1
|
Vanagas L, Alonso AM, Angel SO. Identification of subtelomeric cluster-genes associated to sexual stage in Toxoplasma gondii. Gene 2025; 933:148924. [PMID: 39245231 DOI: 10.1016/j.gene.2024.148924] [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: 07/31/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Toxoplasma gondii is an obligate intracellular parasite with sexual reproduction in the intestinal epithelium of felines. The depletion of two gene repressors, AP2XI-2 and AP2XII-1, induces merozoite formation and gene expression towards sexual commitment. Based on RNA-seq datasets of AP2XI-2 and AP2XII-1 knock downs we identified subtelomeric (ST) TgB12 and hypothetical (HP) genes upregulated. Some of the differentially expressed genes (DEGs) are arranged in ST clusters. These DEG products are characterized by high isoelectric points (pI) and may encode small proteins. The potential roles of these clusters of DEG ST genes in environmental resistance or parasite sexual development of T. gondii is discussed.
Collapse
Affiliation(s)
- Laura Vanagas
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Argentina.
| | - Andres M Alonso
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Argentina.
| | - Sergio O Angel
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Provincia de Buenos Aires, Argentina; Escuela de Bio y Nanotecnologías (UNSAM), Argentina.
| |
Collapse
|
2
|
Cheng X, Meng X, Chen R, Song Z, Li S, Wei S, Lv H, Zhang S, Tang H, Jiang Y, Zhang R. The molecular subtypes of autoimmune diseases. Comput Struct Biotechnol J 2024; 23:1348-1363. [PMID: 38596313 PMCID: PMC11001648 DOI: 10.1016/j.csbj.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by their complexity and a wide range of clinical differences. Despite patients presenting with similar symptoms and disease patterns, their reactions to treatments may vary. The current approach of personalized medicine, which relies on molecular data, is seen as an effective method to address the variability in these diseases. This review examined the pathologic classification of ADs, such as multiple sclerosis and lupus nephritis, over time. Acknowledging the limitations inherent in pathologic classification, the focus shifted to molecular classification to achieve a deeper insight into disease heterogeneity. The study outlined the established methods and findings from the molecular classification of ADs, categorizing systemic lupus erythematosus (SLE) into four subtypes, inflammatory bowel disease (IBD) into two, rheumatoid arthritis (RA) into three, and multiple sclerosis (MS) into a single subtype. It was observed that the high inflammation subtype of IBD, the RA inflammation subtype, and the MS "inflammation & EGF" subtype share similarities. These subtypes all display a consistent pattern of inflammation that is primarily driven by the activation of the JAK-STAT pathway, with the effective drugs being those that target this signaling pathway. Additionally, by identifying markers that are uniquely associated with the various subtypes within the same disease, the study was able to describe the differences between subtypes in detail. The findings are expected to contribute to the development of personalized treatment plans for patients and establish a strong basis for tailored approaches to treating autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Zerun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuai Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuhao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Mafi A, Hedayati N, Kahkesh S, Khoshayand S, Alimohammadi M, Farahani N, Hushmandi K. The landscape of circRNAs in gliomas temozolomide resistance: Insights into molecular pathways. Noncoding RNA Res 2024; 9:1178-1189. [PMID: 39022676 PMCID: PMC11250881 DOI: 10.1016/j.ncrna.2024.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 07/20/2024] Open
Abstract
As the deadliest type of primary brain tumor, gliomas represent a significant worldwide health concern. Circular RNA (circRNA), a unique non-coding RNA molecule, seems to be one of the most alluring target molecules involved in the pathophysiology of many kinds of cancers. CircRNAs have been identified as prospective targets and biomarkers for the diagnosis and treatment of numerous disorders, particularly malignancies. Recent research has established a clinical link between temozolomide (TMZ) resistance and certain circRNA dysregulations in glioma tumors. CircRNAs may play a therapeutic role in controlling or overcoming TMZ resistance in gliomas and may provide guidance for a novel kind of individualized glioma therapy. To address the biological characteristics of circRNAs and their potential to induce resistance to TMZ, this review has highlighted and summarized the possible roles that circRNAs may play in molecular pathways of drug resistance, including the Ras/Raf/ERK PI3K/Akt signaling pathway and metabolic processes in gliomas.
Collapse
Affiliation(s)
- Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sara Khoshayand
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
4
|
Llewellyn J, Baratam R, Culig L, Beerman I. Cellular stress and epigenetic regulation in adult stem cells. Life Sci Alliance 2024; 7:e202302083. [PMID: 39348938 PMCID: PMC11443024 DOI: 10.26508/lsa.202302083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Stem cells are a unique class of cells that possess the ability to differentiate and self-renew, enabling them to repair and replenish tissues. To protect and maintain the potential of stem cells, the cells and the environment surrounding these cells (stem cell niche) are highly responsive and tightly regulated. However, various stresses can affect the stem cells and their niches. These stresses are both systemic and cellular and can arise from intrinsic or extrinsic factors which would have strong implications on overall aging and certain disease states. Therefore, understanding the breadth of drivers, namely epigenetic alterations, involved in cellular stress is important for the development of interventions aimed at maintaining healthy stem cells and tissue homeostasis. In this review, we summarize published findings of epigenetic responses to replicative, oxidative, mechanical, and inflammatory stress on various types of adult stem cells.
Collapse
Affiliation(s)
- Joey Llewellyn
- https://ror.org/049v75w11 Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Rithvik Baratam
- https://ror.org/049v75w11 Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luka Culig
- https://ror.org/049v75w11 Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Isabel Beerman
- https://ror.org/049v75w11 Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
5
|
Lu J, Ma H, Wang Q, Song Z, Wang J. Chemotherapy-mediated lncRNA-induced immune cell plasticity in cancer immunopathogenesis. Int Immunopharmacol 2024; 141:112967. [PMID: 39181018 DOI: 10.1016/j.intimp.2024.112967] [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: 07/02/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Tumor cells engage with the immune system in a complex manner, utilizing evasion and adaptability mechanisms. The development of cancer and resistance to treatment relies on the ability of immune cells to adjust their phenotype and function in response to cues from the tumor microenvironment, known as immunological cell plasticity. This study delves into the role of long non-coding RNAs (lncRNAs) in enhancing immune cell flexibility in cancer, focusing on their regulatory actions in the tumor microenvironment and potential therapeutic implications. Through a comprehensive review of existing literature, the study analyzes the impact of lncRNAs on macrophages, T-cells, and MDSCs, as well as the influence of cytokines and growth factors like TNF, IL-6, HGF, and TGFβ on immunological cell plasticity and tumor immunoediting. LncRNAs exert a strong influence on immune cell plasticity through mechanisms such as transcriptional regulation, post-transcriptional modifications, and chromatin remodeling. These RNA molecules intricately modulate gene expression networks, acting as scaffolding, decoys, guides, and sponges. Moreover, both direct cell-cell interactions and soluble chemicals in the tumor microenvironment contribute to enhancing immune cell activation and survival. Understanding the influence of lncRNAs on immune cell flexibility sheds light on the biological pathways of immune evasion and cancer progression. Targeting long non-coding RNAs holds promise for amplifying anti-tumor immunity and overcoming drug resistance in cancer treatment. However, further research is necessary to determine the therapeutic potential of manipulating lncRNAs in the tumor microenvironment.
Collapse
Affiliation(s)
- Jingyuan Lu
- Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China.
| | - Haowei Ma
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Qian Wang
- Division of Hematology and Solid Tumor Oncology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Zhiheng Song
- Plasma Applied Physics Lab, C&J Nyheim Plasma Institute, Drexel University, 200 Federal St, Suite 500, Camden, NJ 08103.
| | - Jinli Wang
- School of Medicine, Department of Epidemiology and Biochemistry and Molecular & Cellular Biology, Georgetown University, 3700 O ST NW, Washington, DC 20057.
| |
Collapse
|
6
|
Ji M, Yu D, Liu X, Wang L, Zhang D, Yang Z, Huang W, Fan H, Wang L, Sun H. Glutathione-dependent degradation of SMARCA2/4 for targeted lung cancer therapy with improved selectivity. Eur J Med Chem 2024; 277:116751. [PMID: 39128328 DOI: 10.1016/j.ejmech.2024.116751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/20/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
SMARCA2 and SMARCA4 are the mutually exclusive catalytic subunits of the mammalian Switch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex, and have recently been considered as attractive synthetic lethal targets for PROTAC-based cancer therapy. However, the potential off-tissue toxicity towards normal tissues remains a concern. Here, we optimize a GSH-inducible SMARCA2/4-based PROTAC precursor with selective antitumor activity towards lung cancer cells and negligible cytotoxicity towards normal cells in both in vitro and in vivo studies. The precursor is not bioactive or cytotoxic, but preferentially responds to endogenous GSH in GSH-rich lung cancer cells, releasing active PROTAC to degrade SMARCA2/4 via PROTAC-mediated proteasome pathway. Subsequent xenograft model study reveals that selective SMARCA2/4 degradation in lung tumors triggers DNA damage and apoptosis, which significantly inhibits lung cancer cell proliferation without obvious adverse events towards normal tissues. This study exemplifies the targeted degradation of SMARCA2/4 in lung cancer cells by the GSH-responsive PROTAC precursor, highlighting its potential as an encouraging cancer therapeutic strategy.
Collapse
Affiliation(s)
- Ming Ji
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Dehao Yu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Xinmin Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Luo Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Dongli Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Zhengduo Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Wanqiao Huang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Heli Fan
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Lulu Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Huabing Sun
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
7
|
Mendes FC, Garcia-Larsen V, Moreira A. Obesity and Asthma: Implementing a Treatable Trait Care Model. Clin Exp Allergy 2024; 54:881-894. [PMID: 38938020 DOI: 10.1111/cea.14520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Recognition of obesity as a treatable trait of asthma, impacting its development, clinical presentation and management, is gaining widespread acceptance. Obesity is a significant risk factor and disease modifier for asthma, complicating treatment. Epidemiological evidence highlights that obese asthma correlates with poorer disease control, increased severity and persistence, compromised lung function and reduced quality of life. Various mechanisms contribute to the physiological and clinical complexities observed in individuals with obesity and asthma. These encompass different immune responses, including Type IVb, where T helper 2 cells are pivotal and driven by cytokines like interleukins 4, 5, 9 and 13, and Type IVc, characterised by T helper 17 cells and Type 3 innate lymphoid cells producing interleukin 17, which recruits neutrophils. Additionally, Type V involves immune response dysregulation with significant activation of T helper 1, 2 and 17 responses. Finally, Type VI is recognised as metabolic-induced immune dysregulation associated with obesity. Body mass index (BMI) stands out as a biomarker of a treatable trait in asthma, readily identifiable and targetable, with significant implications for disease management. There exists a notable gap in treatment options for individuals with obese asthma, where asthma management guidelines lack specificity. For example, there is currently no evidence supporting the use of incretin mimetics to improve asthma outcomes in asthmatic individuals without Type 2 diabetes mellitus (T2DM). In this review, we advocate for integrating BMI into asthma care models by establishing clear target BMI goals, promoting sustainable weight loss via healthy dietary choices and physical activity and implementing regular reassessment and referral as necessary.
