1
|
Brar HK, Chen E, Chang F, Lu SA, Longowal DK, Moon KM, Foster LJ, Reiner N, Nandan D. Leishmania regulates host YY1: Comparative proteomic analysis identifies infection modulated YY1 dependent proteins. PLoS One 2025; 20:e0323227. [PMID: 40373059 PMCID: PMC12080872 DOI: 10.1371/journal.pone.0323227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/04/2025] [Indexed: 05/17/2025] Open
Abstract
The protein Yin-Yang 1 (YY1) is a ubiquitous multifunctional transcription factor. Interestingly, there are several cellular functions controlled by YY1 that could play a role in Leishmania pathogenesis. Leishmaniasis is a human disease caused by protozoan parasites of the genus Leishmania. This study examined the potential role of macrophage YY1 in promoting Leishmania intracellular survival. Deliberate knockdown of YY1 resulted in attenuated survival of Leishmania in infected macrophages, suggesting a role of YY1 in Leishmania persistence. Biochemical fractionation studies revealed Leishmania infection caused redistribution of YY1 to the cytoplasm from the nucleus where it is primarily located. Inhibition of nuclear transport by leptomycin B attenuates infection-mediated YY1 redistribution and reduces Leishmania survival. This suggests that Leishmania induces the translocation of YY1 from the nucleus to the cytoplasm of infected cells, where it may regulate host molecules to favour parasite survival. A label-free quantitative whole proteome approach showed that the expression of a large number of macrophage proteins was dependent on the YY1 level. Interestingly, several of these proteins were modulated in Leishmania-infected cells, revealing YY1-dependent host response and suggesting their potential role in Leishmania pathogenesis. Together, this study identifies YY1 as a novel virulence factor that promotes Leishmania survival inside host macrophages.
Collapse
Affiliation(s)
- Harsimran Kaur Brar
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eleanor Chen
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fabian Chang
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shawna Angel Lu
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dilraj Kaur Longowal
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J. Foster
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Neil Reiner
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Devki Nandan
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Zhu Q, Yuan Z, Huo Q, Lu Q, Wu Q, Guo J, Fu W, Lu Y, Zhong L, Shang W, Cui D, Li S, Liu X, Tu K, Huang D, Xu Q, Hu X. YY1 induced USP13 transcriptional activation drives the malignant progression of hepatocellular carcinoma by deubiquitinating WWP1. Cell Mol Biol Lett 2025; 30:56. [PMID: 40319251 PMCID: PMC12049795 DOI: 10.1186/s11658-025-00733-7] [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: 08/21/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the sixth most prevalent cancer globally and the third leading cause of cancer-related mortality. Protein ubiquitination and deubiquitination play vital roles in human cancers. Ubiquitin-specific protease 13 (USP13) is a deubiquitinating enzyme (DUB) that is involved in many cellular processes. However, the mechanism by which USP13 regulates deubiquitination remains largely unknown. METHODS Clinical data were analyzed via online databases. USP13 expression in HCC cell lines and tissues was analyzed via western blotting and immunohistochemistry. A lentivirus was used to established stable USP13-knockdown and USP13-overexpression cells. Cell Counting Kit-8, colony formation, wound healing, Transwell, and sphere formation assays were used to detect the malignant behaviors of HCC cells in vitro. A subcutaneous mouse model was used to investigate the function of USP13 in vivo. Co-immunoprecipitation, chromatin immunoprecipitation and dual-luciferase reporter assays were conducted to explore the molecular regulation. RESULTS USP13 was upregulated in HCC cell lines and tissues, which predicted a poor prognosis in patients with HCC. Functional experiments in which USP13 was overexpressed or depleted revealed the oncogenic role of USP13 in driving HCC progression both in vitro and in vivo. Mechanistically, WW domain-containing ubiquitin E3 ligase 1 (WWP1) was identified as a binding protein of USP13. Furthermore, USP13 can interact with WWP1 and then remove the K29- and K48-linked polyubiquitination chains from WWP1 to stabilize the WWP1 protein via the ubiquitin-proteasome pathway. Moreover, Yin Yang 1 (YY1) was explored as a new transcription factor of USP13, and YY1 could also upregulate WWP1 expression through USP13. Moreover, YY1 and WWP1 were shown to participate in the oncogenic role of USP13. CONCLUSIONS Our findings revealed the functional YY1/USP13/WWP1 signaling axis in HCC, identifying a promising therapeutic target for anti-HCC treatment.
Collapse
Affiliation(s)
- Qingwei Zhu
- The Qingdao Medical College of Qingdao University, Qingdao, 266000, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Zibo Yuan
- The Qingdao Medical College of Qingdao University, Qingdao, 266000, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Qiang Huo
- Department of General Surgery, Zhoushan Dinghai Central Hospital (Dinghai District of Zhejiang Provincial People's Hospital), Zhoushan, 316000, China
| | - Qiliang Lu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- The Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Qingsong Wu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Department of Hepatobiliary, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, China
| | - Junwei Guo
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310053, China
| | - Wen Fu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ying Lu
- Department of Haematology, Affiliated People's Hospital of Ningbo University, Ningbo, 315000, China
| | - Lei Zhong
- Department of Laboratory Medicine, Tongxiang Traditional Chinese Medicine Hospital, Tongxiang, 314500, China
| | - Wenzhong Shang
- Department of Hematology, The first People's Hospital of Fuyang Hangzhou, Hangzhou, 311400, China
| | - Di Cui
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Shuangshuang Li
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Xin Liu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Xiaoge Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
3
|
Xia K, Liu S, Wu Z, Jiang JH. Research Status and Applications of Adeno-Associated Virus. Chembiochem 2025; 26:e202400856. [PMID: 39724465 DOI: 10.1002/cbic.202400856] [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: 10/15/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Adeno-associated virus (AAV) has emerged as a powerful and effective tool for the delivery of exogenous genes into various cells or tissues. To improve the gene delivery efficiency, as well as the safety and specificity of AAV's cell-targeting capabilities, extensive investigations have been conducted into its molecular biological characteristics, including capsid structure, cellular tropism, and the mechanisms underlying its entry, replication, DNA packaging, and capsid assembly. Significant differences exist between human and non-human primate AAVs regarding tissue targeting and transduction efficiency. These differences are primarily attributed to the amino acid sequences of AAV capsid proteins, the structural characteristics of these proteins, and the interactions of AAV with surface factors on host cells, such as cell surface receptors, signaling molecules, and associated proteins. This review primarily focuses on several key aspects of AAV, including its genome, coat proteins and their structures, genome replication, virus assembly, and the role of helper viruses. Additionally, it examines the utilization of recombinant adeno-associated viruses (rAAV), detailing their production methods, mechanisms of cell entry and trafficking, and various serotypes. The review further interprets the role of rAAV by analyzing its current applications in research and therapy.
Collapse
Affiliation(s)
- Ke Xia
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Shuangling Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
4
|
Qiu C, Lu Y, Wu S, Guo W, Ni J, Song J, Liu Z, Chang X, Wang K, Sun P, Zhang Q, Yang S, Li K. Blocking Adipocyte YY1 Decouples Thermogenesis From Beneficial Metabolism by Promoting Spermidine Production. Diabetes 2025; 74:295-307. [PMID: 39621859 DOI: 10.2337/db24-0501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
The accumulation of mitochondria in thermogenic adipose tissue (i.e., brown and beige fat) increases energy expenditure, which can aid in alleviating obesity and metabolic disorders. However, recent studies have shown that knocking out key proteins required to maintain mitochondrial function inhibits the energy expenditure in thermogenic fat, and yet the knockout (KO) mice are unexpectedly protected from developing obesity or metabolic disorders when fed a high-fat diet (HFD). In the current study, nonbiased sequencing-based screening revealed the importance of Yin Yang 1 (YY1) in the transcription of electron transport chain genes and the enhancement of mitochondrial function in thermogenic adipose tissue. Specifically, YY1 adipocyte-null (YAKO) mice showed lower energy expenditure and were intolerant to cold stress. Interestingly, YAKO mice showed alleviation of HFD-induced metabolic disorders, which can be attributed to a suppression of adipose tissue inflammation. Metabolomic analysis revealed that blocking YY1 directed glucose metabolism toward lactate, enhanced the uptake of glutamine, and promoted the production of anti-inflammatory spermidine. Conversely, blocking spermidine production in YAKO mice reversed their resistance to HFD-induced disorders. Thus, although blocking adipocyte YY1 impairs the thermogenesis, it promotes spermidine production, alleviates adipose tissue inflammation, and therefore leads to an uncoupling of adipose tissue energy expenditure from HFD-induced metabolic disorders. ARTICLE HIGHLIGHTS Chromatin open atlas profiling in white, beige, and brown adipocytes identified Yin Yang 1 (YY1) as a key transcription factor governing electron transport chain gene expression and mitochondrial function in thermogenic adipocytes. Knocking out adipocyte YY1 leads to impaired thermogenesis under cold stress while protecting the mice from diet-induced obesity and metabolic disorders. YY1-null adipocytes undergo metabolic reprogramming, with increased glutamine use and spermidine generation that combat adipose tissue inflammation and insulin resistance, resulting in an uncoupling of thermogenic capacity and metabolic benefits.
Collapse
Affiliation(s)
- Chen Qiu
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
- Key Laboratory of the Model Animal Research, Animal Core Facility of Nanjing Medical University, Nanjing, China
| | - Yu Lu
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Suyang Wu
- Department of Science and Technology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Wenli Guo
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Jiahao Ni
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Jiyuan Song
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Zichao Liu
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Kai Wang
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital affiliated to Nanjing University Medical School, Nanjing, China
- Department of Medical Technology, Anhui Medical College, Hefei, Anhui, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Qian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Shufang Yang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- School of Medicine, Southeast University, Nanjing, China
| | - Kai Li
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Palo A, Patel SA, Shubhanjali S, Dixit M. Dynamic interplay of Sp1, YY1, and DUX4 in regulating FRG1 transcription with intricate balance. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167636. [PMID: 39708975 DOI: 10.1016/j.bbadis.2024.167636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/16/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Maintaining precise levels of FRG1 is vital. It's over-expression is tied to muscular dystrophy, while reduced levels are linked to tumorigenesis. Despite extensive efforts to characterize FRG1 expression and downstream molecular signaling, a comprehensive understanding of its regulation has remained elusive. This study focused on unravelling the cis -regulatory elements within the FRG1 gene and their interplay. Employing a dual luciferase reporter assay on fragments of the FRG1 promoter upstream of the transcription start site, we observed variations in FRG1 transcription induction. Our in-silico analysis unveiled binding sequences for Sp1 and DUX4 within FRG1 promoter region showing an enhanced luciferase signal. Conversely, we identified a YY1 binding sequence in the FRG1 promoter fragment showing decreased luciferase signal. Confirming these binding sites through site-directed mutagenesis, chromatin immunoprecipitation, and EMSA provided concrete evidence of Sp1, YY1, and DUX4's interaction within the FRG1 promoter. Additionally, interaction between Sp1, YY1, and DUX4 was elucidated using sequential chromatin immunoprecipitation (ChIP re-ChIP) and co-immunoprecipitation assays. Furthermore, alterations in the expression levels of Sp1, YY1, and DUX4 resulted in parallel changes in FRG1 gene expression. Notably, YY1 exhibited the ability to suppress SP1 or DUX4-mediated FRG1 transcription activation, while Sp1 and DUX4 together could counteract YY1-mediated transcription suppression. Our cell proliferation and colony formation assay underscored the tumorigenic properties of these three transcription factors through the modulation of FRG1 expression levels. The in vitro results were verified in vivo using mouse xenograft model. Leveraging RNA sequencing data from various tissues in the GTEx portal, we established a correlation between FRG1, Sp1, and YY1. In essence, this study revealed the vital cis-regulatory components residing in the FRG1 promoter. The combined influence of Sp1, YY1, and DUX4 plays a central role in controlling FRG1 expression.
Collapse
Affiliation(s)
- Ananya Palo
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Saket A Patel
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - S Shubhanjali
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Manjusha Dixit
- National Institute of Science Education and Research, School of Biological Sciences, Bhubaneswar, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
6
|
Rodríguez-Campuzano AG, Castelán F, Hernández-Kelly LC, Felder-Schmittbuhl MP, Ortega A. Yin Yang 1: Function, Mechanisms, and Glia. Neurochem Res 2025; 50:96. [PMID: 39904836 PMCID: PMC11794380 DOI: 10.1007/s11064-025-04345-7] [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: 10/31/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Yin Yang 1 is a ubiquitously expressed transcription factor that has been extensively studied given its particular dual transcriptional regulation. Yin Yang 1 is involved in various cellular processes like cell cycle progression, cell differentiation, DNA repair, cell survival and apoptosis among others. Its malfunction or alteration leads to disease and even to malignant transformation. This transcription factor is essential for the proper central nervous system development and function. The activity of Yin Yang 1 depends on its interacting partners, promoter environment and chromatin structure, however, its mechanistic activity is not completely understood. In this review, we briefly discuss the Yin Yang 1 structure, post-translational modifications, interactions, mechanistic functions and its participation in neurodevelopment. We also discuss its expression and critical involvement in the physiology and physiopathology of glial cells, summarizing the contribution of Yin Yang 1 on different aspects of cellular function.
