1
|
Rousseau L, Hajdu KL, Ho PC. Meta-epigenetic shifts in T cell aging and aging-related dysfunction. J Biomed Sci 2025; 32:51. [PMID: 40410784 PMCID: PMC12101013 DOI: 10.1186/s12929-025-01146-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
Epigenetic regulation, including DNA methylation and histone modifications, play a pivotal role in shaping T cell functionality throughout life. With aging, these epigenetic changes profoundly affect gene expression, altering T cell plasticity, activation, and differentiation. These modifications contribute significantly to immunosenescence, increasing susceptibility to infections, cancer, and autoimmune diseases. In CD8⁺ T cells, chromatin closure at key regulatory regions suppresses activation and migration, while chromatin opening in pro-inflammatory gene loci amplifies inflammation. These changes drive terminal differentiation, characterized by increased expression of senescence-associated markers, impaired migration and loss of epigenetic plasticity. CD4⁺ T cells experience fewer but critical epigenetic alterations, including disrupted pathways, a skewed Th1/Th2 balance, and reduced Treg functionality. These epigenetic changes, compounded by metabolic dysfunctions, such as mitochondrial deficiency and oxidative stress, impair T-cell adaptability and resilience in the aging organism. Therefore, understanding the interplay between epigenetic and metabolic factors in T cell aging offers promising therapeutic opportunities to mitigate immunosenescence and enhance immune function in aging populations. This review explores the interplay between DNA methylation, histone alterations, and metabolic changes underlying T cell aging.
Collapse
Affiliation(s)
- Lorène Rousseau
- Department of Fundamental Oncology, University of Lausanne, 155 Ch. Des Boveresses, 1066, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland
| | - Karina L Hajdu
- Department of Fundamental Oncology, University of Lausanne, 155 Ch. Des Boveresses, 1066, Epalinges, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 155 Ch. Des Boveresses, 1066, Epalinges, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Épalinges, Switzerland.
| |
Collapse
|
2
|
Reed VL, Lalu E, Yoon L, Fultang N, Peethambaran B. Uncovering a Novel Role of ROR1 in the Epigenetic Regulation of Tumor Suppressor Gene CREB3L1 in Triple-Negative Breast Cancer Cells. Biomolecules 2025; 15:734. [PMID: 40427627 PMCID: PMC12109183 DOI: 10.3390/biom15050734] [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: 03/21/2025] [Revised: 04/25/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
A characteristic of triple-negative breast cancer (TNBC) is the epigenetic regulation of tumor suppressor genes, leading to TNBC heterogeneity and treatment resistance in patients. TNBC exhibits high methylation rates, leading to the silencing of numerous tumor suppressor genes. DNA methyltransferase inhibitors (DNMTis) have shown limited clinical efficacy in TNBC treatment. This study aims to uncover a target that could be used to reverse the epigenetic silencing of tumor suppressor genes in TNBC. The Western blot analysis demonstrated that ROR1 knockdown, an oncofetal gene, reduced DNMT3A and DNMT3B protein expression in the TNBC cell lines MDA-MB-231 and HCC1806, as well as a non-malignant breast cell line, MCF10A. The reduced representation bisulfite sequencing (RRBS) analysis identified differential methylation of CREB3L1 when ROR1 is knocked down in TNBC cell lines. CREB3L1 is a transcription factor that plays tumor-suppressive roles in TNBC and is commonly epigenetically silenced in patients. This study shows that ROR1 requires pSTAT3 activation to upregulate DNMT3A and DNMT3B expression to induce CREB3L1 epigenetic silencing in TNBC. ROR1 knockdown resulted in the re-expression of CREB3L1 in TNBC cells. The data provide evidence that ROR1 inhibition, in combination with DNMTis, could enhance patient outcomes as a therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Victoria L. Reed
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| | - Eric Lalu
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| | - Leena Yoon
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
- Cancer Biology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Norman Fultang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Bela Peethambaran
- Department of Biology, St. Joseph’s University, Philadelphia, PA 19131, USA; (V.L.R.); (E.L.); (L.Y.)
| |
Collapse
|
3
|
Baldassarre G, L de la Serna I, Vallette FM. Death-ision: the link between cellular resilience and cancer resistance to treatments. Mol Cancer 2025; 24:144. [PMID: 40375296 PMCID: PMC12080166 DOI: 10.1186/s12943-025-02339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
One of the key challenges in defeating advanced tumors is the ability of cancer cells to evade the selective pressure imposed by chemotherapy, targeted therapies, immunotherapy and cellular therapies. Both genetic and epigenetic alterations contribute to the development of resistance, allowing cancer cells to survive initially effective treatments. In this narration, we explore how genetic and epigenetic regulatory mechanisms influence the state of tumor cells and their responsiveness to different therapeutic strategies. We further propose that an altered balance between cell growth and cell death is a fundamental driver of drug resistance. Cell death programs exist in various forms, shaped by cell type, triggering factors, and microenvironmental conditions. These processes are governed by temporal and spatial constraints and appear to be more heterogeneous than previously understood. To capture the intricate interplay between death-inducing signals and survival mechanisms, we introduce the concept of Death-ision. This framework highlights the dynamic nature of cell death regulation, determining whether specific cancer cell clones evade or succumb to therapy. Building on this understanding offers promising strategies to counteract resistant clones and enhance therapeutic efficacy. For instance, combining DNMT inhibitors with immune checkpoint blockade may counteract YAP1-driven resistance or the use of transcriptional CDK inhibitors could prevent or overcome chemotherapy resistance. Death-ision aims to provide a deeper understanding of the diversity and evolution of cell death programs, not only at diagnosis but also throughout disease progression and treatment adaptation.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, 33081, Italy.
| | - Ivana L de la Serna
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - François M Vallette
- Centre de Recherche en Cancérologie et Immunologie Intégrées Nantes Angers (CRCI2 NA), INSERM UMR1307/CNRS UMR 6075/Nantes Université/Univ. Angers. Nantes, 44007, Nantes, France.
- Institut de Cancérologie de L'Ouest (ICO), 44085, Saint-Herblain, France.
| |
Collapse
|
4
|
Binhassan S, Samman M, Al-Sheikh Y, Alshakweer W, Alhalouli T, Alshamlan N, Abudawood M. Revisiting the molecular landscape of Rosai-Dorfman disease: insights from whole exome sequencing of Saudi patients. Front Oncol 2025; 15:1556830. [PMID: 40406258 PMCID: PMC12094912 DOI: 10.3389/fonc.2025.1556830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Rosai-Dorfman disease (RDD) has traditionally been viewed as a reactive histiocytic disorder, defined by its unique clinical features and immunophenotype. However, recent genetic studies suggest a more complex molecular landscape, challenging the notion of RDD as solely reactive and hinting at a possible neoplastic component. Mutations in MAPK/ERK pathway genes, such as KRAS and MAP2K1, have been observed in up to 33% of cases. Additional genetic alterations in cell cycle regulation, DNA repair, and other processes, along with low-frequency BRAF mutations, further emphasize this complexity. To better understand the molecular basis of RDD and enhance diagnostic precision, we conducted whole exome sequencing (WES) on seven Saudi patients with RDD, comparing their genetic profiles with existing literature. While no kinase driver mutations were detected, our analysis revealed thirteen distinct mutations. Recurrent mutations were observed in CD207 and TDG, each found in six patients. CD207 is linked to antigen processing, while TDG is associated with DNA repair. MUC4 and PDS5A mutations, related to cell cycle regulation, were each identified in three patients. DNMT3A mutation, affecting DNA methylation, was found in two patients. Single mutations were observed in BRCA1, LATS2, ATM, USP35, and CIC, associated with DNA repair, the ubiquitin proteasome pathway, and transcriptional regulation respectively. These findings offer insights into the genetic makeup of RDD, revealing candidate genes and expanding our understanding of the disease's molecular complexities. By uncovering these genetic markers, this study contributes to the ongoing efforts to develop more accurate diagnostic tools and refine the classification of RDD, paving the way for improved patient care and disease management.
Collapse
Affiliation(s)
- Sarah Binhassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| | - Manar Samman
- Molecular Pathology (Genetics) Section, Pathology and Clinical Laboratory Medicine Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Yazeed Al-Sheikh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Wafaa Alshakweer
- Department of Pathology and Laboratory Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tahani Alhalouli
- Department of Pathology and Laboratory Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Najd Alshamlan
- Department of Pathology and Laboratory Medicine, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | - Manal Abudawood
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
- Central Research Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Zhang J, Chen T, Wu W, Hu C, Wang B, Jia X, Ye M. Carbonyl reductase 4 suppresses colorectal cancer progression through the DNMT3B/CBR4/FASN/mTOR axis. Cancer Cell Int 2025; 25:146. [PMID: 40234909 PMCID: PMC11998200 DOI: 10.1186/s12935-025-03776-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
Lipid metabolism is implicated in the initiation and progression of human colorectal cancer (CRC). Carbonyl reductase 4 (CBR4), a member of the carbonyl reductase family, plays a role in the biosynthesis of fatty acids. However, its involvement in CRC remains poorly understood. In this study, we aim to explore the function of CBR4 in CRC. Our findings indicated that the expression of CBR4 was significantly reduced in CRC tissues. Functional analyses revealed that CBR4 functions to inhibit cell proliferation, colony formation, migration, invasion, and tumor growth in vivo. Mechanistically, CBR4 interacts with fatty acid synthase (FASN), activating the ubiquitin-proteasome pathway, which leads to a reduction in FASN expression, thereby inhibiting the mTOR pathway and curtailing CRC development. Orlistat, a known FASN inhibitor, demonstrated anti-cancer properties both in vitro and in vivo. Additionally, DNMT3B, a DNA methyltransferase, contributed to the down-regulation of CBR4 by inducing methylation in the promoter region. In summary, our findings suggest that the DNMT3B/CBR4/FASN/mTOR signaling pathway is crucial in the advancement of CRC, and elucidate the potential mechanism by which enzymatic carbonyl reduction and lipid metabolism may be connected to CRC progression, offering a novel therapeutic strategy for its clinical management.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Medical Image, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, NO.157 Daming Road, Nanjing, 210022, China
| | - Tiaotiao Chen
- Department of Geriatrics, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Wencheng Wu
- Department of Pathology, Changzhou First People's Hospital, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| | - Chunhua Hu
- Neuroendocrine Tumor Diagnosis and Treatment Center of Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University; Neuroendocrine Tumor Diagnosis and Treatment Center of Jiangsu Province; Institute of Neuroendocrine Tumor of Collaborative Innovation Center for Cancer Personalized Medicine of Jiangsu Province; Institute of Neuroendocrine Tumor of Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, 210029, China
| | - Bangting Wang
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofeng Jia
- Department of Medical Image, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, NO.157 Daming Road, Nanjing, 210022, China.