Collapse
Affiliation(s)
- Francisca Castro Mendes
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional Em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Vanessa Garcia-Larsen
- Program in Human Nutrition, Department of International Health, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, USA
| | - André Moreira
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional Em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal
| |
Collapse
|
8
|
Jana K, Rammohan A, Ramani A, Gunasekaran B, Vij M, Ramamoorthi M, Jayakanthan N, Kaliamoorthy I, Ramani A, Rela M. Role of Donor-derived Cell-free DNA In Predicting Short-term Allograft Health In Liver Transplant Recipients. J Clin Exp Hepatol 2024; 14:101477. [PMID: 39170833 PMCID: PMC11334858 DOI: 10.1016/j.jceh.2024.101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024] Open
Abstract
Background/Aims Predicting allograft dysfunction prior to clinical or biochemical evidence remains one of the challenges in transplantation, and a preclinical detection and early management of its cause allows for improved post-transplant outcomes. Donor-derived cell-free DNA (ddcfDNA) has been proposed as an important biomarker of allograft injury and has shown to predict dysfunction prior to any biochemical derangements. We aimed to investigate the diagnostic performance of ddcfDNA in detecting and differentiating the causes of early pre-biochemical detection of graft injury and in predicting the short-term outcomes of graft health using a patented protocol and proprietary set of single-nucleotide polymorphisms. Methods Blood samples were collected on defined postoperative days (1, 3, 7, and at 3 months) and were analysed through relatively economical patented protocol (Trunome™). Biopsy, biochemical tests, and clinical criteria were analysed between various subgroups. Results Of a total 50 patients, percentage ddcfDNA (%ddcfDNA) levels were significantly elevated in the rejection group (n = 8) as compared to that in the non-rejection group (n = 42; median elevation: 12.8% vs 4.3%, respectively), with a significant correlation (r = 0.92, P < 0.0001). Area under the receiver operating characteristic curve (AUC-ROC) analysis revealed that the %ddcfDNA levels can predict graft health more precisely than the conventional liver function tests (AUC for %ddcfDNA: 0.86; P < 0.001; AUC for aspartate transaminase 0.65, P = 0.08; AUC for alanine transaminase: 0.75, P < 0.01). Moreover, %ddcfDNA levels (with a threshold of >10.2%) on post-operative day 7 accurately predicted short-term (3 months) health status of the graft with 93.33% sensitivity, 94.44% specificity, 87.50% positive predictive value, 97.14% negative predictive value, and 94.12% accuracy. Conclusion A single-timepoint ddcfDNA on postoperative day 7 accurately predicts graft health and improves risk stratification in the short-term.
Collapse
Affiliation(s)
- Koustav Jana
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Ashwin Rammohan
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | | | | | - Mukul Vij
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | | | | | - Ilankumaran Kaliamoorthy
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | | | - Mohamed Rela
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| |
Collapse
|
9
|
Pan X, Chandrasekaran S, Woodside JV, Riedel-Heller SG, Scherer M, Wagner M, Ramirez A, Green BD. Development, validation and application of an LC-MS/MS method quantifying free forms of the micronutrients queuine and queuosine in human plasma using a surrogate matrix approach. Anal Bioanal Chem 2024; 416:5711-5719. [PMID: 39160437 PMCID: PMC11493786 DOI: 10.1007/s00216-024-05489-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Queuosine (Q) is a hypermodified 7-deaza-guanosine nucleoside exclusively synthesized by bacteria. This micronutrient and its respective nucleobase form queuine (q) are salvaged by humans either from gut microflora or digested food. Depletion of Q-tRNA in human or mouse cells causes protein misfolding that triggers endoplasmic reticular stress and the activation of the unfolded protein responses. In vivo, this reduces the neuronal architecture of the mouse brain affecting learning and memory. Herein, a sensitive method for quantifying free q and Q in human blood was developed, optimised and validated. After evaluating q/Q extraction efficiency in several different solid-phase sorbents, Bond Elut PBA (phenylboronic acid) cartridges were found to have the highest extraction recovery for q (82%) and Q (71%) from pooled human plasma. PBS with 4% BSA was used as surrogate matrix for method development and validation. An LC-MS/MS method was validated across the concentration range of 0.0003-1 µM for both q and Q, showing excellent linearity (r2 = 0.997 (q) and r2 = 0.998 (Q)), limit of quantification (0.0003 µM), accuracy (100.39-125.71%) and precision (CV% < 15.68%). In a sampling of healthy volunteers (n = 44), there was no significant difference in q levels between male (n = 14; mean = 0.0068 µM) and female (n = 30; mean = 0.0080 µM) participants (p = 0.50). Q was not detected in human plasma. This validated method can now be used to further substantiate the role of q/Q in nutrition, physiology and pathology.
Collapse
Affiliation(s)
- Xiaobei Pan
- Advanced Asset Technology Centre, Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK.
| | - Swathine Chandrasekaran
- Advanced Asset Technology Centre, Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - Jayne V Woodside
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Steffi G Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Martin Scherer
- Department of Primary Medical Care, Center for Psychosocial Medicine, University Medical Center, Hamburg-Eppendorf, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, Medical Faculty, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, Medical Faculty, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer's and Neurodegenerative Diseases, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, 50931, Cologne, Germany
| | - Brian D Green
- Advanced Asset Technology Centre, Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
10
|
Payne K, Suriyanarayanan H, Brooks J, Mehanna H, Nankivell P, Gendoo D. Exploring the impact of intra-tumoural heterogeneity on liquid biopsy cell-free DNA methylation and copy number in head and neck squamous cell carcinoma. Oral Oncol 2024; 158:107011. [PMID: 39236578 DOI: 10.1016/j.oraloncology.2024.107011] [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: 04/17/2024] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Liquid biopsy profiling is gaining increasing promise towards biomarker-led identification and disease stratification of tumours, particularly for tumours displaying significant intra-tumoural heterogeneity (ITH). For head and neck squamous cell carcinoma (HNSCC), which display high levels of genetic ITH, identification of epigenetic modifications and methylation signatures has shown multiple uses in stratification of HNSCC for prognosis, treatment, and HPV status. In this study, we investigated the potential of liquid biopsy methylomics and genomic copy number to profile HNSCC. We conducted multi-region sampling of tumour core, tumour margin and normal adjacent mucosa, as well as plasma cell-free DNA (cfDNA) across 9 HNSCC patients. Collectively, our work highlights the prevalence of methylomic ITH in HNSCC, and demonstrates the potential of cfDNA methylation as a tool for ITH assessment and serial sampling.
Collapse
Affiliation(s)
- Karl Payne
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Harini Suriyanarayanan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jill Brooks
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul Nankivell
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Deena Gendoo
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom; Institute for Interdisciplinary Data Science and AI, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| |
Collapse
|
11
|
Branch MC, Weber M, Li MY, Flora P, Ezhkova E. Overview of chromatin regulatory processes during surface ectodermal development and homeostasis. Dev Biol 2024; 515:30-45. [PMID: 38971398 PMCID: PMC11317222 DOI: 10.1016/j.ydbio.2024.07.001] [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: 11/15/2023] [Revised: 05/02/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
The ectoderm is the outermost of the three germ layers of the early embryo that arise during gastrulation. Once the germ layers are established, the complex interplay of cellular proliferation, differentiation, and migration results in organogenesis. The ectoderm is the progenitor of both the surface ectoderm and the neural ectoderm. Notably, the surface ectoderm develops into the epidermis and its associated appendages, nails, external exocrine glands, olfactory epithelium, and the anterior pituitary. Specification, development, and homeostasis of these organs demand a tightly orchestrated gene expression program that is often dictated by epigenetic regulation. In this review, we discuss the recent discoveries that have highlighted the importance of chromatin regulatory mechanisms mediated by transcription factors, histone and DNA modifications that aid in the development of surface ectodermal organs and maintain their homeostasis post-development.
Collapse
Affiliation(s)
- Meagan C Branch
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Weber
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meng-Yen Li
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pooja Flora
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Elena Ezhkova
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Zheng X, Yang J, Wang Q, Yao P, Xiao J, Mao S, Zhang Z, Zeng Y, Zhu J, Hou J. Characterisation and evolution of the PRC2 complex and its functional analysis under various stress conditions in rice. Int J Biol Macromol 2024; 280:136124. [PMID: 39349087 DOI: 10.1016/j.ijbiomac.2024.136124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
The polycomb repressive complex 2 (PRC2) is a chromatin-associated methyltransferase responsible for catalysing the trimethylation of H3K27, an inhibitory chromatin marker associated with gene silencing. This enzymatic activity is crucial for normal organismal development and the maintenance of gene expression patterns that preserve cellular identity, subsequently influencing plant growth and abiotic stress responses. Therefore, in this study, we investigated the evolutionary characteristics and functional roles of PRC2 in plants. We identified 209 PRC2 genes, including E(z), Su(z), Esc, and Nurf55 families, using 18 representative plant species and revealed that recent gene replication events have led to an expansion in the Nurf55 family, resulting in a greater number of members compared to the E(z), Su(z), and Esc families. Furthermore, protein structure and motif composition analyses highlighted the potential functional site regions within PRC2 members. In addition, we selected rice, a representative monocotyledonous plant, as the model species for food crops. Our findings revealed that SDG711, SDG718, and MSI1-5 genes were induced by abscisic acid (ABA) and/or methyl jasmonate (MeJA) hormones, suggesting that these genes play an important role in abiotic stress and disease resistance. Further experiments involving rice blast fungus treatments confirmed that the expression of SDG711 and MSI1-5 was induced by Magnaporthe oryzae strain GUY11. Multiple protein interaction assays revealed that the M. oryzae effector AvrPiz-t interacts with PRC2 core member SDG711 to increase H3K27me3 levels. Notably, inhibition of PRC2 or mutation of SDG711 enhanced rice resistance to M. oryzae. Collectively, these results provide new insights into PRC2 evolution in plants and its significant functions in rice.
Collapse
Affiliation(s)
- Xueke Zheng
- College of Food Science and Engineering, Xinyang Agriculture and Forestry University, Xinyang 464000, China; State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jieru Yang
- College of Food Science and Engineering, Xinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Qing Wang
- College of Food Science and Engineering, Xinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Peng Yao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jian Xiao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shengxin Mao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zihan Zhang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yan Zeng
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Zhu
- College of Food Science and Engineering, Xinyang Agriculture and Forestry University, Xinyang 464000, China.
| | - Jiaqi Hou
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
13
|
Wu CS, Sun X, Liu L, Cheng L. A Live-Cell Epigenome Manipulation by Photo-Stimuli-Responsive Histone Methyltransferase Inhibitor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404608. [PMID: 39250325 DOI: 10.1002/advs.202404608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Post-translational modifications on the histone H3 tail regulate chromatin structure, impact epigenetics, and hence the gene expressions. Current chemical modulation tools, such as unnatural amino acid incorporation, protein splicing, and sortase-based editing, have allowed for the modification of histones with various PTMs in cellular contexts, but are not applicable for editing native chromatin. The use of small organic molecules to manipulate histone-modifying enzymes alters endogenous histone PTMs but lacks precise temporal and spatial control. To date, there has been no achievement in modulating histone methylation in living cells with spatiotemporal resolution. In this study, a new method is presented for temporally manipulating histone dimethylation H3K9me2 using a photo-responsive inhibitor that specifically targets the methyltransferase G9a on demand. The photo-caged molecule is stable under physiological conditions and cellular environments, but rapidly activated upon exposure to light, releasing the bioactive component that can immediately inhibit the catalytic ability of the G9a in vitro. Besides, this masked compound could also efficiently reactivate the inhibition of methyltransferase activity in living cells, subsequently suppress H3K9me2, a mark that regulates various chromatin functions. Therefore, the chemical system will be a valuable tool for manipulating the epigenome for therapeutic purposes and furthering the understanding of epigenetic mechanisms.
Collapse
Affiliation(s)
- Chuan-Shuo Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Sun
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liang Cheng
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
14
|
Chen B, Wang M, Guo Y, Zhang Z, Zhou W, Cao L, Zhang T, Ali S, Xie L, Li Y, Zinta G, Sun S, Zhang Q. Climate-related naturally occurring epimutation and their roles in plant adaptation in A. thaliana. Mol Ecol 2024; 33:e17356. [PMID: 38634782 DOI: 10.1111/mec.17356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
DNA methylation has been proposed to be an important mechanism that allows plants to respond to their environments sometimes entirely uncoupled from genetic variation. To understand the genetic basis, biological functions and climatic relationships of DNA methylation at a population scale in Arabidopsis thaliana, we performed a genome-wide association analysis with high-quality single nucleotide polymorphisms (SNPs), and found that ~56% on average, especially in the CHH sequence context (71%), of the differentially methylated regions (DMRs) are not tagged by SNPs. Among them, a total of 3235 DMRs are significantly associated with gene expressions and potentially heritable. 655 of the 3235 DMRs are associated with climatic variables, and we experimentally verified one of them, HEI10 (HUMAN ENHANCER OF CELL INVASION NO.10). Such epigenetic loci could be subjected to natural selection thereby affecting plant adaptation, and would be expected to be an indicator of accessions at risk. We therefore incorporated these climate-related DMRs into a gradient forest model, and found that the natural A. thaliana accessions in Southern Europe that may be most at risk under future climate change. Our findings highlight the importance of integrating DNA methylation that is independent of genetic variations, and climatic data to predict plants' vulnerability to future climate change.