Collapse
Affiliation(s)
- Ada G Rodríguez-Campuzano
- Departamento de Biología Celular y Fisiología, Unidad Foránea Tlaxcala, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, Tlaxcala, Mexico
| | - Francisco Castelán
- Departamento de Biología Celular y Fisiología, Unidad Foránea Tlaxcala, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, Tlaxcala, Mexico
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacantenco, G.A. Madero, 07360, Ciudad de Mexico, Mexico
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Université de Strasbourg, Strasbourg, France
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacantenco, G.A. Madero, 07360, Ciudad de Mexico, Mexico.
| |
Collapse
|
7
|
Sirisena DMKP, Kim G, Warnakula WADLR, Jayamali BPMV, Tharanga EMT, Jayasinghe JDHE, Sandeepani RI, Wan Q, Sohn H, Lee J. Interferon regulatory factor 2 of red-spotted grouper (Epinephelus akaara): Insights into its transcriptional profiling, antiviral potential, and function in macrophage polarization. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 163:105323. [PMID: 39848353 DOI: 10.1016/j.dci.2025.105323] [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: 08/15/2024] [Revised: 01/19/2025] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Interferon regulatory factor 2 (IRF2) is a member of the IRF family that is specifically involved in diverse immune responses via interferon (IFN)/IRF-dependent signaling pathways. In this study, IRF2 of Epinephelus akaara (EAIRF2) was identified and characterized by evaluating its structural and functional properties. EAIRF2 showed the highest homology with IRF2 of Epinephelus coioides and clustered with teleosts in the phylogenetic tree. The highest level of EAIRF2 mRNA was found in the blood under normal physiological conditions. In the immune challenge experiment, significant transcriptional modulation of EAIRF2 upon lipopolysaccharide (LPS), polyinosinic: polycytidylic acid (poly I:C), and nervous necrosis virus (NNV) challenge were observed. The subcellular localization assay confirmed the role of EAIRF2 as a transcription factor by revealing its specific nuclear localization. To elucidate its functional implications in antiviral defense, EAIRF2 was overexpressed in fathead minnow cells, which were subsequently infected with viral hemorrhagic septicemia virus (VHSV). Notably, cells overexpressing EAIRF2 exhibited a significant reduction in the transcription of VHSV genes. Concurrently, the genes associated with the IFN/IRF signaling pathway were upregulated. Furthermore, the Hoechst and propidium iodide dual staining assay, water-soluble tetrazolium-1 (WST-1) assay, and transcriptional analysis of B-cell lymphoma 2-associated X protein (Bax)/B-cell lymphoma 2 (Bcl-2) indicated that EAIRF2 possesses anti-apoptotic properties during viral infection and poly I:C treatment. Additionally, EAIRF2 overexpression in murine macrophages induced M1 polarization and augmented relative marker gene expression. Collectively, these findings suggest that EAIRF2 is a pivotal immune-related gene, specifically implicated in the IFN/IRF-mediated antiviral defense mechanism, apoptotic signaling pathway, and activation of macrophage-mediated immune responses in Epinephelus akaara. The finding of this study enhances our understanding of IRF2's function in teleost immunity and presents potential avenues for developing therapeutic strategies against viral infections and other immune-related conditions in aquaculture species.
Collapse
Affiliation(s)
- D M K P Sirisena
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - W A D L R Warnakula
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - B P M Vileka Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - E M T Tharanga
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - J D H E Jayasinghe
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - R I Sandeepani
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Hanchang Sohn
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
8
|
Wang C, Leong MM, Ding W, Narita Y, Liu X, Wang H, Yiu SPT, Lee J, Zhao KRS, Cui A, Gewurz B, Hammerschmidt W, Teng M, Zhao B. Viral oncogene EBNALP regulates YY1 DNA binding and alters host 3D genome organization. EMBO Rep 2025; 26:810-835. [PMID: 39747661 PMCID: PMC11811279 DOI: 10.1038/s44319-024-00357-6] [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: 06/12/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
The Epstein-Barr virus (EBV) nuclear antigen leader protein (EBNALP) is essential for the immortalization of naive B lymphocytes (NBLs). However, the mechanisms remain elusive. To understand EBNALP's role in B-cell transformation, we compare NBLs infected with wild-type EBV and an EBNALP-null mutant EBV using multi-omics techniques. EBNALP inactivation alters enhancer-promoter interactions, resulting in decreased CCND2 and increased CASP1 and BCL2L11 expression. Mechanistically, EBNALP interacts with and colocalizes with the looping factor YY1. Depletion of EBNALP reduces YY1 DNA-binding and enhancer-promoter interactions, similar to effects observed with YY1 depletion. Furthermore, EBNALP colocalizes with DPF2, a protein that binds to H3K14ac and H4K16ac. CRISPR depletion of DPF2 reduces both EBNALP and YY1 DNA binding, suggesting that the DPF2/EBNALP complex may tether YY1 to DNA to increase enhancer-promoter interactions. EBNALP inactivation also increases enhancer-promoter interactions at the CASP1 and BCL2L11 loci, along with elevated DPF2 and YY1 binding and DNA accessibility. Our data suggest that EBNALP regulates YY1 to rewire the host genome, which might facilitate naive B-cell transformation.
Collapse
Affiliation(s)
- Chong Wang
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Merrin Manlong Leong
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Weiyue Ding
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yohei Narita
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiang Liu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Hongbo Wang
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefanie P T Yiu
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica Lee
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katelyn R S Zhao
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amy Cui
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin Gewurz
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Center for Infection Research, Munich, Germany
| | - Mingxiang Teng
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| | - Bo Zhao
- Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Merenstein A, Obeidat L, Zaravinos A, Bonavida B. The Role of YY1 in the Regulation of LAG-3 Expression in CD8 T Cells and Immune Evasion in Cancer: Therapeutic Implications. Cancers (Basel) 2024; 17:19. [PMID: 39796650 PMCID: PMC11718991 DOI: 10.3390/cancers17010019] [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: 11/06/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
The treatment of cancers with immunotherapies has yielded significant milestones in recent years. Amongst these immunotherapeutic strategies, the FDA has approved several checkpoint inhibitors (CPIs), primarily Anti-Programmed Death-1 (PD-1) and Programmed Death Ligand-1/2 (PDL-1/2) monoclonal antibodies, in the treatment of various cancers unresponsive to immune therapeutics. Such treatments resulted in significant clinical responses and the prolongation of survival in a subset of patients. However, not all patients responded to CPIs, due to various mechanisms of immune resistance. One such mechanism is that, in addition to PD-1 expression on CD8 T cells, other inhibitory receptors exist, such as Lymphocyte Activation Gene 3 (LAG-3), T cell Immunoglobulin Mucin 3 (TIM3), and T cell immunoreceptor with Ig and ITIM domains (TIGIT). These inhibitory receptors might be active in the presence of the above approved CPIs. Clearly, it is clinically challenging to block all such inhibitory receptors simultaneously using conventional antibodies. To circumvent this difficulty, we sought to target a potential transcription factor that may be involved in the molecular regulation of more than one inhibitory receptor. The transcription factor Yin Yang1 (YY1) was found to regulate the expression of PD-1, LAG-3, and TIM3. Therefore, we hypothesized that targeting YY1 in CD8 T cells should inhibit the expression of these receptors and, thus, prevent the inactivation of the anti-tumor CD8 T cells by these receptors, by corresponding ligands to tumor cells. This strategy should result in the prevention of immune evasion, leading to the inhibition of tumor growth. In addition, this strategy will be particularly effective in a subset of cancer patients who were unresponsive to approved CPIs. In this review, we discuss the regulation of LAG-3 by YY1 as proof of principle for the potential use of targeting YY1 as an alternative therapeutic approach to preventing the immune evasion of cancer. We present findings on the molecular regulations of both YY1 and LAG-3 expressions, the direct regulation of LAG-3 by YY1, the various approaches to targeting YY1 to evade immune evasion, and their clinical challenges. We also present bioinformatic analyses demonstrating the overexpression of LAG-3, YY1, and PD-L1 in various cancers, their associations with immune infiltrates, and the fact that when LAG-3 is hypermethylated in its promoter region it correlates with a better overall survival. Hence, targeting YY1 in CD8 T cells will result in restoring the anti-tumor immune response and tumor regression. Notably, in addition to the beneficial effects of targeting YY1 in CD8 T cells to inhibit the expression of inhibitory receptors, we also suggest targeting YY1 overexpressed in the tumor cells, which will also inhibit PD-L1 expression and other YY1-associated pro-tumorigenic activities.
Collapse
Affiliation(s)
- Adam Merenstein
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA;
| | - Loiy Obeidat
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus; (L.O.); (A.Z.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 1516 Nicosia, Cyprus
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus; (L.O.); (A.Z.)
- Department of Life Sciences, School of Sciences, European University Cyprus, 1516 Nicosia, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
10
|
Zuurbier KR, Fonseca RS, Arneaud SLB, Wall JM, Kim J, Tatge L, Otuzoglu G, Bali S, Metang P, Douglas PM. Yin Yang 1 and guanine quadruplexes protect dopaminergic neurons from cellular stress via transmissive dormancy. Nat Commun 2024; 15:10592. [PMID: 39632864 PMCID: PMC11618784 DOI: 10.1038/s41467-024-54958-9] [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: 01/17/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Neurons deploy diverse adaptive strategies to ensure survival and neurotransmission amid cellular stress. When these adaptive pathways are overwhelmed, functional impairment or neurodegeneration follows. Here we show that stressed neurons actively induce a state of transmissive dormancy as a protective measure. Extending observations of neurotrauma in C. elegans and mice, human dopaminergic neurons capable of surviving severe cellular challenges both decrease spontaneous activity and modulate dopamine homeostasis through the transcriptional regulator Yin Yang 1 (YY1). To bolster stress resilience and mitigate dopamine toxicity, YY1 increases expression of the vesicular monoamine transporter 2, vMAT2, while coordinately inhibiting dopamine synthesis through stabilization of a guanine quadruplex in intron 10 of tyrosine hydroxylase, TH. This dopaminergic stress response has the potential to cause circuit inactivation, yet safeguards neurons by minimizing the toxic accumulation of cytosolic dopamine and inducing a state of neuronal dormancy. In essence, neurons appear to actively prioritize viability over functionality.
Collapse
Affiliation(s)
- Kielen R Zuurbier
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rene Solano Fonseca
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sonja L B Arneaud
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jordan M Wall
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Juhee Kim
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lexus Tatge
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gupse Otuzoglu
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sofia Bali
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Patrick Metang
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Peter M Douglas
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine; UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
11
|
Zhou S, Zang J, Cai MC, Ye K, Liu J, Ma P, Wu J, Dai C, Lu H, Zhang Q, Jiang J, Chu T, Shen Y, Tan L, Zhuang G, Zhao X, Wang L, Zhuang Y, Fu Y. YY1 downregulation underlies therapeutic response to molecular targeted agents. Cell Death Dis 2024; 15:862. [PMID: 39604408 PMCID: PMC11603335 DOI: 10.1038/s41419-024-07239-8] [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: 06/17/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
During targeted treatment, oncogene-addicted tumor cells often evolve from an initial drug-sensitive state through a drug-tolerant persister bottleneck toward the ultimate emergence of drug-resistant clones. The molecular basis underlying this therapy-induced evolutionary trajectory has not yet been completely elucidated. Here, we employed a multifaceted approach and implicated the convergent role of transcription factor Yin Yang 1 (YY1) in the course of diverse targeted kinase inhibitors. Specifically, pharmacological perturbation of the receptor tyrosine kinase (RTK)/mitogen-activated protein kinase (MAPK) pathway resulted in the downregulation of YY1 transcription, which subsequently resumed upon therapeutic escape. Failure to decrease YY1 subverted cytotoxic effects, whereas elimination of residual YY1 maximized anticancer efficacy and forestalled the emergence of drug resistance. Mechanistically, YY1 was uncovered to dictate cell cycle and autophagic programs. Immunohistochemical analysis on a wide spectrum of clinical specimens revealed that YY1 was ubiquitously expressed across lung adenocarcinomas and exhibited anticipated fluctuation in response to corresponding RTK/MAPK inhibition. These findings advance our understanding of targeted cancer management by highlighting YY1 as a determinant node in the context of genotype-directed agents.
Collapse
Affiliation(s)
- Shichao Zhou
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jingyu Zang
- Department of Radiation Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei-Chun Cai
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaiyan Ye
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Liu
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengfei Ma
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Wu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haijiao Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Zhang
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhong Jiang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianqing Chu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Guanglei Zhuang
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaojing Zhao
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lan Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China.
