately through mitosis. Chen et al. report that at mitotic entry, cyclin B1–CDK1 phosphorylates the focal adhesion protein kindlin to induce its proteasomal degradation and promote focal adhesion disassembly and mitotic rounding.
Collapse
Affiliation(s)
- Nan-Peng Chen
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Jonas Aretz
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
12
|
Gulay KCM, Aoshima K, Maekawa N, Suzuki T, Konnai S, Kobayashi A, Kimura T. Hemangiosarcoma cells induce M2 polarization and PD-L1 expression in macrophages. Sci Rep 2022; 12:2124. [PMID: 35136176 PMCID: PMC8826392 DOI: 10.1038/s41598-022-06203-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Hemangiosarcoma (HSA) is a malignant tumor derived from endothelial cells. Tumor-associated macrophages are one of the major components of tumor microenvironment and crucial for cancer development. The presence and function of macrophages in HSA have not been studied because there is no syngeneic model for HSA. In this study, we evaluated two mouse HSA cell lines and one immortalized mouse endothelial cell line for their usefulness as syngeneic models for canine HSA. Our results showed that the ISOS-1 cell line developed tumors with similar morphology to canine HSA. ISOS-1 cells highly expressed KDM2B and had similar KDM2B target expression patterns with canine HSA. Moreover, we determined that in both ISOS-1 and canine HSA tumors, macrophages were present as a major constituent of the tumor microenvironment. These macrophages were positive for CD204, an M2 macrophage marker, and express PD-L1, an immune checkpoint molecule. Canine HSA with macrophages expressing PD-L1 had a smaller number of T-cells in tumor tissues than tumors with PD-L1 negative macrophages. ISOS-1-conditioned medium could induce M2 polarization and PD-L1 expression in RAW264.7 mouse macrophage cell line and mouse peritoneal macrophages. These results show that ISOS-1 can be used as a syngenic model for canine HSA and suggest that macrophages play an important role in immune evasion in HSA. Using the syngeneic mouse model for canine HSA, we can further study the role of immune cells in the pathology of HSA.
Collapse
Affiliation(s)
- Kevin Christian M Gulay
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan.
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Tamami Suzuki
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Satoru Konnai
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo, Hokkaido, 060-0818, Japan
| |
Collapse
|
13
|
Epigenetics of Cutaneous Sarcoma. Int J Mol Sci 2021; 23:ijms23010422. [PMID: 35008848 PMCID: PMC8745302 DOI: 10.3390/ijms23010422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/25/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Epigenetic changes influence various physiological and pathological conditions in the human body. Recent advances in epigenetic studies of the skin have led to an appreciation of the importance of epigenetic modifications in skin diseases. Cutaneous sarcomas are intractable skin cancers, and there are no curative therapeutic options for the advanced forms of cutaneous sarcomas. In this review, we discuss the detailed molecular effects of epigenetic modifications on skin sarcomas, such as dermatofibrosarcoma protuberans, angiosarcoma, Kaposi's sarcoma, leiomyosarcoma, and liposarcoma. We also discuss the application of epigenetic-targeted therapy for skin sarcomas.
Collapse
|
14
|
Gulay KCM, Aoshima K, Kim S, Kitaguchi R, Kobayashi A, Kimura T. The expression of histone lysine demethylase 2B in canine hemangiosarcoma is associated with disease progression. Vet Comp Oncol 2021; 20:529-534. [DOI: 10.1111/vco.12796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Kevin Christian M. Gulay
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Sangho Kim
- Laboratory of Veterinary Surgery, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Ryusei Kitaguchi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine Hokkaido University Sapporo Japan
| |
Collapse
|