1
|
Ding X, Zhou X, Liu X, Lai Y, Yan W, Cheng Y, Hou A, Chen L, Sun L. Targeting Runx1 in Pathological Retinal Angiogenesis: A Potential Therapeutic Approach. Invest Ophthalmol Vis Sci 2025; 66:40. [PMID: 39946136 PMCID: PMC11827618 DOI: 10.1167/iovs.66.2.40] [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: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/16/2025] Open
Abstract
Purpose Neovascular eye diseases, such as proliferative diabetic retinopathy (PDR), wet age-related macular degeneration (wAMD) and retinopathy of prematurity (ROP), are major causes of vision loss and blindness worldwide. Our transcription factor motif enrichment analysis highlighted RUNX1 as a key regulator in the hypoxic response. The purpose of this study was to characterize how loss of Runx1 affects physiological and pathological retinal vasculature formation. Methods RNA-seq analysis and Transcription factor motif enrichment analysis were conducted in hypoxic and normoxic HUVECs. Conditional deletion of Runx1 in endothelial cells In mice was achieved using recombinase driver Cdh5-CreERT2. Vascular coverage, density, vessel progression, branchpoints, and sprout numbers was measured in retina of Runx1iECKO mice. The expression patterns, functions, and potential therapeutic value of RUNX1 were further explored with clinical samples, as well as in vivo and in vitro experiments. Bioinformatics and high-throughput sequencing were performed to identify potential target genes of Runx1. RT-qPCR and Western blot analyses were carried out to detect the changes of PI3-kinase/AKT/mTOR pathway. Results Loss of Runx1 in mice resulted in a reduction of the vascular coverage, density, vessel progression, branchpoints, and sprouts numbers of the retinal vascular network during its development. Notably, mature blood vessels remained unaffected by Runx1 inhibition. Upregulation of RUNX1 was observed in patients with PDR and ROP. RUNX1 Inhibition reduced endothelial cell proliferation, migration and tubule formation, leading to decreased pathological neovascularization, which is shown in oxygen-induced retinopathy. Mechanistically, in vitro experiments demonstrated that RUNX1 regulates EC angiogenesis through the PI3K/AKT/mTOR signaling pathway. Conclusions Runx1 is essential for physiological retinal vascularization. RUNX1 Inhibition may effectively decrease pathological neovascularization. Our findings suggest that targeting RUNX1 could be a promising therapeutic strategy for retinal neovascular disorders, preserving the integrity of mature blood vessels while selectively inhibiting neovascularization.
Collapse
Affiliation(s)
- Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaodi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xinyu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanting Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Wenjia Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yizhe Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Aohan Hou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Limei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Limei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
2
|
Ma X, Cao Y, Yang D, Dong Z, Wang X. Inhibition of RUNX1 slows the progression of pulmonary hypertension by targeting CBX5. BIOMOLECULES & BIOMEDICINE 2025; 25:472-481. [PMID: 39151099 PMCID: PMC11734815 DOI: 10.17305/bb.2024.10720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/20/2024] [Accepted: 07/20/2024] [Indexed: 08/18/2024]
Abstract
Pulmonary artery smooth muscle cell (PASMC) dysfunction is the central pathogenic mechanism in pulmonary hypertension (PH). This study explored the mechanism of action of RUNX1, a potential therapeutic target for PH, in PASMCs. A PH mouse model was used to investigate the impacts of RUNX1 knockdown on hemodynamics, right ventricular hypertrophy (RVH), and pulmonary artery remodeling (hematoxylin-eosin [H&E] staining). Isolated PASMCs were transfected with RUNX1- or chromobox 5 (CBX5)-related vectors and then subjected to cell function assays. Immunoprecipitation was used to detect molecular binding and ubiquitination. RUNX1 knockdown reduced right ventricular systolic pressure (RVSP), RVH, and pulmonary artery remodeling in mice with PH. Knockdown of RUNX1 or CBX5 suppressed proliferation, invasion, and migration and stimulated apoptosis in PASMCs under hypoxia. RUNX1 enhanced ubiquitin-specific protease 15 (USP15) promoter activity. USP15 bound to CBX5 and reduced CBX5 ubiquitination, thereby promoting CBX5 expression. CBX5 overexpression promoted the proliferation and movement of hypoxic PASMCs with reduced RUNX1 expression and decreased their apoptosis. In conclusion, RUNX1 knockdown inhibits USP15 transcription to promote the ubiquitination and degradation of CBX5, thereby alleviating PH in mice and reducing hypoxia-induced PASMC dysfunction.
Collapse
Affiliation(s)
- Ximiao Ma
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiothoracic Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
- Department of Cardiovascular Surgery, People’s Liberation Army General Hospital of Southern Theater Command, Guangzhou, China
| | - Yiqiu Cao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiac Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Dongpeng Yang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Zhu Dong
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xiaowu Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cardiovascular Surgery, People’s Liberation Army General Hospital of Southern Theater Command, Guangzhou, China
- Department of Cardiovascular Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Sun K, Chen M, Kong X, Hou W, Xu Z, Liu L. Cardiac-specific Suv39h1 knockout ameliorates high-fat diet induced diabetic cardiomyopathy via regulating Hmox1 transcription. Life Sci 2025; 360:123258. [PMID: 39580141 DOI: 10.1016/j.lfs.2024.123258] [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: 09/21/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
AIM Diabetic Cardiomyopathy (DCM), a common complication of Type 2 Diabetic Mellitus (T2DM), has been emerging as one of the leading causes of mortality in T2DM patients. During the past decade, although, clinical studies concerning DCM are increasing at an exponential rate, mechanisms underlying this disease still can't be clearly defined. Here, we aim to recognize the function of Suv39h1 in DCM and to explore underlying mechanisms during this disease, providing new insights into DCM and novel guide for clinical therapy development. MATERIALS AND METHODS We employed cardiac specific Suv39h1 knockout mice to reveal the role of Suv39h1 in high-fat diet induced DCM and using human cardiomyocyte line AC16 cells treated with Suv39h1 siRNA or inhibitor Chaetocin to further explore the mechanism during lipotoxicity condition. KEY FINDINGS Cardiac Suv39h1 knockout ameliorated manifestations of DCM, including cardiac function indexes, cardiomyocyte hypertrophy, interstitial fibrosis, along with improved metabolic disorder in mice. Further, interfering human AC16 cardiomyocytes with siSuv39h1 down-regulated lipotoxicity induced cardiac hypertrophy, inflammation, and fibrosis markers. Subsequent mRNA-seq using siSuv39h1 and SCR AC16 cells discovered a well-recognized cytoprotective, anti-oxidant, and anti-inflammation factor-Hmox1, prominently upregulated in Suv39h1 ablation cells versus SCR under lipotoxicity condition. ChIP assay revealed that Suv39h1 could bind to Hmox1 promoter and reversed by Chaetocin or small interfering RNA. SIGNIFICANCE These results suggested that the protective effects in DCM rendered by Suv39h1 ablation may work through activating Hmox1 transcription and protein function, providing new insights into pathogenesis of DCM and novel epigenetic target for clinical DCM therapies.
Collapse
Affiliation(s)
- Ke Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Rd, Nanjing 210023, China
| | - Maohui Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Rd, Nanjing 210023, China
| | - Xiangyu Kong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Rd, Nanjing 210023, China
| | - Weiyuan Hou
- Department of Cardiac Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Huai'an 223001, China
| | - Zhiwei Xu
- Department of Cardiac Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Huai'an 223001, China.
| | - Li Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Rd, Nanjing 210023, China.
| |
Collapse
|
4
|
Deforzh E, Kharel P, Zhang Y, Karelin A, El Khayari A, Ivanov P, Krichevsky AM. HOXDeRNA activates a cancerous transcription program and super enhancers via genome-wide binding. Mol Cell 2024; 84:3950-3966.e6. [PMID: 39383879 PMCID: PMC11490371 DOI: 10.1016/j.molcel.2024.09.018] [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: 01/09/2024] [Revised: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
The role of long non-coding RNAs (lncRNAs) in malignant cell transformation remains elusive. We previously identified an enhancer-associated lncRNA, LINC01116 (named HOXDeRNA), as a transformative factor converting human astrocytes into glioma-like cells. Employing a combination of CRISPR editing, chromatin isolation by RNA purification coupled with sequencing (ChIRP-seq), in situ mapping RNA-genome interactions (iMARGI), chromatin immunoprecipitation sequencing (ChIP-seq), HiC, and RNA/DNA FISH, we found that HOXDeRNA directly binds to CpG islands within the promoters of 35 glioma-specific transcription factors (TFs) distributed throughout the genome, including key stem cell TFs SOX2, OLIG2, POU3F2, and ASCL1, liberating them from PRC2 repression. This process requires a distinct RNA quadruplex structure and other segments of HOXDeRNA, interacting with EZH2 and CpGs, respectively. Subsequent transformation activates multiple oncogenes (e.g., EGFR, miR-21, and WEE1), driven by the SOX2- and OLIG2-dependent glioma-specific super enhancers. These results help reconstruct the sequence of events underlying the process of astrocyte transformation, highlighting HOXDeRNA's central genome-wide activity and suggesting a shared RNA-dependent mechanism in otherwise heterogeneous and multifactorial gliomagenesis.
Collapse
Affiliation(s)
- Evgeny Deforzh
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Prakash Kharel
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yanhong Zhang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anton Karelin
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Abdellatif El Khayari
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Institute of Biological Sciences (ISSB-P), UM6P Faculty of Medical Sciences, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Pavel Ivanov
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anna M Krichevsky
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Zheng M, Li H, Sun L, Cui S, Zhang W, Gao Y, Gao R. Calcipotriol abrogates TGF-β1/pSmad3-mediated collagen 1 synthesis in pancreatic stellate cells by downregulating RUNX1. Toxicol Appl Pharmacol 2024; 491:117078. [PMID: 39214171 DOI: 10.1016/j.taap.2024.117078] [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: 07/30/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
RUNX1 with CBFβ functions as an activator or repressor of critical mediators regulating cellular function. The aims of this study were to clarify the role of RUNX1 on regulating TGF-β1-induced COL1 synthesis and the mechanism of calcipotriol (Cal) on antagonizing COL1 synthesis in PSCs. RT-qPCR and Western Blot for determining the mRNAs and proteins of RUNX1 and COL1A1/1A2 in rat PSC line (RP-2 cell). Luciferase activities driven by RUNX1 or COL1A1 or COL1A2 promoter, co-immunoprecipitation and immunoblotting for pSmad3/RUNX1 or CBFβ/RUNX1, and knockdown or upregulation of Smad3 and RUNX1 were used. RUNX1 production was regulated by TGF-β1/pSmad3 signaling pathway in RP-2 cells. RUNX1 formed a coactivator with CBFβ in TGF-β1-treated RP-2 cells to regulate the transcriptions of COL1A1/1A2 mRNAs under a fashion of pSmad3/RUNX1/CBFβ complex. However, Cal effectively abrogated the levels of COL1A1/1A2 transcripts in TGF-β1-treated RP-2 cells by downregulating RUNX1 production and hindering the formation of pSmad3/RUNX1/CBFβ complexes. This study suggests that RUNX1 may be a promising antifibrotic target for the treatment of chronic pancreatitis.
Collapse
Affiliation(s)
- Meifang Zheng
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China
| | - Hongyan Li
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China
| | - Li Sun
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China
| | - Shiyuan Cui
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China
| | - Wei Zhang
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China
| | - Yanhang Gao
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.
| | - Runping Gao
- Department of Hepatic biliary Pancreatic Medicine, First Hospital of Jilin University, Changchun, China; Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
6
|
Romero-Reyes J, Vázquez-Martínez ER, Silva CC, Molina-Hernández A, Díaz NF, Camacho-Arroyo I. Navigating glioblastoma complexity: the interplay of neurotransmitters and chromatin. Mol Biol Rep 2024; 51:912. [PMID: 39153092 PMCID: PMC11330389 DOI: 10.1007/s11033-024-09853-3] [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: 05/03/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Glioblastoma is the most aggressive brain cancer with an unfavorable prognosis for patient survival. Glioma stem cells, a subpopulation of cancer cells, drive tumor initiation, self-renewal, and resistance to therapy and, together with the microenvironment, play a crucial role in glioblastoma maintenance and progression. Neurotransmitters such as noradrenaline, dopamine, and serotonin have contrasting effects on glioblastoma development, stimulating or inhibiting its progression depending on the cellular context and through their action on glioma stem cells, perhaps changing the epigenetic landscape. Recent studies have revealed that serotonin and dopamine induce chromatin modifications related to transcriptional plasticity in the mammalian brain and possibly in glioblastoma; however, this topic still needs to be explored because of its potential implications for glioblastoma treatment. Also, it is essential to consider that neurotransmitters' effects depend on the tumor's microenvironment since it can significantly influence the response and behavior of cancer cells. This review examines the possible role of neurotransmitters as regulators of glioblastoma development, focusing on their impact on the chromatin of glioma stem cells.