Collapse
Affiliation(s)
- Bowei Chen
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
- College of Biology Resources and Environmental Sciences, Jishou University, Jishou, China
| | - Min Wang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Yile Guo
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Zihui Zhang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Wei Zhou
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Lesheng Cao
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Tianxu Zhang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Shahid Ali
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Linan Xie
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Yuhua Li
- College of Life Science, Northeast Forestry University, Harbin, China
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Gaurav Zinta
- Integrative Plant AdaptOmics Lab (iPAL), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (CSIR-IHBT), Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shanwen Sun
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Qingzhu Zhang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, China
- The Center for Basic Forestry Research, College of Forestry, Northeast Forestry University, Harbin, China
- College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
15
|
Kim MS, Kim DH, Lee JS. A review of environmental epigenetics in aquatic invertebrates. MARINE POLLUTION BULLETIN 2024; 208:117011. [PMID: 39326327 DOI: 10.1016/j.marpolbul.2024.117011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/30/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024]
Abstract
Aquatic ecosystems face significant challenges due to increasing human-induced environmental stressors. Recent studies emphasize the role of epigenetic mechanisms in the stress responses and adaptations of organisms to those stressors. Epigenetics influences gene expression, enabling phenotypic plasticity and transgenerational effects. Therefore, understanding the epigenetic responses of aquatic invertebrates to environmental stressors is imperative for aquatic ecosystem research. In this study, we organize the mechanisms of epigenetics in aquatic invertebrates and explore their roles in the responses of aquatic invertebrates to environmental stressors. Furthermore, we discuss the inheritance of epigenetic changes and their influence across generations in aquatic invertebrates. A comprehensive understanding of epigenetic responses is crucial for long-term ecosystem management and conservation strategies in the face of irreversible climate change in aquatic environments. In this review, we synthesize existing knowledge about environmental epigenetics in aquatic invertebrates to provide insights and suggest directions for future research.
Collapse
Affiliation(s)
- Min-Sub Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Duck-Hyun Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
16
|
Shokry D, Khan MW, Powell C, Johnson S, Rennels BC, Boyd RI, Sun Z, Fazal Z, Freemantle SJ, Parker MH, Vieson MD, Samuelson JP, Spinella MJ, Singh R. Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B. Cell Commun Signal 2024; 22:528. [PMID: 39482699 PMCID: PMC11529429 DOI: 10.1186/s12964-024-01912-3] [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: 08/27/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024] Open
Abstract
Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with "epidrugs" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4, lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.
Collapse
Affiliation(s)
- Doha Shokry
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Department of Anatomy and Embryology, Alexandria University, Alexandria, Egypt
| | - Mehwish W Khan
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Christine Powell
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Samantha Johnson
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Brayden C Rennels
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Raya I Boyd
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Zhengyang Sun
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Zeeshan Fazal
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Sarah J Freemantle
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Maryanna H Parker
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Miranda D Vieson
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA
| | - Jonathan P Samuelson
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Michael J Spinella
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA.
- Cancer Center of Illinois, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA.
| | - Ratnakar Singh
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
| |
Collapse
|
17
|
Kiliushik D, Goenner C, Law M, Schroeder GM, Srivastava Y, Jenkins JL, Wedekind JE. Knotty is nice: metabolite binding and RNA-mediated gene regulation by the preQ 1 riboswitch family. J Biol Chem 2024:107951. [PMID: 39486689 DOI: 10.1016/j.jbc.2024.107951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
Riboswitches sense specific cellular metabolites, leading to messenger RNA conformational changes that regulate downstream genes. Here we review the three known prequeosine1 (preQ1) riboswitch classes, which encompass five gene-regulatory motifs derived from distinct consensus models of folded RNA pseudoknots. Structural and functional analyses reveal multiple gene-regulation strategies ranging from partial occlusion of the ribosome-binding Shine-Dalgarno sequence (SDS), SDS sequestration driven by kinetic or thermodynamic folding pathways, direct preQ1 recognition by the SDS, and complete SDS burial in the riboswitch architecture. Family members can also induce elemental transcriptional pausing, which depends on ligand-mediated pseudoknot formation. Accordingly, preQ1 family members provide insight into a wide range of gene-regulatory tactics as well as a diverse repertoire of chemical approaches used to recognize the preQ1 metabolite. From a broader perspective, future challenges for the field will include the identification of new riboswitches in messenger RNAs that do not possess an SDS or those that induce ligand-dependent transcriptional pausing. When choosing an antibacterial target, the field must also consider how well a riboswitch accommodates mutations. Investigation of riboswitches in their natural context will also be critical to elucidate how RNA-mediated gene regulation influences organism fitness, thus providing a firm foundation for antibiotic development.
Collapse
Affiliation(s)
- Daniil Kiliushik
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Coleman Goenner
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Matthew Law
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Griffin M Schroeder
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yoshita Srivastava
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jermaine L Jenkins
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Joseph E Wedekind
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for RNA Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
18
|
Dai L, Johnson-Buck A, Laird PW, Tewari M, Walter NG. Ultrasensitive Amplification-Free Quantification of a Methyl CpG-Rich Cancer Biomarker by Single-Molecule Kinetic Fingerprinting. Anal Chem 2024; 96:17209-17216. [PMID: 39425638 DOI: 10.1021/acs.analchem.4c03002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The most well-studied epigenetic marker in humans is the 5-methyl modification of cytosine in DNA, which has great potential as a disease biomarker. Currently, quantification of DNA methylation relies heavily on bisulfite conversion followed by PCR amplification and NGS or microarray analysis. PCR is subject to potential bias in differential amplification of bisulfite-converted methylated versus unmethylated sequences. Here, we combine bisulfite conversion with single-molecule kinetic fingerprinting to develop an amplification-free assay for DNA methylation at the branched-chain amino acid transaminase 1 (BCAT1) promoter. Our assay selectively responds to methylated sequences with a limit of detection below 1 fM and a specificity of 99.9999%. Evaluating complex genomic DNA matrices, we reliably distinguish <5% DNA methylation at the BCAT1 promoter in whole blood DNA from completely unmethylated whole-genome amplified DNA. Taken together, these results demonstrate the feasibility and sensitivity of our amplification-free, single-molecule quantification approach to improve the early detection of methylated cancer DNA biomarkers.
Collapse
Affiliation(s)
- Liuhan Dai
- Single Molecule Analysis Group, Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Alexander Johnson-Buck
- Single Molecule Analysis Group, Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Peter W Laird
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan 49503, United States
| | - Muneesh Tewari
- Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nils G Walter
- Single Molecule Analysis Group, Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
19
|
Shaffer JF, Gupta A, Kharkwal G, Linares EE, Holmes AD, Swartz JR, Katzman S, Sharma U. Epididymis-specific RNase A family genes regulate fertility and small RNA processing. J Biol Chem 2024:107933. [PMID: 39476961 DOI: 10.1016/j.jbc.2024.107933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
Sperm small RNAs are implicated in intergenerational transmission of paternal environmental effects. Small RNAs generated by cleavage of tRNAs, known as tRNA fragments (tRFs) or tRNA-derived RNAs (tDRs or tsRNAs), are an abundant class of RNAs in mature sperm and can be modulated by environmental conditions. The biogenesis of tRFs in the male reproductive tract remains poorly understood. Angiogenin, a member of the Ribonuclease A superfamily (RNase A), cleaves tRNAs to generate tRFs in response to cellular stress. Four paralogs of Angiogenin, namely Rnase9, Rnase10, Rnase11, and Rnase12, are specifically expressed in the epididymis -a long, convoluted tubule where sperm mature and acquire fertility and motility. Here, by generating mice deleted for all four genes (Rnase9-12-/-, termed "KO" for Knock Out), we report that these genes regulate fertility and small RNA levels. KO male mice are sterile; KO sperm fertilized oocytes in vitro but failed to efficiently fertilize oocytes in vivo due to an inability of sperm to pass through the utero-tubular junction. Intriguingly, there were decreased levels of fragments of tRNAs (tRFs) and rRNAs (rRNA-derived small RNAs or rsRNAs) in the KO epididymis and epididymal luminal fluid, although RNases 9-12 did not show ribonucleolytic activity in-vitro. Importantly, KO sperm showed a dramatic decrease in the levels of tRFs, demonstrating a role of epididymis-specific Rnase9-12 genes in regulating sperm small RNA composition. Together, our results reveal an unexpected role of four epididymis-specific non-canonical RNase A family genes in regulating fertility and small RNA processing.
Collapse
Affiliation(s)
- Joshua F Shaffer
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Alka Gupta
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Geetika Kharkwal
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Edgardo E Linares
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Andrew D Holmes
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Julian R Swartz
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064
| | - Sol Katzman
- Genomics Institute, University of California, Santa Cruz, California, 95064
| | - Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, California, 95064.
| |
Collapse
|
20
|
Imre L, Nánási P, Benhamza I, Enyedi KN, Mocsár G, Bosire R, Hegedüs É, Niaki EF, Csóti Á, Darula Z, Csősz É, Póliska S, Scholtz B, Mező G, Bacsó Z, Timmers HTM, Kusakabe M, Balázs M, Vámosi G, Ausio J, Cheung P, Tóth K, Tremethick D, Harata M, Szabó G. Epigenetic modulation via the C-terminal tail of H2A.Z. Nat Commun 2024; 15:9171. [PMID: 39448645 PMCID: PMC11502880 DOI: 10.1038/s41467-024-53514-9] [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: 07/08/2022] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
H2A.Z-nucleosomes are present in both euchromatin and heterochromatin and it has proven difficult to interpret their disparate roles in the context of their stability features. Using an in situ assay of nucleosome stability and DT40 cells expressing engineered forms of the histone variant we show that native H2A.Z, but not C-terminally truncated H2A.Z (H2A.Z∆C), is released from nucleosomes of peripheral heterochromatin at unusually high salt concentrations. H2A.Z and H3K9me3 landscapes are reorganized in H2A.Z∆C-nuclei and overall sensitivity of chromatin to nucleases is increased. These tail-dependent differences are recapitulated upon treatment of HeLa nuclei with the H2A.Z-tail-peptide (C9), with MNase sensitivity being increased genome-wide. Fluorescence correlation spectroscopy revealed C9 binding to reconstituted nucleosomes. When introduced into live cells, C9 elicited chromatin reorganization, overall nucleosome destabilization and changes in gene expression. Thus, H2A.Z-nucleosomes influence global chromatin architecture in a tail-dependent manner, what can be modulated by introducing the tail-peptide into live cells.