| | - Mujie Ye
- Neuroendocrine Tumor Diagnosis and Treatment Center of Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University; Neuroendocrine Tumor Diagnosis and Treatment Center of Jiangsu Province; Institute of Neuroendocrine Tumor of Collaborative Innovation Center for Cancer Personalized Medicine of Jiangsu Province; Institute of Neuroendocrine Tumor of Nanjing Medical University, NO.300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
6
|
Cui J, Ma N, Li X, Chen X, Zhang J, Zhang W, Li H. Morphine Contributes to Epithelial-Mesenchymal Transition in Triple-Negative Breast Cancer Cells by Blocking COX-2 Methylation via Regulating the miR-23a-3p/DNMT3A Feedback. Cell Biochem Biophys 2025:10.1007/s12013-025-01749-8. [PMID: 40227561 DOI: 10.1007/s12013-025-01749-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
To investigate the effects and mechanisms of morphine on epithelial-mesenchymal transformation (EMT) in triple-negative breast cancer (TNBC). The levels of miR-23a-3p, DNMT3A, and COX-2 in tumor tissues from metastatic TNBC patients treated with morphine were assessed using qRT-PCR. Functional assays assessed morphine's impact on TNBC cell malignancy. Dual luciferase reporter and RNA pull-down assays investigated the interaction between miR-23a-3p and DNMT3A. miR-23a-3p inhibitor and DNMT3A siRNA were transfected into TNBC cells. Protein expression was analyzed by Western blot. Methylation status of miR-23a-3p and COX-2 was assessed via methylation-specific PCR. Rescue experiments were performed to research whether morphine modulates EMT in TNBC through COX-2 methylation regulation via the miR-23a-3p/DNMT3A feedback loop. The effects of morphine on TNBC in nude mice xenotransplantation were studied. In metastatic TNBC patients treated with morphine, miR-23a-3p and COX-2 expression were elevated, and DNMT3A levels were reduced. In TNBC cells, morphine enhanced migration, invasion, and EMT, and suppressed apoptosis. It upregulated miR-23a-3p and COX-2; downregulated DNMT3A; and inhibited methylation of miR-23a-3p and COX-2. miR-23a-3p directly inhibited DNMT3A expression. In morphine-treated TNBC cells, silencing DNMT3A reduced methylation of miR-23a-3p and COX-2. miR-23a-3p inhibitor suppressed migration, invasion, and EMT, and promoted apoptosis; however, these effects were reversed by DNMT3A silencing. In vivo, morphine promoted tumor EMT and metastasis in TNBC; reduced miR-23a-3p and COX-2 methylation; and decreased DNMT3A expression. Morphine accelerated EMT in TNBC by inhibiting COX-2 methylation through the miR-23a-3p/DNMT3A loop.
Collapse
Affiliation(s)
- Jian Cui
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Nina Ma
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Xiaohui Li
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Xuexin Chen
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Junxia Zhang
- Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Wenjuan Zhang
- Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Hong Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China.
| |
Collapse
|
7
|
Lorenzo PM, Izquierdo AG, Rodriguez-Carnero G, Costa-Fraga N, Díaz-Lagares A, Porca C, de Luis D, Tejera C, De Paz L, Cueva J, Bellido D, Crujeiras AB. Nutritional ketosis modulates the methylation of cancer-related genes in patients with obesity and in breast cancer cells. J Physiol Biochem 2025:10.1007/s13105-025-01076-9. [PMID: 40140215 DOI: 10.1007/s13105-025-01076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
Scientific evidence demonstrates that a very low-calorie ketogenic diet (VLCKD) is effective and beneficial in the treatment of obesity, capable of reversing the methylome associated with obesity and has immunomodulatory capacity. This effect is in part promoted by nutritional ketosis and could be involved in counteracting obesity-related cancer. The aim of this study was to evaluate the effect of nutritional ketosis on the methylation of genes related to tumor processes in patients with obesity and in breast cancer cells. Based on methylome data (Infinium MethylationEPIC BeadChip, Illumina) from patients with obesity treated with a VLCKD for weight loss (n = 10; n = 5 women, age = 48.8 ± 9.20 years, BMI = 32.9 ± 1.4 kg/m2), genes belonging to cancer-related pathways were specifically evaluated and further validated in vitro in MDA-MB-231 (triple negative) and MCF7 (RE positive) breast tumor cells pretreated for 72 h with βOHB, the main ketone body, secretome from visceral (VATs) or subcutaneous (SATs) adipose tissue of patients with obesity. The cell tumoral phenotype was evaluated by proliferation assay and expression of cancer-related genes. VLCKD-induced nutritional ketosis promoted changes in the methylation of 18 genes (20 CpGs; 17 hypomethylated, 3 hypermethylated) belonged to cancer-related pathways with MAPK10, CCN1, CTNNA2, LAMC3 and GLI2 being the most representative genes. A similar pattern was observed in the MDA-MB-231 cells treated with β-OHB, without changes in MCF7. These epigenetic changes paralleled the tumoral phenotype modulated by the treatments. Taking together these results highlight the potential role of VLCKD as an adjuvant to anticancer treatment in groups more susceptible to the development of cancer such as patients with obesity, exerting epigenetic regulation through nutritional ketosis and weight loss.
Collapse
Affiliation(s)
- Paula M Lorenzo
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBERobn), Madrid, Spain
| | - Andrea G Izquierdo
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBERobn), Madrid, Spain
| | - Gemma Rodriguez-Carnero
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBERobn), Madrid, Spain
- Endocrinology and Nutrition Department, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Nicolas Costa-Fraga
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), ISCIII, Madrid, Spain
| | - Angel Díaz-Lagares
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET), Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), ISCIII, Madrid, Spain
- Department of Clinical Analysis, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Cristina Porca
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- Endocrinology and Nutrition Department, Complejo Hospitalario Universitario de Ferrol (CHUF/SERGAS), Ferrol, Spain
| | - Daniel de Luis
- Center of Investigation of Endocrinology and Nutrition, Medicine School, Department of Endocrinology and Investigation, Hospital Clinico Universitario, University of Valladolid, Valladolid, Spain
| | - Cristina Tejera
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- Endocrinology and Nutrition Department, Complejo Hospitalario Universitario de Ferrol (CHUF/SERGAS), Ferrol, Spain
| | - Laura De Paz
- Medical Oncology Department, Complejo Hospitalario Universitario de Ferrol (CHUF/SERGAS), Ferrol, Spain
| | - Juan Cueva
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), ISCIII, Madrid, Spain
- Medical Oncology Department, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Diego Bellido
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain
- Endocrinology and Nutrition Department, Complejo Hospitalario Universitario de Ferrol (CHUF/SERGAS), Ferrol, Spain
| | - Ana B Crujeiras
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS), Unidad de Epigenomica. Complejo Hospitalario Universitario de Santiago de Compostela (CHUS/SERGAS), Travesía da Choupana Street s/n, Santiago de Compostela, La Coruña, 15706, Spain.
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBERobn), Madrid, Spain.
| |
Collapse
|
8
|
Cortellesi E, Savini I, Veneziano M, Gambacurta A, Catani MV, Gasperi V. Decoding the Epigenome of Breast Cancer. Int J Mol Sci 2025; 26:2605. [PMID: 40141248 PMCID: PMC11942310 DOI: 10.3390/ijms26062605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Breast cancer (BC) is the most prevalent malignancy among women, characterized by extensive heterogeneity stemming from molecular and genetic alterations. This review explores the intricate epigenetic landscape of BC, highlighting the significant role of epigenetic modifications-particularly DNA methylation, histone modifications, and the influence of non-coding RNAs-in the initiation, progression, and prognosis of the disease. Epigenetic alterations drive crucial processes, including gene expression regulation, cell differentiation, and tumor microenvironment interactions, contributing to tumorigenesis and metastatic potential. Notably, aberrations in DNA methylation patterns, including global hypomethylation and hypermethylation of CpG islands, have been associated with distinct BC subtypes, with implications for early detection and risk assessment. Furthermore, histone modifications, such as acetylation and methylation, affect cancer cell plasticity and aggressiveness by profoundly influencing chromatin dynamics and gene transcription. Finally, non-coding RNAs contribute by modulating epigenetic machinery and gene expression. Despite advances in our knowledge, clinical application of epigenetic therapies in BC is still challenging, often yielding limited efficacy when used alone. However, combining epi-drugs with established treatments shows promise for enhancing therapeutic outcomes. This review underscores the importance of integrating epigenetic insights into personalized BC treatment strategies, emphasizing the potential of epigenetic biomarkers for improving diagnosis, prognosis, and therapeutic response in affected patients.