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China.
| | - Yu Zhuang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yujie Fu
- State Key Laboratory of Systems Medicine for Cancer, Department of Thoracic Surgery, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Saha K, Nielsen G, Nandani R, Zhang Y, Kong L, Ye P, An W. YY1 is a transcriptional activator of the mouse LINE-1 Tf subfamily. Nucleic Acids Res 2024; 52:12878-12894. [PMID: 39460630 PMCID: PMC11602158 DOI: 10.1093/nar/gkae949] [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: 12/31/2023] [Revised: 09/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Long interspersed element type 1 (LINE-1, L1) is an active autonomous transposable element in human and mouse genomes. L1 transcription is controlled by an internal RNA polymerase II promoter in the 5' untranslated region (5'UTR) of a full-length L1. It has been shown that transcription factor YY1 binds to a conserved sequence at the 5' end of the human L1 5'UTR and primarily dictates where transcription initiates. Putative YY1-binding motifs have been predicted in the 5'UTRs of two distinct mouse L1 subfamilies, Tf and Gf. Using site-directed mutagenesis, in vitro binding and gene knockdown assays, we experimentally tested the role of YY1 in mouse L1 transcription. Our results indicate that Tf, but not Gf subfamily, harbors functional YY1-binding sites in 5'UTR monomers and YY1 functions as a transcriptional activator for the mouse Tf subfamily. Activation of Tf transcription by YY1 during early embryogenesis is also supported by a reanalysis of published zygotic knockdown data. Furthermore, YY1-binding motifs are solely responsible for the synergistic interaction between Tf monomers, consistent with a model wherein distant monomers act as enhancers for mouse L1 transcription. The abundance of YY1-binding sites in Tf elements also raise important implications for gene regulation across the genome.
Collapse
Affiliation(s)
- Karabi Saha
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Grace I Nielsen
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Raj Nandani
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Yizi Zhang
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Lingqi Kong
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Ping Ye
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| | - Wenfeng An
- Department of Pharmaceutical Sciences, South Dakota State University, 1055 Campanile Ave, Brookings, SD 57007, USA
| |
Collapse
|
13
|
Rezaei S, Timani KA, Liu Y, He JJ. Ectopic USP15 expression inhibits HIV-1 transcription involving changes in YY1 deubiquitination and stability. Front Cell Infect Microbiol 2024; 14:1371655. [PMID: 39624264 PMCID: PMC11609158 DOI: 10.3389/fcimb.2024.1371655] [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: 01/16/2024] [Accepted: 10/21/2024] [Indexed: 01/13/2025] Open
Abstract
Introduction Protein homeostasis is maintained by the opposing action of ubiquitin ligase and deubiquitinase, two important components of the ubiquitin-proteasome pathway, and contributes to both normal physiological and pathophysiological processes. The current study aims to delineate the roles of ubiquitin-specific protease 15 (USP15), a member of the largest deubiquitinase family, in HIV-1 gene expression and replication. Methods We took advantage of highly selective and specific ubiquitin variants (UbV), which were recently designed and developed for USP15, and ascertained the inhibitory effects of USP15 on HIV-1 gene expression and production by transfection and Western blotting. We also used real-time RT-PCR, transcription factor profiling, subcellular fractionation, immunoprecipitation followed by Western blotting to determine the transcription factors involved and the underlying molecular mechanisms. Results We first confirmed the specificity of USP15-mediated HIV-1 gene expression and virus production. We then showed that the inhibition of HIV-1 production by USP15 occurred at the transcription level, associated with an increased protein level of YY1, a known HIV-1 transcription repressor. Moreover, we demonstrated that USP15 regulated YY1 deubiquitination and stability. Lastly, we demonstrated that YY1 siRNA knockdown significantly diminished the inhibition of USP15 on HIV-1 gene expression and virus production. Conclusion These findings together demonstrate that stabilization of YY1 protein by USP15 deubiquitinating activity contributes to USP15-mediated inhibition of HIV-1 transcription and may help the development of USP15-specific UbV inhibitors as an anti-HIV strategy.
Collapse
Affiliation(s)
- Sahar Rezaei
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL, United States
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL, United States
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, United States
| | - Khalid A. Timani
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL, United States
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL, United States
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, United States
| | - Ying Liu
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL, United States
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL, United States
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, United States
| | - Johnny J. He
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL, United States
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL, United States
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, United States
| |
Collapse
|
14
|
Banerjee S, Sanyal S, Hodawadekar S, Naiyer S, Bano N, Banerjee A, Rhoades J, Dong D, Allman D, Atchison ML. YY1 knockout in pro-B cells impairs lineage commitment, enabling unusual hematopoietic lineage plasticity. Genes Dev 2024; 38:887-914. [PMID: 39362773 PMCID: PMC11535188 DOI: 10.1101/gad.351734.124] [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: 03/22/2024] [Accepted: 08/29/2024] [Indexed: 10/05/2024]
Abstract
During B-cell development, cells progress through multiple developmental stages, with the pro-B-cell stage defining commitment to the B-cell lineage. YY1 is a ubiquitous transcription factor that is capable of both activation and repression functions. We found here that knockout of YY1 at the pro-B-cell stage eliminates B lineage commitment. YY1 knockout pro-B cells can generate T lineage cells in vitro using the OP9-DL4 feeder system and in vivo after injection into sublethally irradiated Rag1-/- mice. These T lineage-like cells lose their B lineage transcript profile and gain a T-cell lineage profile. Single-cell RNA-seq experiments showed that as YY1 knockout pro-B cells transition into T lineage cells in vitro, various cell clusters adopt transcript profiles representing a multiplicity of hematopoietic lineages, indicating unusual lineage plasticity. In addition, YY1 KO pro-B cells in vivo can give rise to other hematopoietic lineages in vivo. Evaluation of RNA-seq, scRNA-seq, ChIP-seq, and scATAC-seq data indicates that YY1 controls numerous chromatin-modifying proteins leading to increased accessibility of alternative lineage genes in YY1 knockout pro-B cells. Given the ubiquitous nature of YY1 and its dual activation and repression functions, YY1 may regulate commitment in multiple cell lineages.
Collapse
Affiliation(s)
- Sarmistha Banerjee
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Sulagna Sanyal
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Suchita Hodawadekar
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Sarah Naiyer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Nasreen Bano
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Anupam Banerjee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Joshua Rhoades
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Dawei Dong
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Michael L Atchison
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
15
|
Du S, Chen X, Han X, Wang Y, Yu D, Li Y, Zhu C, Tong Y, Gao S, Wang J, Wei F, Cai Q. Lactate Induces Tumor Progression via LAR Motif-Dependent Yin-Yang 1 Degradation. Mol Cancer Res 2024; 22:957-972. [PMID: 38888574 DOI: 10.1158/1541-7786.mcr-23-0583] [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: 07/21/2023] [Revised: 01/22/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
The metabolic reprogramming of aerobic glycolysis contributes to tumorigenesis. High plasma lactate is a critical regulator in the development of many human malignancies; however, the underlying molecular mechanisms of cancer progression in response to lactate (LA) remain elusive. Here, we show that the reduction of Yin-Yang 1 (YY1) expression correlated with high LA commonly occurs in various cancer cell types, including B-lymphoma and cervical cancer. Mechanistically, LA induces YY1 nuclear export and degradation via HSP70-mediated autophagy adjacent to mitochondria in a histidine (His)-rich LA-responsive (LAR) motif-dependent manner. The mutation of the LAR motif blocks LA-mediated YY1 cytoplasmic accumulation and in turn enhances cell apoptosis. Furthermore, low expression of YY1 promotes colony formation, invasion, angiogenesis, and growth of cancer cells in response to LA in vitro and in vivo using a murine xenograft model. Taken together, our findings reveal a key LAR element and may serve as therapeutic target for intervening cancer progression. Implications: We have shown that lactate can induce YY1 degradation via its His-rich LAR motif and low expression of YY1 promotes cancer cell progression in response to lactate, leading to better prediction of YY1 targeting therapy.
Collapse
Affiliation(s)
- Shujuan Du
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganism and Infection, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Xiaoting Chen
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganism and Infection, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Xiao Han
- Center of Diagnosis and Treatment for Cervical and Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Shanghai, China
| | - Yuyan Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganism and Infection, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Dan Yu
- Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ying Li
- Division of Hematology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Caixia Zhu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganism and Infection, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yin Tong
- Division of Hematology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Shujun Gao
- Center of Diagnosis and Treatment for Cervical and Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Shanghai, China
| | - Junwen Wang
- Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Qiliang Cai
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganism and Infection, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| |
Collapse
|
16
|
Crocini C, Woulfe KC, Ozeroff CD, Perni S, Cardiello J, Walker CJ, Wilson CE, Anseth K, Allen MA, Leinwand LA. Postprandial cardiac hypertrophy is sustained by mechanics, epigenetic, and metabolic reprogramming in pythons. Proc Natl Acad Sci U S A 2024; 121:e2322726121. [PMID: 39159386 PMCID: PMC11388396 DOI: 10.1073/pnas.2322726121] [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: 01/17/2024] [Accepted: 06/18/2024] [Indexed: 08/21/2024] Open
Abstract
Constricting pythons, known for their ability to consume infrequent, massive meals, exhibit rapid and reversible cardiac hypertrophy following feeding. Our primary goal was to investigate how python hearts achieve this adaptive response after feeding. Isolated myofibrils increased force after feeding without changes in sarcomere ultrastructure and without increasing energy cost. Ca2+ transients were prolonged after feeding with no changes in myofibril Ca2+ sensitivity. Feeding reduced titin-based tension, resulting in decreased cardiac tissue stiffness. Feeding also reduced the activity of sirtuins, a metabolically linked class of histone deacetylases, and increased chromatin accessibility. Transcription factor enrichment analysis on transposase-accessible chromatin with sequencing revealed the prominent role of transcription factors Yin Yang1 and NRF1 in postfeeding cardiac adaptation. Gene expression also changed with the enrichment of translation and metabolism. Finally, metabolomics analysis and adenosine triphosphate production demonstrated that cardiac adaptation after feeding not only increased energy demand but also energy production. These findings have broad implications for our understanding of cardiac adaptation across species and hold promise for the development of innovative approaches to address cardiovascular diseases.
Collapse
Affiliation(s)
- Claudia Crocini
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité University Medicine Berlin, Berlin 10115, Germany
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303
| | - Kathleen C Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Christopher D Ozeroff
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303
| | - Stefano Perni
- Department of Physiology and Biophysics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045
| | - Joseph Cardiello
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Cierra J Walker
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Cortney E Wilson
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristi Anseth
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Mary Ann Allen
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303
| |
Collapse
|
17
|
Lam JC, Aboreden NG, Midla SC, Wang S, Huang A, Keller CA, Giardine B, Henderson KA, Hardison RC, Zhang H, Blobel GA. YY1-controlled regulatory connectivity and transcription are influenced by the cell cycle. Nat Genet 2024; 56:1938-1952. [PMID: 39210046 PMCID: PMC11687402 DOI: 10.1038/s41588-024-01871-y] [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: 11/10/2023] [Accepted: 07/16/2024] [Indexed: 09/04/2024]
Abstract
Few transcription factors have been examined for their direct roles in physically connecting enhancers and promoters. Here acute degradation of Yin Yang 1 (YY1) in erythroid cells revealed its requirement for the maintenance of numerous enhancer-promoter loops, but not compartments or domains. Despite its reported ability to interact with cohesin, the formation of YY1-dependent enhancer-promoter loops does not involve stalling of cohesin-mediated loop extrusion. Integrating mitosis-to-G1-phase dynamics, we observed partial retention of YY1 on mitotic chromatin, predominantly at gene promoters, followed by rapid rebinding during mitotic exit, coinciding with enhancer-promoter loop establishment. YY1 degradation during the mitosis-to-G1-phase interval revealed a set of enhancer-promoter loops that require YY1 for establishment during G1-phase entry but not for maintenance in interphase, suggesting that cell cycle stage influences YY1's architectural function. Thus, as revealed here for YY1, chromatin architectural functions of transcription factors can vary in their interplay with CTCF and cohesin as well as by cell cycle stage.
Collapse
Affiliation(s)
- Jessica C Lam
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas G Aboreden
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susannah C Midla
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Siqing Wang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anran Huang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cheryl A Keller
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
- Genomics Research Incubator, Pennsylvania State University, University Park, PA, USA
| | - Belinda Giardine
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Kate A Henderson
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ross C Hardison
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Haoyue Zhang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Catalán-Tatjer D, Tzimou K, Nielsen LK, Lavado-García J. Unravelling the essential elements for recombinant adeno-associated virus (rAAV) production in animal cell-based platforms. Biotechnol Adv 2024; 73:108370. [PMID: 38692443 DOI: 10.1016/j.biotechadv.2024.108370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/05/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
Recombinant adeno-associated viruses (rAAVs) stand at the forefront of gene therapy applications, holding immense significance for their safe and efficient gene delivery capabilities. The constantly increasing and unmet demand for rAAVs underscores the need for a more comprehensive understanding of AAV biology and its impact on rAAV production. In this literature review, we delved into AAV biology and rAAV manufacturing bioprocesses, unravelling the functions and essentiality of proteins involved in rAAV production. We discuss the interconnections between these proteins and how they affect the choice of rAAV production platform. By addressing existing inconsistencies, literature gaps and limitations, this review aims to define a minimal set of genes that are essential for rAAV production, providing the potential to advance rAAV biomanufacturing, with a focus on minimizing the genetic load within rAAV-producing cells.
