1
|
Jiang M, Zhang K, Wei G, Qi F, Yu D, Ma J, Zhang X, Chen L, Xie Y, Yu Z, Chen J, Chen D. HDAC4 super-enhancer drives CEBPB-mediated TWIST2 transcription to promote chemoresistance in LUAD. Cancer Lett 2025; 623:217716. [PMID: 40222483 DOI: 10.1016/j.canlet.2025.217716] [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: 12/16/2024] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Lung cancer remains one of the most prevalent malignancies worldwide. This study investigates the role of histone deacetylase 4 (HDAC4) in mediating chemoresistance in lung adenocarcinoma (LUAD). Super-enhancers (SEs), known to regulate aberrant gene expression, are critical drivers of tumor progression. We identified a specific super-enhancer region associated with HDAC4, referred to as HDAC4-SE. Among its nearby genes, TWIST2 emerged as a key player, strongly linked to chemoresistance and the epithelial-to-mesenchymal transition (EMT). We demonstrated that HDAC4-SE regulates TWIST2 expression, thereby contributing to chemoresistance in LUAD. Through bioinformatics analysis, we identified transcription factors binding to both the promoter of TWIST2 and the activation region of HDAC4-SE, with CCAAT/enhancer-binding protein beta (CEBPB) identified as a central regulator. Chromatin immunoprecipitation (ChIP) assays confirmed that CEBPB binds to both the HDAC4-SE and the TWIST2 promoter. Additionally, our investigation into the involvement of long non-coding RNAs (lncRNAs) revealed that LINC01940 might mediate the regulatory effects of HDAC4-SE on downstream genes. In conclusion, we uncovered a novel HDAC4-SE/LINC01940/CEBPB/TWIST2 signaling pathway that drives chemoresistance and tumor progression in LUAD. This pathway offers promising insights into potential therapeutic targets to overcome chemoresistance in lung cancer.
Collapse
Affiliation(s)
- Min Jiang
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China
| | - Kai Zhang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Guohao Wei
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, 210003, Nanjing, China
| | - Feng Qi
- Department of Pharmacy, the Yancheng Clinical College of Xuzhou Medical University, the First People's Hospital of Yancheng, No.166 West Yulong Road, Yancheng, 224006, Jiangsu, China
| | - Danlei Yu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China
| | - Jingjing Ma
- Department of Pharmacy, Dushu Lake Hospital, Soochow University, No.9 Chongwen Road, Suzhou, 215100, Jiangsu, China
| | - Xiaofei Zhang
- Department of Medical Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.160 Pujian Road, Pudong New District, Shanghai, 200127, China
| | - Longbang Chen
- Department of Medical Oncology, Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Yuhao Xie
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, 11439, USA
| | - Zhengyuan Yu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, No.188 Shizi Street, Gusu District, Suzhou, 215006, Jiangsu, China.
| | - Jing Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, No.138 Xianlin Avenue, Nanjing, 210023, Jiangsu, China; Jiangsu Province Engineering Research Center of TCM Health Preservation, Nanjing, Jiangsu, China.
| | - Dongqin Chen
- Department of Medical Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.160 Pujian Road, Pudong New District, Shanghai, 200127, China; Department of Oncology, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, No. 666 Shengli Road, Nantong, 226000, Jiangsu, China; Department of Medical Oncology, Baoshan Branch, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No.1058 Huanzhen North Road, Baoshan District, Shanghai, 200444, China.
