1
|
Shaikh S, Zhao X, Wagner RT, Pan X, Hlady RA, Wang L, Ho TH, Robertson KD. Deciphering the interplay between SETD2 mediated H3K36me3 and RNA N6-methyladenosine in clear cell renal cell carcinoma (ccRCC). Epigenetics 2025; 20:2456418. [PMID: 39874221 PMCID: PMC11776469 DOI: 10.1080/15592294.2025.2456418] [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: 08/25/2024] [Revised: 12/21/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
RNA N6-methyladenosine (m6A) plays diverse roles in RNA metabolism and its deregulation contributes to tumor initiation and progression. Clear cell renal cell carcinoma (ccRCC) is characterized by near ubiquitous loss of VHL followed by mutations in epigenetic regulators PBRM1, SETD2, and BAP1. Mutations in SETD2, a histone H3 lysine 36 trimethylase (H3K36me3), are associated with reduced survival, greater metastatic propensity, and metabolic reprogramming. While m6A and H3K36me3 deregulation are separately implicated in renal tumorigenesis, H3K36me3 may participate directly in m6A targeting, but the m6A-H3K36me3 interplay has not been investigated in the context of ccRCC. Using RCC-relevant SETD2 isogenic knockout and rescue cell line models, we demonstrate a dynamic redistribution of m6A in the SETD2 depleted transcriptome, with a subset of transcripts involved in metabolic reprogramming demonstrating SETD2 dependent m6A and expression level changes. Using a panel of six histone modifications we show that m6A redistributes to regions enriched in gained active enhancers upon SETD2 inactivation. Finally, we demonstrate a reversal of transcriptomic programs involved in SETD2 loss mediated metabolic reprogramming, and reduced cell viability through pharmacologic inhibition or genetic ablation of m6A writer METTL3 specific to SETD2 deficient cells. Thus, targeting m6A may represent a novel therapeutic vulnerability in SETD2 mutant ccRCC.
Collapse
Affiliation(s)
- Shafiq Shaikh
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ryan T. Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Xiaoyu Pan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ryan A. Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Thai H. Ho
- Division of Hematology and Oncology, Medical University of South Carolina, Charleston, SC, USA
| | - Keith D. Robertson
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Liu Y, Lu B, Yang X, Cui J, Yang T, Zhang H, Zhao Z, Lyu D, Li Y, Yao Y, Huang R, Pan X. Disclosing the development and focus of sequencing and omics studies in kidney neoplasm research. Discov Oncol 2025; 16:928. [PMID: 40418308 DOI: 10.1007/s12672-025-02750-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Kidney cancer is a worldwide prevalent urological malignancy and the leading cause of death. Sequencing and omics studies play a crucial role in unraveling its molecular mechanisms and diagnostic, prognostic, and therapeutic relevance. This study aims to offer a comprehensive review of the evolving trends and hotspots of sequencing and omics studies in kidney neoplasms. METHODS We conducted the retrieval of scientific publications on sequencing and omics studies in kidney neoplasms from the Web of Science Core Collection (WoSCC) on July 3, 2023. The R-based bibliometrix package, VOSviewer, and CiteSpace were utilized to conduct the holistic bibliometric analysis to obtain objective and data-driven results. A comprehensive consultation of papers was then proceeded for an in-depth review. RESULTS Our investigation yielded a dataset containing 1260 records from 509 sources, with 43,404 references, from 1960 to 2023. Publication and citation frequencies have been consistently growing. In country analysis, China and the USA led the research, displaying substantial collaboration. Notable contributors like TEH BT, SAUTER G, and FUTREAL PA shaped this research landscape. Key journals such as PLoS One, Cancer Research, and New England Journal of Medicine actively participated in and significantly influenced this field. Distinguished publications and references were also revealed, along with their historical citation and co-citation relationships. A panel of keywords, including RCC, biomarker, and multi-omics data were identified and clustered. CONCLUSION We obtained a profound understanding of the developing trends and hotspots of research investigating sequencing and omics studies in kidney neoplasms. Specifically, we have highlighted three hotspots: "explore molecular mechanisms of RCC pathogenesis, progression, and metastasis", "identify molecular biomarkers of RCC for diagnosis, prognosis, and therapeutics", "investigate tumor heterogeneity and tailor personalized therapeutic strategies for RCC". Hopefully, our study will serve as a valuable reference for scientific researchers and clinical practitioners.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jinming Cui
- Ulink College of Shanghai, Shanghai, 201615, China
| | - Tianyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Donghao Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuanan Li
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
3
|
Sohel HI, Zahan UF, Kiyono T, Ishikawa M, Razia S, Kanno K, Yamashita H, Sonia SB, Nakayama K, Kyo S. Establishment of Two Novel Ovarian Tumor Cell Lines with Characteristics of Mucinous Borderline Tumors or Dedifferentiated Carcinoma-Implications for Tumor Heterogeneity and the Complex Carcinogenesis of Mucinous Tumors. Cancers (Basel) 2025; 17:1716. [PMID: 40427213 PMCID: PMC12110342 DOI: 10.3390/cancers17101716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND/OBJECTIVE Mucinous borderline tumors of the ovary (MBOTs) are characterized by their unique histological features and intermediate malignant potential; however, the factors underlying their molecular carcinogenesis and tumor biology remain largely unknown. Developing cell lines from these tumors presents an ongoing challenge. The purpose of this study is to establish MBOT cell lines and characterize their biological features. METHODS Epithelial cells were collected and purified from surgically removed MBOT samples and then stably maintained with an extended life span by overexpressing CyclinD1/CDK4 in combination with human telomerase reverse transcriptase. The characterization of resulting cell lines was defined by morphology, growth kinetics, functional analysis, whole-exome sequencing, and tumorigenicity in mice. RESULTS Two independent cell lines, HMucBOT-1 and HMucBOT-2, were successfully established from the tissues of a patient with an MBOT, with the latter showing more aggressive growth capacity. In the patient-derived xenograft model, HMucBOT-1 cells retained the original morphological characteristics of the MBOT, whereas HMucBOT-2 cells displayed a transition to mucinous carcinoma accompanying undifferentiated carcinoma, suggestive of dedifferentiated carcinoma. Genetic analysis of the original tumor sample and HMucBOT-2 cells revealed shared oncogenic mutations. However, KRAS amplification and certain copy number alterations were uniquely observed in the HMucBOT-2 cells. CONCLUSIONS The above results indicate that HMucBOT-1 can serve as a preclinical model for investigating the biological behavior of and potential targeted therapies for human MBOTs, with HMucBOT-2 serving as a valuable tool for studying the heterogeneity and genetic diversity of this tumor and explaining the potential causes of treatment failure or relapse.
Collapse
Affiliation(s)
- Hasibul Islam Sohel
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Umme Farzana Zahan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Tohru Kiyono
- Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Kashiwa 277-8577, Japan;
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Sultana Razia
- Department of Legal Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan;
| | - Kosuke Kanno
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Hitomi Yamashita
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Shahataj Begum Sonia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| | - Kentaro Nakayama
- Department of Obstetrics and Gynecology, East Medical Center, Nagoya City University, Nagoya 464-8547, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan; (H.I.S.); (U.F.Z.); (M.I.); (K.K.); (H.Y.); (S.B.S.)
| |
Collapse
|
4
|
Takai Y, Naito S, Ito H, Horie S, Ushijima M, Narisawa T, Yagi M, Ichiyanagi O, Tsuchiya N. Ankrd1 Promotes Lamellipodia Formation and Cell Motility via Interaction with Talin-1 in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2025; 26:4232. [PMID: 40362467 PMCID: PMC12072362 DOI: 10.3390/ijms26094232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Ankyrin repeat domain 1 (Ankrd1), a transcriptional target of Yes-associated protein (YAP), is linked to cardiomyopathy. However, its role in cancer, particularly in clear cell renal cell carcinoma (ccRCC), remains vague. In this study, we examined the expression, regulation, and function of Ankrd1 in ccRCC. High Ankrd1 expression was related to poor prognosis in patients with ccRCC in The Cancer Genome Atlas cohort. Ankrd1 expression was regulated by YAP in all ccRCC cell lines examined and also by ERK5 in a subset of ccRCC cell lines. Moreover, silencing of Ankrd1 in ccRCC cell lines resulted in decreased cell motility, whereas its overexpression increased the cell motility. Ankrd1 colocalized with F-actin in lamellipodia upon phorbol ester stimulation. Ankrd1 silencing resulted in alterations in the shape of RCC cells and caused a decrease in lamellipodia formation. Ankrd1 also colocalized with talin-1 in lamellipodia. Ankrd1 depletion repressed talin-1-mediated activation of the integrin pathway. Immunohistochemical examination of surgical specimens revealed high expression of Ankrd1 in metastatic RCC tissues compared with that in primary RCC tissues from the same patients. Collectively, these findings suggest that Ankrd1 plays a critical role in the motility of ccRCC cells through lamellipodia formation.
Collapse
Affiliation(s)
- Yuki Takai
- Department of Urology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (S.N.); (H.I.); (S.H.); (M.U.); (T.N.); (M.Y.); (O.I.); (N.T.)
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Górka J, Miękus K. Molecular landscape of clear cell renal cell carcinoma: targeting the Wnt/β-catenin signaling pathway. Discov Oncol 2025; 16:524. [PMID: 40227498 PMCID: PMC11996749 DOI: 10.1007/s12672-025-02228-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma and is characterized by a complex molecular landscape driven by genetic and epigenetic alternations. Among the crucial signaling pathways implicated in ccRCC, the Wnt/β-catenin pathway plays a significant role in tumor progression and prognosis. This review delves into the molecular basis of ccRCC, highlighting the genetic and epigenetic modifications that contribute to its pathogenesis. We explore the significance of the Wnt/β-catenin pathway, focusing on its role in disease development, particularly the nuclear transport of β-catenin and its activation and downstream effects. Furthermore, we examine the role of antagonist genes in regulating this pathway within the context of ccRCC, providing insights into potential therapeutic targets. Dysregulation of this pathway, which is characterized by abnormal activation and nuclear translocation of β-catenin, plays a significant role in promoting tumor growth and metastasis. We explore the intricate molecular aspects of ccRCC, with a particular emphasis on this topic, underscoring the role of the pathway and emphasizing the importance and relevance of antagonist genes. Understanding the intricate interplay between these molecular mechanisms is crucial for developing innovative strategies to improve ccRCC treatment and patient outcomes.
Collapse
Affiliation(s)
- Judyta Górka
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Katarzyna Miękus
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| |
Collapse
|
6
|
Oki R, Takemura K, Urasaki T, Fujiwara R, Numao N, Yonese J, Miura Y, Yuasa T. Prevailing challenges in personalized treatment for metastatic renal cell carcinoma: a narrative review. Expert Rev Anticancer Ther 2025:1-13. [PMID: 40210604 DOI: 10.1080/14737140.2025.2491647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
INTRODUCTION The management of metastatic renal cell carcinoma (mRCC) has advanced with recent therapies, yet optimizing treatment remains challenging due to disease heterogeneity and the growing number of options. Integrating systemic and local treatments requires a multidisciplinary approach to improve outcomes. AREA COVERED This review summarizes recent developments in treatment for mRCC. Upfront immuno-oncology (IO)-based combinations have improved survival, though concerns about overtreatment and toxicity persist. While the role of cytoreductive nephrectomy (CN) has declined to some extent, it may still benefit well-selected patients. Metastasis-directed therapies, including metastasectomy and stereotactic radiotherapy, provide prognostic value, particularly for oligometastatic lesions or brain metastases. Comprehensive genomic profiling (CGP) holds promise for personalized treatment but is currently limited by the lack of actionable mutations and predictive biomarkers. EXPERT OPINION A personalized, multimodal approach is essential for optimizing mRCC management. Careful patient selection is key to balancing the benefits of treatment with the risks of toxicity. While CN and metastasis-directed therapies remain useful in select cases, advancing individualized care requires the development of validated biomarkers and broader application of CGP.