Collapse
Affiliation(s)
- David Catalán-Tatjer
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Konstantina Tzimou
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Lars K Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark; Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia
| | - Jesús Lavado-García
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark.
| |
Collapse
|
19
|
Lu Z, Wang Y, Assumpção ALFV, Liu P, Kopp A, Saka S, Mcilwain SJ, Viny AD, Brand M, Pan X. Yin Yang 1 regulates cohesin complex protein SMC3 in mouse hematopoietic stem cells. Blood Adv 2024; 8:3076-3091. [PMID: 38531064 PMCID: PMC11222949 DOI: 10.1182/bloodadvances.2023011411] [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: 08/09/2023] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
ABSTRACT Yin Yang 1 (YY1) and structural maintenance of chromosomes 3 (SMC3) are 2 critical chromatin structural factors that mediate long-distance enhancer-promoter interactions and promote developmentally regulated changes in chromatin architecture in hematopoietic stem/progenitor cells (HSPCs). Although YY1 has critical functions in promoting hematopoietic stem cell (HSC) self-renewal and maintaining HSC quiescence, SMC3 is required for proper myeloid lineage differentiation. However, many questions remain unanswered regarding how YY1 and SMC3 interact with each other and affect hematopoiesis. We found that YY1 physically interacts with SMC3 and cooccupies with SMC3 at a large cohort of promoters genome wide, and YY1 deficiency deregulates the genetic network governing cell metabolism. YY1 occupies the Smc3 promoter and represses SMC3 expression in HSPCs. Although deletion of 1 Smc3 allele partially restores HSC numbers and quiescence in YY1 knockout mice, Yy1-/-Smc3+/- HSCs fail to reconstitute blood after bone marrow transplant. YY1 regulates HSC metabolic pathways and maintains proper intracellular reactive oxygen species levels in HSCs, and this regulation is independent of the YY1-SMC3 axis. Our results establish a distinct YY1-SMC3 axis and its impact on HSC quiescence and metabolism.
Collapse
Affiliation(s)
- Zhanping Lu
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
| | - Yinghua Wang
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
| | - Anna L. F. V. Assumpção
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
| | - Peng Liu
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Audrey Kopp
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sahitya Saka
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
| | - Sean J. Mcilwain
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Aaron D. Viny
- Division of Hematology & Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY
| | - Marjorie Brand
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Xuan Pan
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI
- Carbone Cancer Center, University of Wisconsin, Madison, WI
- Wisconsin Blood Cancer Research Institute, University of Wisconsin, Madison, WI
| |
Collapse
|
20
|
Shao ZY, Yang WD, Qiu H, He ZH, Lu MR, Shen Q, Ding J, Zheng JN, Bai J. The role of USP7-YY1 interaction in promoting colorectal cancer growth and metastasis. Cell Death Dis 2024; 15:347. [PMID: 38769122 PMCID: PMC11106261 DOI: 10.1038/s41419-024-06740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Colorectal cancer (CRC) remains a significant global health issue with high incidence and mortality. Yin Yang 1 (YY1) is a powerful transcription factor that acts dual roles in gene activation and repression. High expression level of YY1 has been reported in CRC, indicating the existence of stable factors of YY1 in CRC cells. We aimed to identify the key molecules and underlying mechanisms responsible for stabilizing YY1 expression in CRC. Mass spectrometry analysis was utilized to identify USP7 as a potential molecule that interacted with YY1. Mechanically, USP7 stabilizes YY1 expression at the protein level by interfering its K63 linkage ubiquitination. YY1 exerts its oncogenic function through transcriptionally activating TRIAP1 but suppressing LC3B. In addition, at the pathological level, there is a positive correlation between the expression of YY1 and the budding of CRC. This study has revealed the intricate interplay between YY1 and USP7 in CRC, suggesting that they could serve as novel therapeutic targets or predictive biomarkers for CRC patients.
Collapse
Affiliation(s)
- Zhi-Ying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wen-Dong Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhi-Hong He
- Department of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Meng-Ru Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qi Shen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China.
| | - Jun-Nian Zheng
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
21
|
Bowness JS, Almeida M, Nesterova TB, Brockdorff N. YY1 binding is a gene-intrinsic barrier to Xist-mediated gene silencing. EMBO Rep 2024; 25:2258-2277. [PMID: 38654121 PMCID: PMC11094009 DOI: 10.1038/s44319-024-00136-3] [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: 02/23/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
X chromosome inactivation (XCI) in mammals is mediated by Xist RNA which functions in cis to silence genes on a single X chromosome in XX female cells, thereby equalising levels of X-linked gene expression relative to XY males. XCI progresses over a period of several days, with some X-linked genes silencing faster than others. The chromosomal location of a gene is an important determinant of silencing rate, but uncharacterised gene-intrinsic features also mediate resistance or susceptibility to silencing. In this study, we examine mouse embryonic stem cell lines with an inducible Xist allele (iXist-ChrX mESCs) and integrate allele-specific data of gene silencing and decreasing inactive X (Xi) chromatin accessibility over time courses of Xist induction with cellular differentiation. Our analysis reveals that motifs bound by the transcription factor YY1 are associated with persistently accessible regulatory elements, including many promoters and enhancers of slow-silencing genes. We further show that YY1 is evicted relatively slowly from target sites on Xi, and that silencing of X-linked genes is increased upon YY1 degradation. Together our results suggest that YY1 acts as a barrier to Xist-mediated silencing until the late stages of the XCI process.
Collapse
Affiliation(s)
- Joseph S Bowness
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Mafalda Almeida
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | | | - Neil Brockdorff
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
22
|
Xu J, Zhou Y, He S, Wang Y, Ma J, Li C, Liu Z, Zhou X. Activation of the YY1-UGT2B7 Axis Promotes Mammary Estrogen Homeostasis Dysregulation and Exacerbates Breast Tumor Metastasis. Drug Metab Dispos 2024; 52:408-421. [PMID: 38575184 DOI: 10.1124/dmd.124.001640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Metastasis is the most common pathway of cancer death. The lack of effective predictors of breast cancer metastasis is a pressing issue in clinical practice. Therefore, exploring the mechanism of breast cancer metastasis to uncover reliable predictors is very important for the clinical treatment of breast cancer patients. In this study, tandem mass tag quantitative proteomics technology was used to detect protein content in primary breast tumor tissue samples from patients with metastatic and nonmetastatic breast cancer at diagnosis. We found that the high expression of yin-yang 1(YY1) is strongly associated with poor prognosis in high-grade breast cancer. YY1 expression was detected in both clinical tumor tissue samples and tumor tissue samples from mammary-specific polyomavirus middle T antigen overexpression mouse model mice. We demonstrated that upregulation of YY1 expression was closely associated with breast cancer metastasis and that high YY1 expression could promote the migratory invasive ability of breast cancer cells. Mechanistically, YY1 directly binds to the UGT2B7 mRNA initiation sequence ATTCAT, thereby transcriptionally regulating the inhibition of UGT2B7 expression. UGT2B7 can regulate the development of breast cancer by regulating estrogen homeostasis in the breast, and the abnormal accumulation of estrogen, especially 4-OHE2, promotes the migration and invasion of breast cancer cells, ultimately causing the development of breast cancer metastasis. In conclusion, YY1 can regulate the UGT2B7-estrogen metabolic axis and induce disturbances in estrogen metabolism in breast tumors, ultimately leading to breast cancer metastasis. Disturbances in estrogen metabolism in the breast tissue may be an important risk factor for breast tumor progression and metastasis SIGNIFICANCE STATEMENT: In this study, we propose for the first time a regulatory relationship between YY1 and the UGT2B7/estrogen metabolism axis and explore the molecular mechanism. Our study shows that the YY1/UGT2B7/estrogen axis plays an important role in the development and metastasis of breast cancer. This study further elucidates the potential mechanisms of YY1-mediated breast cancer metastasis and the possibility and promise of YY1 as a predictor of cancer metastasis.
Collapse
Affiliation(s)
- Jiahao Xu
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Ying Zhou
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Shiqing He
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Yinghao Wang
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Jiachen Ma
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Changwen Li
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Zhao Liu
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| | - Xueyan Zhou
- 1Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China (J.X., Y.Z., S.H., Y.W., J.M., X.Z.); The First People's Hospital of Changzhou, Changzhou, China (J.X.); Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China (Z.L.); and Department of Breast Surgery, Xuzhou Central Hospital XuZhou Clinical School of Xuzhou Medical University, Xuzhou, China (C.L.)
| |
Collapse
|
23
|
Deng X, Liu Z, Wang B, Ma J, Meng X. The DDX6/KIFC1 signaling axis, as regulated by YY1, contributes to the malignant behavior of pancreatic cancer. FASEB J 2024; 38:e23581. [PMID: 38551642 DOI: 10.1096/fj.202400166r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024]
Abstract
Human DEAD/H box RNA helicase DDX6 acts as an oncogene in several different types of cancer, where it participates in RNA processing. Nevertheless, the role of DDX6 in pancreatic cancer (PC), together with the underlying mechanism, has yet to be fully elucidated. In the present study, compared with adjacent tissues, the level of DDX6 was abnormally increased in human PC tissues, and this increased level of expression was associated with poor prognosis. Furthermore, the role of DDX6 in PC was investigated by overexpressing or silencing the DDX6 in the PC cell lines, SW1990 and PaTu-8988t. A xenograft model was established by injecting nude mice with either DDX6-overexpressing or DDX6-silenced SW1990 cells. DDX6 overexpression promoted the proliferation and cell cycle transition, inhibited the cell apoptosis of PC cells, and accelerated tumor formation, whereas DDX6 knockdown elicited the opposite effects. DDX6 exerted positive effects on PC. RNA immunoprecipitation assay showed that DDX6 bound to kinesin family member C1 (KIFC1) mRNA, which was further confirmed by RNA pull-down assay. These results suggested that DDX6 positively regulated the expression of KIFC1. KIFC1 overexpression enhanced the proliferative capability of PC cells with DDX6 knockdown and inhibited their apoptosis. By contrast, DDX6 overexpression reversed the inhibitory effect of KIFC1 silencing on tumor proliferation. Subsequently, the transcription factor Yin Yang 1 (YY1) was shown to negatively regulate DDX6 at both the mRNA and protein levels. Dual-luciferase reporter assay verified that YY1 targeted the promoter of DDX6 and inhibited its transcription. High expression levels of YY1 decreased the proliferation of PC cells and promoted cell apoptosis, although these effects were reversed by DDX6 overexpression. Taken together, YY1 may target the DDX6/KIFC1 axis, thereby negatively regulating its expression, leading to an inhibitory effect on pancreatic tumor.
Collapse
Affiliation(s)
- Xin Deng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Baosheng Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangpeng Meng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Liang L, Chen S, Su W, Zhang H, Yu R. Integrated Transcriptomic and Proteomic Study of the Mechanism of Action of the Novel Small-Molecule Positive Allosteric Modulator 1 in Targeting PAC1-R for the Treatment of D-Gal-Induced Aging Mice. Int J Mol Sci 2024; 25:3872. [PMID: 38612681 PMCID: PMC11011505 DOI: 10.3390/ijms25073872] [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: 02/26/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Small-molecule positive allosteric modulator 1 (SPAM1), which targets pituitary adenylate cyclase-activating polypeptide receptor 1 (PAC1-R), has been found to have a neuroprotective effect, and the underlying mechanism was explored in this study. First, using a D-galactose (D-gal)-induced aging mouse model, we confirmed that SPAM1 improves the structure of the hippocampal dentate gyrus and restores the number of neurons. Compared with D-gal model mice, SPAM1-treated mice showed up-regulated expression of Sirtuin 6 (SIRT6) and Lamin B1 and down-regulated expression of YinYang 1 (YY1) and p16. A similar tendency was observed in senescent RGC-5 cells induced by long-term culture, indicating that SPAM1 exhibits significant in vitro and in vivo anti-senescence activity in neurons. Then, using whole-transcriptome sequencing and proteomic analysis, we further explored the mechanism behind SPAM1's neuroprotective effects and found that SPAM is involved in the longevity-regulating pathway. Finally, the up-regulation of neurofilament light and medium polypeptides indicated by the proteomics results was further confirmed by Western blotting. These results help to lay a pharmacological network foundation for the use of SPAM1 as a potent anti-aging therapeutic drug to combat neurodegeneration with anti-senescence, neuroprotective, and nerve regeneration activity.
Collapse
Affiliation(s)
- Lili Liang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shang Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Wanlin Su
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Huahua Zhang
- Department of Medical Genetics, Guangdong Medical University, Dongguan 523808, China
| | - Rongjie Yu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
- Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
| |
Collapse
|
25
|
Banerjee S, Sanyal S, Hodawadekar S, Naiyer S, Bano N, Banerjee A, Rhoades J, Dong D, Allman D, Atchison ML. Unusual lineage plasticity revealed by YY1 knockout in pro-B cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586298. [PMID: 38586061 PMCID: PMC10996465 DOI: 10.1101/2024.03.22.586298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
During B cell development, cells progress through multiple developmental stages with the pro-B cell stage defining commitment to the B cell lineage. YY1 is a ubiquitous transcription factor that is capable of both activation and repression functions. We find here that knockout of YY1 at the pro-B cell stage eliminates B lineage commitment. YY1 knockout pro-B cells can generate T lineage cells in vitro using the OP9- DL4 feeder system, as well as in vivo after injection into sub-lethally irradiated Rag1 -/- mice. These T lineage-like cells lose their B lineage transcript profile and gain a T cell lineage profile. Single cell-RNA-seq experiments showed that as YY1 knockout pro-B cells transition into T lineage cells, various cell clusters adopt transcript profiles representing a multiplicity of hematopoietic lineages indicating unusual lineage plasticity. Given the ubiquitous nature of YY1 and its dual activation and repression functions, YY1 likely regulates commitment in multiple cell lineages.