| |
Collapse
|
2
|
Huang B, Li X. Mechanisms of GPM6A in Malignant Tumors. Cancer Rep (Hoboken) 2025; 8:e70137. [PMID: 39957375 PMCID: PMC11831008 DOI: 10.1002/cnr2.70137] [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: 06/28/2024] [Revised: 09/20/2024] [Accepted: 01/25/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Glycoprotein M6A (GPM6A) encodes a transmembrane protein, expressing in large quantities on the cell surface of central nervous system (CNS) neurons. GPM6A acts importantly in neurodevelopment by modulating neuronal differentiation, migration, axon growth, synaptogenesis, and spine formation, but its role in malignancy remains controversial and requires further research. This article reviewed the mechanisms of GPM6A in colorectal cancer, liver cancer, lung cancer, glioblastoma, and other malignant tumors, and made a "one-stop" summary of the relevant mechanisms. RECENT FINDINGS Researches have indicated that GPM6A is related to malignant tumors. It affects epithelial-mesenchymal transition and induces the formation of filopodia, participating in the adhesion, migration, and metastasis of cancer cells. Its role in malignant tumors remains controversial, however. On the one hand, GPM6A may have carcinogenic properties and is related to poor prognosis of malignant tumors. It is highly expressed in lymphoblastic leukemia and is a potential oncogene. It also shows carcinogenic properties in colorectal cancer, glioblastoma, gonadotroph adenomas and so on. On the other hand, the expression of GPM6A decreases in lung adenocarcinoma, liver cancer, thyroid cancer, and so forth as the tumor progresses, and it can inhibit the progression of malignant tumors by inhibiting some signaling pathways, suggesting that it may be a tumor suppressor gene. CONCLUSION Carcinogenic or tumor suppressive? Although the biological function of GPM6A in the development of malignant tumors is still unclear, according to the current research progress, it is still expected to become an effective molecular marker for predicting tumor occurrence, metastasis and prognosis, as well as a new target for diagnosis and treatment.
Collapse
Affiliation(s)
- Bei Huang
- Operation Management and Evaluation Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University)Ministry of EducationChengduSichuanPeople's Republic of China
- Emergency Department, West China Second University HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
3
|
Campanharo CV, Dos Santos Silveira LV, Meira DD, Casotti MC, Altoé LSC, Louro ID, Gonçalves AFM, Machado AM, Paiva BS, de Souza Inocencio E, Rocha FVV, Pesente F, de Castro GDSC, da Paixão JPDS, Bourguignon JHB, Carneiro JS, de Oliveira JR, de Souza Freire P, Zamprogno SB, Dos Santos Uchiya T, de Paula Rezende T, de Pádua Sanders Medeiros V. Pan-cancer and multiomics: advanced strategies for diagnosis, prognosis, and therapy in the complex genetic and molecular universe of cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03819-4. [PMID: 39725831 DOI: 10.1007/s12094-024-03819-4] [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: 09/09/2024] [Accepted: 11/23/2024] [Indexed: 12/28/2024]
Abstract
The pan-cancer and multi-omics approach is motivated by the genetic and molecular complexity inherent in the varied types of cancer. This method presents itself as a crucial resource for advancing early diagnosis, defining prognoses and identifying treatments that share common bases between different forms of tumors. The aim of this article is to explore pan-cancer analysis in conjunction with multi-omics strategies, evaluating laboratory, computational, clinical procedures and their consequences, as well as examining the tumor microenvironment, epigenetics and future directions of these technologies in patient management. To this end, a literature review was conducted using PUBMED, resulting in the selection of 260 articles, of which 81 were carefully chosen to support this analysis. The pan-cancer methodology is applied to the study of this microenvironment with the aim of investigating its common characteristics through multiomics data. The development of new therapies depends on understanding the oncogenic pathways associated with different cancers. Thus, the integration of multi-omics and pan-cancer analyzes offers an innovative perspective in the search for new control points, metabolic pathways and markers, in addition to facilitating the identification of patterns common to multiple cancer types, allowing the development of targeted treatments. In this way, the convergence of multiomics and clinical approaches promotes a broad view of cancer biology, leading to more effective and personalized therapies.