Collapse
Affiliation(s)
- Ryosuke Oki
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kosuke Takemura
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuya Urasaki
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Fujiwara
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noboru Numao
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuji Miura
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Yuasa
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
7
|
Lucain M, Vitobello A, Sadikovic B, Albuisson J, Gaudillat L, Chevarin M, Maraval J, Thauvin-Robinet C, Kerkhof J, Philippe C, Nambot S, Faivre L. Abnormal DNA Methylation Profile Suggests the Extension of the Clinical Spectrum of the SETD2-Related Disorders to a Syndromic Multiple Tumor Phenotype. Am J Med Genet A 2025:e64043. [PMID: 40104911 DOI: 10.1002/ajmg.a.64043] [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: 01/10/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
SETD2 has an essential role in epigenetic regulation. SETD2 pathogenic variants cause neurodevelopmental disorders (SETD2-NDDs) that most commonly include various degrees of intellectual disability and behavioral disorders, macrocephaly, brain malformations, and generalized overgrowth. A distinctive DNA methylation episignature has been identified for Luscan-Lumish syndrome. A less common phenotype, denoted SETD2-NDD with multiple congenital anomalies, failure to thrive, and profound intellectual disability, has been reported in association with a particular pathogenic variant (p.Arg1740Trp). To date, about 50 patients have been described in the literature with SETD2 causative variants. We report here an individual with a phenotype distinct from SETD2-NDDs, including normal cognition, distinctive facial features, and multiple tumor histories, including a sacral osteoblastoma at age 7, a benign femoral bone tumor at age 17, a peritoneal pseudomyxoma at age 27, and a hypophyseal macroadenoma and a low-grade optochiasmatic glioma at age 37 years. Trio exome sequencing identified a de novo heterozygous missense variant of unknown significance (p.Ser1658Leu) in the SETD2 gene. DNA methylation study by EpiSign assay confirmed the presence of an episignature profile compatible with SETD2-related disorders. Given the implication of somatic SETD2 variants in benign and malignant tumors, the implication of these SETD2 constitutional variants in tumorigenesis is discussed.
Collapse
Affiliation(s)
- Marie Lucain
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
| | - Antonio Vitobello
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, Canada, Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | | | - Léa Gaudillat
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Martin Chevarin
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Julien Maraval
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Référence Maladies Rares "Déficiences Intellectuelles de Causes Rares", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, Canada, Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Christophe Philippe
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
| | - Sophie Nambot
- Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, CHU Dijon-Bourgogne, Dijon, France
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Référence Maladies Rares "Déficiences Intellectuelles de Causes Rares", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 GAD, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Génétique, Centre de Référence Maladies Rares "Anomalies du Développement et Syndromes Malformatifs", FHU TRANSLAD et Institut GIMI, CHU Dijon Bourgogne, Dijon, France
| |
Collapse
|
8
|
Pham DX, Hsu T. Tumor-initiating and metastasis-initiating cells of clear-cell renal cell carcinoma. J Biomed Sci 2025; 32:17. [PMID: 39920694 PMCID: PMC11806631 DOI: 10.1186/s12929-024-01111-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common subtype of kidney malignancy. ccRCC is considered a major health concern worldwide because its numbers of incidences and deaths continue to rise and are predicted to continue rising in the foreseeable future. Therefore new strategy for early diagnosis and therapeutics for this disease is urgently needed. The discovery of cancer stem cells (CSCs) offers hope for early cancer detection and treatment. However, there has been no definitive identification of these cancer progenitors for ccRCC. A majority of ccRCC is characterized by the loss of the von Hippel-Lindau (VHL) tumor suppressor gene function. Recent advances in genome analyses of ccRCC indicate that in ccRCC, tumor-initiating cells (TICs) and metastasis-initiating cells (MICs) are two distinct groups of progenitors. MICs result from various genetic changes during subclonal evolution, while TICs reside in the stem of the ccRCC phylogenetic tree of clonal development. TICs likely originate from kidney tubule progenitor cells bearing VHL gene inactivation, including chromatin 3p loss. Recent studies also point to the importance of microenvironment reconstituted by the VHL-deficient kidney tubule cells in promoting ccRCC initiation and progression. These understandings should help define the progenitors of ccRCC and facilitate early detection and treatment of this disease.
Collapse
Affiliation(s)
- Dinh-Xuan Pham
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan, ROC
| | - Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan, ROC.
- Graduate Institute of Biomedical Sciences, China Medical University-Taiwan, No. 91 Hsueh-Shih Road, Taichung, 40402, Taiwan, ROC.
| |
Collapse
|
9
|
Wagner RT, Hlady RA, Pan X, Wang L, Kim S, Zhao X, El Khoury LY, Shaikh S, Zhong J, Lee JH, Grembecka J, Cierpicki T, Ho TH, Robertson KD. SETD2 loss-of-function uniquely sensitizes cells to epigenetic targeting of NSD1-directed H3K36 methylation. Genome Biol 2025; 26:22. [PMID: 39910618 PMCID: PMC11800516 DOI: 10.1186/s13059-025-03483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND SETD2 is the sole epigenetic factor responsible for catalyzing histone 3, lysine 36, tri-methylation (H3K36me3) in mammals. Its role in regulating cellular processes such as RNA splicing, DNA repair, and spurious transcription initiation underlies its broader tumor suppressor function. SETD2 mutation promotes the epithelial-mesenchymal transition and is clinically associated with adverse outcomes highlighting a therapeutic need to develop targeted therapies against this dangerous mutation. RESULTS We employ an unbiased genome-wide synthetic lethal screen, which identifies another H3K36me writer, NSD1, as a synthetic lethal modifier in SETD2-mutant cells. Confirmation of this synthetic lethal interaction is performed in isogenic clear cell renal cell carcinoma and immortalized renal epithelial cell lines, in mouse and human backgrounds. Depletion of NSD1 using a CRISPRi targeting approach promotes the loss of SETD2-mutant cells coincident with elevated levels of DNA damage and apoptosis. Surprisingly, only suppression of NSD1, but not related H3K36-methyltransferases, promotes synthetic lethality in these models. Mapping of genomic H3K36me2 targeting by NSD1 and NSD2 individually highlights the independent functions of these epigenetic writers. Furthermore, as a proof-of-principle, we demonstrate the therapeutic feasibility of targeting this synthetic lethal interaction by recapitulating the phenotype using BT5, a first-in-class pharmacologic inhibitor against NSD1. CONCLUSIONS These findings unify genome-wide screening approaches with the latest genetic and pharmacologic modeling methodologies to reveal an entirely novel epigenetic approach to individualize therapies against a challenging loss-of-function SETD2 mutation in cancer.
Collapse
Affiliation(s)
- Ryan T Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Ryan A Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Xiaoyu Pan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sungho Kim
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Louis Y El Khoury
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA
| | - Shafiq Shaikh
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jian Zhong
- Epigenomics Development Laboratory, Mayo Clinic, CIM Epigenomics Program, Rochester, MN, USA
| | - Jeong-Heon Lee
- Epigenomics Development Laboratory, Mayo Clinic, CIM Epigenomics Program, Rochester, MN, USA
| | | | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Thai H Ho
- Division of Hematology and Oncology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Stabile 12-70, Rochester, MN, 55905, USA.
| |
Collapse
|
10
|
Manoukian P, Kuhnen LC, van Laarhoven HWM, Bijlsma MF. Association of epigenetic landscapes with heterogeneity and plasticity in pancreatic cancer. Crit Rev Oncol Hematol 2025; 206:104573. [PMID: 39581245 DOI: 10.1016/j.critrevonc.2024.104573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Due to a lack of clear symptoms, patients often present with advanced disease, with limited clinical intervention options. The high mortality rate of PDAC is, however, also a result of several other factors that include a high degree of heterogeneity and treatment resistant cellular phenotypes. Molecular subtypes of PDAC have been identified that are thought to represent cellular phenotypes at the tissue level. The epigenetic landscape is an important factor that dictates these subtypes. Permissive epigenetic landscapes serve as drivers of molecular heterogeneity and cellular plasticity in developing crypts as well as metaplastic lesions. Drawing parallels with other cancers, we hypothesize that epigenetic permissiveness is a potential driver of cellular plasticity in PDAC. In this review will explore the epigenetic alterations that underlie PDAC cell states and relate them to cellular plasticity from other contexts. In doing so, we aim to highlight epigenomic drivers of PDAC heterogeneity and plasticity and, with that, offer some insight to guide pre-clinical research.
Collapse
Affiliation(s)
- Paul Manoukian
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - Leo C Kuhnen
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Cesana B, Cochet C, Filhol O. New players in the landscape of renal cell carcinoma bone metastasis and therapeutic opportunities. Int J Cancer 2025; 156:475-487. [PMID: 39306698 PMCID: PMC11622000 DOI: 10.1002/ijc.35181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 12/07/2024]
Abstract
Approximately one-third of advanced renal cell carcinoma (RCC) patients develop osteolytic bone metastases, leading to skeletal complications. In this review, we first provide a comprehensive perspective of seminal studies on bone metastasis of RCC describing the main molecular modulators and growth factor signaling pathways most important for the RCC-stimulated osteoclast-mediated bone destruction. We next focus on newer developments revealing with in-depth details, the bidirectional interplay between renal cancer cells and the immune and stromal microenvironment that can through epigenetic reprogramming, profoundly affect the behaviors of transformed cells. Understanding their mechanistic interactions is of paramount importance for advancing both fundamental and translational research. These new investigations into the landscape of RCC-bone metastasis offer novel insights and identify potential avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Beatrice Cesana
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Claude Cochet
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Odile Filhol
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| |
Collapse
|
12
|
Jasmin MDC, Radhakrishnan N, Prabhu VV, Sakthivel KM, Radha RR. Histone-Lysine N-Methyltransferase 2D (KMT2D) Impending Therapeutic Target for the Management of Cancer: The Giant Rats Tail. J Environ Pathol Toxicol Oncol 2025; 44:31-36. [PMID: 39462447 DOI: 10.1615/jenvironpatholtoxicoloncol.2024053872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
The histone-lysine N-methyltransferase 2D (KMT2D), tumor suppressor gene which is the major component of histone H3K4 mono-methyltransferase in mammals and has significant role in regulation of a gene which are frequently mutated that lead to many different types of cancers that include non-Hodgkin lymphoma, medulloblastoma, prostate carcinoma, renal carcinoma, bladder carcinoma and lung carcinoma. KMT2D gene epigenetic alterations in histone methylation play a significant role for the initiation and progression of cancers from pre-cancerous lesions, yet its complete function in oncogenesis remains unsolved. KMT2D deficiency - loss are thought of initial mediators of cancer development and cell migration such as B-cell lymphoma, medulloblastoma, melanoma, pancreas and lung cancer. The KMT2D loss has know to activate glycolytic genes that promote aggressive tumor progression. Therefore, the present review serves to underline the update on recent research pertaining to KMT2D gene, that could be a potential therapeutic target in downregulating glycolytic genes such as Pgk1, Ldha, Pgam1 and Gapdh; 2, epidermal growth factor receptor tyrosine kinase (EGFR-TK ) - ERBB2, RTK-RAS signaling, RAS activator genes Rgl1, Rasgrp1, Rasgrf1, Rasgrf 2 and Rapgef5 in suppressing the tumor progression that may represent novel targeted therapy for the management of cancer. This review will facilitate to understand the gene expression that inhibits cancer progression and which could serve as a potential molecular target in understanding cancer pathogenesis.
Collapse
Affiliation(s)
- Meshak Dhanashekaran Cecileya Jasmin
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Thandalam, Chennai, Tamil Nadu 602105, India; Department of Biochemistry, Tagore Medical College and Hospital, Rathinamangalam, Melakottaiyur, Chennai, Tamil Nadu 600127, India
| | | | - Venugopal Vinod Prabhu
- Department of Biochemistry, University of Madras, Guindy campus, Chennai-600025, Tamil Nadu, India
| | - Kunnathur Murugesan Sakthivel
- Karunya University, Department of Biotechnology, Karunya Nagar, Coimbatore-641114, Tamil Nadu, India; Laboratory of Cytogenetics and Molecular Diagnostics, Regional Cancer Centre, Medical College Post, Trivandrum 695011, Kerala, India
| | - Rasmi Rajan Radha
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, Tamil Nadu 614014, India
| |
Collapse
|
13
|
Nguyen AL, Facey COB, Boman BM. The Significance of Aldehyde Dehydrogenase 1 in Cancers. Int J Mol Sci 2024; 26:251. [PMID: 39796106 PMCID: PMC11720537 DOI: 10.3390/ijms26010251] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The goal of this paper is to discuss the role of ALDH isozymes in different cancers, review advances in ALDH1-targeting cancer therapies, and explore a mechanism that explains how ALDH expression becomes elevated during cancer development. ALDH is often overexpressed in cancer, and each isoform has a unique expression pattern and a distinct role in different cancers. The abnormal expression of ALDHs in different cancer types (breast, colorectal, lung, gastric, cervical, melanoma, prostate, and renal) is presented and correlated with patient prognosis. ALDH plays a significant role in various cellular functions, such as metabolism, oxidative stress response, detoxification, and cellular differentiation. Among the ALDH families, ALDH1 has gained considerable attention as a cancer stem cell (CSC) marker due to its significant role in the maintenance of stemness and the differentiation of stem cells (SCs), along with its involvement in tumorigenesis. A description of the cellular mechanisms and physiology of ALDH1 that underlies cancer development is provided. Moreover, current advances in ALDH1-targeting cancer therapies are discussed.