Collapse
Affiliation(s)
- László Imre
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Nánási
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ibtissem Benhamza
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kata Nóra Enyedi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE, Supported Research Groups, Research Group of Peptide Chemistry, Budapest, Hungary
| | - Gábor Mocsár
- Damjanovich Cell Analysis Core Facility, Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine, Debrecen, Hungary
| | - Rosevalentine Bosire
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Hegedüs
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erfaneh Firouzi Niaki
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágota Csóti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Darula
- Single Cell Omics Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
- Core Facility, Proteomics Research Group, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Beáta Scholtz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Gábor Mező
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
- HUN-REN-ELTE, Supported Research Groups, Research Group of Peptide Chemistry, Budapest, Hungary
| | - Zsolt Bacsó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - H T Marc Timmers
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between the DKFZ and Medical Center-University of Freiburg and Department of Urology, Medical Center-University of Freiburg, Breisacher Str. 66, Freiburg, Germany
| | - Masayuki Kusakabe
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, 657-8501, Japan
| | - Margit Balázs
- HUN-REN-UD Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vámosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Juan Ausio
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | | | - Katalin Tóth
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - David Tremethick
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Masahiko Harata
- Laboratory of Molecular Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Gábor Szabó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
21
|
Poggiali B, Dupont ME, Jacobsen SB, Smerup MH, Christiansen SNN, Tfelt-Hansen J, Vidaki A, Morling N, Andersen JD. DNA methylation stability in cardiac tissues kept at different temperatures and time intervals. Sci Rep 2024; 14:25170. [PMID: 39448773 PMCID: PMC11502879 DOI: 10.1038/s41598-024-76027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Investigating DNA methylation (DNAm) in cardiac tissues is vital for epigenetic research in cardiovascular diseases (CVDs). During cardiac surgery, biopsies may not be immediately stored due to a lack of human or technical resources at the collection site. Assessing DNAm stability in cardiac samples left in suboptimal conditions is crucial for applying DNAm analysis. We investigated the stability of DNAm in human cardiac tissues kept at 4 °C and 22 °C for periods of 1, 7, 14, and 28 days (exposed samples) using the Illumina Infinium MethylationEPIC v1.0 BeadChip Array. We observed high correlations between samples analysed immediately after tissue collection and exposed ones (R2 > 0.992). Methylation levels were measured as β-values and median absolute β-value differences (|∆β|) ranged from 0.0093 to 0.0119 in all exposed samples. Pairwise differentially methylated position (DMP) analysis revealed no DMPs under 4 °C (fridge temperature) exposure for up to 28 days and 22 °C (room temperature) exposure for one day, while 3,437, 6,918, and 3,824 DMPs were observed for 22 °C samples at 7, 14, and 28 days, respectively. This study provides insights into the stability of genome-wide DNAm, showing that cardiac tissue can be used for reliable DNAm analysis even when stored suboptimally after surgery.
Collapse
Affiliation(s)
- Brando Poggiali
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Mikkel Eriksen Dupont
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stine Bøttcher Jacobsen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Holdgaard Smerup
- Department of Cardiothoracic Surgery, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Steffan Noe Niikanoff Christiansen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Jacob Tfelt-Hansen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Athina Vidaki
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Genetics & Cell Biology, GROW and CARIM Institutes, Maastricht University, Maastricht, The Netherlands
| | - Niels Morling
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Dyrberg Andersen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
22
|
Choudhary RK, Kumar B. V. S, Sekhar Mukhopadhyay C, Kashyap N, Sharma V, Singh N, Salajegheh Tazerji S, Kalantari R, Hajipour P, Singh Malik Y. Animal Wellness: The Power of Multiomics and Integrative Strategies: Multiomics in Improving Animal Health. Vet Med Int 2024; 2024:4125118. [PMID: 39484643 PMCID: PMC11527549 DOI: 10.1155/2024/4125118] [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: 02/12/2024] [Revised: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 11/03/2024] Open
Abstract
The livestock industry faces significant challenges, with disease outbreaks being a particularly devastating issue. These diseases can disrupt the food supply chain and the livelihoods of those involved in the sector. To address this, there is a growing need to enhance the health and well-being of livestock animals, ultimately improving their performance while minimizing their environmental impact. To tackle the considerable challenge posed by disease epidemics, multiomics approaches offer an excellent opportunity for scientists, breeders, and policymakers to gain a comprehensive understanding of animal biology, pathogens, and their genetic makeup. This understanding is crucial for enhancing the health of livestock animals. Multiomic approaches, including phenomics, genomics, epigenomics, metabolomics, proteomics, transcriptomics, microbiomics, and metaproteomics, are widely employed to assess and enhance animal health. High-throughput phenotypic data collection allows for the measurement of various fitness traits, both discrete and continuous, which, when mathematically combined, define the overall health and resilience of animals, including their ability to withstand diseases. Omics methods are routinely used to identify genes involved in host-pathogen interactions, assess fitness traits, and pinpoint animals with disease resistance. Genome-wide association studies (GWAS) help identify the genetic factors associated with health status, heat stress tolerance, disease resistance, and other health-related characteristics, including the estimation of breeding value. Furthermore, the interaction between hosts and pathogens, as observed through the assessment of host gut microbiota, plays a crucial role in shaping animal health and, consequently, their performance. Integrating and analyzing various heterogeneous datasets to gain deeper insights into biological systems is a challenging task that necessitates the use of innovative tools. Initiatives like MiBiOmics, which facilitate the visualization, analysis, integration, and exploration of multiomics data, are expected to improve prediction accuracy and identify robust biomarkers linked to animal health. In this review, we discuss the details of multiomics concerning the health and well-being of livestock animals.
Collapse
Affiliation(s)
- Ratan Kumar Choudhary
- Department of Bioinformatics, Animal Stem Cells Laboratory, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Sunil Kumar B. V.
- Department of Animal Biotechnology, Proteomics & Metabolomics Lab, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Chandra Sekhar Mukhopadhyay
- Department of Bioinformatics, Genomics Lab, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Neeraj Kashyap
- Department of Bioinformatics, Genomics Lab, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Vishal Sharma
- Department of Animal Biotechnology, Reproductive Biotechnology Lab, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Nisha Singh
- Department of Bioinformatics, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| | - Sina Salajegheh Tazerji
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Roozbeh Kalantari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pouneh Hajipour
- Department of Avian Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Yashpal Singh Malik
- Department of Microbial and Environmental Biotechnology, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141004, Punjab, India
| |
Collapse
|
23
|
Hsieh CH, Chang YTS, Yen MR, Hsieh JWA, Chen PY. Predicting protein synergistic effect in Arabidopsis using epigenome profiling. Nat Commun 2024; 15:9160. [PMID: 39448614 PMCID: PMC11502919 DOI: 10.1038/s41467-024-53565-y] [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: 09/05/2023] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Histone modifications can regulate transcription epigenetically by marking specific genomic loci, which can be mapped using chromatin immunoprecipitation sequencing (ChIP-seq). Here we present QHistone, a predictive database of 1534 ChIP-seqs from 27 histone modifications in Arabidopsis, offering three key functionalities. Firstly, QHistone employs machine learning to predict the epigenomic profile of a query protein, characterized by its most associated histone modifications, and uses these modifications to infer the protein's role in transcriptional regulation. Secondly, it predicts synergistic regulatory activities between two proteins by comparing their profiles. Lastly, it detects previously unexplored co-regulating protein pairs by screening all known proteins. QHistone accurately identifies histone modifications associated with specific known proteins, and allows users to computationally validate their results using gene expression data from various plant tissues. These functions demonstrate an useful approach to utilizing epigenome data for gene regulation analysis, making QHistone a valuable resource for the scientific community ( https://qhistone.paoyang.ipmb.sinica.edu.tw ).
Collapse
Affiliation(s)
- Chih-Hung Hsieh
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 115201, Taiwan
| | | | - Ming-Ren Yen
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 115201, Taiwan
| | - Jo-Wei Allison Hsieh
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 115201, Taiwan
| | - Pao-Yang Chen
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, 115201, Taiwan.
| |
Collapse
|
24
|
Soliman HK, Coughlan JM. United by conflict: Convergent signatures of parental conflict in angiosperms and placental mammals. J Hered 2024; 115:625-642. [PMID: 38366852 PMCID: PMC11498613 DOI: 10.1093/jhered/esae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 02/18/2024] Open
Abstract
Endosperm in angiosperms and placenta in eutherians are convergent innovations for efficient embryonic nutrient transfer. Despite advantages, this reproductive strategy incurs metabolic costs that maternal parents disproportionately shoulder, leading to potential inter-parental conflict over optimal offspring investment. Genomic imprinting-parent-of-origin-biased gene expression-is fundamental for endosperm and placenta development and has convergently evolved in angiosperms and mammals, in part, to resolve parental conflict. Here, we review the mechanisms of genomic imprinting in these taxa. Despite differences in the timing and spatial extent of imprinting, these taxa exhibit remarkable convergence in the molecular machinery and genes governing imprinting. We then assess the role of parental conflict in shaping evolution within angiosperms and eutherians using four criteria: 1) Do differences in the extent of sibling relatedness cause differences in the inferred strength of parental conflict? 2) Do reciprocal crosses between taxa with different inferred histories of parental conflict exhibit parent-of-origin growth effects? 3) Are these parent-of-origin growth effects caused by dosage-sensitive mechanisms and do these loci exhibit signals of positive selection? 4) Can normal development be restored by genomic perturbations that restore stoichiometric balance in the endosperm/placenta? Although we find evidence for all criteria in angiosperms and eutherians, suggesting that parental conflict may help shape their evolution, many questions remain. Additionally, myriad differences between the two taxa suggest that their respective biologies may shape how/when/where/to what extent parental conflict manifests. Lastly, we discuss outstanding questions, highlighting the power of comparative work in quantifying the role of parental conflict in evolution.
Collapse
Affiliation(s)
- Hagar K Soliman
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT 06511, United States
- Department of Biotechnology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Jenn M Coughlan
- Department of Ecology & Evolutionary Biology, Yale University, New Haven, CT 06511, United States
| |
Collapse
|
25
|
Wang J, Han K, Lu J. Screening of hub genes for sepsis-induced myopathy by weighted gene co-expression network analysis and protein-protein interaction network construction. BMC Musculoskelet Disord 2024; 25:834. [PMID: 39438952 PMCID: PMC11494751 DOI: 10.1186/s12891-024-07967-0] [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: 06/05/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Sepsis-induced myopathy is one of the serious complications of sepsis, which severely affects the respiratory and peripheral motor systems of patients, reduces their quality of life, and jeopardizes their lives, as evidenced by muscle atrophy, loss of strength, and impaired regeneration after injury. The pathogenesis of sepsis-induced myopathy is complex, mainly including cytokine action, enhances free radical production in muscle, increases muscle protein hydrolysis, and decreases skeletal muscle protein synthesis, etc. The above mechanisms have been demonstrated in existing studies. However, it is still unclear how the overall pattern of gene co-expression affects the pathological process of sepsis-induced myopathy. Therefore, we intend to identify hub genes and signaling pathways. Weighted gene co-expression network analysis was our main approach to study gene expression profiles: skeletal muscle transcriptome in ICU patients with sepsis-induced multi-organ failure (GSE13205). After data pre-processing, about 15,181 genes were used to identify 13 co-expression modules. Then, 16 genes (FEM1B, KLHDC3, GPX3, NIFK, GNL2, EBNA1BP2, PES1, FBP2, PFKP, BYSL, HEATR1, WDR75, TBL3, and WDR43) were selected as the hub genes including 3 up-regulated genes and 13 down-regulated genes. Then, Gene Set Enrichment Analysis was performed to show that the hub genes were closely associated with skeletal muscle dysfunction, necrotic and apoptotic skeletal myoblasts, and apoptosis in sepsis-induced myopathy. Overall, 16 candidate biomarkers were certified as reliable features for more in-depth exploration of sepsis-induced myopathy in basic and clinical studies.
Collapse
Affiliation(s)
- Jianhao Wang
- Postgraduate School, Xinjiang Medical University, Xinjiang, 830000, China
| | - Kun Han
- Postgraduate School, Xinjiang Medical University, Xinjiang, 830000, China
| | - Jinshuai Lu
- Department of Emergency, People's Hospital of Xinjiang Uygur Autonomous Region, No 91, Tian Chi Road, Xinjiang, 830001, China.
| |
Collapse
|
26
|
Chhibber T, Scherzer MT, Prokofyeva A, Becker C, Zitnay RG, Smith E, Khurana N, Skliar M, Deacon DC, VanBrocklin MW, Ghandehari H, Judson-Torres RL, Jafari P. Transdermal Delivery of Ultradeformable Cationic Liposomes Complexed with miR211-5p (UCL-211) Stabilizes BRAFV600E+ Melanocytic Nevi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618694. [PMID: 39484595 PMCID: PMC11527197 DOI: 10.1101/2024.10.17.618694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Small non-coding RNAs (e.g. siRNA, miRNA) are involved in a variety of melanocyte-associated skin conditions and act as drivers for alterations in gene expression within melanocytes. These molecular changes can potentially affect the cellular stability of melanocytes and promote their oncogenic transformation. Thus, small RNAs can be considered as therapeutic targets for these conditions, however, their topical delivery to the melanocytes through the epidermal barrier is challenging. We synthesized and extensively evaluated ultradeformable cationic liposome (UCLs) carriers complexed with synthetic microRNAs (miR211-5p; UCL-211) for transdermal delivery to melanocytes. UCL-211 complexes were characterized for their physicochemical properties, encapsulation efficiency, and deformability, which revealed a significant advantage over conventional liposomal carriers. Increased expression of miR211-5p stabilizes melanocytic nevi and keeps them in growth-arrested state. We did a comprehensive assessment of cellular delivery, and biological activity of the miR211-5p carried by UCL-211 in vitro and their permeation through the epidermis of intact skin using ex vivo human skin tissue explants. We also demonstrated, in vivo, that topical delivery of miR211-5p by UCL-211 stabilized BRAFV600E+ nevi melanocytes in a benign nevi state.