Collapse
Affiliation(s)
- Camilly Victória Campanharo
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Lívia Valle Dos Santos Silveira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil.
| | - Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - André Felipe Monteiro Gonçalves
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - André Manhães Machado
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Breno Sousa Paiva
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Ester de Souza Inocencio
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Fabio Victor Vieira Rocha
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Fellipe Pesente
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Giulia de Souza Cupertino de Castro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - João Pedro Dos Santos da Paixão
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - José Henrique Borges Bourguignon
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Júlia Salarini Carneiro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Juliana Ribeiro de Oliveira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Pâmela de Souza Freire
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Sophia Bridi Zamprogno
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Taissa Dos Santos Uchiya
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Thais de Paula Rezende
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| | - Vinícius de Pádua Sanders Medeiros
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Av. Fernando Ferrari, N. 514, Prédio Ciências Biológicas, Bloco A, Sala 106, Vitória, Espírito Santo, Brasil
| |
Collapse
|
4
|
Sun J, Shi S, Sun C, Wang J, Yang X, Yang Z, Xu J, Zhang S. Predictive nomogram of the clinical outcomes of colorectal cancer based on methylated SEPT9 and intratumoral IL-10 + Tregs infiltration. J Transl Med 2024; 22:861. [PMID: 39334238 PMCID: PMC11430755 DOI: 10.1186/s12967-024-05635-4] [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: 04/03/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Gene methylation and the immune-related tumor microenvironment (TME) are highly correlated in tumor progression and therapeutic efficacy. Although both of them can be used to predict the clinical outcomes of colorectal cancer (CRC) patients, their predictive value is still unsatisfactory. Whether a combination risk model comprising these two prediction parameters performs better predictive effectiveness than independent factor is still unclear. Methylated Septin9 (mSEPT9) is an early diagnosis biomarker of CRC, in this study, we aimed to investigate mSEPT9-related biomarkers of immunosuppressive TME and identify the value of the combination risk model in predicting the clinical outcomes of CRC. METHODS Immunofluorescence staining was performed to clarify the correlation between intratumoral IL-10+ Treg infiltration and mSEPT9 in peripheral blood. Survival time, response to 5-fluorouracil (5-FU)-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis were analyzed in study (197 CRC samples) and validation (195 CRC samples) sets to evaluate the efficacy of combination risk model. Potential mechanisms were explored by mRNA sequencing. RESULTS Hypermethylated SEPT9 in the peripheral blood of patients with CRC (stage I-III, and stage IV with resectable M1) before radical resection was positively correlated with high intratumoral IL-10+ Treg infiltration. The high-risk model revealed poor overall survival and progression-free survival, inferior therapeutic response to 5-FU-based chemotherapy and PD-1 blockade, and high probability of recurrence or metastasis. The underlying mechanisms may be associated with the increase in mSEPT9 and mediation of IL-10 via methionine metabolic reprogramming-induced infiltration of IL-10+ Tregs in the TME, which promotes tumor progression and resistance to 5-FU-based chemotherapy and PD-1 blockade. CONCLUSIONS The combination risk model of peripheral mSETP9 and intratumoral IL-10+ Treg infiltration in CRC can effectively predict prognosis, responsiveness to 5-FU-based chemotherapy and PD-1 blockade, and the probability of recurrence or metastasis. Therefore, this model can be used for precision treatment of CRC.
Collapse
Affiliation(s)
- Jie Sun
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Songli Shi
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300121, People's Republic of China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Zhengduo Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jing Xu
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
- Department of General Surgery, Tianjin University Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China.
| |
Collapse
|
5
|
Wang J, Wang J, Zhang J, Gong H, Li J, Song Y, Huang Y, Ma B, Gu W, Yang R. Association between the methylations of RUNX3 in peripheral blood and lung cancer: a case-control study. Biomarkers 2024; 29:343-351. [PMID: 38923933 DOI: 10.1080/1354750x.2024.2373714] [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: 02/06/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND RUNX3 is hypermethylated in multiple cancers. TIMP2 also functions as a regulator of tumors. However, there are only very few reports on the association of methylation of RUNX3 and TIMP2 with lung cancer (LC) in peripheral blood. METHODS 426 LC patients and 428 age- and sex-matched healthy controls were recruited. DNA methylation in blood was semi-quantitively assessed by mass spectrometry. For the association analysis, binary logistic regression analysis adjusted covariant was applied, and ORs were presented as per +10% methylation. RESULTS Hypermethylation of CpG_1, CpG_5 and CpG_8 in RUNX3 was significantly associated with LC (ORs = 1.45, 1.35 and 1.35, respectively, adjusted p < 0.05), and even stage I LC. The association between the three RUNX3 CpG sites and LC was enhanced by increased age (> 55 years, ORs ranged from 1.43 to 1.75, adjusted p < 0.05), male gender (ORs ranged from 1.47 to 1.59, adjusted p < 0.05) and tumor stage (stage II&III&IV, ORs ranged from 1.86 to 3.03, adjusted p < 0.05). CONCLUSIONS This study suggests a significant association between blood-based RUNX3 hypermethylation and LC, especially in elder people, in males and in LC patients with advanced stage.