Collapse
Affiliation(s)
- Jessica Romero-Reyes
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México
| | - Carlos-Camilo Silva
- Chronobiology of Reproduction Research Lab. Biology of Reproduction Research Unit, Carrera de Biología, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City, México
| | - Anayansi Molina-Hernández
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Mexico City, México
| | - Néstor Fabián Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Mexico City, México.
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, México.
| |
Collapse
|
7
|
Ji Q, Tu Z, Liu J, Zhou Z, Li F, Zhu X, Huang K. RUNX1-PDIA5 Axis Promotes Malignant Progression of Glioblastoma by Regulating CCAR1 Protein Expression. Int J Biol Sci 2024; 20:4364-4381. [PMID: 39247813 PMCID: PMC11379074 DOI: 10.7150/ijbs.92595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 07/28/2024] [Indexed: 09/10/2024] Open
Abstract
PDIA5 is responsible for modification of disulfide bonds of proteins. However, its impact on the malignant progression of glioblastoma multiforme (GBM) remains unknown. We analyzed the expression and prognostic significance of PDIA5 in cohorts of GBM and clinical samples. The PDIA5 protein was significantly overexpressed in GBM tissues, and higher expression of PDIA5 was statistically associated with a worse prognosis in patients with GBM. Transcriptional data from PDIA5 knockdown GBM cells revealed that downstream regulatory genes of PDIA5 were enriched in malignant regulatory pathways and PDIA5 enhanced the proliferative and invasive abilities of GBM cells. By constructing a PDIA5 CXXC motif mutant plasmid, we found CCAR1 was the vital downstream factor of PDIA5 in regulating GBM malignancy in vitro and in vivo. Additionally, RUNX1 bound to the promoter region of PDIA5 and regulated gene transcription, leading to activation of the PDIA5/CCAR1 regulatory axis in GBM. The RUNX1/PDIA5/CCAR1 axis significantly influenced the malignant behavior of GBM cells. In conclusion, this study comprehensively elucidates the crucial role of PDIA5 in the malignant progression of GBM. Downregulating PDIA5 can mitigate the malignant biological behavior of GBM both in vitro and in vivo, potentially improving the efficacy of treatment for clinical patients with GBM.
Collapse
Affiliation(s)
- Qiankun Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan 466000, P. R. China
| | - Zewei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
| | - Junzhe Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
| | - Zhihong Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
| | - Fengze Li
- Queen Mary School, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, P. R. China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, P. R. China
| |
Collapse
|
8
|
Niu W, Yu H, Fan X, Li S, Sun S, Gong M, Zhang S, Bi W, Chen X, Fang Z. Development of stemness-related signature to optimize prognosis prediction and identify XMD8-85 as a novel therapeutic compound for glioma. Cell Signal 2024; 120:111231. [PMID: 38768760 DOI: 10.1016/j.cellsig.2024.111231] [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: 03/10/2024] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Glioma is a highly invasive and aggressive type of brain cancer with poor treatment response. Stemness-related transcription factors form a regulatory network that sustains the malignant phenotype of gliomas. We conducted an integrated analysis of stemness-related transcription factors using The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) datasets, established the characteristics of stemness-related transcription factors, including Octamer-Binding Protein 4 (OCT4), Meis Homeobox 1 (MEIS1), E2F Transcription Factor 1 (E2F1), Transcription Factor CP2 Like 1 (TFCP2L1), and RUNX Family Transcription Factor 1 (RUNX1). The characteristic of stemness-related transcription factors was identified as an independent prognostic factor for glioma patients. Patients in the high-risk group have a worse prognosis than those in the low-risk group. The glioma microenvironment in the high-risk group exhibited a more active immune status. Single-cell level analysis revealed that stem cell-like cells exhibited stronger intercellular communication than glioma cells. Meanwhile, patients in different risk stratification exhibited varying sensitivities to immunotherapy and small molecule drug therapy. XMD8-85 was more effective in the high-risk group, and its antitumor effects were validated both in vivo and in vitro. Our results indicate that this prognostic feature will assist clinicians in predicting the prognosis of glioma patients, guiding immunotherapy and personalized treatment, as well as the potential clinical application of XMD8-85 in glioma treatment, and helping to develop effective treatment strategies.
Collapse
Affiliation(s)
- Wanxiang Niu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China
| | - Huihan Yu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; School of Basic Medical Sciences, Anhui Medical University, No. 81, Meishan Road, Hefei 230032, Anhui, China
| | - Xiaoqing Fan
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China
| | - Shuyang Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; School of Basic Medical Sciences, Anhui Medical University, No. 81, Meishan Road, Hefei 230032, Anhui, China
| | - Suling Sun
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China
| | - Meiting Gong
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; School of Basic Medical Sciences, Anhui Medical University, No. 81, Meishan Road, Hefei 230032, Anhui, China
| | - Siyu Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China
| | - Wenxu Bi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China
| | - Xueran Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China.
| | - Zhiyou Fang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, 230031 Hefei, Anhui, China; Science Island Branch, Graduate School of University of Science and Technology of China, No. 96, Jin Zhai Road, 230026 Hefei, Anhui, China.
| |
Collapse
|
9
|
Xu C, Hou P, Li X, Xiao M, Zhang Z, Li Z, Xu J, Liu G, Tan Y, Fang C. Comprehensive understanding of glioblastoma molecular phenotypes: classification, characteristics, and transition. Cancer Biol Med 2024; 21:j.issn.2095-3941.2023.0510. [PMID: 38712813 PMCID: PMC11131044 DOI: 10.20892/j.issn.2095-3941.2023.0510] [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: 12/27/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Among central nervous system-associated malignancies, glioblastoma (GBM) is the most common and has the highest mortality rate. The high heterogeneity of GBM cell types and the complex tumor microenvironment frequently lead to tumor recurrence and sudden relapse in patients treated with temozolomide. In precision medicine, research on GBM treatment is increasingly focusing on molecular subtyping to precisely characterize the cellular and molecular heterogeneity, as well as the refractory nature of GBM toward therapy. Deep understanding of the different molecular expression patterns of GBM subtypes is critical. Researchers have recently proposed tetra fractional or tripartite methods for detecting GBM molecular subtypes. The various molecular subtypes of GBM show significant differences in gene expression patterns and biological behaviors. These subtypes also exhibit high plasticity in their regulatory pathways, oncogene expression, tumor microenvironment alterations, and differential responses to standard therapy. Herein, we summarize the current molecular typing scheme of GBM and the major molecular/genetic characteristics of each subtype. Furthermore, we review the mesenchymal transition mechanisms of GBM under various regulators.
Collapse
Affiliation(s)
- Can Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Pengyu Hou
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Xiang Li
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Menglin Xiao
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Ziqi Zhang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Ziru Li
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
| | - Jianglong Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Guoming Liu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| | - Yanli Tan
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
- School of Basic Medical Sciences, Hebei University, Baoding 07100, China
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding 07100, China
| | - Chuan Fang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 07100, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding 071000, China
| |
Collapse
|
10
|
Yue Q, Wang Z, Shen Y, Lan Y, Zhong X, Luo X, Yang T, Zhang M, Zuo B, Zeng T, Lu J, Wang Y, Liu B, Guo H. Histone H3K9 Lactylation Confers Temozolomide Resistance in Glioblastoma via LUC7L2-Mediated MLH1 Intron Retention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309290. [PMID: 38477507 PMCID: PMC11109612 DOI: 10.1002/advs.202309290] [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: 11/30/2023] [Revised: 02/03/2024] [Indexed: 03/14/2024]
Abstract
Temozolomide (TMZ) resistance remains the major obstacle in the treatment of glioblastoma (GBM). Lactylation is a novel post-translational modification that is involved in various tumors. However, whether lactylation plays a role in GBM TMZ resistance remains unclear. Here it is found that histone H3K9 lactylation (H3K9la) confers TMZ resistance in GBM via LUC7L2-mediated intron 7 retention of MLH1. Mechanistically, lactylation is upregulated in recurrent GBM tissues and TMZ-resistant cells, and is mainly concentrated in histone H3K9. Combined multi-omics analysis, including CUT&Tag, SLAM-seq, and RNA-seq, reveals that H3K9 lactylation is significantly enriched in the LUC7L2 promoter and activates LUC7L2 transcription to promote its expression. LUC7L2 mediates intron 7 retention of MLH1 to reduce MLH1 expression, and thereby inhibit mismatch repair (MMR), ultimately leading to GBM TMZ resistance. Of note, it is identified that a clinical anti-epileptic drug, stiripentol, which can cross the blood-brain barrier and inhibit lactate dehydrogenase A/B (LDHA/B) activity, acts as a lactylation inhibitor and renders GBM cells more sensitive to TMZ in vitro and in vivo. These findings not only shed light on the mechanism of lactylation in GBM TMZ resistance but also provide a potential combined therapeutic strategy for clinical GBM treatment.
Collapse
Affiliation(s)
- Qu Yue
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Zhao Wang
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Yixiong Shen
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Yufei Lan
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Xiangyang Zhong
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Xin Luo
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Tao Yang
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Manqing Zhang
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Boming Zuo
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Tianci Zeng
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Jiankun Lu
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Yuankai Wang
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Boyang Liu
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Hongbo Guo
- Department of Neurosurgery CenterThe National Key Clinical SpecialtyThe Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular DiseaseGuangdong Provincial Key Laboratory on Brain Function Repair and RegenerationThe Neurosurgery Institute of Guangdong ProvinceZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| |
Collapse
|
11
|
Zhao J, Cui X, Zhan Q, Zhang K, Su D, Yang S, Hong B, Wang Q, Ju J, Cheng C, Li C, Wan C, Wang Y, Zhou J, Kang C. CRISPR-Cas9 library screening combined with an exosome-targeted delivery system addresses tumorigenesis/TMZ resistance in the mesenchymal subtype of glioblastoma. Theranostics 2024; 14:2835-2855. [PMID: 38773970 PMCID: PMC11103500 DOI: 10.7150/thno.92703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/20/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: The large-scale genomic analysis classifies glioblastoma (GBM) into three major subtypes, including classical (CL), proneural (PN), and mesenchymal (MES) subtypes. Each of these subtypes exhibits a varying degree of sensitivity to the temozolomide (TMZ) treatment, while the prognosis corresponds to the molecular and genetic characteristics of the tumor cell type. Tumors with MES features are predominantly characterized by the NF1 deletion/alteration, leading to sustained activation of the RAS and PI3K-AKT signaling pathways in GBM and tend to acquire drug resistance, resulting in the worst prognosis compared to other subtypes (PN and CL). Here, we used the CRISPR/Cas9 library screening technique to detect TMZ-related gene targets that might play roles in acquiring drug resistance, using overexpressed KRAS-G12C mutant GBM cell lines. The study identified a key therapeutic strategy to address the chemoresistance against the MES subtype of GBM. Methods: The CRISPR-Cas9 library screening was used to discover genes associated with TMZ resistance in the U87-KRAS (U87-MG which is overexpressed KRAS-G12C mutant) cells. The patient-derived GBM primary cell line TBD0220 was used for experimental validations in vivo and in vitro. Chromatin isolation by RNA purification (ChIRP) and chromatin immunoprecipitation (ChIP) assays were used to elucidate the silencing mechanism of tumor suppressor genes in the MES-GBM subtype. The small-molecule inhibitor EPIC-0412 was obtained through high-throughput screening. Transmission electron microscopy (TEM) was used to characterize the exosomes (Exos) secreted by GBM cells after TMZ treatment. Blood-derived Exos-based targeted delivery of siRNA, TMZ, and EPIC-0412 was optimized to tailor personalized therapy in vivo. Results: Using the genome-wide CRISPR-Cas9 library screening, we found that the ERBIN gene could be epigenetically regulated in the U87-KRAS cells. ERBIN overexpression inhibited the RAS signaling and downstream proliferation and invasion effects of GBM tumor cells. EPIC-0412 treatment inhibited tumor proliferation and EMT progression by upregulating the ERBIN expression both in vitro and in vivo. Genome-wide CRISPR-Cas9 screening also identified RASGRP1(Ras guanine nucleotide-releasing protein 1) and VPS28(Vacuolar protein sorting-associated protein 28) genes as synthetically lethal in response to TMZ treatment in the U87-KRAS cells. We found that RASGRP1 activated the RAS-mediated DDR pathway by promoting the RAS-GTP transformation. VPS28 promoted the Exos secretion and decreased intracellular TMZ concentration in GBM cells. The targeted Exos delivery system encapsulating drugs and siRNAs together showed a powerful therapeutic effect against GBM in vivo. Conclusions: We demonstrate a new mechanism by which ERBIN is epigenetically silenced by the RAS signaling in the MES subtype of GBM. Restoration of the ERBIN expression with EPIC-0412 significantly inhibits the RAS signaling downstream. RASGRP1 and VPS28 genes are associated with the promotion of TMZ resistance through RAS-GDP to RAS-GTP transformation and TMZ efflux, as well. A quadruple combination therapy based on a targeted Exos delivery system demonstrated significantly reduced tumor burden in vivo. Therefore, our study provides new insights and therapeutic approaches for regulating tumor progression and TMZ resistance in the MES-GBM subtype.