Collapse
Affiliation(s)
- Tanya Chhibber
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | - Carly Becker
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | | | - Eric Smith
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Nitish Khurana
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Mikhail Skliar
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Dekker C Deacon
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Matthew W VanBrocklin
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Robert L Judson-Torres
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Paris Jafari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
27
|
Wang FQ, Dang X, Su H, Lei Y, She CH, Zhang C, Chen X, Yang X, Yang J, Feng H, Yang W. Association of hyperactivated transposon expression with exacerbated immune activation in systemic lupus erythematosus. Mob DNA 2024; 15:23. [PMID: 39427224 PMCID: PMC11490001 DOI: 10.1186/s13100-024-00335-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is a complex autoimmune disorder, and transposable elements (TEs) have been hypothesized to play a significant role in its development. However, limited research has explored this connection. Our study aimed to examine the relationship between TE expression and SLE pathogenesis. METHODS We analyzed whole blood RNA-seq datasets from 198 SLE patients and 84 healthy controls. The REdiscoverTE pipeline was employed to quantify TE and other gene expressions, identifying differentially expressed TEs. A TE score was calculated to measure overall TE expression for each sample. Gene ontology and gene set enrichment analyses were conducted to explore the functional implications of TE upregulation. Independent datasets were utilized to replicate the results and investigate cell type-specific TE expression. RESULTS Our analysis identified two distinct patient groups: one with high TE expression and another with TE expression comparable to controls. Patients with high TE expression exhibited upregulation of pathways involving nucleic acid sensors, and TE expression was strongly correlated with interferon (IFN) signatures. Furthermore, these patients displayed deregulated cell composition, including increased neutrophils and decreased regulatory T cells. Neutrophils were suggested as the primary source of TE expression, contributing to IFN production. CONCLUSIONS Our findings suggest that TE expression may serve as a crucial mediator in maintaining the activation of interferon pathways, acting as an endogenous source of nucleic acid stimulators in SLE patients.
Collapse
Affiliation(s)
- Frank Qingyun Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiao Dang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Huidong Su
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Yao Lei
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Chun Hing She
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Caicai Zhang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Xinxin Chen
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Xingtian Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Jing Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Feng
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Szafron LA, Iwanicka-Nowicka R, Sobiczewski P, Koblowska M, Dansonka-Mieszkowska A, Kupryjanczyk J, Szafron LM. The Diversity of Methylation Patterns in Serous Borderline Ovarian Tumors and Serous Ovarian Carcinomas. Cancers (Basel) 2024; 16:3524. [PMID: 39456618 PMCID: PMC11505613 DOI: 10.3390/cancers16203524] [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: 09/07/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Changes in DNA methylation patterns are a pivotal mechanism of carcinogenesis. In some tumors, aberrant methylation precedes genetic changes, while gene expression may be more frequently modified due to methylation alterations than by mutations. Methods: Herein, 128 serous ovarian tumors were analyzed, including borderline ovarian tumors (BOTS) with (BOT.V600E) and without (BOT) the BRAF V600E mutation, low-grade (lg), and high-grade (hg) ovarian cancers (OvCa). The methylome of the samples was profiled with Infinium MethylationEPIC microarrays. Results: The biggest number of differentially methylated (DM) CpGs and regions (DMRs) was found between lgOvCa and hgOvCa. By contrast, the BOT.V600E tumors had the lowest number of DM CpGs and DMRs compared to all other groups and, in relation to BOT, their genome was strongly downmethylated. Remarkably, the ten most significant DMRs, discriminating BOT from lgOvCa, encompassed the MHC region on chromosome 6. We also identified hundreds of DMRs, being of potential use as predictive biomarkers in BOTS and hgOvCa. DMRs with the best discriminative capabilities overlapped the following genes: BAIAP3, IL34, WNT10A, NEU1, SLC44A4, and HMOX1, TCN2, PES1, RP1-56J10.8, ABR, NCAM1, RP11-629G13.1, AC006372.4, NPTXR in BOTS and hgOvCa, respectively. Conclusions: The global genome-wide hypomethylation positively correlates with the increasing aggressiveness of ovarian tumors. We also assume that the immune system may play a pivotal role in the transition from BOTS to lgOvCa. Given that the BOT.V600E tumors had the lowest number of DM CpGs and DMRs compared to all other groups, when methylome is considered, such tumors might be placed in-between BOT and OvCa.
Collapse
Affiliation(s)
- Laura A. Szafron
- Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Roksana Iwanicka-Nowicka
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland; (R.I.-N.); (M.K.)
- Laboratory for Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Sobiczewski
- Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Marta Koblowska
- Laboratory of Systems Biology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland; (R.I.-N.); (M.K.)
- Laboratory for Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Agnieszka Dansonka-Mieszkowska
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Jolanta Kupryjanczyk
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Lukasz M. Szafron
- Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
29
|
Shokry D, Khan MW, Powell C, Johnson S, Rennels BC, Boyd RI, Sun Z, Fazal Z, Freemantle SJ, Parker MH, Vieson MD, Samuelson JP, Spinella MJ, Singh R. Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B. RESEARCH SQUARE 2024:rs.3.rs-4986186. [PMID: 39483904 PMCID: PMC11527238 DOI: 10.21203/rs.3.rs-4986186/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with "epidrugs" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4 , lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.
Collapse
|
30
|
Jasim SA, Altalbawy FMA, Abohassan M, Oghenemaro EF, Bishoyi AK, Singh RP, Kaur P, Sivaprasad GV, Mohammed JS, Hulail HM. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem Biophys 2024:10.1007/s12013-024-01587-0. [PMID: 39419931 DOI: 10.1007/s12013-024-01587-0] [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] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The histone acetyl transferases (HATs) and histone deacetylases (HDACs), which are mostly recognized for their involvement in regulating chromatin remodeling via histone acetylation/deacetylation, have been shown to also change several non-histone proteins to regulate other cellular processes. Acetylation affects the activity or function of cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors in connection to inflammation. Some small-molecule HDAC inhibitors are utilized as anticancer medications in clinical settings due to their capability to regulate cellular growth arrest, differentiation, and death. Here, we summarize our present knowledge of the innate and adaptive immunological pathways that classical HDAC enzymes control. The aim is to justify the targeted (or non-targeted) use of inhibitors against certain HDAC enzymes in inflammatory diseases such as arthritis, inflammatory bowel diseases (IBD), airways inflammation and neurological diseases.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
31
|
Fagery M, Khorshidi HA, Wong SQ, Karanfil Ö, Emery J, IJzerman MJ. Integrating Multi-Cancer Early Detection (MCED) Tests with Standard Cancer Screening: System Dynamics Model Development and Feasibility Testing. PHARMACOECONOMICS - OPEN 2024:10.1007/s41669-024-00533-3. [PMID: 39424759 DOI: 10.1007/s41669-024-00533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Cancer screening plays a critical role in early disease detection and improving outcomes. In Australia, established screening protocols for colorectal, breast and cervical cancer have significantly contributed to timely cancer detection. However, the recent introduction of multi-cancer early detection (MCED) tests arguably can disrupt current screening, yet the extent to which these tests provide additional benefits remains uncertain. We present the development and initial validation of a system dynamics (SD) model that estimates the additional cancer detections and costs associated with MCED tests. AIM This article describes the development of a simulation model built to evaluate the additional patient diagnoses and the economic impact of incorporating MCED testing alongside Australia's well-established standard of care (SOC) screening programs for colorectal, breast, cervical and lung cancers. The model was designed to estimate the additional number of patients diagnosed at each cancer stage (stage I, II, III, IV, or unknown) and the associated costs. This simulation model allows for the analysis of multiple scenarios under a plausible set of assumptions regarding population-level participation rates. METHODS An SD model was developed to represent the existing SOC national cancer screening pathways and to integrate potential clinical pathways that could be introduced by MCED tests. The SD model was built to investigate three scenarios for the use of MCED testing: firstly, to explore the viability of MCED testing as a substitute among individuals who are not opting for SOC screening for any reason; secondly, to implement MCED testing exclusively for individuals ineligible for SOC screening, yet have high-risk characteristics; and thirdly, to employ MCED testing after SOC screening to serve as a triaging/confirmatory tool for individuals receiving inconclusive test results. The three primary scenarios were constructed by varying diagnostic accuracy and uptake rates of MCED tests. DISCUSSION The clinical utility and outcomes of MCED testing for screening and early detection still lack comprehensive evidence. Nonetheless, this simulation model facilitates a thorough analysis of MCED tests within the Australian healthcare context, providing insights into potential additional detections and costs to the healthcare system, which may help prioritise future evidence development. The adaptable yet novel SD model presented herein is anticipated to be of considerable interest to industry, policymakers, consumers and clinicians involved in informing clinical and economic decisions regarding integrating MCED tests as cancer screening and early detection tools. The expected results of applying this SD model will determine whether using MCED testing in conjunction with SOC screening offers any potential benefits, possibly guiding policy decisions and clinical practices towards the adoption of MCED tests.
Collapse
Affiliation(s)
- Mussab Fagery
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
| | - Hadi A Khorshidi
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Oncology, Sir Peter MacCallum, University of Melbourne, Melbourne, Australia
| | - Özge Karanfil
- College of Administrative Sciences and Economics and School of Medicine, Koç University, Istanbul, Turkey
- MIT Sloan School of Management, Boston, MA, USA
| | - Jon Emery
- Department of General Practice and Primary Care, Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
32
|
Bohrer CH, Fursova NA, Larson DR. Enhancers: A Focus on Synthetic Biology and Correlated Gene Expression. ACS Synth Biol 2024; 13:3093-3108. [PMID: 39276360 DOI: 10.1021/acssynbio.4c00244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Enhancers are central for the regulation of metazoan transcription but have proven difficult to study, primarily due to a myriad of interdependent variables shaping their activity. Consequently, synthetic biology has emerged as the main approach for dissecting mechanisms of enhancer function. We start by reviewing simple but highly parallel reporter assays, which have been successful in quantifying the complexity of the activator/coactivator mechanisms at enhancers. We then describe studies that examine how enhancers function in the genomic context and in combination with other enhancers, revealing that they activate genes through a variety of different mechanisms, working together as a system. Here, we primarily focus on synthetic reporter genes that can quantify the dynamics of enhancer biology through time. We end by considering the consequences of having many genes and enhancers within a 'local environment', which we believe leads to correlated gene expression and likely reports on the general principles of enhancer biology.