Collapse
Affiliation(s)
- Elisa Cortellesi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Isabella Savini
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Matteo Veneziano
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Alessandra Gambacurta
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
- NAST Centre (Nanoscience & Nanotechnology & Innovative Instrumentation), Tor Vergata University of Rome, 00133 Rome, Italy
| | - Maria Valeria Catani
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| | - Valeria Gasperi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (E.C.); (I.S.); (M.V.); (A.G.); (M.V.C.)
| |
Collapse
|
9
|
Wei Z, Kou Z, Luo Y, Cheng Y. DNA methyltransferase 3A: A prognostic biomarker and potential target for immunotherapy in gastric cancer. Medicine (Baltimore) 2025; 104:e41578. [PMID: 39960919 PMCID: PMC11835108 DOI: 10.1097/md.0000000000041578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
DNA methyltransferase 3A (DNMT3A) has been associated with the occurrence or progression of various tumors, including gastric cancer. However, the role of DNMT3A in the efficacy of immune-cell infiltration in the tumor microenvironment and immunotherapy in gastric cancer remains less explored. DNMT3A expression level was analyzed using TIMER 2.0, Sangerbox 3.0, and The Cancer Genome Atlas database and further verified by immunohistochemical staining and RT-qPCR. The UALCAN, chi-square test, and Kaplan-Meier plotter databases were performed to assess the correlation of DNMT3A with clinicopathological characteristics and prognosis. The GeneMANIA database, STRING database, and R package were used to construct a DNMT3A co-expression gene network. Gene set enrichment analysis was used to identify the signaling pathways related to DNMT3A expression. The correlations between DNMT3A and cancer immune infiltrates were investigated using TIMER 2.0, Sangerbox 3.0, Kaplan-Meier Plotter, R package, and TISIDB databases. The TISIDB database and R package were used to construct the correlation between DNMT3A and immunomodulators and Immune cell Proportion Score. The association of DNMT3A expression with tumor mutational burden (TMB), microsatellite instability, and tumor dryness was evaluated using the TMB function of the R package, TIMER 2.0. Finally, the biological function of DNMT3A in gastric cancer cells was further assessed by CCK-8, cloning formation, and transwell assay. DNMT3A expression was remarkably upregulated in gastric cancer. The high expression of DNMT3A was associated with poor clinical features and poor survival in patients with gastric cancer. Moreover, gene set enrichment analyses showed that DNMT3A and its related genes were involved in various pathways that promoted cancer occurrence and progression by influencing the tumor microenvironment. Finally, DNMT3A was significantly related to tumor-infiltrating immune cells, immunomodulators, TMB, microsatellite instability, and immune checkpoints in gastric cancer. Moreover, knockdown of DNMT3A reduced the proliferation and migration of gastric cancer cells. Our findings highlight the potential of DNMT3A as a prognosis biomarker and an immunotherapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Zijie Wei
- College of Basic Medicine, Chengde Medical University, Chengde, Hebei, China
| | - Ziqian Kou
- College of Basic Medicine, Chengde Medical University, Chengde, Hebei, China
| | - Yun Luo
- College of Basic Medicine, Chengde Medical University, Chengde, Hebei, China
| | - Yu Cheng
- College of Basic Medicine, Chengde Medical University, Chengde, Hebei, China
| |
Collapse
|
10
|
Zhang M, Ma X, Wang Z, Han Y, Jia Z, Chen D, Xu Y, Qiao Z, Jiang X, Wang L, Jiang H, Yu M, Li Y, Shen Y. Genome-wide association analysis study on host resistance against the Aeromonas veronii of largemouth bass Micropterus salmoides. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110093. [PMID: 39736405 DOI: 10.1016/j.fsi.2024.110093] [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: 09/01/2024] [Revised: 11/19/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025]
Abstract
Largemouth bass (Micropterus salmoides) has become one of the most important freshwater economic fish farmed almost all over China in recent years. At the same time, the increasing outbreaks of diseases in its aquaculture process have caused substantial economic losses to this industry. However, at present, the genetic basis of disease resistance, including resistance against Aeromonas veronii infection, in largemouth bass is very limited. Therefore, a genome-wide association study (GWAS) on host resistance against the A. veronii of largemouth bass was conducted in the present study. A total of 627 largemouth bass were artificially challenged by A. veronii, among which 160 of the earliest deaths and 173 of the final survivals were genotyped. A total of 3076 high-quality SNPs were used for further analysis employing two analysis models, of which six shared SNPs were finally identified as significant molecular markers with the explaining phenotypic variance ranging from 2.28 % to 8.95 %. Furthermore, seven candidate genes were identified, including one gene, T-cell surface antigen CD2, which is directly involved in T cell activation and the cellular immune response. Additionally, the other identified genes play roles in critical processes such as cell survival, inflammatory responses, and signal transduction. This study lays a genetic foundation for research on largemouth bass disease resistance and studies related to A. veronii. It also contributes significantly to the future development of the commercial production of largemouth bass.
Collapse
Affiliation(s)
- Meng Zhang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China; Observation and Research Station on Water Ecosystem in Danjiangkou Reservoir of Henan Province, Nanyang, 474450, China.
| | - Xiao Ma
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zerui Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yuqing Han
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zhilin Jia
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Dongcai Chen
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yue Xu
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zhigang Qiao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Xinyu Jiang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Lei Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Hongxia Jiang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Miao Yu
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yongjing Li
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yawei Shen
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China; Observation and Research Station on Water Ecosystem in Danjiangkou Reservoir of Henan Province, Nanyang, 474450, China; Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
11
|
Djamgoz MBA. Stemness of Cancer: A Study of Triple-negative Breast Cancer From a Neuroscience Perspective. Stem Cell Rev Rep 2025; 21:337-350. [PMID: 39531198 PMCID: PMC11872763 DOI: 10.1007/s12015-024-10809-0] [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] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Stemness, giving cancer cells massive plasticity enabling them to survive in dynamic (e.g. hypoxic) environments and become resistant to treatment, especially chemotherapy, is an important property of aggressive tumours. Here, we review some essentials of cancer stemness focusing on triple-negative breast cancer (TNBC), the most aggressive form of all breast cancers. TNBC cells express a range of genes and mechanisms associated with stemness, including the fundamental four "Yamanaka factors". Most of the evidence concerns the transcription factor / oncogene c-Myc and an interesting case is the expression of the neonatal splice variant of voltage-gated sodium channel subtype Nav1.5. On the whole, measures that reduce the stemness make cancer cells less aggressive, reducing their invasive/metastatic potential and increasing/restoring their chemosensitivity. Such measures include gene silencing techniques, epigenetic therapies as well as novel approaches like optogenetics aiming to modulate the plasma membrane voltage. Indeed, simply hyperpolarizing their membrane potential can make stem cells differentiate. Finally, we give an overview of the clinical aspects and exploitation of cancer/TNBC stemness, including diagnostics and therapeutics. In particular, personalised mRNA-based therapies and mechanistically meaningful combinations are promising and the emerging discipline of 'cancer neuroscience' is providing novel insights to both fundamental issues and clinical applications.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
12
|
Jamialahmadi K, Noruzi S. Matrix metalloproteinases, chemoresistance and cancer. PATHOPHYSIOLOGICAL ASPECTS OF PROTEASES IN CANCER 2025:385-409. [DOI: 10.1016/b978-0-443-30098-1.00023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Kaušylaitė MM, Jurevičė J, Korobeinikova E, Gudaitienė J, Juozaitytė E, Ugenskienė R. DNMT1 rs2228611, rs2228612 and DNMT3A rs2276598, rs752208 Polymorphisms and Their Association with Breast Cancer Phenotype and Prognosis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1902. [PMID: 39597087 PMCID: PMC11596321 DOI: 10.3390/medicina60111902] [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: 10/23/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: Breast cancer is a leading cause of cancer-related deaths globally. This study investigates the impact of genetic polymorphisms in DNA methyltransferases (DNMT1 and DNMT3A) on breast cancer pathomorphology and patient prognosis. Specifically, we focused on DNMT1 polymorphisms rs2228611 and rs2228612 and DNMT3A polymorphisms rs2276598 and rs752208. Materials and Methods: Conducted at the Institute of Oncology of the Lithuanian University of Health Sciences, this study included 201 Lithuanian women with early-stage breast cancer. DNA was extracted from peripheral blood samples, and genotyping for the specified polymorphisms was performed using the PCR-RFLP assay. Statistical analyses were applied to evaluate associations between polymorphisms and clinicopathological characteristics. Results: The non-carriers of the DNMT1 rs2228611 G allele were less likely to be diagnosed at an older age, while the DNMT3A rs752208 T allele was linked to lower-grade tumors. Survival analysis indicated a potential relationship between DNMT3A rs752208 and overall survival, although no significant findings were observed in progression-free or metastasis-free survival. Conclusions: This study suggests that the DNMT1 and DNMT3A polymorphisms may influence breast cancer pathomorphology and prognosis. The DNMT1 rs2228611 G allele may be associated with earlier onset, and the DNMT3A rs752208 T allele might correlate with less aggressive tumors. These findings underscore the potential of DNMT gene polymorphisms as prognostic biomarkers in breast cancer, warranting further investigation with larger sample sizes.
Collapse
Affiliation(s)
- Meda Marija Kaušylaitė
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| | - Justina Jurevičė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| | - Erika Korobeinikova
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.K.); (J.G.); (E.J.)
| | - Jurgita Gudaitienė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.K.); (J.G.); (E.J.)
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (E.K.); (J.G.); (E.J.)
| | - Rasa Ugenskienė
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| |
Collapse
|
14
|
Shah K, Guo B, Hicks SC. Addressing the mean-variance relationship in spatially resolved transcriptomics data with spoon. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621867. [PMID: 39574747 PMCID: PMC11580860 DOI: 10.1101/2024.11.04.621867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
An important task in the analysis of spatially resolved transcriptomics data is to identify spatially variable genes (SVGs), or genes that vary in a 2D space. Current approaches rank SVGs based on either p-values or an effect size, such as the proportion of spatial variance. However, previous work in the analysis of RNA-sequencing identified a technical bias, referred to as the "mean-variance relationship", where highly expressed genes are more likely to have a higher variance. Here, we demonstrate the mean-variance relationship in spatial transcriptomics data. Furthermore, we propose spoon, a statistical framework using Empirical Bayes techniques to remove this bias, leading to more accurate prioritization of SVGs. We demonstrate the performance of spoon in both simulated and real spatial transcriptomics data. A software implementation of our method is available at https://bioconductor.org/packages/spoon.