Collapse
|
26
|
Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J, Liu B. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B 2024; 14:953-1008. [PMID: 38487001 PMCID: PMC10935242 DOI: 10.1016/j.apsb.2023.12.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer reprogramming is an important facilitator of cancer development and survival, with tumor cells exhibiting a preference for aerobic glycolysis beyond oxidative phosphorylation, even under sufficient oxygen supply condition. This metabolic alteration, known as the Warburg effect, serves as a significant indicator of malignant tumor transformation. The Warburg effect primarily impacts cancer occurrence by influencing the aerobic glycolysis pathway in cancer cells. Key enzymes involved in this process include glucose transporters (GLUTs), HKs, PFKs, LDHs, and PKM2. Moreover, the expression of transcriptional regulatory factors and proteins, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, can also influence cancer progression. Furthermore, lncRNAs, miRNAs, and circular RNAs play a vital role in directly regulating the Warburg effect. Additionally, gene mutations, tumor microenvironment remodeling, and immune system interactions are closely associated with the Warburg effect. Notably, the development of drugs targeting the Warburg effect has exhibited promising potential in tumor treatment. This comprehensive review presents novel directions and approaches for the early diagnosis and treatment of cancer patients by conducting in-depth research and summarizing the bright prospects of targeting the Warburg effect in cancer.
Collapse
Affiliation(s)
- Minru Liao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dahong Yao
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
| | - Lifeng Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chaodan Luo
- Department of Psychology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiwen Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences, Shenzhen Technology University, Shenzhen 518118, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Ji X, Yang Z, Li C, Zhu S, Zhang Y, Xue F, Sun S, Fu T, Ding C, Liu Y, Wan Q. Mitochondrial ribosomal protein L12 potentiates hepatocellular carcinoma by regulating mitochondrial biogenesis and metabolic reprogramming. Metabolism 2024; 152:155761. [PMID: 38104924 DOI: 10.1016/j.metabol.2023.155761] [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: 10/02/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and metabolic reprogramming are key features of hepatocellular carcinoma (HCC). Despite its significance, the precise underlying mechanism behind these processes has not been fully elucidated. The latest investigations, along with our previous discoveries, have substantiated the significant role of mitochondrial ribosomal protein L12 (MRPL12), a newly identified gene involved in mitochondrial transcription regulation, in the modulation of mitochondrial metabolism. Nevertheless, the role of MRPL12 in tumorigenesis has yet to be investigated. METHODS The expression of MRPL12 in HCC was assessed using an online database. Western blot, quantitative real-time polymerase chain reaction (qRT-PCR), and immunohistochemistry (IHC) were employed to determine the expression of MRPL12 in HCC tissues, patient-derived organoid (PDO), and cell lines. The correlation between MRPL12 expression and clinicopathological features, as well as prognosis, was examined using tissue microarray analysis. An in vivo subcutaneous tumor xenograft model, gene knockdown or overexpression assay, chromatin immunoprecipitation (ChIP) assay, Seahorse XF96 assay, and cell function assay were employed to investigate the biological function and potential molecular mechanism of MRPL12 in HCC. RESULTS A significant upregulation of MRPL12 was observed in HCC cells, PDO and patient tissues, which correlated with advanced tumor stage, higher grade and poor prognosis. MRPL12 overexpression promoted cell proliferation, migration, and invasion in vitro, as well as tumorigenicity in vivo, whereas MRPL12 knockdown showed the opposite effect. MRPL12 knockdown also inhibited the capacity of organoids proliferation capacity. Furthermore, MRPL12 was found to be crucial for maintaining mitochondrial homeostasis. Both gain and loss-of-function experiments targeting MRPL12 in HCC cells altered oxidative phosphorylation (OXPHOS) and mitochondrial DNA content. Notably, suppression of OXPHOS effectively mitigates the tumor-promoting effect attributed to MRPL12 overexpression, implying the involvement of MRPL12 in HCC through the modulation of mitochondrial metabolism. Besides, Yin Yang 1 (YY1) was identified as a transcription factor responsible for regulating MRPL12, while the PI3K/mTOR pathway was found to act as an upstream regulator of YY1. MRPL12 knockdown attenuated the YY1 overexpression or PI3K/mTOR activation-induced malignant phenotype in HCC cells. CONCLUSION Our findings provide compelling evidence that MRPL12 is implicated in driving the malignant phenotype of HCC via regulating mitochondrial metabolism. Moreover, the aberrant expression of MRPL12 in HCC is mediated by the upstream PI3K/mTOR/YY1 pathway. These results highlight the potential of targeting MRPL12 as a promising therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Xingzhao Ji
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhen Yang
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Chensheng Li
- Department of Gastroenterological Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Suwei Zhu
- Department of Critical-Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yu Zhang
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Fuyuan Xue
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shengnan Sun
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Tingting Fu
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Can Ding
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Qiang Wan
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
28
|
Hossain MJ, Nyame P, Monde K. Species-Specific Transcription Factors Associated with Long Terminal Repeat Promoters of Endogenous Retroviruses: A Comprehensive Review. Biomolecules 2024; 14:280. [PMID: 38540701 PMCID: PMC10968565 DOI: 10.3390/biom14030280] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/24/2024] [Indexed: 11/11/2024] Open
Abstract
Endogenous retroviruses (ERVs) became a part of the eukaryotic genome through endogenization millions of years ago. Moreover, they have lost their innate capability of virulence or replication. Nevertheless, in eukaryotic cells, they actively engage in various activities that may be advantageous or disadvantageous to the cells. The mechanisms by which transcription is triggered and implicated in cellular processes are complex. Owing to the diversity in the expression of transcription factors (TFs) in cells and the TF-binding motifs of viruses, the comprehensibility of ERV initiation and its impact on cellular functions are unclear. Currently, several factors are known to be related to their initiation. TFs that bind to the viral long-terminal repeat (LTR) are critical initiators. This review discusses the TFs shown to actively associate with ERV stimulation across species such as humans, mice, pigs, monkeys, zebrafish, Drosophila, and yeast. A comprehensive summary of the expression of previously reported TFs may aid in identifying similarities between animal species and endogenous viruses. Moreover, an in-depth understanding of ERV expression will assist in elucidating their physiological roles in eukaryotic cell development and in clarifying their relationship with endogenous retrovirus-associated diseases.
Collapse
Affiliation(s)
| | | | - Kazuaki Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (M.J.H.); (P.N.)
| |
Collapse
|
29
|
Zhang H, Xia T, Xia Z, Zhou H, Li Z, Wang W, Zhai X, Jin B. KIF18A inactivates hepatic stellate cells and alleviates liver fibrosis through the TTC3/Akt/mTOR pathway. Cell Mol Life Sci 2024; 81:96. [PMID: 38372748 PMCID: PMC10876760 DOI: 10.1007/s00018-024-05114-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 02/20/2024]
Abstract
Activation of hepatic stellate cells (HSCs) has been demonstrated to play a pivotal role in the process of liver fibrogenesis. In this study, we observed a decrease in the expression of KIF18A in fibrotic liver tissues compared to healthy liver tissues, which exhibited a negative correlation with the activation of HSCs. To elucidate the molecular mechanisms underlying the involvement of KIF18A, we performed in vitro proliferation experiments and established a CCl4-induced liver fibrosis model. Our results revealed that KIF18A knockdown enhanced HSCs proliferation and reduced HSCs apoptosis in vitro. Mouse liver fibrosis grade was evaluated with Masson's trichrome and alpha-smooth muscle actin (α-SMA) staining. In addition, the expression of fibrosis markers Col1A1, Stat1, and Timp1 were detected. Animal experiments demonstrated that knockdown of KIF18A could promote liver fibrosis, whereas overexpression of KIF18A alleviated liver fibrosis in a CCl4-induced mouse model. Mechanistically, we found that KIF18A suppressed the AKT/mTOR pathway and exhibited direct binding to TTC3. Moreover, TTC3 was found to interact with p-AKT and could promote its ubiquitination and degradation. Our findings provide compelling evidence that KIF18A enhances the protein binding between TTC3 and p-AKT, promoting TTC3-mediated ubiquitination and degradation of p-AKT. These results refine the current understanding of the mechanisms underlying the pathogenesis of liver fibrosis and may offer new targets for treating this patient population.
Collapse
Affiliation(s)
- Hao Zhang
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Tong Xia
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Huaxin Zhou
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Zhipeng Li
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Wei Wang
- Medical Integration and Practice Center, Shandong University, Jinan, China.
| | - Xiangyu Zhai
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| | - Bin Jin
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China.
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, China.
| |
Collapse
|
30
|
Saha K, Nielsen GI, Nandani R, Kong L, Ye P, An W. YY1 is a transcriptional activator of mouse LINE-1 Tf subfamily. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.573552. [PMID: 38260579 PMCID: PMC10802269 DOI: 10.1101/2024.01.03.573552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Long interspersed element type 1 (LINE-1, L1) is an active autonomous transposable element (TE) in the human genome. The first step of L1 replication is transcription, which is controlled by an internal RNA polymerase II promoter in the 5' untranslated region (UTR) of a full-length L1. It has been shown that transcription factor YY1 binds to a conserved sequence motif at the 5' end of the human L1 5'UTR and dictates where transcription initiates but not the level of transcription. Putative YY1-binding motifs have been predicted in the 5'UTRs of two distinct mouse L1 subfamilies, Tf and Gf. Using site-directed mutagenesis, in vitro binding, and gene knockdown assays, we experimentally tested the role of YY1 in mouse L1 transcription. Our results indicate that Tf, but not Gf subfamily, harbors functional YY1-binding sites in its 5'UTR monomers. In contrast to its role in human L1, YY1 functions as a transcriptional activator for the mouse Tf subfamily. Furthermore, YY1-binding motifs are solely responsible for the synergistic interaction between monomers, consistent with a model wherein distant monomers act as enhancers for mouse L1 transcription. The abundance of YY1-binding sites in Tf elements also raise important implications for gene regulation at the genomic level.
Collapse
Affiliation(s)
- Karabi Saha
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Grace I. Nielsen
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Raj Nandani
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Lingqi Kong
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Ping Ye
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Wenfeng An
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| |
Collapse
|
31
|
Lewerissa EI, Nadif Kasri N, Linda K. Epigenetic regulation of autophagy-related genes: Implications for neurodevelopmental disorders. Autophagy 2024; 20:15-28. [PMID: 37674294 PMCID: PMC10761153 DOI: 10.1080/15548627.2023.2250217] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily highly conserved catabolic process that is important for the clearance of cytosolic contents to maintain cellular homeostasis and survival. Recent findings point toward a critical role for autophagy in brain function, not only by preserving neuronal health, but especially by controlling different aspects of neuronal development and functioning. In line with this, mutations in autophagy-related genes are linked to various key characteristics and symptoms of neurodevelopmental disorders (NDDs), including autism, micro-/macrocephaly, and epilepsy. However, the group of NDDs caused by mutations in autophagy-related genes is relatively small. A significant proportion of NDDs are associated with mutations in genes encoding epigenetic regulatory proteins that modulate gene expression, so-called chromatinopathies. Intriguingly, several of the NDD-linked chromatinopathy genes have been shown to regulate autophagy-related genes, albeit in non-neuronal contexts. From these studies it becomes evident that tight transcriptional regulation of autophagy-related genes is crucial to control autophagic activity. This opens the exciting possibility that aberrant autophagic regulation might underly nervous system impairments in NDDs with disturbed epigenetic regulation. We here summarize NDD-related chromatinopathy genes that are known to regulate transcriptional regulation of autophagy-related genes. Thereby, we want to highlight autophagy as a candidate key hub mechanism in NDD-related chromatinopathies.Abbreviations: ADNP: activity dependent neuroprotector homeobox; ASD: autism spectrum disorder; ATG: AutTophaGy related; CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; EHMT: euchromatic histone lysine methyltransferase; EP300: E1A binding protein p300; EZH2: enhancer of zeste 2 polycomb repressive complex 2 subunit; H3K4me3: histone 3 lysine 4 trimethylation; H3K9me1/2/3: histone 3 lysine 9 mono-, di-, or trimethylation; H3K27me2/3: histone 3 lysine 27 di-, or trimethylation; hiPSCs: human induced pluripotent stem cells; HSP: hereditary spastic paraplegia; ID: intellectual disability; KANSL1: KAT8 regulatory NSL complex subunit 1; KAT8: lysine acetyltransferase 8; KDM1A/LSD1: lysine demethylase 1A; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NDD: neurodevelopmental disorder; PHF8: PHD finger protein 8; PHF8-XLID: PHF8-X linked intellectual disability syndrome; PTM: post-translational modification; SESN2: sestrin 2; YY1: YY1 transcription factor; YY1AP1: YY1 associated protein 1.