Collapse
Affiliation(s)
- Christopher H Bohrer
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nadezda A Fursova
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Daniel R Larson
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
33
|
Zhao T, Guan X, Hu Y, Zhang Z, Yang H, Shi X, Han J, Mei H, Wang L, Shao L, Wu H, Chen Q, Zhao Y, Pan J, Hao Y, Dong Z, Long X, Deng Q, Zhao S, Zhang M, Zhu Y, Ma X, Chen Z, Deng Y, Si Z, Li X, Zhang T, Gu F, Gu X, Fang L. Population-wide DNA methylation polymorphisms at single-nucleotide resolution in 207 cotton accessions reveal epigenomic contributions to complex traits. Cell Res 2024:10.1038/s41422-024-01027-x. [PMID: 39420233 DOI: 10.1038/s41422-024-01027-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/01/2024] [Indexed: 10/19/2024] Open
Abstract
DNA methylation plays multiple regulatory roles in crop development. However, the relationships of methylation polymorphisms with genetic polymorphisms, gene expression, and phenotypic variation in natural crop populations remain largely unknown. Here, we surveyed high-quality methylomes, transcriptomes, and genomes obtained from the 20-days-post-anthesis (DPA) cotton fibers of 207 accessions and extended the classical framework of population genetics to epigenetics. Over 287 million single methylation polymorphisms (SMPs) were identified, 100 times more than the number of single nucleotide polymorphisms (SNPs). These SMPs were significantly enriched in intragenic regions while depleted in transposable elements. Association analysis further identified a total of 5,426,782 cis-methylation quantitative trait loci (cis-meQTLs), 5078 cis-expression quantitative trait methylation (cis-eQTMs), and 9157 expression quantitative trait loci (eQTLs). Notably, 36.39% of cis-eQTM genes were not associated with genetic variation, indicating that a large number of SMPs associated with gene expression variation are independent of SNPs. In addition, out of the 1715 epigenetic loci associated with yield and fiber quality traits, only 36 (2.10%) were shared with genome-wide association study (GWAS) loci. The construction of multi-omics regulatory networks revealed 43 cis-eQTM genes potentially involved in fiber development, which cannot be identified by GWAS alone. Among these genes, the role of one encoding CBL-interacting protein kinase 10 in fiber length regulation was successfully validated through gene editing. Taken together, our findings prove that DNA methylation data can serve as an additional resource for breeding purposes and can offer opportunities to enhance and expedite the crop improvement process.
Collapse
Affiliation(s)
- Ting Zhao
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueying Guan
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yan Hu
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ziqian Zhang
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Han Yang
- Damo Academy, Alibaba Group, Hangzhou, Zhejiang, China
| | - Xiaowen Shi
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jin Han
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huan Mei
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Luyao Wang
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China
| | - Lei Shao
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongyu Wu
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qianqian Chen
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongyan Zhao
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiaying Pan
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yupeng Hao
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zeyu Dong
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuan Long
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qian Deng
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shengjun Zhao
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China
| | - Mengke Zhang
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China
| | - Yumeng Zhu
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China
| | - Xiaowei Ma
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zequan Chen
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yayuan Deng
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China
| | - Zhanfeng Si
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Li
- Damo Academy, Alibaba Group, Hangzhou, Zhejiang, China
- Hupan Lab, Hangzhou, Zhejiang, China
| | - Tianzhen Zhang
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China.
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China.
| | - Fei Gu
- Damo Academy, Alibaba Group, Hangzhou, Zhejiang, China.
- Hupan Lab, Hangzhou, Zhejiang, China.
| | - Xiaofeng Gu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.
| | - Lei Fang
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, the Advance Seed Institute, Key Laboratory of Plant FactoryGeneration-adding Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, China.
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, Hainan, China.
| |
Collapse
|
34
|
Nadhan R, Isidoro C, Song YS, Dhanasekaran DN. LncRNAs and the cancer epigenome: Mechanisms and therapeutic potential. Cancer Lett 2024; 605:217297. [PMID: 39424260 DOI: 10.1016/j.canlet.2024.217297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators of epigenome, modulating gene expression through DNA methylation, histone modification, and/or chromosome remodeling. Dysregulated lncRNAs act as oncogenes or tumor suppressors, driving tumor progression by shaping the cancer epigenome. By interacting with the writers, readers, and erasers of the epigenetic script, lncRNAs induce epigenetic modifications that bring about changes in cancer cell proliferation, apoptosis, epithelial-mesenchymal transition, migration, invasion, metastasis, cancer stemness and chemoresistance. This review analyzes and discusses the multifaceted role of lncRNAs in cancer pathobiology, from cancer genesis and progression through metastasis and therapy resistance. It also explores the therapeutic potential of targeting lncRNAs through innovative diagnostic, prognostic, and therapeutic strategies. Understanding the dynamic interplay between lncRNAs and epigenome is crucial for developing personalized therapeutic strategies, offering new avenues for precision cancer medicine.
Collapse
Affiliation(s)
- Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and NanoBioImaging, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 151-921, South Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
35
|
Gorelov R, Hochedlinger K. A cellular identity crisis? Plasticity changes during aging and rejuvenation. Genes Dev 2024; 38:823-842. [PMID: 39293862 PMCID: PMC11535162 DOI: 10.1101/gad.351728.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Cellular plasticity in adult multicellular organisms is a protective mechanism that allows certain tissues to regenerate in response to injury. Considering that aging involves exposure to repeated injuries over a lifetime, it is conceivable that cell identity itself is more malleable-and potentially erroneous-with age. In this review, we summarize and critically discuss the available evidence that cells undergo age-related shifts in identity, with an emphasis on those that contribute to age-associated pathologies, including neurodegeneration and cancer. Specifically, we focus on reported instances of programs associated with dedifferentiation, biased differentiation, acquisition of features from alternative lineages, and entry into a preneoplastic state. As some of the most promising approaches to rejuvenate cells reportedly also elicit transient changes to cell identity, we further discuss whether cell state change and rejuvenation can be uncoupled to yield more tractable therapeutic strategies.
Collapse
Affiliation(s)
- Rebecca Gorelov
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA;
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
36
|
Longhurst AD, Wang K, Suresh HG, Ketavarapu M, Ward HN, Jones IR, Narayan V, Hundley FV, Hassan AZ, Boone C, Myers CL, Shen Y, Ramani V, Andrews BJ, Toczyski DP. The PRC2.1 Subcomplex Opposes G1 Progression through Regulation of CCND1 and CCND2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585604. [PMID: 38562687 PMCID: PMC10983909 DOI: 10.1101/2024.03.18.585604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Progression through the G1 phase of the cell cycle is the most highly regulated step in cellular division. We employed a chemogenetic approach to discover novel cellular networks that regulate cell cycle progression. This approach uncovered functional clusters of genes that altered sensitivity of cells to inhibitors of the G1/S transition. Mutation of components of the Polycomb Repressor Complex 2 rescued proliferation inhibition caused by the CDK4/6 inhibitor palbociclib, but not to inhibitors of S phase or mitosis. In addition to its core catalytic subunits, mutation of the PRC2.1 accessory protein MTF2, but not the PRC2.2 protein JARID2, rendered cells resistant to palbociclib treatment. We found that PRC2.1 (MTF2), but not PRC2.2 (JARID2), was critical for promoting H3K27me3 deposition at CpG islands genome-wide and in promoters. This included the CpG islands in the promoter of the CDK4/6 cyclins CCND1 and CCND2, and loss of MTF2 lead to upregulation of both CCND1 and CCND2. Our results demonstrate a role for PRC2.1, but not PRC2.2, in antagonizing G1 progression in a diversity of cell linages, including CML, breast cancer and immortalized cell lines.
Collapse
Affiliation(s)
- Adam D Longhurst
- University of California, San Francisco, San Francisco, CA 94158, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kyle Wang
- Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Harsha Garadi Suresh
- Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Mythili Ketavarapu
- Gladstone Institute for Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Henry N Ward
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota - Twin Cities Minneapolis MN USA
| | - Ian R Jones
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California
| | - Vivek Narayan
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Frances V Hundley
- University of California, San Francisco, San Francisco, CA 94158, USA
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA 02115, USA
| | - Arshia Zernab Hassan
- Department of Computer Science and Engineering, University of Minnesota - Twin Cities Minneapolis MN USA
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Chad L Myers
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota - Twin Cities Minneapolis MN USA
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA 02115, USA
| | - Yin Shen
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vijay Ramani
- Gladstone Institute for Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Brenda J Andrews
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - David P Toczyski
- University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
37
|
Iona A, Yao P, Pozarickij A, Kartsonaki C, Said S, Wright N, Lin K, Millwood I, Fry H, Mazidi M, Wang B, Chen Y, Du H, Yang L, Avery D, Schmidt D, Sun D, Pei P, Lv J, Yu C, Hill M, Chen J, Bragg F, Bennett D, Walters R, Li L, Clarke R, Chen Z. Proteo-genomic analyses in relatively lean Chinese adults identify proteins and pathways that affect general and central adiposity levels. Commun Biol 2024; 7:1327. [PMID: 39406990 PMCID: PMC11480319 DOI: 10.1038/s42003-024-06984-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/28/2024] [Indexed: 10/19/2024] Open
Abstract
Adiposity is an established risk factor for multiple diseases, but the causal relationships of different adiposity types with circulating protein biomarkers have not been systematically investigated. We examine the causal associations of general and central adiposity with 2923 plasma proteins among 3977 Chinese adults (mean BMI = 23.9 kg/m²). Genetically-predicted body mass index (BMI), body fat percentage (BF%), waist circumference (WC), and waist-to-hip ratio (WHR) are significantly (FDR < 0.05) associated with 399, 239, 436, and 283 proteins, respectively, with 80 proteins associated with all four and 275 with only one adiposity trait. WHR is associated with the most proteins (n = 90) after adjusting for other adiposity traits. These associations are largely replicated in Europeans (mean BMI = 27.4 kg/m²). Two-sample Mendelian randomisation (MR) analyses in East Asians using cis-protein quantitative trait locus (cis-pQTLs) identified in GWAS find 30/2 proteins significantly affect levels of BMI/WC, respectively, with 10 showing evidence of colocalisation, and seven (inter-alpha-trypsin inhibitor heavy chain H3, complement factor B, EGF-containing fibulin-like extracellular matrix protein 1, thioredoxin domain-containing protein 15, alpha-2-antiplasmin, fibronectin, mimecan) are replicated in separate MR using different cis-pQTLs identified in Europeans. These findings identified potential novel mechanisms and targets, to our knowledge, for improved treatment and prevention of obesity and associated diseases.
Collapse
Affiliation(s)
- Andri Iona
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Saredo Said
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hannah Fry
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mohsen Mazidi
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Baihan Wang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dianjianyi Sun
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Ministry of Education, Peking University, Beijing, China
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Jun Lv
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Ministry of Education, Peking University, Beijing, China
| | - Canqing Yu
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Ministry of Education, Peking University, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Fiona Bragg
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Health Data Research UK Oxford, University of Oxford, Oxford, UK
| | - Derrick Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Ministry of Education, Peking University, Beijing, China
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Grisolia P, Tufano R, Iannarone C, De Falco A, Carlino F, Graziano C, Addeo R, Scrima M, Caraglia F, Ceccarelli A, Nuzzo PV, Cossu AM, Forte S, Giuffrida R, Orditura M, Caraglia M, Ceccarelli M. Differential methylation of circulating free DNA assessed through cfMeDiP as a new tool for breast cancer diagnosis and detection of BRCA1/2 mutation. J Transl Med 2024; 22:938. [PMID: 39407254 PMCID: PMC11476115 DOI: 10.1186/s12967-024-05734-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Recent studies have highlighted the importance of the cell-free DNA (cfDNA) methylation profile in detecting breast cancer (BC) and its different subtypes. We investigated whether plasma cfDNA methylation, using cell-free Methylated DNA Immunoprecipitation and High-Throughput Sequencing (cfMeDIP-seq), may be informative in characterizing breast cancer in patients with BRCA1/2 germline mutations for early cancer detection and response to therapy. METHODS We enrolled 23 BC patients with germline mutation of BRCA1 and BRCA2 genes, 19 healthy controls without BRCA1/2 mutation, and two healthy individuals who carried BRCA1/2 mutations. Blood samples were collected for all study subjects at the diagnosis, and plasma was isolated by centrifugation. Cell-free DNA was extracted from 1 mL of plasma, and cfMeDIP-seq was performed for each sample. Shallow whole genome sequencing was performed on the immuno-precipitated samples. Then, the differentially methylated 300-bp regions (DMRs) between 25 BRCA germline mutation carriers and 19 non-carriers were identified. DMRs were compared with tumor-specific regions from public datasets to perform an unbiased analysis. Finally, two statistical classifiers were trained based on the GLMnet and random forest model to evaluate if the identified DMRs could discriminate BRCA-positive from healthy samples. RESULTS We identified 7,095 hypermethylated and 212 hypomethylated regions in 25 BRCA germline mutation carriers compared to 19 controls. These regions discriminate tumors from healthy samples with high accuracy and sensitivity. We show that the circulating tumor DNA of BRCA1/2 mutant breast cancers is characterized by the hypomethylation of genes involved in DNA repair and cell cycle. We uncovered the TFs associated with these DRMs and identified that proteins of the Erythroblast Transformation Specific (ETS) family are particularly active in the hypermethylated regions. Finally, we assessed that these regions could discriminate between BRCA positives from healthy samples with an AUC of 0.95, a sensitivity of 88%, and a specificity of 94.74%. CONCLUSIONS Our study emphasizes the importance of tumor cell-derived DNA methylation in BC, reporting a different methylation profile between patients carrying mutations in BRCA1, BRCA2, and wild-type controls. Our minimally invasive approach could allow early cancer diagnosis, assessment of minimal residual disease, and monitoring of response to therapy.