Collapse
Affiliation(s)
- Kinnary Shah
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, MD, USA
| |
Collapse
|
15
|
Davletgildeeva AT, Kuznetsov NA. The Role of DNMT Methyltransferases and TET Dioxygenases in the Maintenance of the DNA Methylation Level. Biomolecules 2024; 14:1117. [PMID: 39334883 PMCID: PMC11430729 DOI: 10.3390/biom14091117] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
This review deals with the functional characteristics and biological roles of enzymes participating in DNA methylation and demethylation as key factors in epigenetic regulation of gene expression. The set of enzymes that carry out such processes in human cells is limited to representatives of two families, namely DNMT (DNA methyltransferases) and TET (DNA dioxygenases). The review presents detailed information known today about each functionally important member of these families and describes the catalytic activity and roles in the mammalian body while also providing examples of dysregulation of the expression and/or activity of these enzymes in conjunction with the development of some human disorders, including cancers, neurodegenerative diseases, and developmental pathologies. By combining the up-to-date information on the dysfunction of various enzymes that control the DNA "methylome" in the human body, we hope not only to draw attention to the importance of the maintenance of a required DNA methylation level (ensuring epigenetic regulation of gene expression and normal functioning of the entire body) but also to help identify new targets for directed control over the activity of the enzymes that implement the balance between processes of DNA methylation and demethylation.
Collapse
Affiliation(s)
- Anastasiia T Davletgildeeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nikita A Kuznetsov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
16
|
Wang R, Yu X, Ye H, Ao M, Xi M, Hou M. LncRNA FAM83H-AS1 inhibits ferroptosis of endometrial cancer by promoting DNMT1-mediated CDO1 promoter hypermethylation. J Biol Chem 2024; 300:107680. [PMID: 39159808 PMCID: PMC11419805 DOI: 10.1016/j.jbc.2024.107680] [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: 02/18/2024] [Revised: 07/18/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
Endometrial cancer (EC) is the most prevalent gynecological epithelial malignancy. DNA methylation is a promising cancer biomarker but limited use for detecting EC. We previously found that the level of cysteine dioxygenase 1 (CDO1) promoter methylation was elevated in EC patients through methylomics, but the role and mechanism of CDO1 in EC remained unclear. Here, the methylation level of CDO1 promoter was detected by bisulfite-sequencing PCR and methylation-specific PCR (bisulfite conversion-based PCR methods, which remain the most commonly used techniques for methylation detection). Cells were incubated with erastin (the ferroptosis activator). Cell vitality was measured using the cell counting kit-8 assay. FAM83H-AS1 cellular distribution was analyzed by the fluorescence in situ hybridization assay. Lipid reactive oxygen species level was examined by BODIPY-C11 staining. The interactions between FAM83H-AS1, CDO1, and DNA methyltransferase1 (DNMT1) were analyzed by RNA-binding protein immunoprecipitation or chromatin immunoprecipitation assay. The xenograft mouse model was utilized to test CDO1 and FAM83H-AS1's influence on tumor development in vivo. Results showed that CDO1 was hypermethylated and downregulated in EC. CDO1 knockdown reduced erastin-induced ferroptosis in EC cells. Mechanistically, DNMT1 is a DNA methyltransferase, which can transfer methyl groups to cytosine nucleotides in genomic DNA. Long noncoding RNA FAM83H-AS1 increased CDO1 promoter methylation level and inhibited its expression in EC cells by recruiting DNMT1. CDO1 knockdown or FAM83H-AS1 overexpression promoted EC tumor growth in vivo. Long noncoding RNA FAM83H-AS1 inhibited ferroptosis in EC by recruiting DNMT1 to increase CDO1 promoter methylation level and inhibit its expression.
Collapse
Affiliation(s)
- Ruiyu Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Xiuzhang Yu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Hui Ye
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Mengyin Ao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Mingrong Xi
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Minmin Hou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children Sichuan University, Ministry of Education, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
Chen T, Mahdadi S, Vidal M, Desbène-Finck S. Non-nucleoside inhibitors of DNMT1 and DNMT3 for targeted cancer therapy. Pharmacol Res 2024; 207:107328. [PMID: 39079576 DOI: 10.1016/j.phrs.2024.107328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
DNA methylation can deactivate tumor suppressor genes thus causing cancers. Two DNA methylation inhibitors have been approved by the Food and Drug Administration (FDA) and have entered clinical use. However, these inhibitors are nucleoside analogues that can be incorporated into DNA or RNA and induce significant side effects. DNMT1 and DNMT3 are key enzymes involved in DNA methylation. In the acute myeloid leukemia model, a non-nucleoside DNMT1-specific inhibitor has shown lower toxicity and improved pharmacokinetics compared to traditional nucleoside drugs. DNMT3 is also implicated in certain specific cancers. Thus, developing non-nucleoside inhibitors for DNMT1 or DNMT3 can help in understanding their roles in carcinogenesis and provide targeted treatment options in certain cancers. Although no non-nucleoside inhibitors have yet entered clinical trials, in this review, we focus on DNMT1 or DNMT3 selective inhibitors. For DNMT1 selective inhibitors, we have compiled information on the repurposed drugs, derivative compounds and selective inhibitors identified through virtual screening. Additionally, we have outlined potential targets for DNMT1, including protein-protein complex, RNA mimics and aptamers. Compared to DNMT1, research on DNMT3-specific inhibitors has been less extensive. In this context, our exploration has identified a limited number of molecular inhibitors, and we have proposed specific long non-coding RNAs (lncRNAs) as potential contributors to the selective inhibition of DNMT3. This collective effort aims to offer valuable insights into the development of non-nucleoside inhibitors that selectively target DNMT1 or DNMT3.
Collapse
Affiliation(s)
- Ting Chen
- UMR 8038 CNRS, U1268 INSERM, UFR de pharmacie, Université Paris cité, 75270, France
| | - Syrine Mahdadi
- UMR 8038 CNRS, U1268 INSERM, UFR de pharmacie, Université Paris cité, 75270, France
| | - Michel Vidal
- UMR 8038 CNRS, U1268 INSERM, UFR de pharmacie, Université Paris cité, 75270, France; Toxicology, Cochin Hospital, HUPC, APHP, Paris 75014, France
| | | |
Collapse
|
18
|
Yue Y, Ren Y, Lu C, Li P, Zhang G. Epigenetic regulation of human FOXP3+ Tregs: from homeostasis maintenance to pathogen defense. Front Immunol 2024; 15:1444533. [PMID: 39144146 PMCID: PMC11323565 DOI: 10.3389/fimmu.2024.1444533] [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: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Regulatory T cells (Tregs), characterized by the expression of Forkhead Box P3 (FOXP3), constitute a distinct subset of T cells crucial for immune regulation. Tregs can exert direct and indirect control over immune homeostasis by releasing inhibitory factors or differentiating into Th-like Treg (Th-Treg), thereby actively contributing to the prevention and treatment of autoimmune diseases. The epigenetic regulation of FOXP3, encompassing DNA methylation, histone modifications, and post-translational modifications, governs the development and optimal suppressive function of Tregs. In addition, Tregs can also possess the ability to maintain homeostasis in diverse microenvironments through non-suppressive mechanisms. In this review, we primarily focus on elucidating the epigenetic regulation of Tregs as well as their multifaceted roles within diverse physiological contexts while looking forward to potential strategies involving augmentation or suppression of Tregs activity for disease management, particularly in light of the ongoing global COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Ma J, Gong Y, Sun X, Liu C, Li X, Sun Y, Yang D, He J, Wang M, Du J, Zhang J, Xu W, Wang T, Chi X, Tang Y, Song J, Wang Y, Ma F, Chen C, Zhang H, Zhan J. Tumor suppressor FRMD3 controls mammary epithelial cell fate determination via notch signaling pathway. SCIENCE ADVANCES 2024; 10:eadk8958. [PMID: 38959315 PMCID: PMC11221522 DOI: 10.1126/sciadv.adk8958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 05/17/2024] [Indexed: 07/05/2024]
Abstract
The luminal-to-basal transition in mammary epithelial cells (MECs) is accompanied by changes in epithelial cell lineage plasticity; however, the underlying mechanism remains elusive. Here, we report that deficiency of Frmd3 inhibits mammary gland lineage development and induces stemness of MECs, subsequently leading to the occurrence of triple-negative breast cancer. Loss of Frmd3 in PyMT mice results in a luminal-to-basal transition phenotype. Single-cell RNA sequencing of MECs indicated that knockout of Frmd3 inhibits the Notch signaling pathway. Mechanistically, FERM domain-containing protein 3 (FRMD3) promotes the degradation of Disheveled-2 by disrupting its interaction with deubiquitinase USP9x. FRMD3 also interrupts the interaction of Disheveled-2 with CK1, FOXK1/2, and NICD and decreases Disheveled-2 phosphorylation and nuclear localization, thereby impairing Notch-dependent luminal epithelial lineage plasticity in MECs. A low level of FRMD3 predicts poor outcomes for breast cancer patients. Together, we demonstrated that FRMD3 is a tumor suppressor that functions as an endogenous activator of the Notch signaling pathway, facilitating the basal-to-luminal transformation in MECs.