Collapse
Affiliation(s)
- Elly I. Lewerissa
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behavior, Nijmegen, Gelderland, The Netherlands
| | - Katrin Linda
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Gelderland, The Netherlands
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Flemish Brabant, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Flemish Brabant, Belgium
| |
Collapse
|
32
|
Ma L, Song H, Zhang CY, Hou D. MiR-192-5p Ameliorates Hepatic Lipid Metabolism in Non-Alcoholic Fatty Liver Disease by Targeting Yy1. Biomolecules 2023; 14:34. [PMID: 38254634 PMCID: PMC10813355 DOI: 10.3390/biom14010034] [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: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in the liver. Clarifying the molecular mechanism of lipid metabolism is crucial for the treatment of NAFLD. We examined miR-192-5p levels in the livers of mice in which NAFLD was induced via a high-fat diet (HFD), as well as in mouse primary hepatocytes and human HepG2 cells treated with free fatty acids (FFAs). MiR-192-5p inhibitor was administered to NAFLD mice and hepatocytes to verify the specific function of miR-192-5p in NAFLD. We validated the target gene of miR-192-5p and further illustrated the effects of this miRNA on the regulation of triglyceride (TG) metabolism. We found that miR-192-5p was significantly increased in the livers of NAFLD mice and FFA-treated hepatocytes. Inhibition of miR-192-5p increased the accumulation of hepatic TGs and aggravated hepatic steatosis in NAFLD mice. In FFA-treated hepatocytes, miR-192-5p inhibitors markedly increased TG content, whereas overexpression of miR-192-5p reduced TG levels. Yin Yang 1 (Yy1) was identified as the target gene of miR-192-5p, which regulates TG synthesis via the YY1/fatty-acid synthase (FASN) pathway. Our results demonstrated that miR-192-5p should be considered a protective regulator in NAFLD that can inhibit hepatic TG synthesis by targeting Yy1.
Collapse
Affiliation(s)
- Lina Ma
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (L.M.); (H.S.)
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing 210023, China
| | - Huichen Song
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (L.M.); (H.S.)
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing 210023, China
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (L.M.); (H.S.)
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing 210023, China
| | - Dongxia Hou
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (L.M.); (H.S.)
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing 210023, China
| |
Collapse
|
33
|
Chen ZS, Ou M, Taylor S, Dafinca R, Peng SI, Talbot K, Chan HYE. Mutant GGGGCC RNA prevents YY1 from binding to Fuzzy promoter which stimulates Wnt/β-catenin pathway in C9ALS/FTD. Nat Commun 2023; 14:8420. [PMID: 38110419 PMCID: PMC10728118 DOI: 10.1038/s41467-023-44215-w] [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: 11/10/2022] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
The GGGGCC hexanucleotide repeat expansion mutation in the chromosome 9 open reading frame 72 (C9orf72) gene is a major genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). In this study, we demonstrate that the zinc finger (ZF) transcriptional regulator Yin Yang 1 (YY1) binds to the promoter region of the planar cell polarity gene Fuzzy to regulate its transcription. We show that YY1 interacts with GGGGCC repeat RNA via its ZF and that this interaction compromises the binding of YY1 to the FuzzyYY1 promoter sites, resulting in the downregulation of Fuzzy transcription. The decrease in Fuzzy protein expression in turn activates the canonical Wnt/β-catenin pathway and induces synaptic deficits in C9ALS/FTD neurons. Our findings demonstrate a C9orf72 GGGGCC RNA-initiated perturbation of YY1-Fuzzy transcriptional control that implicates aberrant Wnt/β-catenin signalling in C9ALS/FTD-associated neurodegeneration. This pathogenic cascade provides a potential new target for disease-modifying therapy.
Collapse
Affiliation(s)
- Zhefan Stephen Chen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Mingxi Ou
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Stephanie Taylor
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Ruxandra Dafinca
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK
| | - Shaohong Isaac Peng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Kevin Talbot
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, UK.
| | - Ho Yin Edwin Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|
34
|
Metzger DCH, Porter I, Mobley B, Sandkam BA, Fong LJM, Anderson AP, Mank JE. Transposon wave remodeled the epigenomic landscape in the rapid evolution of X-Chromosome dosage compensation. Genome Res 2023; 33:1917-1931. [PMID: 37989601 PMCID: PMC10760456 DOI: 10.1101/gr.278127.123] [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: 05/25/2023] [Accepted: 10/20/2023] [Indexed: 11/23/2023]
Abstract
Sex chromosome dosage compensation is a model to understand the coordinated evolution of transcription; however, the advanced age of the sex chromosomes in model systems makes it difficult to study how the complex regulatory mechanisms underlying chromosome-wide dosage compensation can evolve. The sex chromosomes of Poecilia picta have undergone recent and rapid divergence, resulting in widespread gene loss on the male Y, coupled with complete X Chromosome dosage compensation, the first case reported in a fish. The recent de novo origin of dosage compensation presents a unique opportunity to understand the genetic and evolutionary basis of coordinated chromosomal gene regulation. By combining a new chromosome-level assembly of P. picta with whole-genome bisulfite sequencing and RNA-seq data, we determine that the YY1 transcription factor (YY1) DNA binding motif is associated with male-specific hypomethylated regions on the X, but not the autosomes. These YY1 motifs are the result of a recent and rapid repetitive element expansion on the P. picta X Chromosome, which is absent in closely related species that lack dosage compensation. Taken together, our results present compelling support that a disruptive wave of repetitive element insertions carrying YY1 motifs resulted in the remodeling of the X Chromosome epigenomic landscape and the rapid de novo origin of a dosage compensation system.
Collapse
Affiliation(s)
- David C H Metzger
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada;
| | - Imogen Porter
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Brendan Mobley
- Biology Department, Reed College, Portland, Oregon 97202, USA
| | - Benjamin A Sandkam
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York 14853, USA
| | - Lydia J M Fong
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | | | - Judith E Mank
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| |
Collapse
|
35
|
Shi T, Yuan Z, He Y, Zhang D, Chen S, Wang X, Yao L, Shao J, Wang X. Competition between p53 and YY1 determines PHGDH expression and malignancy in bladder cancer. Cell Oncol (Dordr) 2023; 46:1457-1472. [PMID: 37326803 DOI: 10.1007/s13402-023-00823-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/17/2023] Open
Abstract
PURPOSE Serine metabolism is frequently dysregulated in many types of cancers and the tumor suppressor p53 is recently emerging as a key regulator of serine metabolism. However, the detailed mechanism remains unknown. Here, we investigate the role and underlying mechanisms of how p53 regulates the serine synthesis pathway (SSP) in bladder cancer (BLCA). METHODS Two BLCA cell lines RT-4 (WT p53) and RT-112 (p53 R248Q) were manipulated by applying CRISPR/Cas9 to examine metabolic differences under WT and mutant p53 status. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) and non-targeted metabolomics analysis were adopted to identify metabolomes changes between WT and p53 mutant BLCA cells. Bioinformatics analysis using the cancer genome atlas and Gene Expression Omnibus datasets and immunohistochemistry (IHC) staining was used to investigate PHGDH expression. Loss-of-function of PHGDH and subcutaneous xenograft model was adopted to investigate the function of PHGDH in mice BLCA. Chromatin immunoprecipitation (Ch-IP) assay was performed to analyze the relationships between YY1, p53, SIRT1 and PHGDH expression. RESULTS SSP is one of the most prominent dysregulated metabolic pathways by comparing the metabolomes changes between wild-type (WT) p53 and mutant p53 of BLCA cells. TP53 gene mutation shows a positive correlation with PHGDH expression in TCGA-BLCA database. PHGDH depletion disturbs the reactive oxygen species homeostasis and attenuates the xenograft growth in the mouse model. Further, we demonstrate WT p53 inhibits PHGDH expression by recruiting SIRT1 to the PHGDH promoter. Interestingly, the DNA binding motifs of YY1 and p53 in the PHGDH promoter are partially overlapped which causes competition between the two transcription factors. This competitive regulation of PHGDH is functionally linked to the xenograft growth in mice. CONCLUSION YY1 drives PHGDH expression in the context of mutant p53 and promotes bladder tumorigenesis, which preliminarily explains the relationship between high-frequency mutations of p53 and dysfunctional serine metabolism in bladder cancer.
Collapse
Affiliation(s)
- Tiezhu Shi
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Zhihao Yuan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Yanying He
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
| | - Dongliang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Siteng Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China
| | - Xiongjun Wang
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, 510006, Guangzhou, China
| | - Linli Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiaotong University School of Medicine, 200080, Shanghai, China.
| | - Jialiang Shao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China.
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, 200080, Shanghai, China.
| |
Collapse
|
36
|
Liu R, Xu Z, Huang X, Xu B, Chen M. Yin Yang 1 promotes the neuroendocrine differentiation of prostate cancer cells via the non-canonical WNT pathway (FYN/STAT3). Clin Transl Med 2023; 13:e1422. [PMID: 37771187 PMCID: PMC10539684 DOI: 10.1002/ctm2.1422] [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: 02/28/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND A growing number of studies have shown that Yin Yang 1 (YY1) promotes the development of multiple tumours. The purpose of the current study was to determine the mechanism by which YY1 mediates neuroendocrine differentiation of prostate cancer (NEPC) cells undergoing cellular plasticity. METHODS Using the Cancer Genome Atlas and Gene Expression Omnibus (GEO) databases, we bioinformatically analyzed YY1 expression in prostate cancer (PCa). Aberrant YY1 expression was validated in different PCa tissues and cell lines via quantitative reverse transcription polymerase chain reaction, western blotting, and immunohistochemistry. In vivo and in vitro functional assays verified the oncogenicity of YY1 in PCa. Further functional assays showed that ectopic expression of YY1 promoted cellular plasticity in PCa cells via epithelial-mesenchymal transition induction and neuroendocrine differentiation. RESULTS Androgen deprivation therapy induced a decrease in YY1 protein ubiquitination, enhanced its stability, and thus enhanced the transcriptional activity of FZD8. Castration enhanced FZD8 binding to Wnt9A and mediated cellular plasticity by activating the non-canonical Wnt (FZD8/FYN/STAT3) pathway. CONCLUSIONS We identified YY1 as a novel dysregulated transcription factor that plays an important role in NEPC progression in this study. We believe that an in-depth investigation of the mechanism underlying YY1-mediated disease may lead to improved NEPC therapies.
Collapse
Affiliation(s)
- Rui‐ji Liu
- Department of Urology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
- Surgical Research Center, Institute of UrologySoutheast University Medical SchoolNanjingChina
| | - Zhi‐Peng Xu
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
- Surgical Research Center, Institute of UrologySoutheast University Medical SchoolNanjingChina
| | - Xiang Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Bin Xu
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
- Surgical Research Center, Institute of UrologySoutheast University Medical SchoolNanjingChina
| | - Ming Chen
- Department of UrologyAffiliated Zhongda Hospital of Southeast UniversityNanjingChina
- Surgical Research Center, Institute of UrologySoutheast University Medical SchoolNanjingChina
- Department of Urology, Nanjing Lishui District People's HospitalZhongda Hospital Lishui BranchSoutheast UniversityNanjingChina
| |
Collapse
|
37
|
Wang S, Nie J, Xu K, Liu Y, Tong W, Li A, Zuo W, Liu Z, Yang F. YY1 is regulated by ALKBH5-mediated m6A modification and promotes autophagy and cancer progression through targeting ATG4B. Aging (Albany NY) 2023; 15:9590-9613. [PMID: 37724907 PMCID: PMC10564435 DOI: 10.18632/aging.205037] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023]
Abstract
YY1 affects tumorigenesis and metastasis in multiple ways. However, the function of YY1 and the potential mechanisms through which it operates in gastric cancer (GC) progression by regulating autophagy remains poorly understood. This study aimed to assess the essential transcription factors (TFs) involved in autophagy regulation in GC. Western blot, RFP-GFP-LC3 double fluorescence and transmission electron microscopy (TEM) assays were used to probe autophagy activity in GC cells. Methylated RNA immunoprecipitation (MeRIP) was utilized to evaluate the ALKBH5-regulated m6A levels of YY1. Gain- and loss-of-function assays were employed in the scrutiny of the biological effects of the ALKBH5/YY1/ATG4B axis on cancer cell proliferation and invasion abilities in vitro. Per the findings, YY1 was identified as a crucial transcriptional activator of cancer autophagy-related genes and promoted the proliferation and aggressiveness of cancer cells associated with enhanced ATG4B-mediated autophagy. However, ectopic ALKBH5 expression abolished the YY1-induced effect via m6A modification. Importantly, YTHDF1 facilitated the mRNA stability of YY1 through m6A recognition. Collectively, this study found that YY1 was regulated by ALKBH5 and YTHDF1-mediated m6A modification and served as an autophagy-dependent tumor driver to accelerate cancer progression through ATG4B transactivation, providing an exploitable therapeutic target for GC.