Collapse
Affiliation(s)
- Piera Grisolia
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
| | - Rossella Tufano
- Laboratory of Computational Biology, IRGS, Ariano Irpino, Italy
| | - Clara Iannarone
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
| | | | - Francesca Carlino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 7, 80138, Naples, Italy
- Oncology Unit, San Felice a Cancello Hospital, ASL Caserta, Sanfelice a Cancello, Italy
| | - Cinzia Graziano
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
| | - Raffaele Addeo
- Oncology Unit, S. Giovanni di Dio Hospital, ASL Napoli2 Nord, Frattamaggiore, Italy
| | - Marianna Scrima
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
| | - Francesco Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Anna Ceccarelli
- Medical Oncology, Catholic University of the Sacred Heart, 00168, Rome, RM, Italy
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Alessia Maria Cossu
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 7, 80138, Naples, Italy
| | | | | | - Michele Orditura
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Michele Caraglia
- Laboratory of Molecular and Precision Oncology, Biogem, IRGS, Ariano Irpino, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, 7, 80138, Naples, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
39
|
Tuminello S, Durmus N, Snuderl M, Chen Y, Shao Y, Reibman J, Arslan AA, Taioli E. DNA Methylation as a Molecular Mechanism of Carcinogenesis in World Trade Center Dust Exposure: Insights from a Structured Literature Review. Biomolecules 2024; 14:1302. [PMID: 39456235 PMCID: PMC11506790 DOI: 10.3390/biom14101302] [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: 09/06/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The collapse of the World Trade Center (WTC) buildings in New York City generated a large plume of dust and smoke. WTC dust contained human carcinogens including metals, asbestos, polycyclic aromatic hydrocarbons (PAHs), persistent organic pollutants (POPs, including polychlorinated biphenyls (PCBs) and dioxins), and benzene. Excess levels of many of these carcinogens have been detected in biological samples of WTC-exposed persons, for whom cancer risk is elevated. As confirmed in this structured literature review (n studies = 80), all carcinogens present in the settled WTC dust (metals, asbestos, benzene, PAHs, POPs) have previously been shown to be associated with DNA methylation dysregulation of key cancer-related genes and pathways. DNA methylation is, therefore, a likely molecular mechanism through which WTC exposures may influence the process of carcinogenesis.
Collapse
Affiliation(s)
- Stephanie Tuminello
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nedim Durmus
- Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA
| | - Matija Snuderl
- Department of Pathology, NYU Langone Medical Center, New York, NY 10016, USA;
| | - Yu Chen
- Department of Population Health, NYU Langone Medical Center, New York, NY 10016, USA
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Yongzhao Shao
- Department of Population Health, NYU Langone Medical Center, New York, NY 10016, USA
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
| | - Joan Reibman
- Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA
- Division of Environmental Medicine, Department of Medicine, NYU Langone Medical Center, New York, NY 10016, USA
| | - Alan A. Arslan
- Department of Population Health, NYU Langone Medical Center, New York, NY 10016, USA
- NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA
- Department of Obstetrics and Gynecology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Emanuela Taioli
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
40
|
Xu T, Du T, Zhuang X, He X, Yan Y, Wu J, Zhou H, Li Y, Liao X, He J, Liu C, Dong Y, Ou J, Lin S, Chen D, Huang ZP. Loss of NAT10 Reduces the Translation of Kmt5a mRNA Through ac4C Modification in Cardiomyocytes and Induces Heart Failure. J Am Heart Assoc 2024; 13:e035714. [PMID: 39392166 DOI: 10.1161/jaha.124.035714] [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: 03/24/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND In the past decade, the biological functions of various RNA modifications in mammals have been uncovered. N4-acetylcytidine (ac4C), a highly conserved RNA modification, has been implicated in human diseases. Despite this, the involvement of RNA ac4C modification in cardiac physiology and pathology remains incompletely understood. NAT10 (N-acetyltransferase 10) stands as the sole acetyltransferase known to catalyze RNA ac4C modification. This study aims to explore the role of NAT10 and ac4C modification in cardiac physiology and pathology. METHODS AND RESULTS Cardiac-specific knockout of NAT10, leading to reduced RNA ac4C modification, during both neonatal and adult stages resulted in severe heart failure. NAT10 deficiency induced cardiomyocyte apoptosis, a crucial step in heart failure pathogenesis, supported by in vitro data. Activation of the p53 signaling pathway was closely associated with enhanced apoptosis in NAT10-deficient cardiomyocytes. As ac4C modification on mRNA influences translational efficiency, we employed ribosome footprints coupled with RNA sequencing to explore genome-wide translational efficiency changes caused by NAT10 deficiency. We identified and validated that the translational efficiency of Kmt5a was suppressed in NAT10 knockout hearts due to reduced ac4C modification on its mRNA. This finding was consistent with the observation that Kmt5a protein levels were reduced in heart failure despite unchanged mRNA expression. Knockdown of Kmt5a in cardiomyocytes recapitulated the phenotype of NAT10 deficiency, including increased cardiomyocyte apoptosis and activated p53 signaling. Finally, overexpression of Kmt5a rescued cardiomyocyte apoptosis and p53 activation induced by NAT10 inhibition. CONCLUSIONS Our study highlights the significance of NAT10 in cardiomyocyte physiology, demonstrating that NAT10 loss is sufficient to induce cardiomyocyte apoptosis and heart failure. NAT10 regulates the translational efficiency of Kmt5a, a key mediator, through mRNA ac4C modification during heart failure.
Collapse
Affiliation(s)
- Ting Xu
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Tailai Du
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Xiaodong Zhuang
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Xin He
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Youchen Yan
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Jialing Wu
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Huimin Zhou
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Yan Li
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
- Division of Cardiac Surgery National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- Key Laboratory of Assisted Circulation and Vascular Diseases Chinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Jiangui He
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Chen Liu
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
- Key Laboratory of Assisted Circulation and Vascular Diseases Chinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
| | - Jingsong Ou
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
- Division of Cardiac Surgery National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- Key Laboratory of Assisted Circulation and Vascular Diseases Chinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| | - Shuibin Lin
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| | - Demeng Chen
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University) Guangzhou China
- Division of Cardiac Surgery National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
- Key Laboratory of Assisted Circulation and Vascular Diseases Chinese Academy of Medical Sciences, The First Affiliated Hospital, Sun Yat-sen University Guangzhou China
| |
Collapse
|
41
|
Li J, Huang X, Luo L, Sun J, Guo Q, Yang X, Zhang C, Ni B. The role of p53 in male infertility. Front Endocrinol (Lausanne) 2024; 15:1457985. [PMID: 39469578 PMCID: PMC11513281 DOI: 10.3389/fendo.2024.1457985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
The tumor suppressor p53 is a transcription factor involved in a variety of crucial cellular functions, including cell cycle arrest, DNA repair and apoptosis. Still, a growing number of studies indicate that p53 plays multiple roles in spermatogenesis, as well as in the occurrence and development of male infertility. The representative functions of p53 in spermatogenesis include the proliferation of spermatogonial stem cells (SSCs), spermatogonial differentiation, spontaneous apoptosis, and DNA damage repair. p53 is involved in various male infertility-related diseases. Innovative therapeutic strategies targeting p53 have emerged in recent years. This review focuses on the role of p53 in spermatogenesis and male infertility and analyses the possible underlying mechanism involved. All these conclusions may provide a new perspective on drug intervention targeting p53 for male infertility treatment.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xia Huang
- Department of Human Resource, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Luo
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jialin Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qie Guo
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue Yang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuanzhou Zhang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Beibei Ni
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
42
|
Jiang TQ, Wang H, Cheng WX, Xie C. Modulation of host N6-methyladenosine modification by gut microbiota in colorectal cancer. World J Gastroenterol 2024; 30:4175-4193. [PMID: 39493326 PMCID: PMC11525875 DOI: 10.3748/wjg.v30.i38.4175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
As a research hotspot in the field of molecular biology, N6-methyladenosine (m6A) modification has made progress in the treatment of colorectal cancer (CRC), leukemia and other cancers. Numerous studies have demonstrated that the tumour microenvironment (TME) regulates the level of m6A modification in the host and activates a series of complex epigenetic signalling pathways through interactions with CRC cells, thus affecting the progression and prognosis of CRC. However, with the diversity in the composition of TME factors, this action is reciprocal and complex. Encouragingly, some studies have experimentally revealed that the intestinal flora can alter CRC cell proliferation by directly acting on m6A and thereby altering CRC cell proliferation. This review summarizes the data, supporting the idea that the intestinal flora can influence host m6A levels through pathways such as methyl donor metabolism and thus affect the progression of CRC. We also review the role of m6A modification in the diagnosis, treatment, and prognostic assessment of CRC and discuss the current status, limitations, and potential clinical value of m6A modification in this field. We propose that additional in-depth research on m6A alterations in CRC patients and their TME-related targeted therapeutic issues will lead to better therapeutic outcomes for CRC patients.
Collapse
Affiliation(s)
- Tian-Qi Jiang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Hao Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wang-XinJun Cheng
- Queen Mary College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chuan Xie
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
43
|
Qiao P, Du H, Guo X, Yu M, Zhang C, Shi Y. Serum exosomal miR-200c is a potential diagnostic biomarker for breast cancer. Biomarkers 2024:1-8. [PMID: 39317236 DOI: 10.1080/1354750x.2024.2406520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Breast cancer (BC) is one of the most common malignancies in women. Exosomes are widely found in body fluids and carry microRNAs (miRNAs) that reflect the biological properties of the parental cells. Our study aimed to investigate the differential expression of miR-200c in BC serum exosomes and its diagnostic value. METHODOLOGY miRNA profiles in culture supernatant exosomes of normal mammary epithelial cells MCF-10A and BC cells (MCF-7, MDA-MB-231, MCF-7 Taxol) were examined by miRNA deep sequencing to screen for significantly differentially expressed miRNAs; Transmission electron microscopy (TEM), Nanoparticle tracking analysis (NTA), and Western blot were used to identify exosomes; qPCR was used to detect the expression level of miR-200c in cellular exosomes and serum exosomes; The efficacy of individual and combined tests of each indicator to diagnose BC was evaluated using receiver operating characteristic (ROC) curves. RESULTS We identified typical exosome features by TEM, NTA and Western blot, indicating successful exosome extraction. Then our miRNA sequencing results and qRT-PCR experiments showed that miR-200c was significantly down-regulated in BC cell exosomes. In addition, we divided the clinical serum samples into two cohorts according to region, and in independent cohort I, the serum exosomal miR-200c levels of BC patients were significantly lower than those of healthy controls. In cohort II, serum exosomal miR-200c expression was significantly lower in the BC group than in the control and benign breast disease (BBD) groups, whereas miR-200c expression in the BBD group was not statistically different from that in the control group. ROC analyses in both independent cohorts confirmed that serum exosomal miR-200c could differentiate between patients with and without BC disease and could be used as an early diagnostic marker for BC disease. CONCLUSION Serum exosome miR-200c can be used as a potential biomarker for the diagnosis of BC, and combined with conventional serum diagnostic markers AFP, CA125 and CA153 can help to improve diagnostic efficiency.