Collapse
Affiliation(s)
- Anh L. Nguyen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
| | - Bruce M. Boman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA;
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
14
|
Shen XH, Xiong SP, Wang SP, Lu S, Wan YY, Zhang HQ. Mutation on JmjC domain of UTX impaired its antitumor effects in pancreatic cancer via inhibiting G0S2 expression and activating the Toll-like signaling pathway. Mol Med 2024; 30:258. [PMID: 39707168 DOI: 10.1186/s10020-024-01023-2] [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: 08/02/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Recently, the incidence of pancreatic cancer (PC) has gradually increased. Research has shown that UTX mutants are critical in tumors. However, the underlying mechanisms remain incompletely understood. This study aimed to explore how UTX mutation would affect its related function in PC. METHOD Exome sequencing was used to analyze PC samples. MTT, transwell, and colony formation assays were performed to determine the cellular functions of PC cells. qRT-PCR, Western Blot, TUNEL, immunohistochemistry, CHIP, bioinformatics, and xenograft experiments were used to investigate the mechanism of UTX mutants in PC in vitro and in vivo. RESULTS We compared exome sequencing data from 12 PC samples and found a UTX missense mutation on the JmjC structure. Through cellular functions and xenograft experiments, wild-type UTX was found to significantly inhibit PC malignant progression in vitro and in vivo, while UTX mutation notably impaired this effect. Furthermore, G0S2 was identified as the key target gene for UTX, and wild-type UTX significantly increased its expression, while mutant one lost this function to a certain extent both in vitro and in vivo. More importantly, G0S2 overexpression not only inhibited tumor malignant phenotype and drug resistance for Gemcitabine in PC but also effectively reversed the roles of UTX mutant with Toll-like signaling pathway involved. In terms of mechanism, UTX mutation elevated the H3K27me3 modification level of the G0S2 promoter, which decreased its expression in PC cells. CONCLUSION In conclusion, UTX mutant weakened the antitumor effect of wild-type UTX in PC by inhibiting G0S2 expression and activating the Toll-like signaling pathway.
Collapse
Affiliation(s)
- Xiao-Hua Shen
- Department of Gastrointestinal Medical Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, People's Republic of China
| | - Shu-Ping Xiong
- The Second Department of Radiotherapy, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, People's Republic of China
| | - Sheng-Peng Wang
- The Medical College of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Shan Lu
- Department of Gastrointestinal Medical Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, People's Republic of China
| | - Yi-Ye Wan
- Department of Gastrointestinal Medical Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, People's Republic of China
| | - Hui-Qing Zhang
- Department of Gastrointestinal Medical Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, People's Republic of China.
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, 330029, People's Republic of China.
| |
Collapse
|
15
|
Camp SY, He MX, Cuoco MS, Saad E, Pimenta E, Meli K, Bakouny Z, Labaki C, Titchen BM, Kang YJ, Horst J, Trowbridge R, Shannon E, Helvie K, Thorner AR, Vigneau S, Mayorga A, Kodali J, Lachmayr H, Bemus M, Park J, Choueiri T, Bi K, Van Allen EM. Single-cell epigenetic profiling reveals an interferon response-high program associated with BAP1 deficiency in kidney cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623837. [PMID: 39605676 PMCID: PMC11601305 DOI: 10.1101/2024.11.15.623837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Renal cell carcinoma (RCC) is characterized by recurrent somatic mutations in epigenetic regulators, which stratify patients into clinically significant subgroups with distinct prognoses and treatment responses. However, the cell type-specific epigenetic landscape of RCC-broadly and in the context of these mutations-is incompletely understood. To investigate these open questions, we integrated single nucleus ATAC sequencing data from RCC tumors across four independent cohorts. In clear cell RCC tumors, we identified four shared malignant epigenetic programs related to angiogenesis, proximal tubule-like features, interferon (IFN) signaling, and one that lacked distinct genomic regions with increased accessibility. Among the mutated epigenetic regulators, BAP1 mutation exhibited the most significant impact on chromatin accessibility in tumor cells, and the associated epigenetic changes were linked to IFN response. We identify multiple potential sources of elevated IFN signaling in these lesions, such as increased immune infiltration and increased accessibility and expression of an IFN-associated ERV, ERV3-16A3_LTR. We find that the expression of ERV3-16A3_LTR may itself be a negative prognostic biomarker in ccRCC. Our findings highlight the convergence of malignant epigenetic programs across ccRCC tumors and suggest that BAP1 loss, potentially through ERV3-16A3_LTR dysregulation, is associated with an IFN response-high epigenetic program.
Collapse
Affiliation(s)
- Sabrina Y. Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Boston, MA 02115, USA
| | - Michael S. Cuoco
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA 92093, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Erica Pimenta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Breanna M. Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Yun Jee Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jack Horst
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Rachel Trowbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Karla Helvie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Aaron R. Thorner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sébastien Vigneau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Angie Mayorga
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jahnavi Kodali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hannah Lachmayr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Meredith Bemus
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
16
|
Lu Y, Chen W, Xuan Y, Li X, Wu S, Wang H, Guo T, Wang C, Tian S, Li H, Lai D, Zhao W, Huang X, Zhao X, Wang B, Zhang X, Li H, Huang Y, Ma X. ATF4/NUPR1 axis promotes cancer cell survival and mediates immunosuppression in clear cell renal cell carcinoma. Discov Oncol 2024; 15:607. [PMID: 39480570 PMCID: PMC11528094 DOI: 10.1007/s12672-024-01485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Cancer cells encounter unavoidable stress during tumor growth. The stress-induced transcription factor, activating transcription factor 4 (ATF4), has been reported to upregulate various adaptive genes involved in salvage pathways to alleviate stress and promote tumor progression. However, this effect is unknown in clear cell renal cell carcinoma (ccRCC). In this study, we found that ATF4 expression was remarkably upregulated in tumor tissues and associated with poor ccRCC outcomes. ATF4 depletion significantly impaired ccRCC cell proliferation, migration, and invasion in vitro and in vivo by inhibiting the AKT/mTOR and epithelial-mesenchymal transition (EMT)-related signaling pathway. RNA sequencing and functional studies identified nuclear protein 1 (NUPR1) as a key downstream target of ATF4 for repressing ferroptosis and promoting ccRCC cell survival. In addition, targeting ATF4 or pharmacological inhibition using NUPR1 inhibitor ZZW115 promoted antitumor immunity in syngeneic graft mouse models, represented by increased infiltration of CD4+ and CD8+ T cells. Furthermore, ZZW115 could improve the response to the PD-1 immune checkpoint blockade. The results demonstrate that the ATF4/NUPR1 signaling axis promotes ccRCC survival and facilitates tumor-mediated immunosuppression, providing a set of potential targets and prognostic indicators for ccRCC patients.
Collapse
Affiliation(s)
- Yongliang Lu
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Weihao Chen
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Yundong Xuan
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Xiubin Li
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Shengpan Wu
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Hanfeng Wang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Tao Guo
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Chenfeng Wang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Shuo Tian
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Huaikang Li
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Dong Lai
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Wenlei Zhao
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Xing Huang
- People's Liberation Army Postgraduate Medical School, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Xupeng Zhao
- School of Medicine, Nankai University, Weijin Road 94, Tianjin, 300071, China
| | - Baojun Wang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Xu Zhang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Hongzhao Li
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| | - Yan Huang
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| | - Xin Ma
- State Key Laboratory of Kidney Diseases, Senior Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| |
Collapse
|
17
|
Correa AF, Kalashnikova E, Wu HT, Winters RM, Balcioglu M, Sudhaman S, Connolly DC, Gong Y, Uzzo RG, Sethi H, ElNaggar AC, Aleshin A, Liu MC, Abbosh PH. Association of circulating tumor DNA with patient prognosis in surgically resected renal cell carcinoma. Oncologist 2024; 29:887-893. [PMID: 39013784 PMCID: PMC11449105 DOI: 10.1093/oncolo/oyae180] [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: 02/20/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Despite complete resection, 20%-50% of patients with localized renal cell carcinoma (RCC) experience recurrence within 5 years. Accurate assessment of prognosis in high-risk patients would aid in improving outcomes. Here we evaluate the use of circulating tumor DNA (ctDNA) in RCC using banked samples and clinical data from a single institution. METHODS The cohort consisted of 45 RCC patients (≥pT1b) who underwent complete resection. The presence of ctDNA in plasma was determined using a personalized, tumor-informed ctDNA assay (Signatera RUO, Natera, Inc.). Relationships with outcomes and other relevant clinical variables were assessed. The median follow-up was 62 months. RESULTS Plasma ctDNA was detected in 18 out of 36 patients (50%) pre-operatively and was associated with increased tumor size (mean 9.3 cm vs. 7.0 cm, P < .05) and high Fuhrman grade (60% grades III-IV vs 27% grade II, P = .07). The presence of ctDNA, either pre-operatively or at any time post-operatively, was associated with inferior relapse-free survival (HR = 2.70, P = .046; HR = 3.23, P = .003, respectively). Among patients who were ctDNA positive at any time point, the sensitivity of relapse prediction was 84% with a PPV of 90%. Of note, ctDNA positivity at a post-surgical time point revealed a PPV of 100% and NPV of 64%. The lack of ctDNA detection at both time points yielded an NPV of 80%. CONCLUSIONS Detection of plasma ctDNA using a personalized assay is prognostic of recurrence in patients with resected RCC. Herein, we describe a successful approach for its application and identify potential limitations to be addressed in future studies.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/surgery
- Carcinoma, Renal Cell/blood
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/mortality
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Female
- Male
- Prognosis
- Middle Aged
- Aged
- Kidney Neoplasms/surgery
- Kidney Neoplasms/blood
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/mortality
- Adult
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/genetics
- Aged, 80 and over
Collapse
Affiliation(s)
- Andres F Correa
- Department of Urology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | - Ryan M Winters
- Biosample Repository Facility, Fox Chase Cancer Center, Philadelphia, PA, United States
- WuXi Advanced Therapies, Cheltenham, PA, United States
| | | | | | - Denise C Connolly
- Biosample Repository Facility, Fox Chase Cancer Center, Philadelphia, PA, United States
- Nuclear Dynamics and Cancer, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Yulan Gong
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Robert G Uzzo
- Department of Urology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | | | | | - Philip H Abbosh
- Nuclear Dynamics and Cancer, Fox Chase Cancer Center, Philadelphia, PA, United States
- Department of Urology, Albert Einstein Medical Center, Philadelphia, PA, United States
| |
Collapse
|
18
|
Liu Y, Li H. Epigenetic modifiers: catalytic or noncatalytic, that is the question. Front Med 2024; 18:941-943. [PMID: 39333446 DOI: 10.1007/s11684-024-1104-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 09/29/2024]
Affiliation(s)
- Yimin Liu
- State Key Laboratory of Molecular Oncology, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Haitao Li
- State Key Laboratory of Molecular Oncology, Beijing Frontier Research Center for Biological Structure, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, 030001, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| |
Collapse
|
19
|
Chen S, Liu D, Chen B, Li Z, Chang B, Xu C, Li N, Feng C, Hu X, Wang W, Zhang Y, Xie Y, Huang Q, Wang Y, Nimer SD, Chen S, Chen Z, Wang L, Sun X. Catalytic activity of Setd2 is essential for embryonic development in mice: establishment of a mouse model harboring patient-derived Setd2 mutation. Front Med 2024; 18:831-849. [PMID: 39115793 DOI: 10.1007/s11684-024-1095-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/08/2024] [Indexed: 11/01/2024]
Abstract
SETD2 is the only enzyme responsible for transcription-coupled histone H3 lysine 36 trimethylation (H3K36me3). Mutations in SETD2 cause human diseases including cancer and developmental defects. In mice, Setd2 is essential for embryonic vascular remodeling. Given that many epigenetic modifiers have recently been found to possess noncatalytic functions, it is unknown whether the major function(s) of Setd2 is dependent on its catalytic activity or not. Here, we established a site-specific knockin mouse model harboring a cancer patient-derived catalytically dead Setd2 (Setd2-CD). We found that the essentiality of Setd2 in mouse development is dependent on its methyltransferase activity, as the Setd2CD/CD and Setd2-/- mice showed similar embryonic lethal phenotypes and largely comparable gene expression patterns. However, compared with Setd2-/-, the Setd2CD/CD mice showed less severe defects in allantois development, and single-cell RNA-seq analysis revealed differentially regulated allantois-specific 5' Hoxa cluster genes in these two models. Collectively, this study clarifies the importance of Setd2 catalytic activity in mouse development and provides a new model for comparative study of previously unrecognized Setd2 functions.