Collapse
Affiliation(s)
- Jixing Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Xiaoteng Cui
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qi Zhan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Kailiang Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
| | - Dongyuan Su
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Shixue Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Biao Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qixue Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Jiasheng Ju
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chunchao Cheng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunfei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Junhu Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| |
Collapse
|
12
|
BV H, Jolly MK. Proneural-mesenchymal antagonism dominates the patterns of phenotypic heterogeneity in glioblastoma. iScience 2024; 27:109184. [PMID: 38433919 PMCID: PMC10905000 DOI: 10.1016/j.isci.2024.109184] [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: 11/28/2023] [Revised: 12/31/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024] Open
Abstract
The aggressive nature of glioblastoma (GBM) - one of the deadliest forms of brain tumors - is majorly attributed to underlying phenotypic heterogeneity. Early attempts to classify this heterogeneity at a transcriptomic level in TCGA GBM cohort proposed the existence of four distinct molecular subtypes: Proneural, Neural, Classical, and Mesenchymal. Further, a single-cell RNA sequencing (scRNA-seq) analysis of primary tumors also reported similar four subtypes mimicking neurodevelopmental lineages. However, it remains unclear whether these four subtypes identified via bulk and single-cell transcriptomics are mutually exclusive or not. Here, we perform pairwise correlations among individual genes and gene signatures corresponding to these proposed subtypes and show that the subtypes are not distinctly mutually antagonistic in either TCGA or scRNA-seq data. We observed that the proneural (or neural progenitor-like)-mesenchymal axis is the most prominent antagonistic pair, with the other two subtypes lying on this spectrum. These results are reinforced through a meta-analysis of over 100 single-cell and bulk transcriptomic datasets as well as in terms of functional association with metabolic switching, cell cycle, and immune evasion pathways. Finally, this proneural-mesenchymal antagonistic trend percolates to the association of relevant transcription factors with patient survival. These results suggest rethinking GBM phenotypic characterization for more effective therapeutic targeting efforts.
Collapse
Affiliation(s)
- Harshavardhan BV
- IISc Mathematics Initiative, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
13
|
Cai R, Shan Y, Du F, Miao Z, Zhu L, Hang L, Xiao L, Wang Z. Injectable hydrogels as promising in situ therapeutic platform for cartilage tissue engineering. Int J Biol Macromol 2024; 261:129537. [PMID: 38278383 DOI: 10.1016/j.ijbiomac.2024.129537] [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: 11/06/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Injectable hydrogels are gaining prominence as a biocompatible, minimally invasive, and adaptable platform for cartilage tissue engineering. Commencing with their synthesis, this review accentuates the tailored matrix formulations and cross-linking techniques essential for fostering three-dimensional cell culture and melding with complex tissue structures. Subsequently, it spotlights the hydrogels' enhanced properties, highlighting their augmented functionalities and broadened scope in cartilage tissue repair applications. Furthermore, future perspectives are advocated, urging continuous innovation and exploration to surmount existing challenges and harness the full clinical potential of hydrogels in regenerative medicine. Such advancements are crucial for validating the long-term efficacy and safety of hydrogels, positioning them as a promising direction in regenerative medicine to address cartilage-related ailments.
Collapse
Affiliation(s)
- Rong Cai
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Yisi Shan
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Fengyi Du
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212013, China
| | - Zhiwei Miao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Like Zhu
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Li Hang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China
| | - Long Xiao
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| | - Zhirong Wang
- Translational Medical Innovation Center, The Affiliated Zhangjiagang TCM Hospital of Yangzhou University, Zhangjiagang 215600, Jiangsu, China.
| |
Collapse
|
14
|
Fedele M, Cerchia L, Battista S. Subtype Transdifferentiation in Human Cancer: The Power of Tissue Plasticity in Tumor Progression. Cells 2024; 13:350. [PMID: 38391963 PMCID: PMC10887430 DOI: 10.3390/cells13040350] [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: 01/19/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
The classification of tumors into subtypes, characterized by phenotypes determined by specific differentiation pathways, aids diagnosis and directs therapy towards targeted approaches. However, with the advent and explosion of next-generation sequencing, cancer phenotypes are turning out to be far more heterogenous than initially thought, and the classification is continually being updated to include more subtypes. Tumors are indeed highly dynamic, and they can evolve and undergo various changes in their characteristics during disease progression. The picture becomes even more complex when the tumor responds to a therapy. In all these cases, cancer cells acquire the ability to transdifferentiate, changing subtype, and adapt to changing microenvironments. These modifications affect the tumor's growth rate, invasiveness, response to treatment, and overall clinical behavior. Studying tumor subtype transitions is crucial for understanding tumor evolution, predicting disease outcomes, and developing personalized treatment strategies. We discuss this emerging hallmark of cancer and the molecular mechanisms involved at the crossroads between tumor cells and their microenvironment, focusing on four different human cancers in which tissue plasticity causes a subtype switch: breast cancer, prostate cancer, glioblastoma, and pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Monica Fedele
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council—CNR, 80131 Naples, Italy; (L.C.); (S.B.)
| | | | | |
Collapse
|
15
|
Ariffin NS. Increased RUNX1 mutations in breast cancer disease progression. Pathol Res Pract 2024; 254:155076. [PMID: 38219493 DOI: 10.1016/j.prp.2023.155076] [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: 09/07/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
Despite advances in screening, therapy and surveillance, breast cancer remains threatening to women. Worst, patients suffer from side effects of treatments and cancer cells become resistant. The emergence of RUNX1 in breast cancer has put it in a spotlight due to its roles in the disease progression. It also plays important roles in normal mammary glands such as for cell growth, proliferation, migration and stemness. However, mutations in the RUNX1 gene have changed the regulation of these phenotypes and the full spectrum of its implications in breast cancer patients is unknown. In this study therefore, the pattern of RUNX1 mutations in breast cancer patients was examined to understand its fundamental impacts on the disease. The perturbation of RUNX1 and its mutations in breast cancer was elucidated through different studies reported in cBioPortal in the past ten years. From our analyses, the majority of RUNX1 mutations were found in the primary breast cancer, with women constituted most of the mutations, especially on the left side of the breast. Similarly, increased number of mutations was observed in ER-positive breast cancer patients and this was also the case at the early stage of the disease development. The level of RUNX1 mutations also increased gradually as patients got older and the peak was highest in the patients of 60-70 years old. Altogether, these data indicated that the mutated RUNX1 gene contributed to the progression of breast cancer and understanding of its regulatory mechanisms is crucial to therapeutically target this gene in the future.
Collapse
Affiliation(s)
- Nur Syamimi Ariffin
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
16
|
Yu X, Zhao P, Luo Q, Wu X, Wang Y, Nan Y, Liu S, Gao W, Li B, Liu Z, Cui Z. RUNX1-IT1 acts as a scaffold of STAT1 and NuRD complex to promote ROS-mediated NF-κB activation and ovarian cancer progression. Oncogene 2024; 43:420-433. [PMID: 38092960 DOI: 10.1038/s41388-023-02910-4] [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: 06/14/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 02/04/2024]
Abstract
Dysregulated expression of long-stranded non-coding RNAs is strongly associated with carcinogenesis. However, the precise mechanisms underlying their involvement in ovarian cancer pathogenesis remain poorly defined. Here, we found that lncRNA RUNX1-IT1 plays a crucial role in the progression of ovarian cancer. Patients with high RUNX1-IT1 expression had shorter survival and poorer outcomes. Notably, knockdown of RUNX1-IT1 suppressed the proliferation, migration and invasion of ovarian cancer cells in vitro, and reduced the formation of peritoneum metastasis in vivo. Mechanistically, RUNX1-IT1 bound to HDAC1, the core component of the NuRD complex, and STAT1, acting as a molecular scaffold of the STAT1 and NuRD complex to regulate intracellular reactive oxygen homeostasis by altering the histone modification status of downstream targets including GPX1. Consequently, RUNX1-IT1 activated NF-κB signaling and altered the biology of ovarian cancer cells. In conclusion, our findings demonstrate that RUNX1-IT1 promotes ovarian malignancy and suggest that targeting RUNX1-IT1 represents a promising therapeutic strategy for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiao Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Pengfei Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qingyu Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Xiaowei Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Yating Wang
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yabing Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shi Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenyan Gao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bin Li
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhumei Cui
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
17
|
Cui X, Huo D, Wang Q, Wang Y, Liu X, Zhao K, You Y, Zhang J, Kang C. RUNX1/NPM1/H3K4me3 complex contributes to extracellular matrix remodeling via enhancing FOSL2 transcriptional activation in glioblastoma. Cell Death Dis 2024; 15:98. [PMID: 38286983 PMCID: PMC10825180 DOI: 10.1038/s41419-024-06481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 01/31/2024]
Abstract
Extracellular matrix (ECM) remodeling has been implicated in the tumor malignant progression and immune escape in glioblastoma (GBM). Runt-related transcription factor 1 (RUNX1) is a vital transcriptional factor for promoting tumorigenesis and invasion in mesenchymal subtype of GBM. But the correlation between RUNX1 and ECM genes expression and regulatory mechanism of RUNX1 on ECM genes expression remain poorly understood to date. In this study, by using integral analysis of chromatin immunoprecipitation-sequencing and RNA sequencing, we reported that RUNX1 positively regulated the expression of various ECM-related genes, including Fibronectin 1 (FN1), Collagen type IV alpha 1 chain (COL4A1), and Lumican (LUM), in GBM. Mechanistically, we demonstrated that RUNX1 interacted with Nucleophosmin 1 (NPM1) to maintain the chromatin accessibility and facilitate FOS Like 2, AP-1 Transcription Factor Subunit (FOSL2)-mediated transcriptional activation of ECM-related genes, which was independent of RUNX1's transcriptional function. ECM remodeling driven by RUNX1 promoted immunosuppressive microenvironment in GBM. In conclusion, this study provides a novel mechanism of RUNX1 binding to NPM1 in driving the ECM remodeling and GBM progression.
Collapse
Affiliation(s)
- Xiaoteng Cui
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Dawei Huo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Laboratory, Institute of Hematology, Zhejiang University, Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310003, China
| | - Qixue Wang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Yunfei Wang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Nankai University, Tianjin, 300350, China
| | - Kai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China.
| | - Chunsheng Kang
- Lab of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.
| |
Collapse
|
18
|
Seaton G, Smith H, Brancale A, Westwell AD, Clarkson R. Multifaceted roles for BCL3 in cancer: a proto-oncogene comes of age. Mol Cancer 2024; 23:7. [PMID: 38195591 PMCID: PMC10775530 DOI: 10.1186/s12943-023-01922-8] [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: 10/27/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
In the early 1990's a group of unrelated genes were identified from the sites of recurring translocations in B-cell lymphomas. Despite sharing the nomenclature 'Bcl', and an association with blood-borne cancer, these genes have unrelated functions. Of these genes, BCL2 is best known as a key cancer target involved in the regulation of caspases and other cell viability mechanisms. BCL3 on the other hand was originally identified as a non-canonical regulator of NF-kB transcription factor pathways - a signaling mechanism associated with important cell outcomes including many of the hallmarks of cancer. Most of the early investigations into BCL3 function have since focused on its role in NF-kB mediated cell proliferation, inflammation/immunity and cancer. However, recent evidence is coming to light that this protein directly interacts with and modulates a number of other signaling pathways including DNA damage repair, WNT/β-catenin, AKT, TGFβ/SMAD3 and STAT3 - all of which have key roles in cancer development, metastatic progression and treatment of solid tumours. Here we review the direct evidence demonstrating BCL3's central role in a transcriptional network of signaling pathways that modulate cancer biology and treatment response in a range of solid tumour types and propose common mechanisms of action of BCL3 which may be exploited in the future to target its oncogenic effects for patient benefit.