Collapse
Affiliation(s)
- Ji Ma
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yuqing Gong
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoran Sun
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
- Department of Pathology, Peking University Health Science Center, Beijing 100191, China
| | - Cheng Liu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xueying Li
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yi Sun
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Decao Yang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Junming He
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Mengyuan Wang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Juan Du
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jing Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Weizhi Xu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Tianzhuo Wang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaochun Chi
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yan Tang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jiagui Song
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yunling Wang
- Institute of Cardiovascular Research, Peking University Health Science Center, Beijing 100191, China
| | - Fei Ma
- National Cancer Center, State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Hongquan Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jun Zhan
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
20
|
Zhou L, Yu CW. Epigenetic modulations in triple-negative breast cancer: Therapeutic implications for tumor microenvironment. Pharmacol Res 2024; 204:107205. [PMID: 38719195 DOI: 10.1016/j.phrs.2024.107205] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/23/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype lacking estrogen receptors, progesterone receptors and lacks HER2 overexpression. This absence of critical molecular targets poses significant challenges for conventional therapies. Immunotherapy, remarkably immune checkpoint blockade, offers promise for TNBC treatment, but its efficacy remains limited. Epigenetic dysregulation, including altered DNA methylation, histone modifications, and imbalances in regulators such as BET proteins, plays a crucial role in TNBC development and resistance to treatment. Hypermethylation of tumor suppressor gene promoters and the imbalance of histone methyltransferases such as EZH2 and histone deacetylases (HDACs) profoundly influence tumor cell proliferation, survival, and metastasis. In addition, epigenetic alterations critically shape the tumor microenvironment (TME), including immune cell composition, cytokine signaling, and immune checkpoint expression, ultimately contributing to immune evasion. Targeting these epigenetic mechanisms with specific inhibitors such as EZH2 and HDAC inhibitors in combination with immunotherapy represents a compelling strategy to remodel the TME, potentially overcoming immune evasion and enhancing therapeutic outcomes in TNBC. This review aims to comprehensively elucidate the current understanding of epigenetic modulation in TNBC, its influence on the TME, and the potential of combining epigenetic therapies with immunotherapy to overcome the challenges posed by this aggressive breast cancer subtype.
Collapse
Affiliation(s)
- Linlin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China; School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Chen-Wei Yu
- Department of Statistics and Information Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
21
|
Lin S, Wang S, Xu B. Fragmentation patterns of cell-free DNA and somatic mutations in the urine of metastatic breast cancer patients. J Cancer Res Ther 2024; 20:563-569. [PMID: 38454812 DOI: 10.4103/jcrt.jcrt_1359_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/08/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Urinary cell-free deoxyribonucleic acid (DNA) (ucfDNA) holds promise as a biomarker; however, its potential remains largely unexplored. We examined the fragmentation pattern of ucfDNA and identified somatic mutations within urine samples from metastatic breast cancer (MBC) patients. METHODS Urine and blood specimens were collected before treatment from 45 MBC patients and posttreatment urine samples from 16 of the 45 patients at the China National Cancer Center. Somatic mutations and tumor mutational burden (TMB) in the urine and plasma of 10 patients were analyzed by next-generation sequencing (NGS). Fragmentation patterns of cfDNA were displayed using electropherograms. Differences in the extracted amount of cfDNA, length of cfDNA fragments, and TMB between urine and plasma were compared using a Wilcoxon test. RESULTS The fragmentation patterns of ucfDNA were categorized as follows: (1) profile A (n = 26) containing a short peak (100-200 bp) and a long peak (>1500 bp); (2) profile B (n = 8) containing only a long peak; and (3) profile C (n = 11) containing flat pattern. For profile A patients, the short-peaked ucfDNA circulating in the bloodstream was much shorter compared with plasma cfDNA (149 vs. 171 bp, Wilcoxon test, P = 0.023). The fragmentation patterns in lung metastasis patients exhibited a higher propensity toward profile C ( P = 0.002). After treatment, 87.5% of the patients exhibited consistent fragmentation patterns. The concordance rate for somatic mutations in the plasma and urine was 30%, and the median TMB of urine and plasma was not significantly different. CONCLUSIONS This study established a fragmentation pattern for ucfDNA and detected somatic mutations in the urine of MBC patients. These results suggest the potential application of ucfDNA as a biomarker for MBC.
Collapse
Affiliation(s)
- Shaoyan Lin
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Shusen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
22
|
Bagheri M, Lee MK, Muller KE, Miller TW, Pattabiraman DR, Christensen BC. Alteration of DNA methyltransferases by eribulin elicits broad DNA methylation changes with potential therapeutic implications for triple-negative breast cancer. Epigenomics 2024; 16:293-308. [PMID: 38356412 PMCID: PMC10910603 DOI: 10.2217/epi-2023-0339] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive disease with limited treatment options. Eribulin, a chemotherapeutic drug, induces epigenetic changes in cancer cells, suggesting a unique mechanism of action. Materials & methods: MDA-MB 231 cells were treated with eribulin and paclitaxel, and the samples from 53 patients treated with neoadjuvant eribulin were compared with those from 14 patients who received the standard-of-care treatment using immunohistochemistry. Results: Eribulin treatment caused significant DNA methylation changes in drug-tolerant persister TNBC cells, and it also elicited changes in the expression levels of epigenetic modifiers (DNMT1, TET1, DNMT3A/B) in vitro and in primary TNBC tumors. Conclusion: These findings provide new insights into eribulin's mechanism of action and potential biomarkers for predicting TNBC treatment response.
Collapse
Affiliation(s)
- Meisam Bagheri
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Min Kyung Lee
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Kristen E Muller
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
- Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon NH 03756, USA
| | - Todd W Miller
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Diwakar R Pattabiraman
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Dartmouth Cancer Center, Lebanon, NH 03756, USA
| | - Brock C Christensen
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Department of Community & Family Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
23
|
Feng Y, Dong H, Tan B. Endometriotic mesenchymal stem cells promote the fibrosis process of endometriosis through paracrine TGF-β1 mediated RASAL1 inhibition. J Obstet Gynaecol Res 2024; 50:467-477. [PMID: 38113862 DOI: 10.1111/jog.15851] [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: 08/01/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Endometrial-derived stem cells are key players in endometriosis (EMs) pathogenesis, while the mechanism involved is still unclear. Herein, the role and regulatory mechanism of endometriotic mesenchymal stem cells (ecto-MSCs) in regulating fibrosis during EMs progression were investigated. METHODS The mRNA and protein expressions were assessed using qRT-PCR, western blot, and immunofluorescence. Flow cytometry was adopted to analyze the markers of MSCs. Transwell assay was adopted to examine endometriotic stromal cells (ESCs) migration and invasion. The interactions between DNMT3A and RASAL1 were analyzed by ChIP assay. In addition, MSP was employed to detect RASAL1 promoter methylation level. RESULTS Ecto-MSCs promoted ESCs migration, invasion, and fibrosis process by TGF-β1 paracrine. It was subsequently revealed that TGF-β1 upregulated DNMT3A in ESCs in a SMAD3-dependent manner. As expected, DNMT3A knockdown abolished ecto-MSCs' facilitation on ESCs migration, invasion, and fibrosis process. DNMT3A, as a methyltransferase, reduced RASAL1 expression in TGF-β1-treated ESCs by increasing RASAL1 promoter methylation level. RASAL1, as an antifibrotic protein, was lowly expressed in TGF-β1-treated ESCs, and its overexpression ameliorated TGF-β1-induced increase in ESCs migration, invasion, and fibrosis process. CONCLUSION TGF-β1 secreted by ecto-MSCs facilitated fibrogenesis in EMs through SMAD3/DNMT3A-mediated RASAL1 inhibition.
Collapse
Affiliation(s)
- Ying Feng
- The Second Affiliated Hospital of Nanchang University, The Department of Obstetrics and Gynecology, Nanchang, Jiangxi, China
| | - Han Dong
- Gynecology Women and Children's Hospital of Jinzhou, The Department of Obstetrics and Gynecology, Jinzhou, Liaoning, China
| | - Buzhen Tan
- The Second Affiliated Hospital of Nanchang University, The Department of Obstetrics and Gynecology, Nanchang, Jiangxi, China
| |
Collapse
|
24
|
Zhang T, Liu J, Wang M, Liu X, Qu J, Zhang H. Prognosis stratification and response to treatment in breast cancer based on one-carbon metabolism-related signature. Front Oncol 2024; 13:1288909. [PMID: 38239641 PMCID: PMC10794736 DOI: 10.3389/fonc.2023.1288909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Breast cancer (BC) is the most common malignant tumor in the female population. Despite staging and treatment consensus guidelines, significant heterogeneity exists in BC patients' prognosis and treatment efficacy. Alterations in one-carbon (1C) metabolism are critical for tumor growth, but the value of the role of 1C metabolism in BC has not been fully investigated. Methods To investigate the prognostic value of 1C metabolism-related genes in BC, 72 1C metabolism-related genes from GSE20685 dataset were used to construct a risk-score model via univariate Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) regression algorithm, which was validated on three external datasets. Based on the risk score, all BC patients were categorized into high-risk and low-risk groups. The predictive ability of the model in the four datasets was verified by plotting Kaplan-Meier curve and receiver operating characteristic (ROC) curve. The candidate genes were then analyzed in relation to gene mutations, gene enrichment pathways, immune infiltration, immunotherapy, and drug sensitivity. Results We identified a 7-gene 1C metabolism-related signature for prognosis and structured a prognostic model. ROC analysis demonstrated that the model accurately predicted the 2-, 3-, and 5-year overall survival rate of BC patients in the four cohorts. Kaplan-Meier analysis revealed that survival time of high-risk patients was markedly shorter than that of low-risk patients (p < 0.05). Meanwhile, high-risk patients had a higher tumor mutational burden (TMB), enrichment of tumor-associated pathways such as the IL-17 signaling pathway, lower levels of T follicular helper (Tfh) and B cells naive infiltration, and poorer response to immunotherapy. Furthermore, a strong correlation was found between MAT2B and CHKB and immune checkpoints. Discussion These findings offer new insights into the effect of 1C metabolism in the onset, progression, and therapy of BC and can be used to assess BC patients' prognosis, study immune infiltration, and develop potentially more effective clinical treatment options.