Collapse
Affiliation(s)
- Jun Wang
- Research and Development Department, TANTICA Biotechnology (Shanghai) Co., Ltd, Shanghai, China
| | - Jue Wang
- Research and Development Department, TANTICA Biotechnology (Shanghai) Co., Ltd, Shanghai, China
| | - Jie Zhang
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Haixia Gong
- Department of Respiratory and Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinchang Li
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yakang Song
- Research and Development Department, TANTICA Biotechnology (Shanghai) Co., Ltd, Shanghai, China
| | - Yuyang Huang
- Department of Respiratory and Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Boyue Ma
- Department of Respiratory and Sleep Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Rongxi Yang
- Research and Development Department, TANTICA Biotechnology (Shanghai) Co., Ltd, Shanghai, China
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Shafi O, Siddiqui G, Jaffry HA. The benign nature and rare occurrence of cardiac myxoma as a possible consequence of the limited cardiac proliferative/ regenerative potential: a systematic review. BMC Cancer 2023; 23:1245. [PMID: 38110859 PMCID: PMC10726542 DOI: 10.1186/s12885-023-11723-3] [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: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions. METHODS Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes. RESULTS The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others. The Nkx2-5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2-5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential. CONCLUSION The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan.
| | - Ghazia Siddiqui
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| | - Hassam A Jaffry
- Sindh Medical College - Jinnah Sindh Medical University / Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
7
|
Vazana-Netzarim R, Elmalem Y, Sofer S, Bruck H, Danino N, Sarig U. Distinct HAND2/HAND2-AS1 Expression Levels May Fine-Tune Mesenchymal and Epithelial Cell Plasticity of Human Mesenchymal Stem Cells. Int J Mol Sci 2023; 24:16546. [PMID: 38003736 PMCID: PMC10672054 DOI: 10.3390/ijms242216546] [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: 09/21/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
We previously developed several successful decellularization strategies that yielded porcine cardiac extracellular matrices (pcECMs) exhibiting tissue-specific bioactivity and bioinductive capacity when cultured with various pluripotent and multipotent stem cells. Here, we study the tissue-specific effects of the pcECM on seeded human mesenchymal stem cell (hMSC) phenotypes using reverse transcribed quantitative polymerase chain reaction (RT-qPCR) arrays for cardiovascular related gene expression. We further corroborated interesting findings at the protein level (flow cytometry and immunological stains) as well as bioinformatically using several mRNA sequencing and protein databases of normal and pathologic adult and embryonic (organogenesis stage) tissue expression. We discovered that upon the seeding of hMSCs on the pcECM, they displayed a partial mesenchymal-to-epithelial transition (MET) toward endothelial phenotypes (CD31+) and morphologies, which were preceded by an early spike (~Day 3 onward after seeding) in HAND2 expression at both the mRNA and protein levels compared to that in plate controls. The CRISPR-Cas9 knockout (KO) of HAND2 and its associated antisense long non-coding RNA (HAND2-AS1) regulatory region resulted in proliferation arrest, hypertrophy, and senescent-like morphology. Bioinformatic analyses revealed that HAND2 and HAND2-AS1 are highly correlated in expression and are expressed in many different tissue types albeit at distinct yet tightly regulated expression levels. Deviation (downregulation or upregulation) from these basal tissue expression levels is associated with a long list of pathologies. We thus suggest that HAND2 expression levels may possibly fine-tune hMSCs' plasticity through affecting senescence and mesenchymal-to-epithelial transition states, through yet unknown mechanisms. Targeting this pathway may open up a promising new therapeutic approach for a wide range of diseases, including cancer, degenerative disorders, and aging. Nevertheless, further investigation is required to validate these findings and better understand the molecular players involved, potential inducers and inhibitors of this pathway, and eventually potential therapeutic applications.