Collapse
Affiliation(s)
- Shubei Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Dianjia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Bingyi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zijuan Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Binhe Chang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chunhui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ningzhe Li
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Changzhou Feng
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Clinical Laboratory, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China
| | - Xibo Hu
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Weiying Wang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yuanliang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qiuhua Huang
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yingcai Wang
- Sylvester Comprehensive Cancer Center and Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center and Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiaojian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Omics and Diseases, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, and School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
20
|
Mondal V, Higgins PJ, Samarakoon R. Emerging Role of Hippo-YAP (Yes-Associated Protein)/TAZ (Transcriptional Coactivator with PDZ-Binding Motif) Pathway Dysregulation in Renal Cell Carcinoma Progression. Cancers (Basel) 2024; 16:2758. [PMID: 39123485 PMCID: PMC11312123 DOI: 10.3390/cancers16152758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Although Hippo-YAP/TAZ pathway involvement has been extensively studied in the development of certain cancers, the involvement of this cascade in kidney cancer progression is not well-established and, therefore, will be the focus of this review. Renal cell carcinoma (RCC), the most prevalent kidney tumor subtype, has a poor prognosis and a high mortality rate. Core Hippo signaling inactivation (e.g., LATS kinases) leads to the nuclear translocation of YAP/TAZ where they bind to co-transcriptional factors such as TEAD promoting transcription of genes which initiates various fibrotic and neoplastic diseases. Loss of expression of LATS1/2 kinase and activation of YAP/TAZ correlates with poor survival in RCC patients. Renal-specific ablation of LATS1 in mice leads to the spontaneous development of several subtypes of RCC in a YAP/TAZ-dependent manner. Genetic and pharmacological inactivation of YAP/TAZ reverses the oncogenic potential in LATS1-deficient mice, highlighting the therapeutic benefit of network targeting in RCC. Here, we explore the unique upstream controls and downstream consequences of the Hippo-YAP/TAZ pathway deregulation in renal cancer. This review critically evaluates the current literature on the role of the Hippo pathway in RCC progression and highlights the recent scientific evidence designating YAP/TAZ as novel therapeutic targets against kidney cancer.
Collapse
Affiliation(s)
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208-3479, USA;
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208-3479, USA;
| |
Collapse
|
21
|
Tong T, Huang M, Yan B, Lin B, Yu J, Teng Q, Li P, Pang J. Hippo signaling modulation and its biological implications in urological malignancies. Mol Aspects Med 2024; 98:101280. [PMID: 38870717 DOI: 10.1016/j.mam.2024.101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/15/2024]
Abstract
Although cancer diagnosis and treatment have rapidly advanced in recent decades, urological malignancies, which have high morbidity and mortality rates, are among the most difficult diseases to treat. The Hippo signaling is an evolutionarily conserved pathway in organ size control and tissue homeostasis maintenance. Its downstream effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are key modulators of numerous physiological and pathological processes. Recent work clearly indicates that Hippo signaling is frequently altered in human urological malignancies. In this review, we discuss the disparate viewpoints on the upstream regulators of YAP/TAZ and their downstream targets and systematically summarize the biological implications. More importantly, we highlight the molecular mechanisms involved in Hippo-YAP signaling to improve our understanding of its role in every stage of prostate cancer, bladder cancer and kidney cancer progression. A better understanding of the biological outcomes of YAP/TAZ modulation will contribute to the establishment of future therapeutic approaches.
Collapse
Affiliation(s)
- Tongyu Tong
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Mengjun Huang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Binyuan Yan
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Bingbiao Lin
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, Guangdong, 515041, China
| | - Jiaying Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Qiliang Teng
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Jun Pang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
22
|
Zhang H, Huang W, Chen M, Liu Y, Yan B, Mou S, Jiang W, Mei H. Research on molecular characteristics of ADME-related genes in kidney renal clear cell carcinoma. Sci Rep 2024; 14:16834. [PMID: 39039118 PMCID: PMC11263354 DOI: 10.1038/s41598-024-67516-6] [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: 02/17/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024] Open
Abstract
Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) are named ADME genes. However, the comprehensive role of ADME genes in kidney renal clear cell carcinoma (KIRC) remains unclear. Using the clinical and gene expression data of KIRC patients downloaded from The Cancer Genome Atlas (TCGA), ArrayExpress, and the Gene Expression Omnibus (GEO) databases, we cluster patients into two patterns, and the population with a relatively poor prognosis demonstrated higher level of immunosuppressive cell infiltration and higher proportion of glycolytic subtypes. Then, 17 ADME genes combination identified through the least absolute shrinkage and selection operator algorithm (LASSO, 1000 times) was utilized to calculate the ADME score. The ADME score was found to be an independent predictor of prognosis in KIRC and to be tightly associated with the infiltration level of immune cells, metabolic properties, tumor-related signaling pathways, genetic variation, and responses to chemotherapeutics. Our work revealed the characteristics of ADME in KIRC. Assessing the ADME profiles of individual patients can deepen our comprehension of tumor microenvironment (TME) features in KIRC and can aid in developing more personalized and effective therapeutic strategies.
Collapse
Affiliation(s)
- Haiyu Zhang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weisheng Huang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mutong Chen
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuhan Liu
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bing Yan
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shuanzhu Mou
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wendong Jiang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
- Department of Urology, Shantou University Medical College, Shantou, China.
- Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
23
|
Isoda B, Kandori S, Sazuka T, Kojima T, Nitta S, Shiga M, Nagumo Y, Fujimoto A, Arai T, Sato H, Mathis BJ, Wu CL, Jan YH, Ichikawa T, Nishiyama H. TNFSF9 Is Associated with Favorable Tumor Immune Microenvironment in Patients with Renal Cell Carcinoma Who Are Treated with the Combination Therapy of Nivolumab and Ipilimumab. Int J Mol Sci 2024; 25:7444. [PMID: 39000552 PMCID: PMC11242552 DOI: 10.3390/ijms25137444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Combination therapy of nivolumab and ipilimumab (NIVO + IPI) for metastatic renal cell carcinoma (mRCC) has shown efficacy, but approximately 20% of patients experience disease progression in the early stages of treatment. No useful biomarkers have been reported to date. Therefore, it is desirable to identify biomarkers to predict treatment responses in advance. We examined the tumor microenvironment (TME)-related gene expression in mRCC patients treated with NIVO + IPI, between the response and non-response groups, using tumor tissues, before administering NIVO + IPI. In TME-related genes, TNFSF9 expression was identified as a candidate for the predictive biomarker. Its expression discriminated between the response and non-response groups with 88.89% sensitivity and 87.50% specificity (AUC = 0.9444). We further analyzed the roles of TNFSF9 in TME using bioinformatics from The Cancer Genome Atlas (TCGA) cohort. An adaptive immune response was activated in the TNFSF9-high-expression tumors. Indeed, T follicular helper cells, plasma B cells, and tumor-infiltrating CD8+ T cells were increased in the tumors, which indicates the promotion of humoral immunity due to enhanced T-B interactions. However, as the number of regulatory T cells (Treg) increased in the tumors, the percentage of dysfunctional T cells also increased. This suggests that not only PD-1 but also CTLA-4 inhibition may have suppressed Treg activation and improved the therapeutic effect in the TNFSF9 high-expression tumors. Therefore, TNFSF9 may predict the therapeutic efficacy of NIVO + IPI for mRCC and allow more appropriate patient selection.
Collapse
Affiliation(s)
- Bunpei Isoda
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| | - Shuya Kandori
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| | - Tomokazu Sazuka
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Chiba, Japan; (T.S.); (A.F.); (T.A.); (H.S.); (T.I.)
| | - Takahiro Kojima
- Department of Urology, Aichi Cancer Center, Nagoya 464-8681, Aichi, Japan;
| | - Satoshi Nitta
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| | - Masanobu Shiga
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| | - Yoshiyuki Nagumo
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| | - Ayumi Fujimoto
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Chiba, Japan; (T.S.); (A.F.); (T.A.); (H.S.); (T.I.)
| | - Takayuki Arai
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Chiba, Japan; (T.S.); (A.F.); (T.A.); (H.S.); (T.I.)
| | - Hiroaki Sato
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Chiba, Japan; (T.S.); (A.F.); (T.A.); (H.S.); (T.I.)
| | - Bryan J. Mathis
- International Medical Center, University of Tsukuba Affiliated Hospital, Tsukuba 305-8576, Ibaraki, Japan;
| | - Chia-Ling Wu
- ACT Genomics, Co., Ltd., Taipei 114, Taiwan; (C.-L.W.); (Y.-H.J.)
| | - Yi-Hua Jan
- ACT Genomics, Co., Ltd., Taipei 114, Taiwan; (C.-L.W.); (Y.-H.J.)
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Chiba, Japan; (T.S.); (A.F.); (T.A.); (H.S.); (T.I.)
| | - Hiroyuki Nishiyama
- Department of Urology, Institute of Medicine, University of Tsukuba, Tsukuba 305-8577, Ibaraki, Japan; (B.I.); (S.N.); (M.S.); (Y.N.); (H.N.)
| |
Collapse
|
24
|
Maldonado E, Rathmell WK, Shapiro GI, Takebe N, Rodon J, Mahalingam D, Trikalinos NA, Kalebasty AR, Parikh M, Boerner SA, Balido C, Krings G, Burns TF, Bergsland EK, Munster PN, Ashworth A, LoRusso P, Aggarwal RR. A Phase II Trial of the WEE1 Inhibitor Adavosertib in SETD2-Altered Advanced Solid Tumor Malignancies (NCI 10170). CANCER RESEARCH COMMUNICATIONS 2024; 4:1793-1801. [PMID: 38920407 PMCID: PMC11264598 DOI: 10.1158/2767-9764.crc-24-0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
UNLABELLED We sought to evaluate the efficacy of WEE1 inhibitor adavosertib in patients with solid tumor malignancies (cohort A) and clear cell renal cell carcinoma (ccRCC; cohort B). NCT03284385 was a parallel cohort, Simon two-stage, phase II study of adavosertib (300 mg QDAY by mouth on days 1-5 and 8-12 of each 21-day cycle) in patients with solid tumor malignancies harboring a pathogenic SETD2 mutation. The primary endpoint was the objective response rate. Correlative assays evaluated the loss of H3K36me3 by IHC, a downstream consequence of SETD2 loss, in archival tumor tissue. Eighteen patients were enrolled (9/cohort). The median age was 60 years (range 45-74). The median duration of treatment was 1.28 months (range 0-24+). No objective responses were observed in either cohort; accrual was halted following stage 1. Minor tumor regressions were observed in 4/18 (22%) evaluable patients. Stable disease (SD) was the best overall response in 10/18 (56%) patients, including three patients with SD > 4 months. One patient with ccRCC remains on treatment for >24 months. The most common adverse events of any grade were nausea (59%), anemia (41%), diarrhea (41%), and neutropenia (41%). Nine patients (50%) experienced a Grade ≥3 adverse event. Of eight evaluable archival tissue samples, six (75%) had a loss of H3K36me3 by IHC. Adavosertib failed to exhibit objective responses in SETD2-altered ccRCC and other solid tumor malignancies although prolonged SD was observed in a subset of patients. Combination approaches may yield greater depth of tumor response. SIGNIFICANCE WEE1 inhibition with adavosertib monotherapy demonstrated limited clinical activity in patients with SETD2-altered solid tumors despite compelling preclinical data indicating a synthetic lethal effect, which did not translate into robust tumor regression. Loss of the H3K36me3 trimethylation mark caused by SETD2-deficiency was confirmed in the majority of evaluable tumors. A subset of patients derived clinical benefit as manifested by minor tumor regressions and prolonged SD.
Collapse
Affiliation(s)
- Edward Maldonado
- University of California, San Francisco, San Francisco, California.
| | | | | | | | - Jordi Rodon
- Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, Texas.
| | | | | | | | - Mamta Parikh
- University of California, Davis, Davis, California.
| | | | - Celene Balido
- University of California, San Francisco, San Francisco, California.
| | - Gregor Krings
- University of California, San Francisco, San Francisco, California.
| | - Timothy F. Burns
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| | | | | | - Alan Ashworth
- University of California, San Francisco, San Francisco, California.
| | | | | |
Collapse
|
25
|
Li L, Hossain SM, Eccles MR. The Role of the PAX Genes in Renal Cell Carcinoma. Int J Mol Sci 2024; 25:6730. [PMID: 38928435 PMCID: PMC11203709 DOI: 10.3390/ijms25126730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Renal cell carcinoma (RCC) is a significant oncological challenge due to its heterogeneous nature and limited treatment options. The PAX developmental gene family encodes nine highly conserved transcription factors that play crucial roles in embryonic development and organogenesis, which have been implicated in the occurrence and development of RCC. This review explores the molecular landscape of RCC, with a specific focus on the role of the PAX gene family in RCC tumorigenesis and disease progression. Of the various RCC subtypes, clear cell renal cell carcinoma (ccRCC) is the most prevalent, characterized by the loss of the von Hippel-Lindau (VHL) tumor suppressor gene. Here, we review the published literature on the expression patterns and functional implications of PAX genes, particularly PAX2 and PAX8, in the three most common RCC subtypes, including ccRCC, papillary RCC (PRCC), and chromophobe RCC (ChRCC). Further, we review the interactions and potential biological mechanisms involving PAX genes and VHL loss in driving the pathogenesis of RCC, including the key signaling pathways mediated by VHL in ccRCC and associated mechanisms implicating PAX. Lastly, concurrent with our update regarding PAX gene research in RCC, we review and comment on the targeting of PAX towards the development of novel RCC therapies.