Collapse
Affiliation(s)
- Ping Qiao
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hua Du
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
- Department of Pathology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xin Guo
- Clinical Laboratory Center, Inner Mongolia International Mongolian Hospital, Hohhot, China
| | - Mingxuan Yu
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Caihong Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yingxu Shi
- Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
44
|
Fischetti I, De Luca M, Jachetti E. Exploring the role of EZH2 modulation in shaping the tumor microenvironment. Epigenomics 2024; 16:1265-1268. [PMID: 39387445 PMCID: PMC11534139 DOI: 10.1080/17501911.2024.2410693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Irene Fischetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michele De Luca
- Integrated Biology of Rare Tumors Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
45
|
Taryma-Leśniak O, Bińkowski J, Przybylowicz PK, Sokolowska KE, Borowski K, Wojdacz TK. Methylation patterns at the adjacent CpG sites within enhancers are a part of cell identity. Epigenetics Chromatin 2024; 17:30. [PMID: 39385277 PMCID: PMC11465701 DOI: 10.1186/s13072-024-00555-5] [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: 05/30/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND It is generally accepted that methylation status of CpG sites spaced up to 50 bp apart is correlated, and accumulation of locally disordered methylation at adjacent CpG sites is involved in neoplastic transformation, acting in similar way as stochastic accumulation of mutations. RESULTS We used EPIC microarray data from 596 samples, representing 12 healthy tissue and cell types, as well as 572 blood cancer specimens to analyze methylation status of adjacent CpG sites across human genome, and subsequently validated our findings with NGS and Sanger sequencing. Our analysis showed that there is a subset of the adjacent CpG sites in human genome, with cytosine at one CpG site methylated and the other devoid of methyl group. These loci map to enhancers that are targeted by families of transcription factors involved in cell differentiation. Moreover, our results suggest that the methylation at these loci differ between alleles within a cell, what allows for remarkable level of heterogeneity of methylation patterns. However, different types of specialized cells acquire only one specific and stable pattern of methylation at each of these loci and that pattern is to a large extent lost during neoplastic transformation. CONCLUSIONS We identified a substantial number of adjacent CpG loci in human genome that display remarkably stable and cell type specific methylation pattern. The methylation pattern at these loci appears to reflect different methylation of alleles in cells. Furthermore, we showed that changes of methylation status at those loci are likely to be involved in regulation of the activity of enhancers and contribute to neoplastic transformation.
Collapse
Affiliation(s)
- Olga Taryma-Leśniak
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Jan Bińkowski
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Patrycja Kamila Przybylowicz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Katarzyna Ewa Sokolowska
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Konrad Borowski
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland
| | - Tomasz Kazimierz Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, 71-252, Szczecin, Poland.
| |
Collapse
|
46
|
Chiec L, Bruno DS. Immunotherapy for Treatment of Pleural Mesothelioma: Current and Emerging Therapeutic Strategies. Int J Mol Sci 2024; 25:10861. [PMID: 39409190 PMCID: PMC11477297 DOI: 10.3390/ijms251910861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/20/2024] Open
Abstract
Pleural mesothelioma is a rare malignancy associated with asbestos exposure and very poor prognosis, with a 5-year overall survival of 12%. Outcomes may vary according to stage at time of diagnosis and histologic subtype. Most recently, clinical trials utilizing dual checkpoint inhibitor regimens and chemotherapy in combination with immune oncologic agents have demonstrated impactful changes in outcomes. In this article, we review studies that have led to the successful implementation of immunotherapy in clinical practice for the treatment of this disease and highlight ongoing clinical trials exploring the use of different immunotherapy strategies for the treatment of pleural mesothelioma. We also discuss the challenges of immunotherapy-based approaches in the context of mesothelioma and future strategies currently being investigated to overcome them.
Collapse
Affiliation(s)
- Lauren Chiec
- University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Debora S. Bruno
- University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
47
|
Heraghty SD, Rahman SR, Verble KM, Lozier JD. Examining the Effects of Environment, Geography, and Elevation on Patterns of DNA Methylation Across Populations of Two Widespread Bumble Bee Species. Genome Biol Evol 2024; 16:evae207. [PMID: 39327899 PMCID: PMC11474243 DOI: 10.1093/gbe/evae207] [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: 06/07/2024] [Revised: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024] Open
Abstract
Understanding the myriad avenues through which spatial and environmental factors shape evolution is a major focus in biological research. From a molecular perspective, much work has been focused on genomic sequence variation; however, recently there has been increased interest in how epigenetic variation may be shaped by different variables across the landscape. DNA methylation has been of particular interest given that it is dynamic and can alter gene expression, potentially offering a path for a rapid response to environmental change. We utilized whole genome enzymatic methyl sequencing to evaluate the distribution of CpG methylation across the genome and to analyze patterns of spatial and environmental association in the methylomes of two broadly distributed montane bumble bees (Bombus vancouverensis Cresson and Bombus vosnesenskii Radoszkowski) across elevational gradients in the western US. Methylation patterns in both species are similar at the genomic scale with ∼1% of CpGs being methylated and most methylation being found in exons. At the landscape scale, neither species exhibited strong spatial or population structuring in patterns of methylation, although some weak relationships between methylation and distance or environmental variables were detected. Differential methylation analysis suggests a stronger environment association in B. vancouverensis given the larger number of differentially methylated CpG's compared to B. vosnesenskii. We also observed only a handful of genes with both differentially methylated CpGs and previously detected environmentally associated outlier SNPs. Overall results reveal a weak but present pattern in variation in methylation over the landscape in both species.
Collapse
Affiliation(s)
- Sam D Heraghty
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Sarthok Rasique Rahman
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
- Department of Ecology and Evolutionary Biology, 106A Guyot Hall, Princeton University, Princeton, NJ 08544
| | - Kelton M Verble
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Jeffrey D Lozier
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
48
|
Shoer S, Reicher L, Zhao C, Pollard KS, Pilpel Y, Segal E. Pangenomes of human gut microbiota uncover links between genetic diversity and stress response. Cell Host Microbe 2024; 32:1744-1757.e2. [PMID: 39353429 DOI: 10.1016/j.chom.2024.08.017] [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: 04/22/2024] [Revised: 07/11/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024]
Abstract
The genetic diversity of the gut microbiota has a central role in host health. Here, we created pangenomes for 728 human gut prokaryotic species, quadrupling the genes of strain-specific genomes. Each of these species has a core set of a thousand genes, differing even between closely related species, and an accessory set of genes unique to the different strains. Functional analysis shows high strain variability associates with sporulation, whereas low variability is linked with antibiotic resistance. We further map the antibiotic resistome across the human gut population and find 237 cases of extreme resistance even to last-resort antibiotics, with a predominance among Enterobacteriaceae. Lastly, the presence of specific genes in the microbiota relates to host age and sex. Our study underscores the genetic complexity of the human gut microbiota, emphasizing its significant implications for host health. The pangenomes and antibiotic resistance map constitute a valuable resource for further research.
Collapse
Affiliation(s)
- Saar Shoer
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Lee Reicher
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel; Lis Maternity and Women's Hospital, Sourasky Medical Center, Tel Aviv, Israel
| | - Chunyu Zhao
- Gladstone Institute for Data Science and Biotechnology, San Francisco, CA, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Katherine S Pollard
- Gladstone Institute for Data Science and Biotechnology, San Francisco, CA, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Yitzhak Pilpel
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
49
|
Zhang M, Cai R, Liu J, Wang Y, He S, Wang Q, Song X, Wu J, Zhao J. Multi-omics integration analysis reveals the role of N6-methyladenosine in lncRNA translation during glioma stem cell differentiation. Brief Funct Genomics 2024:elae037. [PMID: 39377261 DOI: 10.1093/bfgp/elae037] [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: 05/31/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024] Open
Abstract
Glioblastoma is one of the most lethal brain diseases in humans. Although recent studies have shown reciprocal interactions between N6-methyladenosine (m6A) modifications and long noncoding RNAs (lncRNAs) in gliomagenesis and malignant progression, the mechanism of m6A-mediated lncRNA translational regulation in glioblastoma remains unclear. Herein, we profiled the transcriptomes, translatomes, and epitranscriptomics of glioma stem cells and differentiated glioma cells to investigate the role of m6A in lncRNA translation comprehensively. We found that lncRNAs with numerous m6A peaks exhibit reduced translation efficiency. Transcript-level expression analysis demonstrates an enrichment of m6A around short open reading frames (sORFs) of translatable lncRNA transcripts. Further comparison analysis of m6A modifications in different RNA regions indicates that m6A peaks downstream of sORFs inhibit lncRNA translation more than those upstream. Observations in glioma-associated lncRNAs H19, LINC00467, and GAS5 further confirm the negative effect of m6A methylation on lncRNA translation. Overall, these findings elucidate the dynamic profiles of the m6A methylome and enhance the understanding of the complexity of lncRNA translational regulation.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Runqiu Cai
- Equipment Department, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Qinhuai District, Nanjing 210029, Jiangsu Province, China
| | - Jingjing Liu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Yulan Wang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Shan He
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Quan Wang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Xiaofeng Song
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| | - Jing Wu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, No. 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu Province, China
| | - Jian Zhao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing 211106, Jiangsu Province, China
| |
Collapse
|
50
|
Vieira AA, Almada-Correia I, Inácio J, Costa-Reis P, da Rocha ST. Female-bias in systemic lupus erythematosus: How much is the X chromosome to blame? Biol Sex Differ 2024; 15:76. [PMID: 39375734 PMCID: PMC11460073 DOI: 10.1186/s13293-024-00650-y] [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: 02/13/2024] [Accepted: 08/30/2024] [Indexed: 10/09/2024] Open
Abstract
Systemic lupus erythematosus (SLE or lupus) is an immune-mediated disease associated with substantial medical burden. Notably, lupus exhibits a striking female bias, with women having significantly higher susceptibility compared to men, up to 14-fold higher in some ethnicities. Supernumerary X chromosome syndromes, like Klinefelter (XXY) and Triple X syndrome (XXX), also present higher SLE prevalence, whereas Turner syndrome (XO) displays lower prevalence. Taken together, SLE prevalence in different X chromosome dosage sceneries denotes a relationship between the number of X chromosomes and the risk of developing lupus. The dosage of X-linked genes, many of which play roles in the immune system, is compensated between males and females through the inactivation of one of the two X chromosomes in female cells. X-chromosome inactivation (XCI) initiates early in development with a random selection of which X chromosome to inactivate, a choice that is then epigenetically maintained in the daughter cells. This process is regulated by the X-Inactive-Specific Transcript (XIST), encoding for a long non-coding RNA, exclusively expressed from the inactive X chromosome (Xi). XIST interacts with various RNA binding proteins and chromatin modifiers to form a ribonucleoprotein (RNP) complex responsible for the transcriptional silencing and heterochromatinization of the Xi. This ensures stable silencing of most genes on the X chromosome, with only a few genes able to escape this process. Recent findings suggest that the molecular components involved in XCI, or their dysregulation, contribute to the pathogenesis of lupus. Indeed, nonrandom XCI, elevated gene escape from XCI, and the autoimmune potential of the XIST RNP complex have been suggested to contribute to auto-immune diseases, such as lupus. This review examines these current hypotheses concerning how this dosage compensation mechanism might impact the development of lupus, shedding light on potential mechanisms underlying the pathogenesis of the disease.
Collapse
Affiliation(s)
- Adriana A Vieira
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
| | - Inês Almada-Correia
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Inácio
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
- Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Patrícia Costa-Reis
- Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Pediatric Rheumatology Unit, Pediatrics Department, Hospital de Santa Maria, Lisbon, Portugal
| | - S T da Rocha
- iBB - Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|