Collapse
Affiliation(s)
| | | | | | | | - Jia Qu
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
25
|
Wang D, Zhang Z, Zhao L, Yang L, Lou C. Recent advances in natural polysaccharides against hepatocellular carcinoma: A review. Int J Biol Macromol 2023; 253:126766. [PMID: 37689300 DOI: 10.1016/j.ijbiomac.2023.126766] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor of the digestive system that poses a serious threat to human life and health. Chemotherapeutic drugs commonly used in the clinic have limited efficacy and heavy adverse effects. Therefore, it is imperative to find effective and safe alternatives, and natural polysaccharides (NPs) fit the bill. This paper summarizes in detail the anti-HCC activity of NPs in vitro, animal and clinical trials. Furthermore, the addition of NPs can reduce the deleterious effects of chemotherapeutic drugs such as immunotoxicity, bone marrow suppression, oxidative stress, etc. The potential mechanisms are related to induction of apoptosis and cell cycle arrest, block of angiogenesis, invasion and metastasis, stimulation of immune activity and targeting of MircoRNA. And on this basis, we further elucidate that the anti-HCC activity may be related to the monosaccharide composition, molecular weight (Mw), conformational features and structural modifications of NPs. In addition, due to its good physicochemical properties, it is widely used as a drug carrier in the delivery of chemotherapeutic drugs and small molecule components. This review provides a favorable theoretical basis for the application of the anti-HCC activity of NPs.
Collapse
Affiliation(s)
- Dazhen Wang
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Zhengfeng Zhang
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Lu Zhao
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Liu Yang
- Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - ChangJie Lou
- Harbin Medical University Cancer Hospital, Harbin 150081, China.
| |
Collapse
|
26
|
Zou Q, Cao S. miR-4270 suppresses hepatocellular carcinoma progression by inhibiting DNMT3A-mediated methylation of HGFAC promoter. PeerJ 2023; 11:e16566. [PMID: 38077422 PMCID: PMC10704985 DOI: 10.7717/peerj.16566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/12/2023] [Indexed: 12/18/2023] Open
Abstract
Background miR-4270 is a regulatory factor has been linked with the progression of various cancers, such as nasopharyngeal carcinoma, hepatocellular carcinoma (HCC), and gastric cancer. However, the underlying mechanisms through which miR-4270 modulates HCC development are not fully understood. Methods miR-4270 expression levels were analyzed in various HCC cell lines and tissue samples. An online bioinformatics tool was then utilized to predict the miR-4270 target gene. The binding relationship between miR-4270 and its target gene DNMT3A was verified using dual-luciferase reporter and Ago2-RIP assays. Then, co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (ChIP) assays were conducted to investigate the association between DNMT3A and the hepatocyte growth factor activator (HGFAC) promoter region. To assess the methylation level of the HGFAC promoter, methylation-specific PCR (MSP) was employed. Furthermore, rescue analyses were carried out to evaluate the functional relevance of miR-4270 and HGFAC in the modulation of the malignant properties of HCC cells. Finally, HepG2 cells overexpressing miR-4270 were subcutaneously injected into nude mice to estimate the impact of miR-4270 on the xenograft tumor growth of HCC. Results A substantial miR-4270 downregulation was revealed in HCC patient samples and cell lines. miR-4270 upregulation suppressed both cell proliferation and invasion while promoting apoptosis. At the molecular level, miR-4270 was found to bind to the 3'untranslated region (3'UTR) of DNMT3A, thereby inhibiting DNMT3A-mediated methylation of the HGFAC promoter. Functional assays indicated that inhibition of miR-4270 stimulated HCC cell growth, an effect counteracted by overexpression of HGFAC. In vivo assays further verified that miR-4270 effectively suppressed the progression of HCC xenograft tumors. Conclusions miR-4270 was found to mitigate the malignant characteristics of HCC by inhibiting DNMT3A-mediated methylation of the HGFAC promoter, suggesting a potential therapeutic avenue for the management of HCC.
Collapse
Affiliation(s)
- Qiang Zou
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Shasha Cao
- Department of Neonatology, Zibo Maternal and Child Health Hospital, Zibo, China
| |
Collapse
|
27
|
Wu G, Wang Q, Wang D, Xiong F, Liu W, Chen J, Wang B, Huang W, Wang X, Chen Y. Targeting polycomb repressor complex 2-mediated bivalent promoter epigenetic silencing of secreted frizzled-related protein 1 inhibits cholangiocarcinoma progression. Clin Transl Med 2023; 13:e1502. [PMID: 38050190 PMCID: PMC10696163 DOI: 10.1002/ctm2.1502] [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: 06/14/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) refers to a collection of malignancies that are associated with a dismal prognosis. Currently, surgical resection is the only way to cure patients with CCA. Available systemic therapy is limited to gemcitabine plus cisplatin; however, this treatment is palliative in nature. Therefore, there is still a need to explore new effective therapeutic targets to intervene against CCA. METHODS We analyzed the expression of EZH2 and the prognosis of patients in CCA. The proliferation, migration and invasion of CCA cells after gene knockdown and overexpression were examined and validated by a xenograft model and a primary CCA mouse model with corresponding gene intervention. Targeting DNA methylation, and RNA-sequencing-based transcriptomic analysis in EZH2 and SUZ12 knockout CCA cells was performed. Bisulfite sequencing polymerase chain reaction (PCR), chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR) and reverse-ChIP assays were performed for research purposes. RESULTS Increased expression of EZH2 in CCA exhibited a significantly poorer prognosis. DNA hypomethylation of the promoter and increased mRNA levels of secreted frizzled-related protein 1 (SFRP1) were observed in CCA cells following the inhibition of polycomb repressor complex 2 (PRC2), which was achieved through a knockout of EZH2, SUZ12 and EED, respectively, or treatment with GSK126 and GSK343. Targeting the SFRP1 promoter DNA hypermethylation with dCas9-DNMT3a decreased the mRNA level of SFRP1. The expression of SFRP1 is regulated by both H3K27me3 and DNA methylation and H3K27me3 plays a crucial role in promoting SFRP1 promotor DNA methylation. GSK343 is a small molecule inhibitor that targets the catalytic activity of EZH2. It effectively inhibits the progression and development of subcutaneous xenografts and primary CCA mouse models. CONCLUSION Overall, our data strongly suggested that targeting PRC2 promotes the expression of SFRP1, thereby inhibiting the progression of CCA. KEY POINTS/HEADLIGHTS Cholangiocarcinoma (CCA) exhibits elevated expression of EZH2, SUZ12 and EED, resulting in increased levels of H3K27me3. Targeting polycomb repressor complex 2 (PRC2) leads to the removal of H3K27me3 from the secreted frizzled-related protein 1 (SFRP1) promoter and DNA hypomethylation, thereby activating the transcription of SFRP1. Inhibiting PRC2, including the use of EZH2 inhibitors, holds promise as a potential strategy for developing anti-cancer drugs for CCA.
Collapse
Affiliation(s)
- Guanhua Wu
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Qi Wang
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Da Wang
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Fei Xiong
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Wenzheng Liu
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Junsheng Chen
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Bing Wang
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Wenhua Huang
- Department of EmergencyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Xin Wang
- Departement of Pediatric SurgeryWuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| | - Yongjun Chen
- Department of Biliary‐Pancreatic SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanP. R. China
| |
Collapse
|
28
|
Zheng X, Zhao D, Liu Y, Jin Y, Liu T, Li H, Liu D. Regeneration and anti-inflammatory effects of stem cells and their extracellular vesicles in gynecological diseases. Biomed Pharmacother 2023; 168:115739. [PMID: 37862976 DOI: 10.1016/j.biopha.2023.115739] [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: 08/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
There are many gynecological diseases, among which breast cancer (BC), cervical cancer (CC), endometriosis (EMs), and polycystic ovary syndrome (PCOS) are common and difficult to cure. Stem cells (SCs) are a focus of regenerative medicine. They are commonly used to treat organ damage and difficult diseases because of their potential for self-renewal and multidirectional differentiation. SCs are also commonly used for difficult-to-treat gynecological diseases because of their strong directional differentiation ability with unlimited possibilities, their tendency to adhere to the diseased tissue site, and their use as carriers for drug delivery. SCs can produce exosomes in a paracrine manner. Exosomes can be produced in large quantities and have the advantage of easy storage. Their safety and efficacy are superior to those of SCs, which have considerable potential in gynecological treatment, such as inhibiting endometrial senescence, promoting vascular reconstruction, and improving anti-inflammatory and immune functions. In this paper, we review the mechanisms of the regenerative and anti-inflammatory capacity of SCs and exosomes in incurable gynecological diseases and the current progress in their application in genetic engineering to provide a foundation for further research.