Collapse
Affiliation(s)
- Gillian Seaton
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Hannah Smith
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Andrea Brancale
- UCT Prague, Technická 5, 166 28, 6 - Dejvice, IČO: 60461337, Prague, Czech Republic
| | - Andrew D Westwell
- Cardiff University School of Pharmacy and Pharmaceutical Sciences, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Richard Clarkson
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
19
|
Lettieri S, Bertuccio FR, del Frate L, Perrotta F, Corsico AG, Stella GM. The Plastic Interplay between Lung Regeneration Phenomena and Fibrotic Evolution: Current Challenges and Novel Therapeutic Perspectives. Int J Mol Sci 2023; 25:547. [PMID: 38203718 PMCID: PMC10779349 DOI: 10.3390/ijms25010547] [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: 12/13/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Interstitial lung diseases (ILDs) are a heterogeneous group of pulmonary disorders characterized by variable degrees of inflammation, interstitial thickening, and fibrosis leading to distortion of the pulmonary architecture and gas exchange impairment. Among them, idiopathic pulmonary fibrosis (IPF) displays the worst prognosis. The only therapeutic options consist of the two antifibrotic drugs, pirfenidone and nintedanib, which limit fibrosis progression but do not reverse the lung damage. The shift of the pathogenetic paradigm from inflammatory disease to epithelium-derived disease has definitively established the primary role of type II alveolar cells, which lose their epithelial phenotype and acquire a mesenchymal phenotype with production of collagen and extracellular matrix (EMC) deposition. Some predisposing environmental and genetic factors (e.g., smoke, pollution, gastroesophageal reflux, variants of telomere and surfactant genes) leading to accelerated senescence set a pro-fibrogentic microenvironment and contribute to the loss of regenerative properties of type II epithelial cells in response to pathogenic noxae. This review provides a complete overview of the different pathogenetic mechanisms leading to the development of IPF. Then, we summarize the currently approved therapies and the main clinical trials ongoing. Finally, we explore the potentialities offered by agents not only interfering with the processes of fibrosis but also restoring the physiological properties of alveolar regeneration, with a particular focus on potentialities and concerns about cell therapies based on mesenchymal stem cells (MSCs), whose anti-inflammatory and immunomodulant properties have been exploited in other fibrotic diseases, such as graft versus host disease (GVHD) and COVID-19-related ARDS.
Collapse
Affiliation(s)
- Sara Lettieri
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Francesco R. Bertuccio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Lucia del Frate
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fabio Perrotta
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, 80055 Naples, Italy;
| | - Angelo G. Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia M. Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy; (S.L.); (F.R.B.); (L.d.F.); (A.G.C.)
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
20
|
Brandt VP, Holland H, Wallenborn M, Koschny R, Frydrychowicz C, Richter M, Holland L, Nestler U, Sander C. SNP array genomic analysis of matched pairs of brain and liver metastases in primary colorectal cancer. J Cancer Res Clin Oncol 2023; 149:18173-18183. [PMID: 38010391 PMCID: PMC10725338 DOI: 10.1007/s00432-023-05505-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Brain metastasis formation is a rare and late event in colorectal cancer (CRC) patients and associated with poor survival. In contrast to other metastatic sites, the knowledge on chromosomal aberrations in brain metastases is very limited. METHODS Therefore, we carried out single nucleotide polymorphism (SNP) array analyses on matched primary CRC and brain metastases of four patients as well as on liver metastases of three patients. RESULTS Brain metastases showed more chromosomal aberrations than primary tumors or liver metastases. Commonly occurring aberrations were gain of 8q11.1-q24.3 (primary CRC), gain of 13q12.13-q12.3 (liver metastases), and gain of 20q11.1-q13.33 (brain metastases). Furthermore, we found one copy-neutral loss of heterozygosity (cn-LOH) region on chromosome 3 in primary CRC, three cn-LOH regions in liver metastases and 23 cn-LOH regions in brain metastases, comprising 26 previously undescribed sites. CONCLUSION The more frequent occurrence of cn-LOHs and subsequently affected genes in brain metastases shed light on the pathophysiology of brain metastasis formation. Further pairwise genetic analyses between primary tumors and their metastases will help to define the role of affected genes in cn-LOH regions.
Collapse
Affiliation(s)
- Vivian-Pascal Brandt
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Saxony, Germany.
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Saxony, Germany.
| | - Heidrun Holland
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Saxony, Germany
| | - Marco Wallenborn
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Saxony, Germany
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Saxony, Germany
| | - Ronald Koschny
- Interdisciplinary Endoscopy Center (IEZ), Department of Gastroenterology and Hepatology, University Hospital Heidelberg, Heidelberg, Baden-Wuerttemberg, Germany
| | - Clara Frydrychowicz
- Paul Flechsig Institute of Neuropathology, University Medicine Leipzig, Leipzig, Saxony, Germany
| | - Mandy Richter
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Saxony, Germany
| | - Lydia Holland
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Saxony, Germany
| | - Ulf Nestler
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Saxony, Germany
| | - Caroline Sander
- Department of Neurosurgery, University Hospital Leipzig, Leipzig, Saxony, Germany
| |
Collapse
|
21
|
Zhang Y, Sun S, Qi Y, Dai Y, Hao Y, Xin M, Xu R, Chen H, Wu X, Liu Q, Kong C, Zhang G, Wang P, Guo Q. Characterization of tumour microenvironment reprogramming reveals invasion in epithelial ovarian carcinoma. J Ovarian Res 2023; 16:200. [PMID: 37817210 PMCID: PMC10563280 DOI: 10.1186/s13048-023-01270-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/29/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Patients with epithelial ovarian carcinoma (EOC) are usually diagnosed at an advanced stage with tumour cell invasion. However, identifying the underlying molecular mechanisms and biomarkers of EOC proliferation and invasion remains challenging. RESULTS Herein, we explored the relationship between tumour microenvironment (TME) reprogramming and tissue invasion based on single-cell RNA sequencing (scRNA-seq) datasets. Interestingly, hypoxia, oxidative phosphorylation (OXPHOS) and glycolysis, which have biologically active trajectories during epithelial mesenchymal transition (EMT), were positively correlated. Moreover, energy metabolism and anti-apoptotic activity were found to be critical contributors to intratumor heterogeneity. In addition, HMGA1, EGR1 and RUNX1 were found to be critical drivers of the EMT process in EOC. Experimental validation revealed that suppressing EGR1 expression inhibited tumour cell invasion, significantly upregulated the expression of E-cadherin and decreased the expression of N-cadherin. In cell components analysis, cancer-associated fibroblasts (CAFs) were found to significantly contribute to immune infiltration and tumour invasion, and the accumulation of CAFs was associated with poorer patient survival. CONCLUSION We revealed the molecular mechanism and biomarkers of tumour invasion and TME reprogramming in EOC, which provides effective targets for the suppression of tumour invasion.
Collapse
Affiliation(s)
- Yuanfu Zhang
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Shu Sun
- Department Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yue Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yifan Dai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yangyang Hao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Mengyu Xin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Rongji Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Hongyan Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xiaoting Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qian Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Congcong Kong
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Guangmei Zhang
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| | - Peng Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Qiuyan Guo
- Department of Gynecology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
22
|
Santamarina‐Ojeda P, Tejedor JR, Pérez RF, López V, Roberti A, Mangas C, Fernández AF, Fraga MF. Multi-omic integration of DNA methylation and gene expression data reveals molecular vulnerabilities in glioblastoma. Mol Oncol 2023; 17:1726-1743. [PMID: 37357610 PMCID: PMC10483606 DOI: 10.1002/1878-0261.13479] [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: 01/13/2023] [Revised: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive types of cancer and exhibits profound genetic and epigenetic heterogeneity, making the development of an effective treatment a major challenge. The recent incorporation of molecular features into the diagnosis of patients with GBM has led to an improved categorization into various tumour subtypes with different prognoses and disease management. In this work, we have exploited the benefits of genome-wide multi-omic approaches to identify potential molecular vulnerabilities existing in patients with GBM. Integration of gene expression and DNA methylation data from both bulk GBM and patient-derived GBM stem cell lines has revealed the presence of major sources of GBM variability, pinpointing subtype-specific tumour vulnerabilities amenable to pharmacological interventions. In this sense, inhibition of the AP-1, SMAD3 and RUNX1/RUNX2 pathways, in combination or not with the chemotherapeutic agent temozolomide, led to the subtype-specific impairment of tumour growth, particularly in the context of the aggressive, mesenchymal-like subtype. These results emphasize the involvement of these molecular pathways in the development of GBM and have potential implications for the development of personalized therapeutic approaches.
Collapse
Affiliation(s)
- Pablo Santamarina‐Ojeda
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
| | - Juan Ramón Tejedor
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
- Nanomaterials and Nanotechnology Research Centre (CINN‐CSIC)Principality of AsturiasSpain
| | - Raúl F. Pérez
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
- Nanomaterials and Nanotechnology Research Centre (CINN‐CSIC)Principality of AsturiasSpain
| | - Virginia López
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
| | - Annalisa Roberti
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Nanomaterials and Nanotechnology Research Centre (CINN‐CSIC)Principality of AsturiasSpain
| | - Cristina Mangas
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
| | - Agustín F. Fernández
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
- Nanomaterials and Nanotechnology Research Centre (CINN‐CSIC)Principality of AsturiasSpain
| | - Mario F. Fraga
- Health Research Institute of Asturias (ISPA)Spain
- Foundation for Biomedical Research and Innovation in Asturias (FINBA)Spain
- University Institute of Oncology of Asturias (IUOPA)Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)MadridSpain
- Nanomaterials and Nanotechnology Research Centre (CINN‐CSIC)Principality of AsturiasSpain
| |
Collapse
|
23
|
Chen Y, He Y, Liu S. RUNX1-Regulated Signaling Pathways in Ovarian Cancer. Biomedicines 2023; 11:2357. [PMID: 37760803 PMCID: PMC10525517 DOI: 10.3390/biomedicines11092357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 09/29/2023] Open
Abstract
Ovarian cancer is the leading cause of gynecological death worldwide, and its poor prognosis and high mortality seriously affect the life of ovarian cancer patients. Runt-related transcription factor 1 (RUNX1) has been widely studied in hematological diseases and plays an important role in the occurrence and development of hematological diseases. In recent years, studies have reported the roles of RUNX1 in solid tumors, including the significantly increased expression of RUNX1 in ovarian cancer. In ovarian cancer, the dysregulation of the RUNX1 signaling pathway has been implicated in tumor progression, metastasis, and response to therapy. At the same time, the decreased expression of RUNX1 in ovarian cancer can significantly improve the sensitivity of clinical chemotherapy and provide theoretical support for the subsequent diagnosis and treatment target of ovarian cancer, providing prognosis and treatment options to patients with ovarian cancer. However, the role of RUNX1 in ovarian cancer remains unclear. Therefore, this article reviews the relationship between RUNX1 and the occurrence and development of ovarian cancer, as well as the closely regulated signaling pathways, to provide some inspiration and theoretical support for future research on RUNX1 in ovarian cancer and other diseases.