Collapse
Affiliation(s)
- Shijiang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Jiangbo Nie
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Kaiying Xu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Yangyang Liu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Weilai Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Anan Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Wei Zuo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Zhili Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| | - Feng Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People’s Republic of China
| |
Collapse
|
38
|
Romero-Estrada JH, Montaño LF, Rendón-Huerta EP. Binding of YY1/CREB to an Enhancer Region Triggers Claudin 6 Expression in H. pylori LPS-Stimulated AGS Cells. Int J Mol Sci 2023; 24:13974. [PMID: 37762277 PMCID: PMC10531490 DOI: 10.3390/ijms241813974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Aberrant expression of the tight junction protein claudin 6 (CLDN6) is a hallmark of gastric cancer progression. Its expression is regulated by the cAMP response element-binding protein (CREB). In gastric cancer induced by Helicobacter pylori (H. pylori) there is no information regarding what transcription factors induce/upregulate the expression of CLDN6. We aimed to identify whether CREB and Yin Yang1 (YY1) regulate the expression of CLDN6 and the site where they bind to the promoter sequence. Bioinformatics analysis, H. pylori lipopolysaccharide (LPS), YY1 and CREB silencing, Western blot, luciferase assays, and chromatin immunoprecipitation experiments were performed using the stomach gastric adenocarcinoma cell line AGS. A gen reporter assay suggested that the initial 2000 bp contains the regulatory sequence associated with CLDN6 transcription; the luciferase assay demonstrated three different regions with transcriptional activity, but the -901 to -1421 bp region displayed the maximal transcriptional activity in response to LPS. Fragment 1279-1421 showed CREB and, surprisingly, YY1 occupancy. Sequential Chromatin Immunoprecipitation (ChIP) experiments confirmed that YY1 and CREB interact in the 1279-1421 region. Our results suggest that CLDN6 expression is regulated by the binding of YY1 and CREB in the 901-1421 enhancer, in which a non-described interaction of YY1 with CREB was established in the 1279-1421 region.
Collapse
Affiliation(s)
| | - Luis F. Montaño
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Tisular, Facultad de Medicina, Ciudad Universitaria, Ciudad de México 04510, Mexico;
| | - Erika P. Rendón-Huerta
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Tisular, Facultad de Medicina, Ciudad Universitaria, Ciudad de México 04510, Mexico;
| |
Collapse
|
39
|
Sun L, Dong X, Song X. PtrABR1 Increases Tolerance to Drought Stress by Enhancing Lateral Root Formation in Populus trichocarpa. Int J Mol Sci 2023; 24:13748. [PMID: 37762051 PMCID: PMC10530772 DOI: 10.3390/ijms241813748] [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: 08/12/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Roots are the main organ for water uptake and the earliest part of a plant's response to drought, making them of great importance to our understanding of the root system's response to drought. However, little is known about the underlying molecular mechanisms that control root responses to drought stress. Here, we identified and functionally characterized the AP2/ERF family transcription factor (TF) PtrABR1 and the upstream target gene zinc-finger protein TF PtrYY1, which respond to drought stress by promoting the growth and development of lateral roots in Populus trichocarpa. A root-specific induction of PtrABR1 under drought stress was explored. The overexpression of PtrABR1 (PtrABR1-OE) promoted root growth and development, thereby increasing tolerance to drought stress. In addition, PtrYY1 is directly bound to the promoter of PtrABR1 under drought stress, and the overexpression of PtrYY1 (PtrYY1-OE) promoted lateral root growth and development and increased tolerance to drought stress. An RNA-seq analysis of PtrABR1-OE with wild-type (WT) poplar identified PtrGH3.6 and PtrPP2C44, which share the same pattern of expression changes as PtrABR1. A qRT-PCR and cis-element analysis further suggested that PtrGH3.6 and PtrPP2C44 may act as potential downstream targets of PtrABR1 genes in the root response pathway to drought stress. In conclusion, these results reveal a novel drought regulatory pathway in which PtrABR1 regulates the network through the upstream target gene PtrYY1 and the potential downstream target genes PtrGH3.6 and PtrPP2C44, thereby promoting root growth and development and improving tolerance to drought stress.
Collapse
Affiliation(s)
- Lijiao Sun
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin 150040, China; (L.S.); (X.D.)
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Xinxin Dong
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin 150040, China; (L.S.); (X.D.)
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Xingshun Song
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin 150040, China; (L.S.); (X.D.)
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
40
|
Zhou W, Feng Y, Lin C, CHAO CK, He Z, Zhao S, Xue J, Zhao X, Cao W. Yin Yang 1-Induced Long Noncoding RNA DUXAP9 Drives the Progression of Oral Squamous Cell Carcinoma by Blocking CDK1-Mediated EZH2 Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207549. [PMID: 37401236 PMCID: PMC10477890 DOI: 10.1002/advs.202207549] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/04/2023] [Indexed: 07/05/2023]
Abstract
LncRNAs play a critical role in oral squamous cell carcinoma (OSCC) progression. However, the function and detailed molecular mechanism of most lncRNAs in OSCC are not fully understood. Here, a novel nuclear-localized lncRNA, DUXAP9 (DUXAP9), that is highly expressed in OSCC is identified. A high level of DUXAP9 is positively associated with lymph node metastasis, poor pathological differentiation, advanced clinical stage, worse overall survival, and worse disease-specific survival in OSCC patients. Overexpression of DUXAP9 significantly promotes OSCC cell proliferation, migration, invasion, and xenograft tumor growth and metastasis, and upregulates N-cadherin, Vimentin, Ki67, PCNA, and EZH2 expression and downregulates E-cadherin in vitro and in vivo, whereas knockdown of DUXAP9 remarkably suppresses OSCC cell proliferation, migration, invasion, and xenograft tumor growth in vitro and in vivo in an EZH2-dependent manner. Yin Yang 1 (YY1) is found to activate the transcriptional expression of DUXAP9 in OSCC. Furthermore, DUXAP9 physically interacts with EZH2 and inhibits EZH2 degradation via the suppression of EZH2 phosphorylation, thereby blocking EZH2 translocation from the nucleus to the cytoplasm. Thus, DUXAP9 can serve as a promising target for OSCC therapy.
Collapse
Affiliation(s)
- Wenkai Zhou
- Department of Oral and Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Yisheng Feng
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Chengzhong Lin
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
- The 2nd Dental CenterShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Chi Kuan CHAO
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Ziqi He
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Shiyao Zhao
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Jieyuan Xue
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030China
| | - Xu‐Yun Zhao
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationKey Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Wei Cao
- Department of Oral and Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| |
Collapse
|
41
|
Jung M, Bui I, Bonavida B. Role of YY1 in the Regulation of Anti-Apoptotic Gene Products in Drug-Resistant Cancer Cells. Cancers (Basel) 2023; 15:4267. [PMID: 37686541 PMCID: PMC10486809 DOI: 10.3390/cancers15174267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer is a leading cause of death among the various diseases encountered in humans. Cancer is not a single entity and consists of numerous different types and subtypes that require various treatment regimens. In the last decade, several milestones in cancer treatments were accomplished, such as specific targeting agents or revitalizing the dormant anti-tumor immune response. These milestones have resulted in significant positive clinical responses as well as tumor regression and the prolongation of survival in subsets of cancer patients. Hence, in non-responding patients and non-responding relapsed patients, cancers develop intrinsic mechanisms of resistance to cell death via the overexpression of anti-apoptotic gene products. In parallel, the majority of resistant cancers have been reported to overexpress a transcription factor, Yin Yang 1 (YY1), which regulates the chemo-immuno-resistance of cancer cells to therapeutic anticancer cytotoxic agents. The relationship between the overexpression of YY1 and several anti-apoptotic gene products, such as B-cell lymphoma 2 protein (Bcl-2), B-cell lymphoma extra-large (Bcl-xL), myeloid cell leukemia 1 (Mcl-1) and survivin, is investigated in this paper. The findings demonstrate that these anti-apoptotic gene products are regulated, in part, by YY1 at the transcriptional, epigenetic, post-transcriptional and translational levels. While targeting each of the anti-apoptotic gene products individually has been examined and clinically tested for some, this targeting strategy is not effective due to compensation by other overexpressed anti-apoptotic gene products. In contrast, targeting YY1 directly, through small interfering RNAs (siRNAs), gene editing or small molecule inhibitors, can be therapeutically more effective and generalized in YY1-overexpressed resistant cancers.
Collapse
Affiliation(s)
| | | | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
Wu Y, Li X, Dong L, Liu T, Tang Z, Lin R, Norvienyeku J, Xing M. A New Insight into 6-Pentyl-2H-pyran-2-one against Peronophythora litchii via TOR Pathway. J Fungi (Basel) 2023; 9:863. [PMID: 37623635 PMCID: PMC10515317 DOI: 10.3390/jof9080863] [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: 07/05/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/26/2023] Open
Abstract
The litchi downy blight disease of litchi caused by Peronophythora litchii accounts for severe losses in the field and during storage. While ample quantitative studies have shown that 6-pentyl-2H-pyran-2-one (6PP) possesses antifungal activities against multiple plant pathogenic fungi, the regulatory mechanisms of 6PP-mediated inhibition of fungal pathogenesis and growth are still unknown. Here, we investigated the potential molecular targets of 6PP in the phytopathogenic oomycetes P. litchii through integrated deployment of RNA-sequencing, functional genetics, and biochemical techniques to investigate the regulatory effects of 6PP against P. litchii. Previously we demonstrated that 6PP exerted significant oomyticidal activities. Also, comparative transcriptomic evaluation of P. litchii strains treated with 6PP Revealed significant up-regulations in the expression profile of TOR pathway-related genes, including PlCytochrome C and the transcription factors PlYY1. We also noticed that 6PP treatment down-regulated putative negative regulatory genes of the TOR pathway, including PlSpm1 and PlrhoH12 in P. litchii. Protein-ligand binding analyses revealed stable affinities between PlYY1, PlCytochrome C, PlSpm1, PlrhoH12 proteins, and the 6PP ligand. Phenotypic characterization of PlYY1 targeted gene deletion strains generated in this study using CRISPR/Cas9 and homologous recombination strategies significantly reduced the vegetative growth, sporangium, encystment, zoospore release, and pathogenicity of P. litchii. These findings suggest that 6PP-mediated activation of PlYY1 expression positively regulates TOR-related responses and significantly influences vegetative growth and the virulence of P. litchii. The current investigations revealed novel targets for 6PP and underscored the potential of deploying 6PP in developing management strategies for controlling the litchi downy blight pathogen.
Collapse
Affiliation(s)
- Yinggu Wu
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Xinyu Li
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Li Dong
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Tong Liu
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Zhengbin Tang
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Runmao Lin
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Justice Norvienyeku
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| | - Mengyu Xing
- Key Laboratory of Green Prevention and Control of Tropical Diseases and Pests, Ministry of Education, School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; (Y.W.); (X.L.); (L.D.); (T.L.); (Z.T.); (R.L.)
- Sanya Nanfan Research Institute, Hainan University, Sanya 572025, China
| |
Collapse
|
43
|
Hosea R, Hillary S, Wu S, Kasim V. Targeting Transcription Factor YY1 for Cancer Treatment: Current Strategies and Future Directions. Cancers (Basel) 2023; 15:3506. [PMID: 37444616 DOI: 10.3390/cancers15133506] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer represents a significant and persistent global health burden, with its impact underscored by its prevalence and devastating consequences. Whereas numerous oncogenes could contribute to cancer development, a group of transcription factors (TFs) are overactive in the majority of tumors. Targeting these TFs may also combat the downstream oncogenes activated by the TFs, making them attractive potential targets for effective antitumor therapeutic strategy. One such TF is yin yang 1 (YY1), which plays crucial roles in the development and progression of various tumors. In preclinical studies, YY1 inhibition has shown efficacy in inhibiting tumor growth, promoting apoptosis, and sensitizing tumor cells to chemotherapy. Recent studies have also revealed the potential of combining YY1 inhibition with immunotherapy for enhanced antitumor effects. However, clinical translation of YY1-targeted therapy still faces challenges in drug specificity and delivery. This review provides an overview of YY1 biology, its role in tumor development and progression, as well as the strategies explored for YY1-targeted therapy, with a focus on their clinical implications, including those using small molecule inhibitors, RNA interference, and gene editing techniques. Finally, we discuss the challenges and current limitations of targeting YY1 and the need for further research in this area.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| |
Collapse
|
44
|
Dejosez M, Dall'Agnese A, Ramamoorthy M, Platt J, Yin X, Hogan M, Brosh R, Weintraub AS, Hnisz D, Abraham BJ, Young RA, Zwaka TP. Regulatory architecture of housekeeping genes is driven by promoter assemblies. Cell Rep 2023; 42:112505. [PMID: 37182209 PMCID: PMC10329844 DOI: 10.1016/j.celrep.2023.112505] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/22/2023] [Accepted: 04/28/2023] [Indexed: 05/16/2023] Open
Abstract
Genes that are key to cell identity are generally regulated by cell-type-specific enhancer elements bound by transcription factors, some of which facilitate looping to distant gene promoters. In contrast, genes that encode housekeeping functions, whose regulation is essential for normal cell metabolism and growth, generally lack interactions with distal enhancers. We find that Ronin (Thap11) assembles multiple promoters of housekeeping and metabolic genes to regulate gene expression. This behavior is analogous to how enhancers are brought together with promoters to regulate cell identity genes. Thus, Ronin-dependent promoter assemblies provide a mechanism to explain why housekeeping genes can forgo distal enhancer elements and why Ronin is important for cellular metabolism and growth control. We propose that clustering of regulatory elements is a mechanism common to cell identity and housekeeping genes but is accomplished by different factors binding distinct control elements to establish enhancer-promoter or promoter-promoter interactions, respectively.