Collapse
Affiliation(s)
- Xu Zheng
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dan Zhao
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Yang Liu
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Ye Jin
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianjia Liu
- Changchun University of Chinese Medicine, Changchun 130117, China; Baicheng Medical College, Baicheng 137000, China.
| | - Huijing Li
- Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Da Liu
- Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
29
|
Dou R, Kang S, Yang H, Zhang W, Zhang Y, Liu Y, Ping Y, Pang B. Identifying the driver miRNAs with somatic copy number alterations driving dysregulated ceRNA networks in cancers. Biol Direct 2023; 18:79. [PMID: 37993951 PMCID: PMC10666415 DOI: 10.1186/s13062-023-00438-x] [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: 09/21/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play critical roles in cancer initiation and progression, which were critical components to maintain the dynamic balance of competing endogenous RNA (ceRNA) networks. Somatic copy number alterations (SCNAs) in the cancer genome could disturb the transcriptome level of miRNA to deregulate this balance. However, the driving effects of SCNAs of miRNAs were insufficiently understood. METHODS In this study, we proposed a method to dissect the functional roles of miRNAs under different copy number states and identify driver miRNAs by integrating miRNA SCNAs profile, miRNA-target relationships and expression profiles of miRNA, mRNA and lncRNA. RESULTS Applying our method to 813 TCGA breast cancer (BRCA) samples, we identified 29 driver miRNAs whose SCNAs significantly and concordantly regulated their own expression levels and further inversely dysregulated expression levels of their targets or disturbed the miRNA-target networks they directly involved. Based on miRNA-target networks, we further constructed dynamic ceRNA networks driven by driver SCNAs of miRNAs and identified three different patterns of SCNA interference in the miRNA-mediated dynamic ceRNA networks. Survival analysis of driver miRNAs showed that high-level amplifications of four driver miRNAs (including has-miR-30d-3p, has-mir-30b-5p, has-miR-30d-5p and has-miR-151a-3p) in 8q24 characterized a new BRCA subtype with poor prognosis and contributed to the dysfunction of cancer-associated hallmarks in a complementary way. The SCNAs of driver miRNAs across different cancer types contributed to the cancer development by dysregulating different components of the same cancer hallmarks, suggesting the cancer specificity of driver miRNA. CONCLUSIONS These results demonstrate the efficacy of our method in identifying driver miRNAs and elucidating their functional roles driven by endogenous SCNAs, which is useful for interpreting cancer genomes and pathogenic mechanisms.
Collapse
Affiliation(s)
- Renjie Dou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Shaobo Kang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Huan Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Wanmei Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yijing Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yuanyuan Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
30
|
Wu G, Wang D, Xiong F, Liu W, Wang Q, Chen J, Wang B, Chen Y. Upregulation of RSPO3 via targeted promoter DNA demethylation inhibits the progression of cholangiocarcinoma. Clin Epigenetics 2023; 15:177. [PMID: 37932819 PMCID: PMC10629118 DOI: 10.1186/s13148-023-01592-9] [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: 08/07/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) refers to a collection of malignant tumors that develop from the biliary epithelium. Extensive clinical evidence and epidemiological observations indicate a concerning increase in both the incidence and mortality rates of CCA. Surgical resection is currently the sole available cure for CCA. However, it is unfortunate that only a fraction of patients has access to surgery at the time of diagnosis. Moreover, there is a high incidence of cancer recurrence after resection, and systemic treatments have limited efficacy. Therefore, the identification of novel biomarkers for CCA-targeted molecular therapy remains a crucial task in oncology research. RESULTS Our study demonstrated that low expression of RSPO3 was associated with poorer survival rates in patients with CCA. We found that the RSPO3 promoter DNA was hypermethylated in CCA, which was correlated with the low expression of RSPO3. The expression of RSPO3 was influenced by the balance between the DNA methyltransferase DNMT3a and the DNA demethylase TET1 in CCA. In vitro and in vivo experiments showed that targeting RSPO3 promoter DNA methylation using dCas9DNMT3a promoted tumorigenicity of CCA, while targeted RSPO3 promoter DNA demethylation using dCas9TET1CD inhibited CCA tumorigenicity. Additionally, in our primary CCA model, knockdown of Rspo3 promoted CCA progression, whereas overexpression of Rspo3 inhibited CCA progression. CONCLUSIONS Our findings suggest that increased methylation and decreased expression of RSPO3 may indicate a poor prognosis in CCA. Restoring RSPO3 expression by targeting promoter DNA demethylation could offer insights for precise treatment of CCA.
Collapse
Affiliation(s)
- Guanhua Wu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Da Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Fei Xiong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Wenzheng Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Qi Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Junsheng Chen
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China
| | - Bing Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China.
| | - Yongjun Chen
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430074, Hubei, China.
| |
Collapse
|
31
|
Xie J, Gan L, Xue B, Wang X, Pei X. Emerging roles of interactions between ncRNAs and other epigenetic modifications in breast cancer. Front Oncol 2023; 13:1264090. [PMID: 37901333 PMCID: PMC10602744 DOI: 10.3389/fonc.2023.1264090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Up till the present moment, breast cancer is still the leading cause of cancer-related death in women worldwide. Although the treatment methods and protocols for breast cancer are constantly improving, the long-term prognosis of patients is still not optimistic due to the complex heterogeneity of the disease, multi-organ metastasis, chemotherapy and radiotherapy resistance. As a newly discovered class of non-coding RNAs, ncRNAs play an important role in various cancers. Especially in breast cancer, lncRNAs have received extensive attention and have been confirmed to regulate cancer progression through a variety of pathways. Meanwhile, the study of epigenetic modification, including DNA methylation, RNA methylation and histone modification, has developed rapidly in recent years, which has greatly promoted the attention to the important role of non-coding RNAs in breast cancer. In this review, we carefully and comprehensively describe the interactions between several major classes of epigenetic modifications and ncRNAs, as well as their different subsequent biological effects, and discuss their potential for practical clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Xinhong Pei
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Zheng X, Zhao D, Jin Y, Liu Y, Liu D. Role of the NLRP3 inflammasome in gynecological disease. Biomed Pharmacother 2023; 166:115393. [PMID: 37660654 DOI: 10.1016/j.biopha.2023.115393] [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: 07/04/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023] Open
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Collapse
Affiliation(s)
- Xu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Dan Zhao
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Yang Liu
- Acupuncture department,Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
33
|
Deng QF, Li C, Liu J, Ji XX, Wan XY, Wang CY, Sun H, Fang QY, Gu WQ, Ma C, Wang HY, Zhou CC, Li YX, Xie BX, Zhou SW. DNMT3A governs tyrosine kinase inhibitors responses through IAPs and in a cell senescence-dependent manner in non-small cell lung cancer. Am J Cancer Res 2023; 13:3517-3530. [PMID: 37693159 PMCID: PMC10492131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/15/2023] [Indexed: 09/12/2023] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) treated with tyrosine kinase inhibitors (TKIs) inevitably exhibit drug resistance, which diminishes therapeutic effects. Nonetheless, the molecular mechanisms of TKI resistance in NSCLC remain obscure. In this study, data from clinical and TCGA databases revealed an increase in DNMT3A expression, which was correlated with a poor prognosis. Using NSCLC organoid models, we observed that high DNMT3A levels reduced TKI susceptibility of NSCLC cells via upregulating inhibitor of apoptosis proteins (IAPs). Simultaneously, the DNMT3Ahigh subset, which escaped apoptosis, underwent an early senescent-like state in a CDKN1A-dependent manner. Furthermore, the cellular senescence induced by TKIs was observed to be reversible, whereas DNMT3Ahigh cells reacquired their proliferative characteristics in the absence of TKIs, resulting in subsequent tumour recurrence and growth. Notably, the blockade of DNMT3A/IAPs signals enhanced the efficacy of TKIs in DNMT3Ahigh tumour-bearing mice, which represented a promising strategy for the effective treatment of NSCLC.
Collapse
Affiliation(s)
- Qin-Fang Deng
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Chao Li
- Smartquerier Gene Technology (Shanghai) Co., Ltd.Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical TranslationShanghai, China
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai, China
| | - Jing Liu
- Smartquerier Gene Technology (Shanghai) Co., Ltd.Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical TranslationShanghai, China
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai, China
| | - Xian-Xiu Ji
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Xiao-Ying Wan
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Chun-Yan Wang
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Hui Sun
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Qi-Yu Fang
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Wei-Qin Gu
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Chao Ma
- Smartquerier Gene Technology (Shanghai) Co., Ltd.Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical TranslationShanghai, China
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai, China
| | - Hui-Yong Wang
- Smartquerier Gene Technology (Shanghai) Co., Ltd.Shanghai, China
- Shanghai Engineering Research Center of Pharmaceutical TranslationShanghai, China
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai, China
| | - Cai-Cun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Yi-Xue Li
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of SciencesHangzhou, Zhejiang, China
- Guangzhou LaboratoryGuangzhou, Guangdong, China
- Collaborative Innovation Center for Genetics and Development, Fudan UniversityShanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong UniversityShanghai, China
- Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghai, China
| | - Bo-Xiong Xie
- Department of Thoracic, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Song-Wen Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| |
Collapse
|
34
|
Santarosa M, Baldazzi D, Armellin M, Maestro R. In Silico Identification of a BRCA1:miR-29:DNMT3 Axis Involved in the Control of Hormone Receptors in BRCA1-Associated Breast Cancers. Int J Mol Sci 2023; 24:9916. [PMID: 37373065 DOI: 10.3390/ijms24129916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Germline inactivating mutations in the BRCA1 gene lead to an increased lifetime risk of ovarian and breast cancer (BC). Most BRCA1-associated BC are triple-negative tumors (TNBC), aggressive forms of BC characterized by a lack of expression of estrogen and progesterone hormone receptors (HR) and HER2. How BRCA1 inactivation may favor the development of such a specific BC phenotype remains to be elucidated. To address this question, we focused on the role of miRNAs and their networks in mediating BRCA1 functions. miRNA, mRNA, and methylation data were retrieved from the BRCA cohort of the TCGA project. The cohort was divided into a discovery set (Hi-TCGA) and a validation set (GA-TCGA) based on the platform used for miRNA analyses. The METABRIC, GSE81002, and GSE59248 studies were used as additional validation data sets. BCs were differentiated into BRCA1-like and non-BRCA1-like based on an established signature of BRCA1 pathway inactivation. Differential expression of miRNAs, gene enrichment analysis, functional annotation, and methylation correlation analyses were performed. The miRNAs downregulated in BRCA1-associated BC were identified by comparing the miRNome of BRCA1-like with non-BRCA1-like tumors from the Hi-TCGA discovery cohort. miRNAs:gene-target anticorrelation analyses were then performed. The target genes of miRNAs downregulated in the Hi-TCGA series were enriched in the BRCA1-like tumors from the GA-TCGA and METABRIC validation data sets. Functional annotation of these genes revealed an over-representation of several biological processes ascribable to BRCA1 activity. The enrichment of genes related to DNA methylation was particularly intriguing, as this is an aspect of BRCA1 functions that has been poorly explored. We then focused on the miR-29:DNA methyltransferase network and showed that the miR-29 family, which was downregulated in BRCA1-like tumors, was associated with poor prognosis in these BCs and inversely correlated with the expression of the DNA methyltransferases DNMT3A and DNMT3B. This, in turn, correlated with the methylation extent of the promoter of HR genes. These results suggest that BRCA1 may control the expression of HR via a miR-29:DNMT3:HR axis and that disruption of this network may contribute to the receptor negative phenotype of tumors with dysfunctional BRCA1.