Collapse
Affiliation(s)
- Rachel Vazana-Netzarim
- The Dr. Miriam and Sheldon Adelson School of Medicine, Department of Morphological Sciences and Teratology, Ariel University, Ariel 4070000, Israel; (R.V.-N.); (N.D.)
| | - Yishay Elmalem
- Department of Chemical Engineering, Faculty of Engineering, Ariel University, Ariel 4070000, Israel (S.S.); (H.B.)
| | - Shachar Sofer
- Department of Chemical Engineering, Faculty of Engineering, Ariel University, Ariel 4070000, Israel (S.S.); (H.B.)
| | - Hod Bruck
- Department of Chemical Engineering, Faculty of Engineering, Ariel University, Ariel 4070000, Israel (S.S.); (H.B.)
| | - Naama Danino
- The Dr. Miriam and Sheldon Adelson School of Medicine, Department of Morphological Sciences and Teratology, Ariel University, Ariel 4070000, Israel; (R.V.-N.); (N.D.)
| | - Udi Sarig
- The Dr. Miriam and Sheldon Adelson School of Medicine, Department of Morphological Sciences and Teratology, Ariel University, Ariel 4070000, Israel; (R.V.-N.); (N.D.)
- Department of Chemical Engineering, Faculty of Engineering, Ariel University, Ariel 4070000, Israel (S.S.); (H.B.)
| |
Collapse
|
8
|
Ma W, Han X, Shasaltaneh MD, Hosseinifard H, Maghsoudloo M, Zhang Y, Weng Q, Wang Q, Wen Q, Imani S. The p110α/ΔNp63α complex mutations in triple-negative breast cancer: Potential targets for transcriptional-based therapies. Tumour Biol 2023; 45:127-146. [PMID: 37980588 DOI: 10.3233/tub-230013] [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] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Hotspot mutations occurring in the p110α domain of the PIK3CA gene, specifically p110αH1047R/L increase tumor metastasis and cell motility in triple-negative breast cancer (TNBC). These mutations also affect the transcriptional regulation of ΔNp63α, a significant isoform of the p53 protein involved in cancer progression. This study attempts to investigate the transcriptional impact of p110αH1047R/L mutations on the PIK3CA/ΔNp63α complex in TNBC carcinogenesis. METHODS We performed site-directed mutagenesis to introduce p110αH1047R/L mutations and evaluated their oncogenic effects on the growth, invasion, migration, and apoptosis of three different TNBC cell lines in vitro. We investigated the impact of these mutations on the p110α/ΔNp63α complex and downstream transcriptional signaling pathways at the gene and protein levels. Additionally, we used bioinformatics techniques such as molecular dynamics simulations and protein-protein docking to gain insight into the stability and structural changes induced by the p110αH1047R/L mutations in the p110α/ΔNp63α complex and downstream signaling pathway. RESULTS The presence of PIK3CA oncogenic hotspot mutations in the p110α/ΔNp63α complex led to increased scattering of TNBC cells during growth, migration, and invasion. Our in vitro mutagenesis assay showed that the p110αH1047R/L mutations activated the PI3K-Akt-mTOR and tyrosine kinase receptor pathways, resulting in increased cell proliferation, invasion, and apoptosis in TNBC cells. These mutations decreased the repressing effect of ΔNp63α on the p110α kinase domain, leading to the enhancement of downstream signaling pathways of PI3K and tyrosine kinase receptors and oncogenic transformation in TNBC. Additionally, our findings suggest that the physical interaction between the DNA binding domain of ΔNp63α and the kinase domain of p110α may be partially impaired, potentially leading to alterations in the conformation of the p110α/ΔNp63α complex. CONCLUSION Our findings suggest that targeting the p110αH1047R/L mutations in TNBC could be a promising strategy for developing transcriptional-based therapies. Restoring the interaction between ΔNp63α and the p110α kinase domain, which is disrupted by these mutations, may provide a new approach to treating TNBC.