Collapse
Affiliation(s)
- Marion Dejosez
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA
| | - Alessandra Dall'Agnese
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Mahesh Ramamoorthy
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA
| | - Jesse Platt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Xing Yin
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA
| | - Megan Hogan
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA
| | - Ran Brosh
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA
| | - Abraham S Weintraub
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Denes Hnisz
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Brian J Abraham
- St. Jude Research Children's Hospital, Memphis, TN 38105, USA
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Thomas P Zwaka
- Black Family Stem Cell Institute, Huffington Center for Cell-based Research in Parkinson's Disease, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10502, USA.
| |
Collapse
|
45
|
Wu T, Zhao B, Cai C, Chen Y, Miao Y, Chu J, Sui Y, Li F, Chen W, Cai Y, Wang F, Jin J. The Males Absent on the First (MOF) Mediated Acetylation Alters the Protein Stability and Transcriptional Activity of YY1 in HCT116 Cells. Int J Mol Sci 2023; 24:ijms24108719. [PMID: 37240065 DOI: 10.3390/ijms24108719] [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: 04/23/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Yin Yang 1 (YY1) is a well-known transcription factor that controls the expression of many genes and plays an important role in the occurrence and development of various cancers. We previously found that the human males absent on the first (MOF)-containing histone acetyltransferase (HAT) complex may be involved in regulating YY1 transcriptional activity; however, the precise interaction between MOF-HAT and YY1, as well as whether the acetylation activity of MOF impacts the function of YY1, has not been reported. Here, we present evidence that the MOF-containing male-specific lethal (MSL) HAT complex regulates YY1 stability and transcriptional activity in an acetylation-dependent manner. First, the MOF/MSL HAT complex was bound to and acetylated YY1, and this acetylation further promoted the ubiquitin-proteasome degradation pathway of YY1. The MOF-mediated degradation of YY1 was mainly related to the 146-270 amino acid residues of YY1. Further research clarified that acetylation-mediated ubiquitin degradation of YY1 mainly occurred through lysine 183. A mutation at the YY1K183 site was sufficient to alter the expression level of p53-mediated downstream target genes, such as CDKN1A (encoding p21), and it also suppressed the transactivation of YY1 on CDC6. Furthermore, a YY1K183R mutant and MOF remarkably antagonized the clone-forming ability of HCT116 and SW480 cells facilitated by YY1, suggesting that the acetylation-ubiquitin mode of YY1 plays an important role in tumor cell proliferation. These data may provide new strategies for the development of therapeutic drugs for tumors with high expression of YY1.
Collapse
Affiliation(s)
- Tingting Wu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bingxin Zhao
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chengyu Cai
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yuyang Chen
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yujuan Miao
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jinmeng Chu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yi Sui
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fuqiang Li
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenqi Chen
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fei Wang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
46
|
Liu X, Xing Q, Liu X, Müller-Xing R. Expression of the Populus Orthologues of AtYY1, YIN and YANG Activates the Floral Identity Genes AGAMOUS and SEPALLATA3 Accelerating Floral Transition in Arabidopsis thaliana. Int J Mol Sci 2023; 24:ijms24087639. [PMID: 37108801 PMCID: PMC10146089 DOI: 10.3390/ijms24087639] [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: 02/10/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
YIN YANG 1 (YY1) encodes a dual-function transcription factor, evolutionary conserved between the animal and plant kingdom. In Arabidopsis thaliana, AtYY1 is a negative regulator of ABA responses and floral transition. Here, we report the cloning and functional characterization of the two AtYY1 paralogs, YIN and YANG (also named PtYY1a and PtYY1b) from Populus (Populus trichocarpa). Although the duplication of YY1 occurred early during the evolution of the Salicaceae, YIN and YANG are highly conserved in the willow tree family. In the majority of Populus tissues, YIN was more strongly expressed than YANG. Subcellular analysis showed that YIN-GFP and YANG-GFP are mainly localized in the nuclei of Arabidopsis. Stable and constitutive expression of YIN and YANG resulted in curled leaves and accelerated floral transition of Arabidopsis plants, which was accompanied by high expression of the floral identity genes AGAMOUS (AG) and SEPELLATA3 (SEP3) known to promote leaf curling and early flowering. Furthermore, the expression of YIN and YANG had similar effects as AtYY1 overexpression to seed germination and root growth in Arabidopsis. Our results suggest that YIN and YANG are functional orthologues of the dual-function transcription factor AtYY1 with similar roles in plant development conserved between Arabidopsis and Populus.
Collapse
Affiliation(s)
- Xinying Liu
- Institute of Genetics, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Qian Xing
- Lushan Botanical Garden, Chinese Academy of Sciences (CAS), Jiujiang 332900, China
| | - Xuemei Liu
- Institute of Genetics, College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Ralf Müller-Xing
- Lushan Botanical Garden, Chinese Academy of Sciences (CAS), Jiujiang 332900, China
| |
Collapse
|
47
|
Wang X, Fan W, Li N, Ma Y, Yao M, Wang G, He S, Li W, Tan J, Lu Q, Hou S. YY1 lactylation in microglia promotes angiogenesis through transcription activation-mediated upregulation of FGF2. Genome Biol 2023; 24:87. [PMID: 37085894 PMCID: PMC10120156 DOI: 10.1186/s13059-023-02931-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 04/07/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Ocular neovascularization is a leading cause of blindness. Retinal microglia have been implicated in hypoxia-induced angiogenesis and vasculopathy, but the underlying mechanisms are not entirely clear. Lactylation is a novel lactate-derived posttranslational modification that plays key roles in multiple cellular processes. Since hypoxia in ischemic retinopathy is a precipitating factor for retinal neovascularization, lactylation is very likely to be involved in this process. The present study aimed to explore the role of lactylation in retinal neovascularization and identify new therapeutic targets for retinal neovascular diseases. RESULTS Microglial depletion by the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX3397 suppresses retinal neovascularization in oxygen-induced retinopathy. Hypoxia increased lactylation in microglia and accelerates FGF2 expression, promoting retinal neovascularization. We identify 77 sites of 67 proteins with increased lactylation in the context of increased lactate under hypoxia. Our results show that the nonhistone protein Yin Yang-1 (YY1), a transcription factor, is lactylated at lysine 183 (K183), which is regulated by p300. Hyperlactylated YY1 directly enhances FGF2 transcription and promotes angiogenesis. YY1 mutation at K183 eliminates these effects. Overexpression of p300 increases YY1 lactylation and enhances angiogenesis in vitro and administration of the p300 inhibitor A485 greatly suppresses vascularization in vivo and in vitro. CONCLUSIONS Our results suggest that YY1 lactylation in microglia plays an important role in retinal neovascularization by upregulating FGF2 expression. Targeting the lactate/p300/YY1 lactylation/FGF2 axis may provide new therapeutic targets for proliferative retinopathies.
Collapse
Affiliation(s)
- Xiaotang Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Wei Fan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Na Li
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Ma
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Mudi Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Guoqing Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Siyuan He
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Wanqian Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Jun Tan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China
- Chongqing Eye Institute, Chongqing, China
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Qi Lu
- The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Ophthalmology, Chongqing, China.
- Chongqing Eye Institute, Chongqing, China.
- Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China.
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
48
|
Hu Y, Mu H, Deng Z. RBM14 as a novel epigenetic-activated tumor oncogene is implicated in the reprogramming of glycolysis in lung cancer. World J Surg Oncol 2023; 21:132. [PMID: 37060064 PMCID: PMC10105460 DOI: 10.1186/s12957-023-02928-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 02/05/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND RNA-binding motif protein 14 (RBM14) is upregulated in a variety of tumors. However, the expression and biological role of RBM14 in lung cancer remain unclear. METHODS Chromatin immunoprecipitation and PCR were carried out to measure the levels of sedimentary YY1, EP300, H3K9ac, and H3K27ac in the RBM14 promoter. Co-immunoprecipitation was used to verify the interaction between YY1 and EP300. Glycolysis was investigated according to glucose consumption, lactate production, and the extracellular acidification rate (ECAR). RESULTS RBM14 level is increased in lung adenocarcinoma (LUAD) cells. The increased RBM14 expression was correlated with TP53 mutation and individual cancer stages. A high level of RBM14 predicted a poorer overall survival of LUAD patients. The upregulated RBM14 in LUAD is induced by DNA methylation and histone acetylation. The transcription factor YY1 directly binds to EP300 and recruits EP300 to the promoter regions of RBM14, which further enhances H3K27 acetylation and promotes RBM14 expression. YY1-induced upregulation of RBM14 promoted cell growth and inhibited apoptosis by affecting the reprogramming of glycolysis. CONCLUSIONS These results indicated that epigenetically activated RBM14 regulated growth and apoptosis by regulating the reprogramming of glycolysis and RBM14 may serve as a promising biomarker and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Yan Hu
- Department of Respiratory, The First People's Hospital of Zigong City, No.42, Shangyihao Road, Ziliujing District, Zigong City, 643000, Sichuan, China
| | - Hanshuo Mu
- Medical School, Nantong University, Nantong, 226001, Jiangsu, China
| | - Zhiping Deng
- Department of Respiratory, The First People's Hospital of Zigong City, No.42, Shangyihao Road, Ziliujing District, Zigong City, 643000, Sichuan, China.
| |
Collapse
|
49
|
Sun H, Pratt RE, Dzau VJ, Hodgkinson CP. Neonatal and adult cardiac fibroblasts exhibit inherent differences in cardiac regenerative capacity. J Biol Chem 2023; 299:104694. [PMID: 37044217 DOI: 10.1016/j.jbc.2023.104694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
Directly reprogramming fibroblasts into cardiomyocytes improves cardiac function in the infarcted heart. However, the low efficacy of this approach hinders clinical applications. Unlike the adult mammalian heart, the neonatal heart has an intrinsic regenerative capacity. Consequently, we hypothesized that birth imposes fundamental changes on cardiac fibroblasts which limit their regenerative capabilities. In support, we found that reprogramming efficacy in vitro was markedly lower with fibroblasts derived from adult mice versus those derived from neonatal mice. Notably, fibroblasts derived from adult mice expressed significantly higher levels of pro-angiogenic genes. Moreover, under conditions which promote angiogenesis, only fibroblasts derived from adult mice differentiated into tube-like structures. Targeted knockdown screening studies suggested a possible role for the transcription factor Epas1. Epas1 expression was higher in fibroblasts derived from adult mice and Epas1 knockdown improved reprogramming efficacy in cultured adult cardiac fibroblasts. Promoter activity assays indicated that Epas1 functions as both a transcription repressor and activator, inhibiting cardiomyocyte genes while activating angiogenic genes. Finally, the addition of an Epas1 targeting siRNA to the reprogramming cocktail markedly improved reprogramming efficacy in vivo with both the number of reprogramming events as well as cardiac function being markedly improved. Collectively, our results highlight differences between neonatal and adult cardiac fibroblasts and the dual transcriptional activities of Epas1 related to reprogramming efficacy.
Collapse
Affiliation(s)
- Hualing Sun
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710; Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Hubei Province, China
| | - Richard E Pratt
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| | - Victor J Dzau
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710.
| |
Collapse
|
50
|
Li M, Wei J, Xue C, Zhou X, Chen S, Zheng L, Duan Y, Deng H, Xiong W, Tang F, Li G, Zhou M. Dissecting the roles and clinical potential of YY1 in the tumor microenvironment. Front Oncol 2023; 13:1122110. [PMID: 37081988 PMCID: PMC10110844 DOI: 10.3389/fonc.2023.1122110] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Yin-Yang 1 (YY1) is a member of the GLI-Kruppel family of zinc finger proteins and plays a vital dual biological role in cancer as an oncogene or a tumor suppressor during tumorigenesis and tumor progression. The tumor microenvironment (TME) is identified as the “soil” of tumor that has a critical role in both tumor growth and metastasis. Many studies have found that YY1 is closely related to the remodeling and regulation of the TME. Herein, we reviewed the expression pattern of YY1 in tumors and summarized the function and mechanism of YY1 in regulating tumor angiogenesis, immune and metabolism. In addition, we discussed the potential value of YY1 in tumor diagnosis and treatment and provided a novel molecular strategy for the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- MengNa Li
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - JianXia Wei
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - ChangNing Xue
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - XiangTing Zhou
- The First Clinical College of Changsha Medical University, Changsha, China
| | - ShiPeng Chen
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - LeMei Zheng
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - YuMei Duan
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - HongYu Deng
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - FaQing Tang
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - GuiYuan Li
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Ming Zhou
- Key Laboratory of Carcinogenesis, National Health Commission, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Ming Zhou,
| |
Collapse
|