Collapse
Affiliation(s)
- Manuela Santarosa
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Davide Baldazzi
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Michela Armellin
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Roberta Maestro
- Unit of Oncogenetics and Functional Oncogenomics, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| |
Collapse
|
35
|
Pham TMQ, Phan TH, Jasmine TX, Tran TTT, Huynh LAK, Vo TL, Nai THT, Tran TT, Truong MH, Tran NC, Nguyen VTC, Nguyen TH, Nguyen THH, Le NDK, Nguyen TD, Nguyen DS, Truong DK, Do TTT, Phan MD, Giang H, Nguyen HN, Tran LS. Multimodal analysis of genome-wide methylation, copy number aberrations, and end motif signatures enhances detection of early-stage breast cancer. Front Oncol 2023; 13:1127086. [PMID: 37223690 PMCID: PMC10200909 DOI: 10.3389/fonc.2023.1127086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/24/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Breast cancer causes the most cancer-related death in women and is the costliest cancer in the US regarding medical service and prescription drug expenses. Breast cancer screening is recommended by health authorities in the US, but current screening efforts are often compromised by high false positive rates. Liquid biopsy based on circulating tumor DNA (ctDNA) has emerged as a potential approach to screen for cancer. However, the detection of breast cancer, particularly in early stages, is challenging due to the low amount of ctDNA and heterogeneity of molecular subtypes. Methods Here, we employed a multimodal approach, namely Screen for the Presence of Tumor by DNA Methylation and Size (SPOT-MAS), to simultaneously analyze multiple signatures of cell free DNA (cfDNA) in plasma samples of 239 nonmetastatic breast cancer patients and 278 healthy subjects. Results We identified distinct profiles of genome-wide methylation changes (GWM), copy number alterations (CNA), and 4-nucleotide oligomer (4-mer) end motifs (EM) in cfDNA of breast cancer patients. We further used all three signatures to construct a multi-featured machine learning model and showed that the combination model outperformed base models built from individual features, achieving an AUC of 0.91 (95% CI: 0.87-0.95), a sensitivity of 65% at 96% specificity. Discussion Our findings showed that a multimodal liquid biopsy assay based on analysis of cfDNA methylation, CNA and EM could enhance the accuracy for the detection of early- stage breast cancer.
Collapse
Affiliation(s)
- Thi Mong Quynh Pham
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Thanh Hai Phan
- Ultrasound Department Medic Medical Center, Ho Chi Minh, Vietnam
| | | | - Thuy Thi Thu Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Le Anh Khoa Huynh
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Department of Biostatistics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Thi Loan Vo
- Ultrasound Department Medic Medical Center, Ho Chi Minh, Vietnam
| | | | - Thuy Trang Tran
- Ultrasound Department Medic Medical Center, Ho Chi Minh, Vietnam
| | - My Hoang Truong
- Ultrasound Department Medic Medical Center, Ho Chi Minh, Vietnam
| | - Ngan Chau Tran
- Ultrasound Department Medic Medical Center, Ho Chi Minh, Vietnam
| | - Van Thien Chi Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Trong Hieu Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Thi Hue Hanh Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Nguyen Duy Khang Le
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Thanh Dat Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Duy Sinh Nguyen
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
- Faculty of Medicine Nguyen Tat Thanh University, Ho Chi Minh, Vietnam
| | | | | | - Minh-Duy Phan
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Hoai-Nghia Nguyen
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
- Research and Development Department Gene Solutions, Ho Chi Minh, Vietnam
| |
Collapse
|
36
|
Sibuh BZ, Quazi S, Panday H, Parashar R, Jha NK, Mathur R, Jha SK, Taneja P, Jha AK. The Emerging Role of Epigenetics in Metabolism and Endocrinology. BIOLOGY 2023; 12:256. [PMID: 36829533 PMCID: PMC9953656 DOI: 10.3390/biology12020256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
Each cell in a multicellular organism has its own phenotype despite sharing the same genome. Epigenetics is a somatic, heritable pattern of gene expression or cellular phenotype mediated by structural changes in chromatin that occur without altering the DNA sequence. Epigenetic modification is an important factor in determining the level and timing of gene expression in response to endogenous and exogenous stimuli. There is also growing evidence concerning the interaction between epigenetics and metabolism. Accordingly, several enzymes that consume vital metabolites as substrates or cofactors are used during the catalysis of epigenetic modification. Therefore, altered metabolism might lead to diseases and pathogenesis, including endocrine disorders and cancer. In addition, it has been demonstrated that epigenetic modification influences the endocrine system and immune response-related pathways. In this regard, epigenetic modification may impact the levels of hormones that are important in regulating growth, development, reproduction, energy balance, and metabolism. Altering the function of the endocrine system has negative health consequences. Furthermore, endocrine disruptors (EDC) have a significant impact on the endocrine system, causing the abnormal functioning of hormones and their receptors, resulting in various diseases and disorders. Overall, this review focuses on the impact of epigenetics on the endocrine system and its interaction with metabolism.
Collapse
Affiliation(s)
- Belay Zeleke Sibuh
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore 560043, India
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK
- Clinical Bioinformatics, School of Health Sciences, The University of Manchester, Manchester M13 9P, UK
- SCAMT Institute, ITMO University, St. Petersburg 197101, Russia
| | - Hrithika Panday
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Ritika Parashar
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Runjhun Mathur
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
| | - Pankaj Taneja
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| |
Collapse
|
37
|
Peripheral mRNA Expression and Prognostic Significance of Emotional Stress Biomarkers in Metastatic Breast Cancer Patients. Int J Mol Sci 2022; 23:ijms232214097. [PMID: 36430579 PMCID: PMC9694977 DOI: 10.3390/ijms232214097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/12/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Emotional stress is believed to be associated with increased tumor progression. Stress-induced epigenetic modifications can contribute to the severity of disease and poor prognosis in cancer patients. The current study aimed to investigate the expression profiles along with the prognostic significance of psychological stress-related genes in metastatic breast cancer patients, to rationalize the molecular link between emotional stress and cancer progression. We profiled the expression of selected stress-associated genes (5-HTT, NR3C1, OXTR, and FKBP5) in breast cancer including the stress evaluation of all participants using the Questionnaire on Distress in Cancer Patients-short form (QSC-R10). A survival database, the Kaplan-Meier Plotter, was used to explore the prognostic significance of these genes in breast cancer. Our results showed relatively low expressions of 5-HTT (p = 0.02) and OXTR (p = 0.0387) in metastatic breast cancer patients as compared to the non-metastatic group of patients. The expression of NR3C1 was low in tumor grade III as compared to grade II (p = 0.04). Additionally, the expression of NR3C1 was significantly higher in patients with positive estrogen receptor status. However, no significant difference was found regarding FKBP5 expression in breast cancer. The results suggest a potential implication of these genes in breast cancer pathology and prognosis.
Collapse
|
38
|
Luo JF, Zhou H, Lio CK. Akebia Saponin D Inhibits the Inflammatory Reaction by Inhibiting the IL-6-STAT3-DNMT3b Axis and Activating the Nrf2 Pathway. Molecules 2022; 27:molecules27196236. [PMID: 36234773 PMCID: PMC9614599 DOI: 10.3390/molecules27196236] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
Akebia saponin D (ASD) is derived from the Dipsacus asper Wall. ex Henry, which is a traditional Chinese medicine commonly used to treat rheumatic arthritis (RA). However, the in-depth mechanism of the anti-inflammatory effect of ASD is still unclear. This study aimed to preliminarily explore the anti-inflammatory effect of ASD and the underlying mechanisms from the perspective of DNA methylation and inflammation-related pathways. We found that ASD significantly reduced the production of multiple inflammatory mediators, including nitric oxide (NO) and prostaglandin E2 (PGE2), in LPS-induced RAW264.7 cells. The expression of DNA methyltransferase (DNMT) 3b and inducible nitric oxide synthase (iNOS) was also obviously inhibited by the ASD treatment. The protein and mRNA levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were also significantly inhibited by ASD. ASD inhibited the macrophage M1 phenotype, inhibited the high level of DNMT3b, and downregulated the signal transducer and activator of the transcription 3 (STAT3) pathway to exert its anti-inflammatory activity. Furthermore, DNMT3b siRNA and Nrf2 siRNA significantly promoted the anti-inflammatory effect of ASD. Our study demonstrates for the first time that ASD inhibits the IL-6-STAT3-DNMT3b axis and activates the nuclear factor-E2-related factor 2 (Nrf2) signaling pathway to achieve its inhibitory effect on inflammatory reactions.
Collapse
Affiliation(s)
- Jin-Fang Luo
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guian District, Guiyang 550025, China
| | - Hua Zhou
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
- Correspondence:
| | - Chon-Kit Lio
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medcine and Immune Disease Research, Guangzhou 510006, China
| |
Collapse
|