Collapse
Affiliation(s)
- Lei Li
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
| | - Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
26
|
Huang Y, Yang Z, Tang Y, Chen H, Liu T, Peng G, Huang X, He X, Mei M, Du C. Identification of a signature of histone modifiers in kidney renal clear cell carcinoma. Aging (Albany NY) 2024; 16:10489-10511. [PMID: 38888515 PMCID: PMC11236308 DOI: 10.18632/aging.205944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
Kidney renal clear cell carcinoma (KIRC) is a cancer that is closely associated with epigenetic alterations, and histone modifiers (HMs) are closely related to epigenetic regulation. Therefore, this study aimed to comprehensively explore the function and prognostic value of HMs-based signature in KIRC. HMs were first obtained from top journal. Then, the mRNA expression profiles and clinical information in KIRC samples were downloaded from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) datasets. Cox regression analysis and least absolute shrinkage and selection operator (Lasso) analysis were implemented to find prognosis-related HMs and construct a risk model related to the prognosis in KIRC. Kaplan-Meier analysis was used to determine prognostic differences between high- and low-risk groups. Immune infiltration and drug sensitivity analysis were also performed between high- and low-risk groups. Eventually, 8 HMs were successfully identified for the construction of a risk model in KIRC. The results of the correlation analysis between risk signature and the prognosis showed HMs-based signature has good prognostic value in KIRC. Results of immune analysis of risk models showed there were significant differences in the level of immune cell infiltration and expression of immune checkpoints between high- and low-risk groups. The results of the drug sensitivity analysis showed that the high-risk group was more sensitive to several chemotherapeutic agents such as Sunitinib, Tipifarnib, Nilotinib and Bosutinib than the low-risk group. In conclusion, we successfully constructed HMs-based prognostic signature that can predict the prognosis of KIRC.
Collapse
Affiliation(s)
- Yongming Huang
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Zhongsheng Yang
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Ying Tang
- Department of Day Ward, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hua Chen
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Tairong Liu
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Guanghua Peng
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Xin Huang
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Xiaolong He
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Ming Mei
- Department of Day Ward, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chuance Du
- Department of Urology, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| |
Collapse
|
27
|
Feng M, Chai C, Hao X, Lai X, Luo Y, Zhang H, Tang W, Gao N, Pan G, Liu X, Wang Y, Xiong W, Wu Q, Wang J. Inherited KDM6A A649T facilitates tumor-immune escape and exacerbates colorectal signet-ring cell carcinoma outcomes. Oncogene 2024; 43:1757-1768. [PMID: 38622203 DOI: 10.1038/s41388-024-03029-w] [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: 07/09/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
Childhood onset of colorectal signet-ring cell carcinoma (CR-SRCC) is extremely rare and featured as highly malignant with poor prognosis. Here we reported a CR-SRCC case of 11-year-old boy with a novel inherited X-linked KDM6AA694T mutation. The H3K27me3 demethylase KDM6A was frequently mutated in varieties of tumors and acts as a tumor suppressor. In vivo H3K27me3 demethylation assay demonstrated that KDM6AA694T had dampened H3K27me3 demethylase activity. Overexpression of KDM6AA694T in SRCC cell line KATO3 promoted cell proliferation, invasion and migration, which were further confirmed in vivo by constructing orthotopic tumor growth and lung metastasis model. Besides, expression of KDM6AA694T in immune cells suppresses inflammatory macrophage response and effector T cell response. In conclusion, we characterized a novel inherited KDM6AA694T mutant from a childhood-onset SRCC case and demonstrated that the mutant with impaired H3K27me3 demethylase activity could potentiate tumor malignancy and suppress antitumor immunity.
Collapse
Affiliation(s)
- Maoxiao Feng
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Chengwei Chai
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Department of Pediatric General Surgery, Guangdong Women and Children Hospital, Guangzhou, 511442, China.
| | - Xiaodong Hao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China
| | - Xiaojiang Lai
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yuanyuan Luo
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hong Zhang
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenzhu Tang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ningxin Gao
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Guihong Pan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaojie Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Wenjing Xiong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Qiang Wu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Jun Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
28
|
Tong D, Tang Y, Zhong P. The emerging roles of histone demethylases in cancers. Cancer Metastasis Rev 2024; 43:795-821. [PMID: 38227150 DOI: 10.1007/s10555-023-10160-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Modulation of histone methylation status is regarded as an important mechanism of epigenetic regulation and has substantial clinical potential for the therapy of diseases, including cancer and other disorders. The present study aimed to provide a comprehensive introduction to the enzymology of histone demethylases, as well as their cancerous roles, molecular mechanisms, therapeutic possibilities, and challenges for targeting them, in order to advance drug design for clinical therapy and highlight new insight into the mechanisms of these enzymes in cancer. A series of clinical trials have been performed to explore potential roles of histone demethylases in several cancer types. Numerous targeted inhibitors associated with immunotherapy, chemotherapy, radiotherapy, and targeted therapy have been used to exert anticancer functions. Future studies should evaluate the dynamic transformation of histone demethylases leading to carcinogenesis and explore individual therapy.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urological Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| | - Ying Tang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China.
| |
Collapse
|
29
|
Gulati S, Barata PC, Elliott A, Bilen MA, Burgess EF, Choueiri TK, Darabi S, Dawson NA, Gartrell BA, Hammers HJ, Heath EI, Magee D, Rao A, Ryan CJ, Twardowski P, Wei S, Brugarolas J, Zhang T, Zibelman MR, Nabhan C, McKay RR. Molecular analysis of primary and metastatic sites in patients with renal cell carcinoma. J Clin Invest 2024; 134:e176230. [PMID: 39007269 PMCID: PMC11245151 DOI: 10.1172/jci176230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 05/17/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUNDMetastases are the hallmark of lethal cancer, though underlying mechanisms that drive metastatic spread to specific organs remain poorly understood. Renal cell carcinoma (RCC) is known to have distinct sites of metastases, with lung, bone, liver, and lymph nodes being more common than brain, gastrointestinal tract, and endocrine glands. Previous studies have shown varying clinical behavior and prognosis associated with the site of metastatic spread; however, little is known about the molecular underpinnings that contribute to the differential outcomes observed by the site of metastasis.METHODSWe analyzed primary renal tumors and tumors derived from metastatic sites to comprehensively characterize genomic and transcriptomic features of tumor cells as well as to evaluate the tumor microenvironment at both sites.RESULTSWe included a total of 657 tumor samples (340 from the primary site [kidney] and 317 from various sites of metastasis). We show distinct genomic alterations, transcriptomic signatures, and immune and stromal tumor microenvironments across metastatic sites in a large cohort of patients with RCC.CONCLUSIONWe demonstrate significant heterogeneity among primary tumors and metastatic sites and elucidate the complex interplay between tumor cells and the extrinsic tumor microenvironment that is vital for developing effective anticancer therapies.
Collapse
Affiliation(s)
- Shuchi Gulati
- UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Pedro C Barata
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | | | | | | | | | - Sourat Darabi
- Hoag Memorial Hospital Presbyterian, Newport Beach, California, USA
| | - Nancy Ann Dawson
- Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Benjamin Adam Gartrell
- Montefiore Medical Center and Albert Einstein College of Medicine, New York, New York, USA
| | | | - Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | - Arpit Rao
- Baylor College of Medicine, Houston, Texas, USA
| | | | - Przemyslaw Twardowski
- Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, California, USA
| | - Shuanzeng Wei
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | | - Tian Zhang
- UT Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Rana R McKay
- University of California San Diego, La Jolla, California, USA
| |
Collapse
|
30
|
Qiang Y, Fan J, Xie C, Yan L, Song X, Zhang N, Lin Y, Xiong J, Zhang W, Liu Y, Wei L, Li Y, Chen S, Liang K, Li F. KDM5C-Mediated Recruitment of BRD4 to Chromatin Regulates Enhancer Activation and BET Inhibitor Sensitivity. Cancer Res 2024; 84:1252-1269. [PMID: 38285760 DOI: 10.1158/0008-5472.can-23-2888] [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: 09/20/2023] [Revised: 12/05/2023] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
The BET family member BRD4 is a bromodomain-containing protein that plays a vital role in driving oncogene expression. Given their pivotal role in regulating oncogenic networks in various cancer types, BET inhibitors (BETi) have been developed, but the clinical application has been impeded by dose-limiting toxicity and resistance. Understanding the mechanisms of BRD4 activity and identifying predictive biomarkers could facilitate the successful clinical use of BETis. Herein, we show that KDM5C and BRD4 cooperate to sustain tumor cell growth. Mechanistically, KDM5C interacted with BRD4 and stimulated BRD4 enhancer recruitment. Moreover, binding of the BRD4 C-terminus to KDM5C stimulated the H3K4 demethylase activity of KDM5C. The abundance of both KDM5C-associated BRD4 and H3K4me1/3 determined the transcriptional activation of many oncogenes. Notably, depletion or pharmacologic degradation of KDM5C dramatically reduced BRD4 chromatin enrichment and significantly increased BETi efficacy across multiple cancer types in both tumor cell lines and patient-derived organoid models. Furthermore, targeting KDM5C in combination with BETi suppressed tumor growth in vivo in a xenograft mouse model. Collectively, this work reveals a KDM5C-mediated mechanism by which BRD4 regulates transcription, providing a rationale for incorporating BETi into combination therapies with KDM5C inhibitors to enhance treatment efficacy. SIGNIFICANCE BRD4 is recruited to enhancers in a bromodomain-independent manner by binding KDM5C and stimulates KDM5C H3K4 demethylase activity, leading to synergistic effects of BET and KDM5C inhibitor combinations in cancer.
Collapse
Affiliation(s)
- Yulong Qiang
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Jiachen Fan
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Chuanshuai Xie
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Leilei Yan
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Xiaofei Song
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Nan Zhang
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Yan Lin
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wei Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yu Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Shizhen Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Kaiwei Liang
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Feng Li
- Department of Medical Genetics, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| |
Collapse
|
31
|
Waldum H, Slupphaug G. Correctly identifying the cells of origin is essential for tailoring treatment and understanding the emergence of cancer stem cells and late metastases. Front Oncol 2024; 14:1369907. [PMID: 38660133 PMCID: PMC11040596 DOI: 10.3389/fonc.2024.1369907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Malignancy manifests itself by deregulated growth and the ability to invade surrounding tissues or metastasize to other organs. These properties are due to genetic and/or epigenetic changes, most often mutations. Many aspects of carcinogenesis are known, but the cell of origin has been insufficiently focused on, which is unfortunate since the regulation of its growth is essential to understand the carcinogenic process and guide treatment. Similarly, the concept of cancer stem cells as cells having the ability to stop proliferation and rest in a state of dormancy and being resistant to cytotoxic drugs before "waking up" and become a highly malignant tumor recurrence, is not fully understood. Some tumors may recur after decades, a phenomenon probably also connected to cancer stem cells. The present review shows that many of these questions are related to the cell of origin as differentiated cells being long-term stimulated to proliferation.