Collapse
Affiliation(s)
- Yuanzhi Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingying He
- School of Chemical Science & Technology, Yunnan University, Kunming 650091, China
| | - Shubai Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
24
|
Muniyandi A, Hartman GD, Song Y, Mijit M, Kelley MR, Corson TW. Beyond VEGF: Targeting Inflammation and Other Pathways for Treatment of Retinal Disease. J Pharmacol Exp Ther 2023; 386:15-25. [PMID: 37142441 PMCID: PMC10289243 DOI: 10.1124/jpet.122.001563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Neovascular eye diseases include conditions such as retinopathy of prematurity, proliferative diabetic retinopathy, and neovascular age-related macular degeneration. Together, they are a major cause of vision loss and blindness worldwide. The current therapeutic mainstay for these diseases is intravitreal injections of biologics targeting vascular endothelial growth factor (VEGF) signaling. Lack of universal response to these anti-VEGF agents coupled with the challenging delivery method underscore a need for new therapeutic targets and agents. In particular, proteins that mediate both inflammatory and proangiogenic signaling are appealing targets for new therapeutic development. Here, we review agents currently in clinical trials and highlight some promising targets in preclinical and early clinical development, focusing on the redox-regulatory transcriptional activator APE1/Ref-1, the bioactive lipid modulator soluble epoxide hydrolase, the transcription factor RUNX1, and others. Small molecules targeting each of these proteins show promise for blocking neovascularization and inflammation. The affected signaling pathways illustrate the potential of new antiangiogenic strategies for posterior ocular disease. SIGNIFICANCE STATEMENT: Discovery and therapeutic targeting of new angiogenesis mediators is necessary to improve treatment of blinding eye diseases like retinopathy of prematurity, diabetic retinopathy, and neovascular age-related macular degeneration. Novel targets undergoing evaluation and drug discovery work include proteins important for both angiogenesis and inflammation signaling, including APE1/Ref-1, soluble epoxide hydrolase, RUNX1, and others.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Gabriella D Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Yang Song
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mahmut Mijit
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark R Kelley
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Timothy W Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
25
|
Deforzh E, Kharel P, Karelin A, Ivanov P, Krichevsky AM. HOXDeRNA activates a cancerous transcription program and super-enhancers genome-wide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547275. [PMID: 37425921 PMCID: PMC10327164 DOI: 10.1101/2023.06.30.547275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background The origin and genesis of highly malignant and heterogenous glioblastoma brain tumors remain unknown. We previously identified an enhancer-associated long non-coding RNA, LINC01116 (named HOXDeRNA here), that is absent in the normal brain but is commonly expressed in malignant glioma. HOXDeRNA has a unique capacity to transform human astrocytes into glioma-like cells. This work aimed to investigate molecular events underlying the genome-wide function of this lncRNA in glial cell fate and transformation. Results Using a combination of RNA-Seq, ChIRP-Seq, and ChIP-Seq, we now demonstrate that HOXDeRNA binds in trans to the promoters of genes encoding 44 glioma-specific transcription factors distributed throughout the genome and derepresses them by removing the Polycomb repressive complex 2 (PRC2). Among the activated transcription factors are the core neurodevelopmental regulators SOX2, OLIG2, POU3F2, and SALL2. This process requires an RNA quadruplex structure of HOXDeRNA that interacts with EZH2. Moreover, HOXDeRNA-induced astrocyte transformation is accompanied by the activation of multiple oncogenes such as EGFR, PDGFR, BRAF, and miR-21, and glioma-specific super-enhancers enriched for binding sites of glioma master transcription factors SOX2 and OLIG2. Conclusions Our results demonstrate that HOXDeRNA overrides PRC2 repression of glioma core regulatory circuitry with RNA quadruplex structure. These findings help reconstruct the sequence of events underlying the process of astrocyte transformation and suggest a driving role for HOXDeRNA and a unifying RNA-dependent mechanism of gliomagenesis.
Collapse
Affiliation(s)
- Evgeny Deforzh
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Prakash Kharel
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anton Karelin
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Pavel Ivanov
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anna M. Krichevsky
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Qiu W, Xiao Z, Yang Y, Jiang L, Song S, Qi X, Chen Y, Yang H, Liu J, Chu L. USP10 deubiquitinates RUNX1 and promotes proneural-to-mesenchymal transition in glioblastoma. Cell Death Dis 2023; 14:207. [PMID: 36949071 PMCID: PMC10033651 DOI: 10.1038/s41419-023-05734-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 03/03/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
The mesenchymal (MES) subtype of glioblastoma (GBM) is a highly aggressive, malignant and proliferative cancer that is resistant to chemotherapy. Runt-related transcription factor 1 (RUNX1) was shown to support MES GBM, however, its underlying mechanisms are unclear. Here, we identified USP10 as a deubiquitinating enzyme that regulates RUNX1 stabilization and is mainly expressed in MES GBM. Overexpression of USP10 upregulated RUNX1 and induced proneural-to-mesenchymal transition (PMT), thus maintaining MES properties in GBM. Conversely, USP10 knockdown inhibited RUNX1 and resulted in the loss of MES properties. USP10 was shown to interact with RUNX1, with RUNX1 being stabilized upon deubiquitylation. Moreover, we found that USP10 inhibitor Spautin-1 induced RUNX1 degradation and inhibited MES properties in vitro and in vivo. Furthermore, USP10 was strongly correlated with RUNX1 expression in samples of different subtypes of human GBM and had prognostic value for GBM patients. We identified USP10 as a key deubiquitinase for RUNX1 protein stabilization. USP10 maintains MES properties of GBM, and promotes PMT of GBM cells. Our study indicates that the USP10/RUNX1 axis may be a potential target for novel GBM treatments.
Collapse
Affiliation(s)
- Wenjin Qiu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Zumu Xiao
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Yushi Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Lishi Jiang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Shibin Song
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yimin Chen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Hua Yang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China
| | - Jian Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, 550001, Guizhou, China.
| | - Liangzhao Chu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, Guizhou, China.
| |
Collapse
|
27
|
EGFRvIII Promotes the Proneural–Mesenchymal Transition of Glioblastoma Multiforme and Reduces Its Sensitivity to Temozolomide by Regulating the NF-κB/ALDH1A3 Axis. Genes (Basel) 2023; 14:genes14030651. [PMID: 36980923 PMCID: PMC10048499 DOI: 10.3390/genes14030651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
(1) Background: Glioblastoma multiforme (GBM) is the most common and malignant intracranial tumor in adults. At present, temozolomide (TMZ) is recognized as the preferred chemotherapeutic drug for GBM, but some patients have low sensitivity to TMZ or chemotherapy resistance to TMZ. Our previous study found that GBM patients with EGFRvIII (+) have low sensitivity to TMZ. However, the reasons and possible mechanisms of the chemoradiotherapy resistance in GBM patients with EGFRvIII (+) are not clear. (2) Methods: In this study, tissue samples of patients with GBM, GBM cell lines, glioma stem cell lines, and NSG mice were used to explore the causes and possible mechanisms of low sensitivity to TMZ in patients with EGFRvIII (+)-GBM. (3) Results: The study found that EGFRvIII promoted the proneural–mesenchymal transition of GBM and reduced its sensitivity to TMZ, and EGFRvIII regulated of the expression of ALDH1A3. (4) Conclusions: EGFRvIII activated the NF-κB pathway and further regulated the expression of ALDH1A3 to promote the proneural–mesenchymal transition of GBM and reduce its sensitivity to TMZ, which will provide an experimental basis for the selection of clinical drugs for GBM patients with EGFRvIII (+).
Collapse
|
28
|
Mancini A, Colapietro A, Cristiano L, Rossetti A, Mattei V, Gravina GL, Perez-Montoyo H, Yeste-Velasco M, Alfon J, Domenech C, Festuccia C. Anticancer effects of ABTL0812, a clinical stage drug inducer of autophagy-mediated cancer cell death, in glioblastoma models. Front Oncol 2022; 12:943064. [PMID: 36408162 PMCID: PMC9668006 DOI: 10.3389/fonc.2022.943064] [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: 05/13/2022] [Accepted: 10/13/2022] [Indexed: 11/30/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant adult brain tumor. Current standard of care treatments have very limited efficacy, being the patients´ overall survival 14 months and the 2-year survival rate less than 10%. Therefore, the treatment of GBM is an urgent unmet clinical need. Methods The aim of this study was to investigate in vitro and in vivo the potential of ABTL0812, an oral anticancer compound currently in phase II clinical stage, as a novel therapy for GBM. Results We showed that ABTL0812 inhibits cell proliferation in a wide panel of GBM cell lines and patient-derived glioblastoma stem cells (GSCs) with half maximal inhibitory concentrations (IC50s) ranging from 15.2 µM to 46.9 µM. Additionally, ABTL0812 decreased GSCs neurosphere formation. GBM cells aggressiveness is associated with a trans-differentiation process towards a less differentiated phenotype known as proneural to mesenchymal transition (PMT). ABTL0812 was shown to revert PMT and induce cell differentiation to a less malignant phenotype in GBM cell lines and GSCs, and consequently reduced cell invasion. As previously shown in other cancer types, we demonstrated that the molecular mechanism of action of ABTL0812 in glioblastoma involves the inhibition of Akt/mTORC1 axis by overexpression of TRIB3, and the activation of endoplasmic reticulum (ER) stress/unfolded protein response (UPR). Both actions converge to induce autophagy-mediated cell death. ABTL0812 anticancer efficacy was studied in vivo using subcutaneous and orthotopic intra-brain xenograft tumor models. We demonstrated that ABTL0812 impairs tumor growth and increases disease-free survival and overall survival of mice. Furthermore, the histological analysis of tumors indicated that ABTL0812 decreases angiogenesis. Finally, we investigated the combination of ABTL0812 with the standard of care treatments for GBM radiotherapy and temozolomide in an orthotopic model, detecting that ABTL0812 potentiates the efficacy of both treatments and that the strongest effect is obtained with the triple combination of ABTL0812+radiotherapy+temozolomide. Conclusions Overall, the present study demonstrated the anticancer efficacy of ABTL0812 as single agent and in combination with the GBM standard of care treatments in models of glioblastoma and supports the clinical investigation of ABTL0812 as a potential novel therapy for this aggressive brain tumor type.
Collapse
Affiliation(s)
- Andrea Mancini
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Colapietro
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Department of Clinical Medicine, Public Health, Life Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandra Rossetti
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, “Sabina Universitas”, Rieti, Italy
| | - Giovanni Luca Gravina
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Division of Radiation Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L’Aquila, Italy
| | - Héctor Perez-Montoyo
- R&D Department, Ability Pharmaceuticals, Parc Tecnològic del Vallès, Cerdanyola del Vallès, Barcelona, Spain
| | - Marc Yeste-Velasco
- R&D Department, Ability Pharmaceuticals, Parc Tecnològic del Vallès, Cerdanyola del Vallès, Barcelona, Spain
| | - Jose Alfon
- R&D Department, Ability Pharmaceuticals, Parc Tecnològic del Vallès, Cerdanyola del Vallès, Barcelona, Spain
| | - Carles Domenech
- R&D Department, Ability Pharmaceuticals, Parc Tecnològic del Vallès, Cerdanyola del Vallès, Barcelona, Spain
| | - Claudio Festuccia
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
29
|
Hsia T, Yekula A, Batool SM, Rosenfeld YB, You DG, Weissleder R, Lee H, Carter BS, Balaj L. Glioblastoma-derived extracellular vesicle subpopulations following 5-aminolevulinic acid treatment bear diagnostic implications. J Extracell Vesicles 2022; 11:e12278. [PMID: 36404434 PMCID: PMC9676504 DOI: 10.1002/jev2.12278] [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: 05/25/2022] [Revised: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
Liquid biopsy is a minimally invasive alternative to surgical biopsy, encompassing different analytes including extracellular vesicles (EVs), circulating tumour cells (CTCs), circulating tumour DNA (ctDNA), proteins, and metabolites. EVs are released by virtually all cells, but at a higher rate by faster cycling, malignant cells. They encapsulate cargo native to the originating cell and can thus provide a window into the tumour landscape. EVs are often analysed in bulk which hinders the analysis of rare, tumour-specific EV subpopulations from the large host EV background. Here, we fractionated EV subpopulations in vitro and in vivo and characterized their phenotype and generic cargo. We used 5-aminolevulinic acid (5-ALA) to induce release of endogenously fluorescent tumour-specific EVs (EVPpIX ). Analysis of five different subpopulations (EVPpIX , EVCD63 , EVCD9 , EVEGFR , EVCFDA ) from glioblastoma (GBM) cell lines revealed unique transcriptome profiles, with the EVPpIX transcriptome demonstrating closer alignment to tumorigenic processes over the other subpopulations. Similarly, isolation of tumour-specific EVs from GBM patient plasma showed enrichment in GBM-associated genes, when compared to bulk EVs from plasma. We propose that fractionation of EV populations facilitates detection and isolation of tumour-specific EVs for disease monitoring.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Anudeep Yekula
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - S. Maheen Batool
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yulia B. Rosenfeld
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Dong Gil You
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Systems BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Hakho Lee
- Center for Systems BiologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Bob S. Carter
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Leonora Balaj
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
30
|
Jin Y, Zhang J, Pan Y, Shen W. Berberine Suppressed the Progression of Human Glioma Cells by Inhibiting the TGF-β1/SMAD2/3 Signaling Pathway. Integr Cancer Ther 2022; 21:15347354221130303. [PMID: 36255058 PMCID: PMC9583234 DOI: 10.1177/15347354221130303] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Previous studies have shown that berberine can inhibit glioma progression,
although the underlying molecular mechanisms needed to be explored further.