Collapse
Affiliation(s)
- Wenqiong Ma
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xingping Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | | - Hossein Hosseinifard
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuqin Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiao Weng
- Department of Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qingjing Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - QingLian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Moradi A, Shahsavari M, Gowdini E, Mohammadian K, Alizamir A, Khalilollahi M, Abgarmi ZM, Ganji SM. Consequences of aberrated DNA methylation in Colon Adenocarcinoma: a bioinformatic-based multi-approach. BMC Genom Data 2022; 23:83. [PMID: 36443682 PMCID: PMC9706923 DOI: 10.1186/s12863-022-01100-7] [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: 05/28/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION The biology of colorectal cancer (CRC) is remained to be elucidated. Numerous genetic and epigenetic modifications are in concert to create and progress CRC. DNA methylation as a principal epigenetic factor has gained increased attention and could be utilized for biological studies. This study aims to find novel methylated and downregulated genes with a focus on HAND2 in CRC and decipher the biological consequences. MATERIAL AND METHOD Data on DNA methylation from GEO and SMART databases and the expression GEPIA2 database were downloaded. Afterward, a set of hypermethylated and downregulated genes in CRC was chosen by overlapping genes. Consequently, HAND2 was selected as a key gene for further investigation and confirmed with cell lines methylation and expression data. The functions of HAND2 were further analyzed using gene ontology analyses and the protein-protein interaction network. RESULTS The methylation (p < 0.01) and expression (p < 0.01) of HAND2 are significantly varied in CRC compared to normal control. The correlation analysis (Pearson's correlation coefficient = -0.44, p = 6.6e-14) conveys that HAND2 significantly downregulated and has a reverse correlation with the methylation status of CpG islands. The biological process analysis of HAND2 target genes conveyed that disruption in HAND2 expression could dysregulate ERK1 and ERK2 signaling pathways. CONCLUSION Together, the findings showed that DNA hypermethylation of HAND2 was critical evidence in CRC. Further validation and prospective studies are needed to utilize HAND2 methylation as a promising biomarker.
Collapse
Affiliation(s)
- Arash Moradi
- grid.419420.a0000 0000 8676 7464Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran - Karaj Highway, Tehran, Iran
| | - Milad Shahsavari
- grid.411463.50000 0001 0706 2472Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Erfan Gowdini
- grid.419420.a0000 0000 8676 7464Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran - Karaj Highway, Tehran, Iran
| | - Kamal Mohammadian
- grid.411950.80000 0004 0611 9280Department of Radiation Oncology, Hamadan University of Medical Sciences, Mahdieh Center, Hamadan, Iran
| | - Aida Alizamir
- grid.411950.80000 0004 0611 9280Department of Pathology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Khalilollahi
- grid.411463.50000 0001 0706 2472Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Zahara Mohammadi Abgarmi
- grid.412266.50000 0001 1781 3962Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Shahla Mohammad Ganji
- grid.419420.a0000 0000 8676 7464Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran - Karaj Highway, Tehran, Iran
| |
Collapse
|
10
|
Yuan Z, Yu X, Chen W, Chen D, Cai J, Jiang Y, Liu X, Wu Z, Wang L, Grady WM, Wang H. Correction: Epigenetic silencing and tumor suppressor gene of HAND2 by targeting ERK signaling in colorectal cancer. Cell Commun Signal 2022; 20:113. [PMID: 35897069 PMCID: PMC9327259 DOI: 10.1186/s12964-022-00944-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Zixu Yuan
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China.
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D4-100, Seattle, WA, 98109, USA.
| | - Xihu Yu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenle Chen
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Daici Chen
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jian Cai
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingming Jiang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Xiaoxia Liu
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhijie Wu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Lei Wang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - William M Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D4-100, Seattle, WA, 98109, USA.
| | - Hui Wang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China.
- Guangdong Institute of Gastroenterology, The Sixth Afliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|