Collapse
Affiliation(s)
- Helge Waldum
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
32
|
Gamallat Y, Felipe Lima J, Seyedi S, Li Q, Rokne JG, Alhajj R, Ghosh S, Bismar TA. Exploring The Prognostic Significance of SET-Domain Containing 2 (SETD2) Expression in Advanced and Castrate-Resistant Prostate Cancer. Cancers (Basel) 2024; 16:1436. [PMID: 38611113 PMCID: PMC11010867 DOI: 10.3390/cancers16071436] [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: 12/31/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
SET-domain containing 2 (SETD2) is a histone methyltransferase and an epigenetic modifier with oncogenic functionality. In the current study, we investigated the potential prognostic role of SETD2 in prostate cancer. A cohort of 202 patients' samples was assembled on tissue microarrays (TMAs) containing incidental, advanced, and castrate-resistant CRPCa cases. Our data showed significant elevated SETD2 expression in advanced and castrate-resistant disease (CRPCa) compared to incidental cases (2.53 ± 0.58 and 2.21 ± 0.63 vs. 1.9 ± 0.68; p < 0.001, respectively). Interestingly, the mean intensity of SETD2 expression in deceased vs. alive patients was also significantly different (2.31 ± 0.66 vs. 2 ± 0.68; p = 0.003, respectively). Overall, high SETD2 expression was found to be considered high risk and was significantly associated with poor prognosis and worse overall survival (OS) (HR 1.80; 95% CI: 1.28-2.53, p = 0.001) and lower cause specific survival (CSS) (HR 3.14; 95% CI: 1.94-5.08, p < 0.0001). Moreover, combining high-intensity SETD2 with PTEN loss resulted in lower OS (HR 2.12; 95% CI: 1.22-3.69, p = 0.008) and unfavorable CSS (HR 3.74; 95% CI: 1.67-8.34, p = 0.001). Additionally, high SETD2 intensity with ERG positive expression showed worse prognosis for both OS (HR 1.99, 95% CI 0.87-4.59; p = 0.015) and CSS (HR 2.14, 95% CI 0.98-4.68, p = 0.058). We also investigated the protein expression database TCPA, and our results showed that high SETD2 expression is associated with a poor prognosis. Finally, we performed TCGA PRAD gene set enrichment analysis (GSEA) data for SETD2 overexpression, and our data revealed a potential association with pathways involved in tumor progression such as the AMPK signaling pathway, the cAMP signaling pathway, and the PI3K-Akt signaling pathway, which are potentially associated with tumor progression, chemoresistance, and a poor prognosis.
Collapse
Affiliation(s)
- Yaser Gamallat
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (Y.G.); (J.F.L.); (S.S.)
- Departments of Oncology, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Joema Felipe Lima
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (Y.G.); (J.F.L.); (S.S.)
- Departments of Oncology, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Sima Seyedi
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (Y.G.); (J.F.L.); (S.S.)
- Departments of Oncology, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Qiaowang Li
- Department of Computer Science, University of Calgary, Calgary, AB T2N 1N4, Canada; (Q.L.)
| | - Jon George Rokne
- Department of Computer Science, University of Calgary, Calgary, AB T2N 1N4, Canada; (Q.L.)
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, AB T2N 1N4, Canada; (Q.L.)
- Department of Computer Engineering, Istanbul Medipol University, 34810 Istanbul, Turkey
- Department of Health Informatics, University of Southern Denmark, 5230 Odense, Denmark
| | - Sunita Ghosh
- Department of Medical Oncology, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R7, Canada
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | - Tarek A. Bismar
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (Y.G.); (J.F.L.); (S.S.)
- Departments of Oncology, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Tom Baker Cancer Center, Alberta Health Services, Calgary, AB T2N 4N1, Canada
- Prostate Cancer Centre, Alberta Health Services, Calgary, AB T2V 1P9, Canada
- Department of Pathology, Alberta Precision Labs, Calgary, AB T2V 1P9, Canada
| |
Collapse
|
33
|
He H, Xie Y, Song F, Feng Z, Rong P. Radiogenomic analysis based on lipid metabolism-related subset for non-invasive prediction for prognosis of renal clear cell carcinoma. Eur J Radiol 2024; 175:111433. [PMID: 38554673 DOI: 10.1016/j.ejrad.2024.111433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024]
Abstract
PURPOSE Multiple lipid metabolism pathways alterations are associated with clear cell renal cell carcinoma (ccRCC) development and aggressiveness. In this study, we aim to develop a novel radiogenomics signature based on lipid metabolism-related genes (LMRGs) that may accurately predict ccRCC patients' survival. MATERIALS AND METHODS First, 327 ccRCC were used to screen survival-related LMRGs and construct a gene signature based on The Cancer Genome Atlas (TCGA) database. Then, 182 ccRCC were analyzed to establish radiogenomics signature linking LMRGs signature to radiomic features in The Cancer Imaging Archive (TCIA) database included enhanced CT images and transcriptome sequencing data. Lastly, we validated the prognostic power of the identified radiogenomics signature using these patients of TCIA and the Third Xiangya Hospital. RESULTS We identified the LMRGs signature, consisting of 13 genes, which could efficiently discriminate between low-risk and high-risk patients and serve as an independent and reliable predictor of overall survival (OS). Radiogenomics signature, comprised of 9 radiomic features, was created and could accurately predict the expression level of LMRGs signature (low- or high-risk) for patients. The predictive performance of this radiogenomics signature was demonstrated through AUC values of 0.75 and 0.74 for the training and validation sets (at a ratio of 7:3), respectively. Radiogenomics signature was proven to be an independent risk factor for OS by multivariable analysis (HR = 4.98, 95 % CI:1.72-14.43, P = 0.003). CONCLUSIONS The LMRGs radiogenomics signature could serve as a novel prognostic predictor.
Collapse
Affiliation(s)
- Haifeng He
- Department of Radiology, The Third Xiangya Hospital Central South University, Changsha, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital Central South University, Changsha, China
| | - Fulong Song
- Department of Radiology, The Third Xiangya Hospital Central South University, Changsha, China
| | - Zhichao Feng
- Department of Radiology, The Third Xiangya Hospital Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital Central South University, Changsha, China.
| |
Collapse
|
34
|
Zhang X, Wang H, Zhang Y, Wang X. Advances in epigenetic alterations of chronic lymphocytic leukemia: from pathogenesis to treatment. Clin Exp Med 2024; 24:54. [PMID: 38492089 PMCID: PMC10944427 DOI: 10.1007/s10238-023-01268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/01/2023] [Indexed: 03/18/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a heterogeneous disease with alterations in genetic expression and epigenetic modifications. In recent years, the new insight into epigenetics in the pathogenesis of CLL has been developed considerably, including DNA methylation, histone modification, RNA methylation, non-coding RNAs as well as chromatin remodeling. Epigenetic modification regulates various processes such as stem cell biology, cell growth, and tumorigenesis without altering gene sequence. Growing evidence indicates that the disturbance of gene expression profiles which were regulated by epigenetic modifications exerts vital roles in the development and progress in CLL, which provides novel perspectives to explore the etiology of CLL. In addition, the integration with epigenetic therapeutic targets and the in-depth understanding of epigenetic therapy contribute to develop new therapeutic strategies for CLL. Herein, the present review discusses the advances of epigenetic alterations in the pathogenesis, diagnosis, and prognostic assessment of CLL patients and also highlights existing and emerging agents targeting epigenetic regulators.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Hua Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Taishan Scholars Program of Shandong Province, Jinan, 250021, Shandong, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Taishan Scholars Program of Shandong Province, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
35
|
Dupéré-Richer D, Riva A, Maji S, Barwick BG, Román HC, Sobh A, Quickstad G, Li J, De U, Piper C, Kulis M, Ezponda T, Martin-Subero JI, Tonon G, Zhang W, Mitsiades CS, Boise LH, Bennett RL, Licht JD. KDM6A Regulates Immune Response Genes in Multiple Myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579179. [PMID: 38405853 PMCID: PMC10888870 DOI: 10.1101/2024.02.12.579179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The histone H3K27 demethylase KDM6A is a tumor suppressor in multiple cancers, including multiple myeloma (MM). We created isogenic MM cells disrupted for KDM6A and tagged the endogenous protein to facilitate genome wide studies. KDM6A binds genes associated with immune recognition and cytokine signaling. Most importantly, KDM6A binds and activates NLRC5 and CIITA encoding regulators of Major Histocompatibility Complex (MHC) genes. Patient data indicate that NLRC5 and CIITA, are downregulated in MM with low KDM6A expression. Chromatin analysis shows that KDM6A binds poised and active enhancers and KDM6A loss led to decreased H3K27ac at enhancers, increased H3K27me3 levels in body of genes bound by KDM6A and decreased gene expression. Reestablishing histone acetylation with an HDAC3 inhibitor leads to upregulation of MHC expression, offering a strategy to restore immunogenicity of KDM6A deficient tumors. Loss of Kdm6a in murine RAS-transformed fibroblasts led to increased growth in vivo associated with decreased T cell infiltration. Statement of significance We show that KDM6A participates in immune recognition of myeloma tumor cells by directly regulating the expression of the master regulators of MHC-I and II, NLRC5 and CIITA. The expression of these regulators can by rescued by the HDAC3 inhibitors in KDM6A-null cell lines.
Collapse
|
36
|
Zhang C, Zeng J, Ye C, Tian K, Xian Z. Construction and validation of a chromatin regulator-related gene signature for prognostic and therapeutic significance of clear cell renal cell carcinoma. Transl Cancer Res 2024; 13:150-172. [PMID: 38410230 PMCID: PMC10894348 DOI: 10.21037/tcr-23-1383] [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: 08/03/2023] [Accepted: 11/29/2023] [Indexed: 02/28/2024]
Abstract
Background Epigenetic alterations driven by chromatin regulators (CRs) are well-recognized cancer hallmarks. Growing evidence suggests that the imbalance of CRs may lead to the occurrence of various diseases including tumors. However, the role and prognostic value of CRs in clear cell renal cell carcinoma (ccRCC) remain undefined. Methods Consensus clustering analysis was used to identify different subtypes. Univariate and multivariate Cox regression analysis were performed to identify prognosis-related CRs and constructed a risk model. Transcriptome sequencing was used to verify gene expression levels. Kaplan-Meier survival analysis was used to compare overall survival (OS) between high- and low-risk groups. The area under the curve (AUC) value of the receiver operating characteristic (ROC) curve was used to evaluate the performance of the model. The ESTIMATE algorithm and single-sample gene set enrichment analysis (ssGSEA) were executed to evaluate the immune characteristics of samples. Correlation analysis was used to assess the relationship between risk score and immune checkpoint genes, the relationship between expression levels of CRs and immune cell infiltration and drug therapeutic response. Finally, we also compared differences in drug sensitivity between low- and high-risk groups. Results We identified three CRs-related subtypes with different characteristics. A prognostic model was built with four CRs and can precisely predict the OS of patients in different risk groups. The model has good stability and applicability and was further verified in the internal and external dataset. The transcriptomic levels of the four CRs were also validated, and the risk score was an independent prognostic factor for ccRCC. Obvious differences in the immune microenvironment and the expression levels of immune checkpoints were observed in low- and high-risk group. Higher immune activity and immune cell infiltration were found in the high-risk group. Besides, the expression levels of CRs were associated with drug therapeutic response. Patients with high-risk score may be more sensitive to gemcitabine, vinblastine, paclitaxel, axitinib, sunitinib, and temsirolimus. Conclusions CRs were strongly associated with the occurrence and development of ccRCC. Targeting CRs may become a new therapeutic strategy for ccRCC. Besides, CRs-related gene signature can predict the prognosis and therapeutic significance of ccRCC, which provides an important reference for clinical decision-making.
Collapse
Affiliation(s)
- Changzheng Zhang
- Department of Urology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiayi Zeng
- Department of Urology, Guangdong Provincial People’s Hospital’s Nanhai Hospital, Foshan, China
| | - Chujin Ye
- Department of Urology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Kaiwen Tian
- Department of Urology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhiyong Xian
- Department of Urology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Urology, Guangdong Provincial People’s Hospital’s Nanhai Hospital, Foshan, China
| |
Collapse
|
37
|
Yu F, Li L, Gu Y, Wang S, Zhou L, Cheng X, Jiang H, Huang Y, Zhang Y, Qian W, Li X, Liu Z. Lysine demethylase 5C inhibits transcription of prefoldin subunit 5 to activate c-Myc signal transduction and colorectal cancer progression. Mol Med 2024; 30:9. [PMID: 38216914 PMCID: PMC10785505 DOI: 10.1186/s10020-023-00775-7] [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: 11/09/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Lysine demethylase 5C (KDM5C) has been implicated in the development of several human cancers. This study aims to investigate the role of KDM5C in the progression of colorectal cancer (CRC) and explore the associated molecular mechanism. METHODS Bioinformatics tools were employed to predict the target genes of KDM5C in CRC. The expression levels of KDM5C and prefoldin subunit 5 (PFDN5) in CRC cells were determined by RT-qPCR and western blot assays. The interaction between KDM5C, H3K4me3, and PFDN5 was validated by chromatin immunoprecipitation. Expression and prognostic values of KDM5C and PFDN5 in CRC were analyzed in a cohort of 72 patients. The function of KDM5C/PFDN5 in c-Myc signal transduction was analyzed by luciferase assay. Silencing of KDM5C and PFDN5 was induced in CRC cell lines to analyze the cell malignant phenotype in vitro and tumorigenic activity in nude mice. RESULTS KDM5C exhibited high expression, while PFDN5 displayed low expression in CRC cells and clinical CRC samples. High KDM5C levels correlated with poor survival and unfavorable clinical presentation, whereas elevated PFDN5 correlated with improved patient outcomes. KDM5C mediated demethylation of H3K4me3 on the PFDN5 promoter, suppressing its transcription and thereby enhancing the transcriptional activity of c-Myc. KDM5C knockdown in CRC cells suppressed cell proliferation, migration and invasion, epithelial-mesenchymal transition, and tumorigenic activity while increasing autophagy and apoptosis rates. However, the malignant behavior of cells was restored by the further silencing of PFDN5. CONCLUSION This study demonstrates that KDM5C inhibits PFDN5 transcription, thereby activating c-Myc signal transduction and promoting CRC progression.