The aim of this study was to evaluate the suppressive effects of berberine
on human glioma cells, and identify the underlying signaling pathways. Material and Methods: The cytotoxic effect of different concentrations of berberine against normal
human glial cells (HEB) and 4 glioma cell lines was evaluated by the CCK-8
assay. Apoptosis was assayed by flow cytometry. In vitro migration and
invasion were analyzed by the wound healing and transwell assays. The
expression levels of specific proteins were measured by western blotting and
ELISA. Results: Berberine significantly inhibited the proliferation of human glioma U-87
cells, and induced apoptosis in the U-87 and LN229 cells by downregulating
Bcl-2, and upregulating Bax and caspase-3. In addition, berberine also
inhibited migration and invasion of the glioma cells. Furthermore, berberine
exerted its effects on the proliferation, migration, invasion, and apoptosis
of glioma cells by inhibiting the TGF-β1/SMAD2/3 signaling pathway, and
exogenous TGF-β abrogated the pro-apoptotic and anti-oncogenic effects of
berberine. Conclusions: Berberine inhibits glioma progression by targeting the TGF-β1/SMAD2/3
signaling pathway.
Collapse
Affiliation(s)
- Yun Jin
- Tongxiang First People’s Hospital,
Tongxiang, Zhejiang, China
| | - Jiawei Zhang
- Tongxiang First People’s Hospital,
Tongxiang, Zhejiang, China
| | - Yunfeng Pan
- Tongxiang First People’s Hospital,
Tongxiang, Zhejiang, China
| | - Wangzhen Shen
- Tongxiang First People’s Hospital,
Tongxiang, Zhejiang, China,Wangzhen Shen, Department of Neurosurgery,
Tongxiang First People’s Hospital, No. 1918, Jiaochang East Road, Zhendong New
District, Tongxiang City, Zhejiang 314500, China.
| |
Collapse
|
31
|
Xiao Y, Wang Z, Zhao M, Deng Y, Yang M, Su G, Yang K, Qian C, Hu X, Liu Y, Geng L, Xiao Y, Zou Y, Tang X, Liu H, Xiao H, Fan R. Single-Cell Transcriptomics Revealed Subtype-Specific Tumor Immune Microenvironments in Human Glioblastomas. Front Immunol 2022; 13:914236. [PMID: 35669791 PMCID: PMC9163377 DOI: 10.3389/fimmu.2022.914236] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Human glioblastoma (GBM), the most aggressive brain tumor, comprises six major subtypes of malignant cells, giving rise to both inter-patient and intra-tumor heterogeneity. The interaction between different tumor subtypes and non-malignant cells to collectively shape a tumor microenvironment has not been systematically characterized. Herein, we sampled the cellular milieu of surgically resected primary tumors from 7 GBM patients using single-cell transcriptome sequencing. A lineage relationship analysis revealed that a neural-progenitor-2-like (NPC2-like) state with high metabolic activity was associated with the tumor cells of origin. Mesenchymal-1-like (MES1-like) and mesenchymal-2-like (MES2-like) tumor cells correlated strongly with immune infiltration and chronic hypoxia niche responses. We identified four subsets of tumor-associated macrophages/microglia (TAMs), among which TAM-1 co-opted both acute and chronic hypoxia-response signatures, implicated in tumor angiogenesis, invasion, and poor prognosis. MES-like GBM cells expressed the highest number of M2-promoting ligands compared to other cellular states while all six states were associated with TAM M2-type polarization and immunosuppression via a set of 10 ligand–receptor signaling pathways. Our results provide new insights into the differential roles of GBM cell subtypes in the tumor immune microenvironment that may be deployed for patient stratification and personalized treatment.
Collapse
Affiliation(s)
- Yong Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhen Wang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Mengjie Zhao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Mingyu Yang
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Kun Yang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chunfa Qian
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xinhua Hu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yong Liu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Liangyuan Geng
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yang Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Yuanjie Zou
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xianglong Tang
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hongyi Liu
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- *Correspondence: Hongyi Liu, ; Hong Xiao, ; Rong Fan,
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- *Correspondence: Hongyi Liu, ; Hong Xiao, ; Rong Fan,
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Hongyi Liu, ; Hong Xiao, ; Rong Fan,
| |
Collapse
|
32
|
Omatsu Y, Aiba S, Maeta T, Higaki K, Aoki K, Watanabe H, Kondoh G, Nishimura R, Takeda S, Chung UI, Nagasawa T. Runx1 and Runx2 inhibit fibrotic conversion of cellular niches for hematopoietic stem cells. Nat Commun 2022; 13:2654. [PMID: 35551452 PMCID: PMC9098511 DOI: 10.1038/s41467-022-30266-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/08/2022] [Indexed: 01/01/2023] Open
Abstract
In bone marrow, special microenvironments, known as niches, are essential for the maintenance of hematopoietic stem cells (HSCs). A population of mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells or leptin receptor-expressing cells are the major cellular component of HSC niches. The molecular regulation of HSC niche properties is not fully understood. The role of Runx transcription factors, Runx1 and Runx2 in HSC cellular niches remains unclear. Here we show that Runx1 is predominantly expressed in CAR cells and that mice lacking both Runx1 and Runx2 in CAR cells display an increase in fibrosis and bone formation with markedly reduced hematopoietic stem and progenitor cells in bone marrow. In vitro, Runx1 is induced by the transcription factor Foxc1 and decreases fibrotic gene expression in CAR cells. Thus, HSC cellular niches require Runx1 or Runx2 to prevent their fibrotic conversion and maintain HSCs and hematopoiesis in adults. The transcription factors, Runx1 and Runx2 are critical embryonically for generation of HSCs and osteoblasts, respectively. Here the authors show that adult mice lacking Runx1 and Runx2 in HSC-supporting CAR cells displayed an increase in fibrosis with reduced HSCs in bone marrow.
Collapse
Affiliation(s)
- Yoshiki Omatsu
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shota Aiba
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tomonori Maeta
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kei Higaki
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazunari Aoki
- Laboratory of Stem Cell Genetics, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hitomi Watanabe
- Laboratory of Animal Experiments for Regeneration, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Gen Kondoh
- Laboratory of Animal Experiments for Regeneration, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shu Takeda
- Endocrinology Division, Toranomon Hospital, Minato-ku, Tokyo, 105-8470, Japan
| | - Ung-Il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan. .,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan. .,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
33
|
Xu C, Zhao J, Song J, Xiao M, Cui X, Xin L, Xu J, Zhang Y, Yi K, Hong B, Tong F, Tian S, Tan Y, Kang C, Fang C. lncRNA PRADX is a Mesenchymal Glioblastoma Biomarker for Cellular Metabolism Targeted Therapy. Front Oncol 2022; 12:888922. [PMID: 35574370 PMCID: PMC9106305 DOI: 10.3389/fonc.2022.888922] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal type of primary malignant central nervous system (CNS) tumor with an extremely poor prognosis, and the mesenchymal subtype of GBM has the worst prognosis. Here, we found that lncRNA PRADX was overexpressed in the mesenchymal GBM and was transcriptionally regulated by RUNX1-CBFβ complex, overexpressed PRADX suppressed BLCAP expression via interacting with EZH2 and catalyzing trimethylation of lysine 27 on histone H3 (H3K27me3). Moreover, we showed that BLCAP interacted with STAT3 and reduced STAT3 phosphorylation, overexpressed PRADX activated STAT3 phosphorylation, and promoted ACSL1 expression via suppressing BLCAP expression, accelerating tumor metabolism. Finally, we determined that combined of ACSL1 and CPT1 inhibitors could reverse the accelerated cellular metabolism and tumor growth induced by PRADX overexpression in vivo and in vitro. Collectively, PRADX/PRC2 complex activated the STAT3 pathway and energy metabolism in relation to mesenchymal GBM progression. Furthermore, our findings provided a novel therapeutic strategy targeting the energy metabolism activity of GBM.
Collapse
Affiliation(s)
- Can Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Jixing Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Jia Song
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Menglin Xiao
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Xiaoteng Cui
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Lei Xin
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Jianglong Xu
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Yuhao Zhang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Kaikai Yi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Biao Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Fei Tong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Shaohui Tian
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| | - Yanli Tan
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin, China
- Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Chuan Fang
- School of Clinical Medicine, Hebei University, Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
- Hebei Key Laboratory of Precise Diagnosis and Treatment of Glioma, Baoding, China
| |
Collapse
|
34
|
Ariffin NS. Healthcare Resource Utilization and Costs of Steroid-Associated Complications in Patients With Graft-Versus-Host Disease. Clin Breast Cancer 2022; 22:499-506. [DOI: 10.1016/j.clbc.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/03/2022] [Accepted: 04/18/2022] [Indexed: 11/03/2022]
|
35
|
Ren W, Jin W, Liang Z. Construction and Validation of an Immune-Related Risk Score Model for Survival Prediction in Glioblastoma. Front Neurol 2022; 13:832944. [PMID: 35370869 PMCID: PMC8965766 DOI: 10.3389/fneur.2022.832944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
BackgroundAs one of the most important brain tumors, glioblastoma (GBM) has a poor prognosis, especially in adults. Immune-related genes (IRGs) and immune cell infiltration are responsible for the pathogenesis of GBM. This study aimed to identify new tumor markers to predict the prognosis of patients with GBM.MethodsThe Cancer Genome Atlas (TCGA) database and ImmPort database were used for model construction. The Wilcoxon rank-sum test was applied to identify the differentially expressed IRGs (DEIRGs) between the GBM and normal samples. Univariate Cox regression analysis and Kaplan–Meier analysis was performed to investigate the relationship between each DEIRG and overall survival. Next, multivariate Cox regression analysis was exploited to further explore the prognostic potential of DEIRGs. A risk-score model was constructed based on the above results. The area under the curve (AUC) values were calculated to assess the effect of the model prediction. Furthermore, the Chinese Glioma Genome Atlas (CGGA) dataset was used for model validation. STRING database and functional enrichment analysis were used for exploring the gene interactions and the underlying functions and pathways. The CIBERSORT algorithm was used for correlation analysis of the marker genes and the tumor-infiltrating immune cells.ResultsThere were 198 DEIRGs in GBM, including 153 upregulated genes and 45 downregulated genes. Seven marker genes (LYNX1, PRELID1P4, MMP9, TCF12, RGS14, RUNX1, and CCR2) were filtered out by sequential screening for DEIRGs. The regression coefficients (0.0410, 1.335, 0.005, −0.021, 0.123, 0.142, and −0.329) and expression data of the marker genes were used to construct the model. The AUC values for 1, 2, and 3 years were 0.744, 0.737, and 0.749 in the TCGA–GBM cohort and 0.612, 0.602, and 0.594 in the CGGA-GBM cohort, respectively, which indicated a high predictive power. The results of enrichment analysis revealed that these genes were enriched in the activation of T cell and cytokine receptor interaction pathways. The interaction network map demonstrated a close relationship between the marker genes MMP9 and CCR2. Infiltration analysis of the immune cells showed that dendritic cells (DCs) could identify GBM, while LYNX1, RUNX1, and CCR2 were significantly positively correlated with DCs expression.ConclusionThis study analyzed the expression of IRGs in GBM and identified seven marker genes for the construction of an immune-related risk score model. These marker genes were found to be associated with DCs and were enriched in similar immune response pathways. These findings are likely to provide new insights for the immunotherapy of patients with GBM.
Collapse
Affiliation(s)
- Wei Ren
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zehua Liang
- School of Humanities and Management, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Zehua Liang
| |
Collapse
|
36
|
Lin TC. RUNX1 and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188715. [DOI: 10.1016/j.bbcan.2022.188715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
|
37
|
Calori IR, Alves SR, Bi H, Tedesco AC. Type-I Collagen/Collagenase Modulates the 3D Structure and Behavior of Glioblastoma Spheroid Models. ACS APPLIED BIO MATERIALS 2022; 5:723-733. [PMID: 35068151 DOI: 10.1021/acsabm.1c01138] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Multicellular tumor spheroids have emerged as well-structured, three-dimensional culture models that resemble and mimic the complexity of the dense and hypoxic cancer microenvironment. However, in brain tumor studies, a variety of glioblastoma multiforme (GBM) cell lines only self-assemble into loose cellular aggregates, lacking the properties of actual glioma tumors in humans. In this study, we used type-I collagen as an extracellular matrix component to promote the compaction of GBM aggregates forming tight spheroids to understand how collagen influences the properties of tumors, such as their growth, proliferation, and invasion, and collagenase to promote collagen degradation. The GBM cell lines U87MG, T98G, and A172, as well as the medulloblastoma cell line UW473, were used as standard cell lines that do not spontaneously self-assemble into spheroids, and GBM U251 was used as a self-assembling cell line. According to the findings, all cell lines formed tight spheroids at collagen concentrations higher than 15.0 μg mL-1. Collagen was distributed along the spheroid, similarly to that observed in invasive GBM tumors, and decreased cell migration with no effect on the cellular uptake of small active molecules, as demonstrated by uptake studies using the photosensitizer verteporfin. The enzymatic cleavage of collagen affected spheroid morphology and increased cell migration while maintaining cell viability. Such behaviors are relevant to the physiological models of GBM tumors and are useful for better understanding cell migration and the in vivo infiltration path, drug screening, and kinetics of progression of GBM tumors.
Collapse
Affiliation(s)
- Italo Rodrigo Calori
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Samara Rodrigues Alves
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Hong Bi
- School of Chemistry and Chemical Engineering, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei 230601, China
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil.,School of Chemistry and Chemical Engineering, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei 230601, China
| |
Collapse
|
38
|
Fan X, Fan J, Yang H, Zhao C, Niu W, Fang Z, Chen X. Heterogeneity of subsets in glioblastoma mediated by Smad3 palmitoylation. Oncogenesis 2021; 10:72. [PMID: 34707087 PMCID: PMC8551152 DOI: 10.1038/s41389-021-00361-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/25/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common and deadly of the primary intracranial tumors and is comprised of subsets that show plasticity and marked heterogeneity, contributing to the lack of success in genomic profiling to guide development of precision medicine for these tumors. In this study, a mutation in isocitrate dehydrogenase 1 was found to suppress the transforming growth factor-beta signaling pathway and E2F4 interacted with Smad3 to inhibit expression of mesenchymal markers. However, palmitoylation of Smad3 mediated by palmitoyltransferase ZDHHC19 promoted activation of the transforming growth factor-beta signaling pathway, and its interaction with EP300 promoted expression of mesenchymal markers in the mesenchymal subtype of GBM. Smad3 and hypoxia-inducible factor 1-alpha may be important molecular targets for treatment of glioma because they appear to coordinate the basic aspects of cancer stem cell biology.
Collapse
Affiliation(s)
- Xiaoqing Fan
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China.,University of Science and Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230031, China.,Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), No. 17, Lu Jiang Road, Hefei, Anhui, 230001, China
| | - Junqi Fan
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China.,University of Science and Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230031, China
| | - Haoran Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China.,Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), No. 17, Lu Jiang Road, Hefei, Anhui, 230001, China.,Department of Laboratory Medicine, Hefei Cancer Hospital, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui, 230031, China
| | - Chenggang Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China.,University of Science and Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230031, China
| | - Wanxiang Niu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China.,University of Science and Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230031, China
| | - Zhiyou Fang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China. .,Department of Laboratory Medicine, Hefei Cancer Hospital, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui, 230031, China.
| | - Xueran Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui 230031, China. MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, No. 96, Jin Zhai Road, Hefei, Anhui, 230027, China. .,Department of Laboratory Medicine, Hefei Cancer Hospital, Chinese Academy of Sciences, No. 350, Shushan Hu Road, Hefei, Anhui, 230031, China.
| |
Collapse
|
39
|
Zhao J, Jin W, Yi K, Wang Q, Zhou J, Tan Y, Xu C, Xiao M, Hong B, Xu F, Zhang K, Kang C. Combination LSD1 and HOTAIR-EZH2 inhibition disrupts cell cycle processes and induces apoptosis in glioblastoma cells. Pharmacol Res 2021; 171:105764. [PMID: 34246782 DOI: 10.1016/j.phrs.2021.105764] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
Glioblastoma (GBM) is the most common primary central nervous system tumor and has a poor prognosis, with a median survival time of only 14 months from diagnosis. Abnormally expressed long noncoding RNAs (lncRNAs) are important epigenetic regulators of chromatin modification and gene expression regulation in tumors, including GBM. We previously showed that the lncRNA HOTAIR is related to the cell cycle progression and can be used as an independent predictor in GBM. Lysine-specific demethylase 1 (LSD1), binding to 3' domain of HOTAIR, specifically removes mono- and di-methyl marks from H3 lysine 4 (H3K4) and plays key roles during carcinogenesis. In this study, we combined a HOTAIR-EZH2 disrupting agent and an LSD1 inhibitor, AC1Q3QWB (AQB) and GSK-LSD1, respectively, to block the two functional domains of HOTAIR and potentially provide therapeutic benefit in the treatment of GBM. Using an Agilent Human ceRNA Microarray, we identified tumor suppressor genes upregulated by AQB and GSK-LSD1, followed by Chromatin immunoprecipitation (ChIP) assays to explore the epigenetic mechanisms of genes activation. Microarray analysis showed that AQB and GSK-LSD1 regulate cell cycle processes and induces apoptosis in GBM cell lines. Furthermore, we found that the combination of AQB and GSK-LSD1 showed a powerful effect of inhibiting cell cycle processes by targeting CDKN1A, whereas apoptosis promoting effects of combination therapy were mediated by BBC3 in vitro. ChIP assays revealed that GSK-LSD1 and AQB regulate P21 and PUMA, respectively via upregulating H3K4me2 and downregulating H3K27me3. Combination therapy with AQB and GSK-LSD1 on tumor malignancy in vitro and GBM patient-derived xenograft (PDX) models shows enhanced anti-tumor efficacy and appears to be a promising new strategy for GBM treatment through its effects on epigenetic regulation.
Collapse
Affiliation(s)
- Jixing Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Weili Jin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Kaikai Yi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Qixue Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Junhu Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Yanli Tan
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Can Xu
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Menglin Xiao
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Biao Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Fenfen Xu
- Department of Pediatrics, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250013, Shandong, China
| | - Kailiang Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, Shandong, China.
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China.
| |
Collapse
|
40
|
He Y, Chen Y, Tong Y, Long W, Liu Q. Identification of a circRNA-miRNA-mRNA regulatory network for exploring novel therapeutic options for glioma. PeerJ 2021; 9:e11894. [PMID: 34434651 PMCID: PMC8351580 DOI: 10.7717/peerj.11894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
Background Glioma is the most common brain neoplasm with a poor prognosis. Circular RNA (circRNA) and their associated competing endogenous RNA (ceRNA) network play critical roles in the pathogenesis of glioma. However, the alteration of the circRNA-miRNA-mRNA regulatory network and its correlation with glioma therapy haven't been systematically analyzed. Methods With GEO, GEPIA2, circBank, CSCD, CircInteractome, mirWalk 2.0, and mirDIP 4.1, we constructed a circRNA-miRNA-mRNA network in glioma. LASSO regression and multivariate Cox regression analysis established a hub mRNA signature to assess the prognosis. GSVA was used to estimate the immune infiltration level. Potential anti-glioma drugs were forecasted using the cMap database and evaluated with GSEA using GEO data. Results A ceRNA network of seven circRNAs (hsa_circ_0030788/0034182/0000227/ 0018086/0000229/0036592/0002765), 15 miRNAs(hsa-miR-1200/1205/1248/ 1303/3925-5p/5693/581/586/599/607/640/647/6867-5p/767-3p/935), and 46 mRNAs (including 11 hub genes of ARHGAP11A, DRP2, HNRNPA3, IGFBP5, IP6K2, KLF10, KPNA4, NRP2, PAIP1, RCN1, and SEMA5A) was constructed. Functional enrichment showed they influenced majority of the hallmarks of tumors. Eleven hub genes were proven to be decent prognostic signatures for glioma in both TCGA and CGGA datasets. Forty-six LASSO regression significant genes were closely related to immune infiltration. Finally, five compounds (fulvestrant, tanespimycin, mifepristone, tretinoin, and harman) were predicted as potential treatments for glioma. Among them, mifepristone and tretinoin were proven to inhibit the cell cycle and DNA repair in glioma. Conclusion This study highlights the potential pathogenesis of the circRNA-miRNA-mRNA regulatory network and identifies novel therapeutic options for glioma.
Collapse
Affiliation(s)
- Yi He
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yihong Chen
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yuxin Tong
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Wenyong Long
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Qing Liu
- Neurosurgery Department, Xiangya Hospital Central South University, Changsha, Hunan, China
| |
Collapse
|
41
|
Huang C, Wang M, Wang J, Wu D, Gao Y, Huang K, Yao X. Suppression MGP inhibits tumor proliferation and reverses oxaliplatin resistance in colorectal cancer. Biochem Pharmacol 2021; 189:114390. [PMID: 33359068 DOI: 10.1016/j.bcp.2020.114390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022]
Abstract
Matrix Gla protein (MGP), an extracellular matrix protein, has been widely reported to participate in the tumorigenic process and is abnormally expressed in several tumors. However, the role of MGP in colorectal cancer (CRC) remains unknown. Chemotherapy resistance represents a significant limitation in the treatment of CRC. Here, a comprehensive bioinformatics analysis revealed that MGP, which is overexpressed in CRC, might act as one of the critical genes conferring resistance to oxaliplatin (OXA). Furthermore, we found that MGP overexpression in tumor tissue might be correlated with cancer stage and patient prognosis, consistent with the bioinformatics analysis. The upregulation of MGP may act as an independent risk factor for CRC. The knockdown of MGP or inhibition of MGP expression significantly increased the sensitivity of the CRC cell lines to OXA. Suppression of MGP may reverse OXA resistance by upregulating copper transporter 1 (CTR1) and downregulating ATP7A and ATP7B. When used in combination with OXA, the inhibition of MGP reduced cancer cell proliferation, invasion, and migration and increased cell apoptosis in vitro. Suppression of MGP or OXA treatment alone significantly inhibited tumor growth in the CRC mouse model. Additionally, we found that OXA might promote the antitumor immune response in vivo. In summary, our study is the first to provide evidence that MGP expression confers OXA chemotherapy resistance in CRC and provides novel strategies to overcome chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Chengzhi Huang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Minjia Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Deqing Wu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Xueqing Yao
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
42
|
Yeini E, Ofek P, Pozzi S, Albeck N, Ben-Shushan D, Tiram G, Golan S, Kleiner R, Sheinin R, Israeli Dangoor S, Reich-Zeliger S, Grossman R, Ram Z, Brem H, Hyde TM, Magod P, Friedmann-Morvinski D, Madi A, Satchi-Fainaro R. P-selectin axis plays a key role in microglia immunophenotype and glioblastoma progression. Nat Commun 2021; 12:1912. [PMID: 33771989 PMCID: PMC7997963 DOI: 10.1038/s41467-021-22186-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GB) is a highly invasive type of brain cancer exhibiting poor prognosis. As such, its microenvironment plays a crucial role in its progression. Among the brain stromal cells, the microglia were shown to facilitate GB invasion and immunosuppression. However, the reciprocal mechanisms by which GB cells alter microglia/macrophages behavior are not fully understood. We propose that these mechanisms involve adhesion molecules such as the Selectins family. These proteins are involved in immune modulation and cancer immunity. We show that P-selectin mediates microglia-enhanced GB proliferation and invasion by altering microglia/macrophages activation state. We demonstrate these findings by pharmacological and molecular inhibition of P-selectin which leads to reduced tumor growth and increased survival in GB mouse models. Our work sheds light on tumor-associated microglia/macrophage function and the mechanisms by which GB cells suppress the immune system and invade the brain, paving the way to exploit P-selectin as a target for GB therapy.
Collapse
Affiliation(s)
- Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitzan Albeck
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Golan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ron Sheinin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Rachel Grossman
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Prerna Magod
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Sherman Building, Tel Aviv University, Tel Aviv, Israel
| | - Dinorah Friedmann-Morvinski
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Sherman Building, Tel Aviv University, Tel Aviv, Israel
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
43
|
Geng X, Zhang Y, Li Q, Xi W, Yu W, Shi L, Lin X, Sun S, Wang H. Screening and functional prediction of differentially expressed circular RNAs in human glioma of different grades. Aging (Albany NY) 2020; 13:1989-2014. [PMID: 33323543 PMCID: PMC7880344 DOI: 10.18632/aging.202192] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
Circular RNAs (circRNAs) have a critical regulatory function in human glioma. However, novel circRNAs related to different pathological grades of glioma and their crucial potential function are worth screening and prediction. CircRNA expression profiling was performed for 6 paired high- and low-grade glioma tissues and 5 adjacent normal brain tissues through next-generation sequencing. Quantitative real-time PCR (qRT-PCR) was conducted to validate circRNA expression. Bioinformatics analysis was performed, and circRNA-miRNA-mRNA networks were constructed. The expression and survival data of miRNAs and target genes were examined by GEPIA, Chinese Glioma Genome Atlas (CGGA), ONCOMINE, and cBioPortal databases. The RNA binding proteins (RBPs), open reading frames (ORFs) and N6-methyladenosine (m6A) modifications of the identified circRNAs were also predicted. Through multilevel research screening, 4 circRNAs (hsa_circ_0000915, hsa_circ_0127664, hsa_circ_0008362, and hsa_circ_0001467) were associated with glioma of different pathological grades and could be preferred candidates for subsequent functional analysis. Therefore, circRNAs are associated with the different pathological grades of glioma and reveal their potential critical regulatory function. CircRNAs might provide vital molecular biomarkers and potential therapeutic targets for glioma.