Collapse
Affiliation(s)
- Fulong Yu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Liang Li
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Yimei Gu
- Emergency ICU, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui, People's Republic of China
| | - Song Wang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Lianbang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Xiaohu Cheng
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Heng Jiang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Yang Huang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Yingfeng Zhang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China
| | - Wenbao Qian
- Department of Molecular Pathology, Hefei Da'an Medical Laboratory Co., Ltd., Hefei, 230012, Anhui, People's Republic of China
| | - Xianghua Li
- Department of Molecular Pathology, Hefei Da'an Medical Laboratory Co., Ltd., Hefei, 230012, Anhui, People's Republic of China.
| | - Zhining Liu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, People's Republic of China.
| |
Collapse
|
38
|
Hillen H, Candi A, Vanderhoydonck B, Kowalczyk W, Sansores-Garcia L, Kesikiadou EC, Van Huffel L, Spiessens L, Nijs M, Soons E, Haeck W, Klaassen H, Smets W, Spieser SA, Marchand A, Chaltin P, Ciesielski F, Debaene F, Chen L, Kamal A, Gwaltney SL, Versele M, Halder GA. A Novel Irreversible TEAD Inhibitor, SWTX-143, Blocks Hippo Pathway Transcriptional Output and Causes Tumor Regression in Preclinical Mesothelioma Models. Mol Cancer Ther 2024; 23:3-13. [PMID: 37748190 DOI: 10.1158/1535-7163.mct-22-0681] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/13/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
The Hippo pathway and its downstream effectors, the YAP and TAZ transcriptional coactivators, are deregulated in multiple different types of human cancer and are required for cancer cell phenotypes in vitro and in vivo, while largely dispensable for tissue homeostasis in adult mice. YAP/TAZ and their main partner transcription factors, the TEAD1-4 factors, are therefore promising anticancer targets. Because of frequent YAP/TAZ hyperactivation caused by mutations in the Hippo pathway components NF2 and LATS2, mesothelioma is one of the prime cancer types predicted to be responsive to YAP/TAZ-TEAD inhibitor treatment. Mesothelioma is a devastating disease for which currently no effective treatment options exist. Here, we describe a novel covalent YAP/TAZ-TEAD inhibitor, SWTX-143, that binds to the palmitoylation pocket of all four TEAD isoforms. SWTX-143 caused irreversible and specific inhibition of the transcriptional activity of YAP/TAZ-TEAD in Hippo-mutant tumor cell lines. More importantly, YAP/TAZ-TEAD inhibitor treatment caused strong mesothelioma regression in subcutaneous xenograft models with human cells and in an orthotopic mesothelioma mouse model. Finally, SWTX-143 also selectively impaired the growth of NF2-mutant kidney cancer cell lines, suggesting that the sensitivity of mesothelioma models to these YAP/TAZ-TEAD inhibitors can be extended to other tumor types with aberrations in Hippo signaling. In brief, we describe a novel and specific YAP/TAZ-TEAD inhibitor that has potential to treat multiple Hippo-mutant solid tumor types.
Collapse
Affiliation(s)
- Hanne Hillen
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | - Weronika Kowalczyk
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leticia Sansores-Garcia
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena C Kesikiadou
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leen Van Huffel
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Lore Spiessens
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | - Patrick Chaltin
- Cistim Leuven vzw, Leuven, Belgium
- Center for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | | | | | - Lei Chen
- SpringWorks Therapeutics, Stamford, Connecticut
| | | | | | | | - Georg A Halder
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Sun W, Lee KL, Poellinger L, Masai H, Kato H. Catalytic domain-dependent and -independent transcriptional activities of the tumour suppressor histone H3K27 demethylase UTX/KDM6A in specific cancer types. Epigenetics 2023; 18:2222245. [PMID: 37300822 DOI: 10.1080/15592294.2023.2222245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/10/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
The histone H3K27 demethylase, UTX/KDM6A, plays a critical role in the early development of vertebrates, and mutations are frequently found in various cancers. Several studies on developmental and cancer biology have focused on preferential transcriptional regulation by UTX independently of its H3K27 demethylase catalytic activity. Here, we analysed gene expression profiles of wild-type (WT) UTX and a catalytic activity-defective mutant in 786-O and HCT116 cells and confirmed that catalytic activity-dependent and -independent regulation contributes to the expression of most of the target genes. Indeed, the catalytic activity-defective mutant indeed suppressed colony formation similar to the WT in our assay system. However, the expression of several genes was significantly dependent on the catalytic activity of UTX in a cell type-specific manner, which could account for the inherent variation in the transcriptional landscape of various cancer types. The promoter/enhancer regions of the catalytic activity-dependent genes identified here were found to be preferentially modified with H3K4me1 and less with H3K27me3 than those of the independent genes. These findings, combined with previous reports, highlight not only the understanding of determinants for the catalytic activity dependency but also the development and application of pharmaceutical agents targeting the H3K27 or H3K4 modifications.
Collapse
Affiliation(s)
- Wendi Sun
- Cancer Science Institute of Singapore, National University of Singapore, Singapoe, Republic of Singapore
| | - Kian Leong Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapoe, Republic of Singapore
- Cancer & Stem Cell Biology Signature Research Programme, Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Lorenz Poellinger
- Cancer Science Institute of Singapore, National University of Singapore, Singapoe, Republic of Singapore
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hisao Masai
- Genome Dynamics Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroyuki Kato
- Cancer Science Institute of Singapore, National University of Singapore, Singapoe, Republic of Singapore
| |
Collapse
|
40
|
Liu Y, Wu G. The utilization of single-cell sequencing technology in investigating the immune microenvironment of ccRCC. Front Immunol 2023; 14:1276658. [PMID: 38090562 PMCID: PMC10715415 DOI: 10.3389/fimmu.2023.1276658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
The growth and advancement of ccRCC are strongly associated with the presence of immune infiltration and the tumor microenvironment, comprising tumor cells, immune cells, stromal cells, vascular cells, myeloid-derived cells, and extracellular matrix (ECM). Nevertheless, as a result of the diverse and constantly evolving characteristics of the tumor microenvironment, prior advanced sequencing methods have frequently disregarded specific less prevalent cellular traits at varying intervals, thereby concealing their significance. The advancement and widespread use of single-cell sequencing technology enable us to comprehend the source of individual tumor cells and the characteristics of a greater number of individual cells. This, in turn, minimizes the impact of intercellular heterogeneity and temporal heterogeneity of the same cell on experimental outcomes. This review examines the attributes of the tumor microenvironment in ccRCC and provides an overview of the progress made in single-cell sequencing technology and its particular uses in the current focus of immune infiltration in ccRCC.
Collapse
Affiliation(s)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
41
|
Lee SCES, Pyo AHA, Koritzinsky M. Longitudinal dynamics of the tumor hypoxia response: From enzyme activity to biological phenotype. SCIENCE ADVANCES 2023; 9:eadj6409. [PMID: 37992163 PMCID: PMC10664991 DOI: 10.1126/sciadv.adj6409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Poor oxygenation (hypoxia) is a common spatially heterogeneous feature of human tumors. Biological responses to tumor hypoxia are orchestrated by the decreased activity of oxygen-dependent enzymes. The affinity of these enzymes for oxygen positions them along a continuum of oxygen sensing that defines their roles in launching reactive and adaptive cellular responses. These responses encompass regulation of all steps in the central dogma, with rapid perturbation of the metabolome and proteome followed by more persistent reprogramming of the transcriptome and epigenome. Core hypoxia response genes and pathways are commonly regulated at multiple inflection points, fine-tuning the dependencies on oxygen concentration and hypoxia duration. Ultimately, shifts in the activity of oxygen-sensing enzymes directly or indirectly endow cells with intrinsic hypoxia tolerance and drive processes that are associated with aggressive phenotypes in cancer including angiogenesis, migration, invasion, immune evasion, epithelial mesenchymal transition, and stemness.
Collapse
Affiliation(s)
- Sandy Che-Eun S. Lee
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Hye An Pyo
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Guo CC, Xu HE, Ma X. ARID3a from the ARID family: structure, role in autoimmune diseases and drug discovery. Acta Pharmacol Sin 2023; 44:2139-2150. [PMID: 37488425 PMCID: PMC10618457 DOI: 10.1038/s41401-023-01134-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/09/2023] [Indexed: 07/26/2023]
Abstract
The AT-rich interaction domain (ARID) family of DNA-binding proteins is a group of transcription factors and chromatin regulators with a highly conserved ARID domain that recognizes specific AT-rich DNA sequences. Dysfunction of ARID family members has been implicated in various human diseases including cancers and intellectual disability. Among them, ARID3a has gained increasing attention due to its potential involvement in autoimmunity. In this article we provide an overview of the ARID family, focusing on the structure and biological functions of ARID3a. It explores the role of ARID3a in autoreactive B cells and its contribution to autoimmune diseases such as systemic lupus erythematosus and primary biliary cholangitis. Furthermore, we also discuss the potential for drug discovery targeting ARID3a and present a plan for future research in this field.
Collapse
Affiliation(s)
- Cheng-Cen Guo
- Department of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, 200001, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Xiong Ma
- Department of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, 200001, China.
| |
Collapse
|
43
|
Meng J, Jiang A, Lu X, Gu D, Ge Q, Bai S, Zhou Y, Zhou J, Hao Z, Yan F, Wang L, Wang H, Du J, Liang C. Multiomics characterization and verification of clear cell renal cell carcinoma molecular subtypes to guide precise chemotherapy and immunotherapy. IMETA 2023; 2:e147. [PMID: 38868222 PMCID: PMC10989995 DOI: 10.1002/imt2.147] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/21/2023] [Indexed: 06/14/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a heterogeneous tumor with different genetic and molecular alterations. Schemes for ccRCC classification system based on multiomics are urgent, to promote further biological insights. Two hundred and fifty-five ccRCC patients with paired data of clinical information, transcriptome expression profiles, copy number alterations, DNA methylation, and somatic mutations were collected for identification. Bioinformatic analyses were performed based on our team's recently developed R package "MOVICS." With 10 state-of-the-art algorithms, we identified the multiomics subtypes (MoSs) for ccRCC patients. MoS1 is an immune exhausted subtype, presented the poorest prognosis, and might be caused by an exhausted immune microenvironment, activated hypoxia features, but can benefit from PI3K/AKT inhibitors. MoS2 is an immune "cold" subtype, which represented more mutation of VHL and PBRM1, favorable prognosis, and is more suitable for sunitinib therapy. MoS3 is the immune "hot" subtype, and can benefit from the anti-PD-1 immunotherapy. We successfully verified the different molecular features of the three MoSs in external cohorts GSE22541, GSE40435, and GSE53573. Patients that received Nivolumab therapy helped us to confirm that MoS3 is suitable for anti-PD-1 therapy. E-MTAB-3267 cohort also supported the fact that MoS2 patients can respond more to sunitinib treatment. We also confirm that SETD2 is a tumor suppressor in ccRCC, along with the decreased SETD2 protein level in advanced tumor stage, and knock-down of SETD2 leads to the promotion of cell proliferation, migration, and invasion. In summary, we provide novel insights into ccRCC molecular subtypes based on robust clustering algorithms via multiomics data, and encourage future precise treatment of ccRCC patients.