Collapse
Affiliation(s)
- Xiuchao Geng
- Faculty of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, PR China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050091, PR China
| | - Yuhao Zhang
- School of Clinical Medicine, Hebei University, Baoding 071000, PR China.,Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Qiang Li
- Faculty of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, PR China
| | - Wang Xi
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Wentao Yu
- Faculty of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, PR China
| | - Liang Shi
- Endoscope Room, Department of General Surgery, Cangzhou Central Hospital, Cangzhou 061001, PR China
| | - Xiaomeng Lin
- Departments of Breast Surgery, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Shaoguang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Hong Wang
- Faculty of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, PR China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050091, PR China.,School of Clinical Medicine, Hebei University, Baoding 071000, PR China
| |
Collapse
|
44
|
Delgado-Tirado S, Amarnani D, Zhao G, Rossin EJ, Eliott D, Miller JB, Greene WA, Ramos L, Arevalo-Alquichire S, Leyton-Cifuentes D, Gonzalez-Buendia L, Isaacs-Bernal D, Whitmore HAB, Chmielewska N, Duffy BV, Kim E, Wang HC, Ruiz-Moreno JM, Kim LA, Arboleda-Velasquez JF. Topical delivery of a small molecule RUNX1 transcription factor inhibitor for the treatment of proliferative vitreoretinopathy. Sci Rep 2020; 10:20554. [PMID: 33257736 PMCID: PMC7705016 DOI: 10.1038/s41598-020-77254-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is the leading cause of retinal detachment surgery failure. Despite significant advances in vitreoretinal surgery, it still remains without an effective prophylactic or therapeutic medical treatment. After ocular injury or retinal detachment, misplaced retinal cells undergo epithelial to mesenchymal transition (EMT) to form contractile membranes within the eye. We identified Runt-related transcription factor 1 (RUNX1) as a gene highly expressed in surgically-removed human PVR specimens. RUNX1 upregulation was a hallmark of EMT in primary cultures derived from human PVR membranes (C-PVR). The inhibition of RUNX1 reduced proliferation of human C-PVR cells in vitro, and curbed growth of freshly isolated human PVR membranes in an explant assay. We formulated Ro5-3335, a lipophilic small molecule RUNX1 inhibitor, into a nanoemulsion that when administered topically curbed the progression of disease in a novel rabbit model of mild PVR developed using C-PVR cells. Mass spectrometry analysis detected 2.67 ng/mL of Ro5-3335 within the vitreous cavity after treatment. This work shows a critical role for RUNX1 in PVR and supports the feasibility of targeting RUNX1 within the eye for the treatment of an EMT-mediated condition using a topical ophthalmic agent.
Collapse
Affiliation(s)
- Santiago Delgado-Tirado
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Guannan Zhao
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Elizabeth J Rossin
- Retina Service, Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Dean Eliott
- Retina Service, Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - John B Miller
- Retina Service, Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Whitney A Greene
- Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, USA
| | - Leslie Ramos
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Said Arevalo-Alquichire
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
- Energy, Materials and Environment Group, Faculty of Engineering, Universidad de La Sabana, Chia, Colombia
| | - David Leyton-Cifuentes
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
- Universidad EIA, Envigado, Colombia
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Daniela Isaacs-Bernal
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
- Energy, Materials and Environment Group, Faculty of Engineering, Universidad de La Sabana, Chia, Colombia
| | - Hannah A B Whitmore
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Natalia Chmielewska
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
- Boston College, Boston, USA
| | - Brandon V Duffy
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
- Harvard College, Cambridge, USA
| | - Eric Kim
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA
| | - Heuy-Ching Wang
- Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, San Antonio, USA
| | - Jose M Ruiz-Moreno
- Department of Ophthalmology, Castilla La Mancha University, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
- Vissum Corporation, Alicante, Spain
| | - Leo A Kim
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA.
- Retina Service, Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA.
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Massachusetts Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, USA.
- Universidad EIA, Envigado, Colombia.
| |
Collapse
|
45
|
Liu S, Xie F, Gan L, Peng T, Xu X, Guo S, Fu W, Wang Y, Ouyang Y, Yang J, Wang X, Zheng Y, Zhang J, Wang H. Integration of transcriptome and cistrome analysis identifies RUNX1-target genes involved in pancreatic cancer proliferation. Genomics 2020; 112:5343-5355. [PMID: 33189780 DOI: 10.1016/j.ygeno.2020.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/09/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
The extremely high proliferation rate of tumor cells contributes to pancreatic cancer (PC) progression. Runt-related transcription factor 1(RUNX1), a key factor in hematopoiesis that was correlated with tumor progression. However, the role of RUNX1 in PC proliferation was still unclear. We found that RUNX1 was significantly upregulated in PC tissues and its expression was negatively associated with prognosis of PC patients in a multicenter analysis according to immunohistochemical (IHC). RUNX1 downregulation in PC resulted in a significantly reduced cell proliferation rate, which was consistent with in vivo subcutaneous tumor formation assay results. RNA-seq and ChIP-seq results revealed that a portion of target genes, including HAP1, GPRC5B, PTPN21, VHL and EN2, were regulated by RUNX1, a finding successfully validated by ChIP-qPCR, qRT-PCR and Western blot. Subsequently, IHC and proliferation assays showed these target genes to be dysregulated in PC, affecting tumor growth. Our data suggest that RUNX1 plays an oncogenic role in tumor proliferation and is a potential prognostic biomarker and therapeutic target for PC.
Collapse
Affiliation(s)
- Songsong Liu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Fuming Xie
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Lang Gan
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Tao Peng
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xuejun Xu
- Department of Hepatobiliary Surgery, General Hospital of Xinjiang Military Region of PLA, Xinjiang, PR China
| | - Shixiang Guo
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Wen Fu
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Yunchao Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yongsheng Ouyang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Jiali Yang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Xianxing Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Yao Zheng
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China.
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China; Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China.
| |
Collapse
|
46
|
Park M, Kim D, Moon K, Park T. Integrative Analysis of Multi-Omics Data Based on Blockwise Sparse Principal Components. Int J Mol Sci 2020; 21:E8202. [PMID: 33147797 PMCID: PMC7663540 DOI: 10.3390/ijms21218202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 01/14/2023] Open
Abstract
The recent development of high-throughput technology has allowed us to accumulate vast amounts of multi-omics data. Because even single omics data have a large number of variables, integrated analysis of multi-omics data suffers from problems such as computational instability and variable redundancy. Most multi-omics data analyses apply single supervised analysis, repeatedly, for dimensional reduction and variable selection. However, these approaches cannot avoid the problems of redundancy and collinearity of variables. In this study, we propose a novel approach using blockwise component analysis. This would solve the limitations of current methods by applying variable clustering and sparse principal component (sPC) analysis. Our approach consists of two stages. The first stage identifies homogeneous variable blocks, and then extracts sPCs, for each omics dataset. The second stage merges sPCs from each omics dataset, and then constructs a prediction model. We also propose a graphical method showing the results of sparse PCA and model fitting, simultaneously. We applied the proposed methodology to glioblastoma multiforme data from The Cancer Genome Atlas. The comparison with other existing approaches showed that our proposed methodology is more easily interpretable than other approaches, and has comparable predictive power, with a much smaller number of variables.
Collapse
Affiliation(s)
- Mira Park
- Department of Preventive Medicine, Eulji University, Daejeon 34824, Korea;
| | - Doyoen Kim
- Department of Statistics, Korea University, Seoul 02841, Korea; (D.K.); (K.M.)
| | - Kwanyoung Moon
- Department of Statistics, Korea University, Seoul 02841, Korea; (D.K.); (K.M.)
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
47
|
Critical role of HOX transcript antisense intergenic RNA (HOTAIR) in gliomas. J Mol Med (Berl) 2020; 98:1525-1546. [PMID: 32978667 DOI: 10.1007/s00109-020-01984-x] [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: 06/05/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Despite extensive research, gliomas are associated with high morbidity and mortality, mainly attributed to the rapid growth rate, excessive invasiveness, and molecular heterogeneity, as well as regenerative potential of cancer stem cells. Therefore, elucidation of the underlying molecular mechanisms and the identification of potential molecular diagnostic and prognostic biomarkers are of paramount importance. HOX transcript antisense intergenic RNA (HOTAIR) is a well-studied long noncoding RNA, playing an emerging role in tumorigenesis of several human cancers. A growing amount of preclinical and clinical evidence highlights the pro-oncogenic role of HOTAIR in gliomas, mainly attributed to the enhancement of proliferation and migration, as well as inhibition of apoptosis. In vitro and in vivo studies demonstrate that HOTAIR modulates the activity of specific transcription factors, such as MXI1, E2F1, ATF5, and ASCL1, and regulates the expression of cell cycle-associated genes along with related signaling pathways, like the Wnt/β-catenin axis. Moreover, it can interact with specific miRNAs, including miR-326, miR-141, miR-148b-3p, miR-15b, and miR-126-5p. Of importance, HOTAIR has been demonstrated to enhance angiogenesis and affect the permeability of the blood-tumor barrier, thus modulating the efficacy of chemotherapeutic agents. Herein, we provide evidence on the functional role of HOTAIR in gliomas and discuss the benefits of its targeting as a novel approach toward glioma treatment.
Collapse
|
48
|
Computational analysis and verification of molecular genetic targets for glioblastoma. Biosci Rep 2020; 40:225082. [PMID: 32469390 PMCID: PMC7298167 DOI: 10.1042/bsr20201401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant brain tumor with a poor prognosis. The initial treatment for high-grade gliomas is surgical excision. However, even with concomitant use of radiation or chemotherapy, patients are still prone to recurrence. The specific pathogenesis of GBM is still controversial. METHODS Differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) between GBM and normal brain tissues were screened. P-value was obtained by Bayes test based on the limma package. Statistical significance was set as P-value <0.05 and |Fold change (FC)| > 0.2 (GSE90886); P-value <0.05 and |FC| > 1 (GSE116520, GSE103228). Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG), protein-protein interaction (PPI) network were performed. Hub genes were selected from miRNA target genes and DEGs. GBM and normal brain tissues were extracted to verify the expression. RESULTS A total of 100 DEGs were overlapped in both datasets. Analysis of pathways and process enrichment tests indicated that ion transport, positive regulation of macromolecule metabolic process, cell cycle, axon guidance were enriched in the GBM. Sixteen hub genes were identified. Hub genes ADARB1 and neuropilin 1 (NRP1) were significantly associated with overall survival (OS) and disease-free survival (DFS) (P<0.05). Eukaryotic translation termination factor 1 (ETF1) was associated with DFS (P<0.05). CONCLUSIONS DEGs and DEMs were found between GBM tumor tissues and normal brain tissues. These biomarkers may be used as targets for early diagnosis and specific treatment.
Collapse
|
49
|
Na Y, Huang G, Wu J. The Role of RUNX1 in NF1-Related Tumors and Blood Disorders. Mol Cells 2020; 43:153-159. [PMID: 31940719 PMCID: PMC7057834 DOI: 10.14348/molcells.2019.0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/12/2019] [Indexed: 11/27/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder. NF1 patients are predisposed to formation of several type solid tumors as well as to juvenile myelomonocytic leukemia. Loss of NF1 results in dysregulation of MAPK, PI3K and other signaling cascades, to promote cell proliferation and to inhibit cell apoptosis. The RUNX1 gene is associated with stem cell function in many tissues, and plays a key role in the fate of stem cells. Aberrant RUNX1 expression leads to context-dependent tumor development, in which RUNX1 may serve as a tumor suppressor or an oncogene in specific tissue contexts. The co-occurrence of mutation of NF1 and RUNX1 is detected rarely in several cancers and signaling downstream of RAS-MAPK can alter RUNX1 function. Whether aberrant RUNX1 expression contributes to NF1-related tumorigenesis is not fully understood. This review focuses on the role of RUNX1 in NF1-related tumors and blood disorders, and in sporadic cancers.
Collapse
Affiliation(s)
- Youjin Na
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Gang Huang
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Pathology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 459, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 5267, USA
| |
Collapse
|