Collapse
Affiliation(s)
- Jialin Meng
- Department of UrologyThe First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefeiChina
| | - Aimin Jiang
- Department of Urology, Changhai HospitalNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Xiaofan Lu
- Department of Cancer and Functional GenomicsInstitute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRAIllkirchFrance
| | - Di Gu
- Department of Urology, Changhai HospitalNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Qintao Ge
- Department of UrologyThe First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefeiChina
| | - Suwen Bai
- The Second Affiliated Hospital, School of MedicineThe Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of ShenzhenShenzhenChina
| | - Yundong Zhou
- Department of Surgery, Ningbo Medical Center Lihuili HospitalNingbo UniversityNingboZhejiangChina
| | - Jun Zhou
- Department of UrologyThe First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefeiChina
| | - Zongyao Hao
- Department of UrologyThe First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefeiChina
| | - Fangrong Yan
- Research Center of Biostatistics and Computational PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Linhui Wang
- Department of Urology, Changhai HospitalNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Haitao Wang
- Cancer Center, Faculty of Health SciencesUniversity of MacauMacau SARChina
- Present address:
Center for Cancer ResearchBethesdaMarylandUSA
| | - Juan Du
- The Second Affiliated Hospital, School of MedicineThe Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of ShenzhenShenzhenChina
| | - Chaozhao Liang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary DiseasesAnhui Medical UniversityHefeiChina
| |
Collapse
|
44
|
Shirole NH, Kaelin WG. von-Hippel Lindau and Hypoxia-Inducible Factor at the Center of Renal Cell Carcinoma Biology. Hematol Oncol Clin North Am 2023; 37:809-825. [PMID: 37270382 PMCID: PMC11315268 DOI: 10.1016/j.hoc.2023.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The most common form of kidney cancer is clear cell renal cell carcinoma (ccRCC). Biallelic VHL tumor suppressor gene inactivation is the usual initiating event in both hereditary (VHL Disease) and sporadic ccRCCs. The VHL protein, pVHL, earmarks the alpha subunits of the HIF transcription factor for destruction in an oxygen-dependent manner. Deregulation of HIF2 drives ccRCC pathogenesis. Drugs inhibiting the HIF2-responsive growth factor VEGF are now mainstays of ccRCC treatment. A first-in-class allosteric HIF2 inhibitor was recently approved for treating VHL Disease-associated neoplasms and appears active against sporadic ccRCC in early clinical trials.
Collapse
Affiliation(s)
- Nitin H Shirole
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - William G Kaelin
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Brigham and Women's Hospital, Harvard Medical School; Howard Hughes Medical Institute.
| |
Collapse
|
45
|
Braun DA, Chakraborty AA. Immunobiology and Metabolic Pathways of Renal Cell Carcinoma. Hematol Oncol Clin North Am 2023; 37:827-840. [PMID: 37246090 DOI: 10.1016/j.hoc.2023.04.012] [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] [Indexed: 05/30/2023]
Abstract
The treatment of advanced renal cell carcinoma (RCC) has changed dramatically with immune checkpoint inhibitors, yet most patients do not have durable responses. There is consequently a tremendous need for novel therapeutic development. RCC, and particularly the most common histology clear cell RCC, is an immunobiologically and metabolically distinct tumor. An improved understanding of RCC-specific biology will be necessary for the successful identification of new treatment targets for this disease. In this review, we discuss the current understanding of RCC immune pathways and metabolic dysregulation, with a focus on topics important for future clinical development.
Collapse
Affiliation(s)
- David A Braun
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, 300 George Street (Suite 6400), New Haven, CT 06511, USA.
| | - Abhishek A Chakraborty
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinical, 9500 Euclid Avenue (NB40), Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
46
|
Khan A, Metts JM, Collins LC, Mills CA, Li K, Brademeyer AL, Bowman BM, Major MB, Aubé J, Herring LE, Davis IJ, Strahl BD. SETD2 maintains nuclear lamina stability to safeguard the genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.560032. [PMID: 37808753 PMCID: PMC10557632 DOI: 10.1101/2023.09.28.560032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Histone methyltransferases play essential roles in the organization and function of chromatin. They are also frequently mutated in human diseases including cancer1. One such often mutated methyltransferase, SETD2, associates co-transcriptionally with RNA polymerase II and catalyzes histone H3 lysine 36 trimethylation (H3K36me3) - a modification that contributes to gene transcription, splicing, and DNA repair2. While studies on SETD2 have largely focused on the consequences of its catalytic activity, the non-catalytic functions of SETD2 are largely unknown. Here we report a catalysis-independent function of SETD2 in maintaining nuclear lamina stability and genome integrity. We found that SETD2, via its intrinsically disordered N-terminus, associates with nuclear lamina proteins including lamin A/C, lamin B1, and emerin. Depletion of SETD2, or deletion of its N-terminus, resulted in widespread nuclear morphology abnormalities and genome stability defects that were reminiscent of a defective nuclear lamina. Mechanistically, the N-terminus of SETD2 facilitates the association of the mitotic kinase CDK1 with lamins, thereby promoting lamin phosphorylation and depolymerization required for nuclear envelope disassembly during mitosis. Taken together, our findings reveal an unanticipated link between the N-terminus of SETD2 and nuclear lamina organization that may underlie how SETD2 acts as a tumor suppressor.
Collapse
Affiliation(s)
- Abid Khan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - James M. Metts
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Lucas C. Collins
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - C. Allie Mills
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, 27599, USA
| | - Kelin Li
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Amanda L. Brademeyer
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Brittany M. Bowman
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Cell Biology & Physiology, Washington University in St. Louis
| | - M. Ben Major
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Cell Biology & Physiology, Washington University in St. Louis
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Laura E. Herring
- UNC Proteomics Core Facility, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, 27599, USA
| | - Ian J. Davis
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina 27514
| | - Brian D. Strahl
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
47
|
Davidson G, Helleux A, Vano YA, Lindner V, Fattori A, Cerciat M, Elaidi RT, Verkarre V, Sun CM, Chevreau C, Bennamoun M, Lang H, Tricard T, Fridman WH, Sautes-Fridman C, Su X, Plassard D, Keime C, Thibault-Carpentier C, Barthelemy P, Oudard SM, Davidson I, Malouf GG. Mesenchymal-like Tumor Cells and Myofibroblastic Cancer-Associated Fibroblasts Are Associated with Progression and Immunotherapy Response of Clear Cell Renal Cell Carcinoma. Cancer Res 2023; 83:2952-2969. [PMID: 37335139 DOI: 10.1158/0008-5472.can-22-3034] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/24/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
Immune checkpoint inhibitors (ICI) represent the cornerstone for the treatment of patients with metastatic clear cell renal cell carcinoma (ccRCC). Despite a favorable response for a subset of patients, others experience primary progressive disease, highlighting the need to precisely understand the plasticity of cancer cells and their cross-talk with the microenvironment to better predict therapeutic response and personalize treatment. Single-cell RNA sequencing of ccRCC at different disease stages and normal adjacent tissue (NAT) from patients identified 46 cell populations, including 5 tumor subpopulations, characterized by distinct transcriptional signatures representing an epithelial-to-mesenchymal transition gradient and a novel inflamed state. Deconvolution of the tumor and microenvironment signatures in public data sets and data from the BIONIKK clinical trial (NCT02960906) revealed a strong correlation between mesenchymal-like ccRCC cells and myofibroblastic cancer-associated fibroblasts (myCAF), which are both enriched in metastases and correlate with poor patient survival. Spatial transcriptomics and multiplex immune staining uncovered the spatial proximity of mesenchymal-like ccRCC cells and myCAFs at the tumor-NAT interface. Moreover, enrichment in myCAFs was associated with primary resistance to ICI therapy in the BIONIKK clinical trial. These data highlight the epithelial-mesenchymal plasticity of ccRCC cancer cells and their relationship with myCAFs, a critical component of the microenvironment associated with poor outcome and ICI resistance. SIGNIFICANCE Single-cell and spatial transcriptomics reveal the proximity of mesenchymal tumor cells to myofibroblastic cancer-associated fibroblasts and their association with disease outcome and immune checkpoint inhibitor response in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Guillaume Davidson
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
| | - Alexandra Helleux
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
| | - Yann A Vano
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, APHP, Université Paris Cité, Paris, France
| | - Véronique Lindner
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Antonin Fattori
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Marie Cerciat
- Genomeast platform, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France
| | - Reza T Elaidi
- Association pour la Recherche sur les Thérapeutiques Innovantes en Cancérologie, Paris, France
| | - Virginie Verkarre
- Department of Pathology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, APHP, Université Paris Cité, Paris, France
| | - Cheng-Ming Sun
- Centre des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Christine Chevreau
- Department of Medical Oncology, Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| | - Mostefa Bennamoun
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - Hervé Lang
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - Thibault Tricard
- Department of Urology, Strasbourg University Hospital, Strasbourg, France
| | - Wolf H Fridman
- Centre des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Catherine Sautes-Fridman
- Centre des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Damien Plassard
- Genomeast platform, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France
| | - Celine Keime
- Genomeast platform, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France
| | - Christelle Thibault-Carpentier
- Genomeast platform, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France
| | - Philippe Barthelemy
- Department of Medical Oncology, Strasbourg University, Institut de Cancérologie de Strasbourg, Strasbourg, France
| | - Stéphane M Oudard
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, APHP, Université Paris Cité, Paris, France
| | - Irwin Davidson
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
| | - Gabriel G Malouf
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, Illkirch, France
- Department of Medical Oncology, Strasbourg University, Institut de Cancérologie de Strasbourg, Strasbourg, France
| |
Collapse
|
48
|
Hong LYQ, Yeung ESH, Tran DT, Yerra VG, Kaur H, Kabir MDG, Advani SL, Liu Y, Batchu SN, Advani A. Altered expression, but small contribution, of the histone demethylase KDM6A in obstructive uropathy in mice. Dis Model Mech 2023; 16:dmm049991. [PMID: 37655466 PMCID: PMC10482012 DOI: 10.1242/dmm.049991] [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: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Epigenetic processes have emerged as important modulators of kidney health and disease. Here, we studied the role of KDM6A (a histone demethylase that escapes X-chromosome inactivation) in kidney tubule epithelial cells. We initially observed an increase in tubule cell Kdm6a mRNA in male mice with unilateral ureteral obstruction (UUO). However, tubule cell knockout of KDM6A had relatively minor consequences, characterized by a small reduction in apoptosis, increase in inflammation and downregulation of the peroxisome proliferator-activated receptor (PPAR) signaling pathway. In proximal tubule lineage HK-2 cells, KDM6A knockdown decreased PPARγ coactivator-1α (PGC-1α) protein levels and mRNA levels of the encoding gene, PPARGC1A. Tubule cell Kdm6a mRNA levels were approximately 2-fold higher in female mice than in male mice, both under sham and UUO conditions. However, kidney fibrosis after UUO was similar in both sexes. The findings demonstrate Kdm6a to be a dynamically regulated gene in the kidney tubule, varying in expression levels by sex and in response to injury. Despite the context-dependent variation in Kdm6a expression, knockout of tubule cell KDM6A has subtle (albeit non-negligible) effects in the adult kidney, at least in males.
Collapse
Affiliation(s)
- Lisa Y. Q. Hong
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Emily S. H. Yeung
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Duc Tin Tran
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Harmandeep Kaur
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - M. D. Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Suzanne L. Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Sri Nagarjun Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
49
|
Anwaier A, Xu W, Liu W, Wei S, Tian X, Qu Y, Yang J, Zhang H, Ye D. Tumor microenvironment-based signatures distinguish intratumoral heterogeneity, prognosis, and immunogenomic features of clear cell renal cell carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:236-249. [PMID: 39035192 PMCID: PMC11256720 DOI: 10.1016/j.jncc.2023.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 07/23/2024] Open
Abstract
Background The tumor microenvironment (TME) performs a crucial function in the tumorigenesis and response to immunotherapies of clear cell renal cell carcinoma (ccRCC). However, a lack of recognized pre-clinical TME-based risk models poses a great challenge to investigating the risk factors correlated with prognosis and treatment responses for patients with ccRCC. Methods Stromal and immune contexture were assessed to calculate the TMErisk score of a large sample of patients with ccRCC from public and real-world cohorts using machine-learning algorithms. Next, analyses for prognostic efficacy, correlations with clinicopathological features, functional enrichment, immune cell distributions, DNA variations, immune response, and heterogeneity were performed and validated. Results Clinical hub genes, including INAFM2, SRPX, DPYSL3, VSIG4, APLNR, FHL5, A2M, SLFN11, ADAMTS4, IFITM1, NOD2, CCR4, HLA-DQB2, and PLAUR, were identified and incorporated to develop the TMErisk signature. Patients in the TMEhigh risk group (category) exhibited a considerably grim prognosis, and the TMErisk model was shown to independently function as a risk indicator for the overall survival (OS) of ccRCC patients. Expression levels of immune checkpoint genes were substantially increased in TMEhigh risk group, while those of the human leukocyte antigen (HLA) family genes were prominently decreased. In addition, tumors in the TMEhigh group showed significantly high infiltration levels of tumor-infiltrated lymphocytes, including M2 macrophages, CD8+ T cells, B cells, and CD4+ T cells. In heterogeneity analysis, more frequent somatic mutations, including pro-tumorigenic BAP1 and PBRM1, were observed in the TMEhigh group. Importantly, 19.3% of patients receiving immunotherapies in the TMEhigh group achieved complete or partial response compared with those with immune tolerance in the TMElow group, suggesting that TMErisk prominently differentiates prognosis and responses to immunotherapy for patients with ccRCC. Conclusions We first established the TMErisk score of ccRCC using machine-learning algorithms based on a large-scale population. The TMErisk score can be utilized as an innovative independent prognosis predictive marker with high sensitivity and accuracy. Our discovery also predicted the efficacy of immunotherapy in ccRCC patients, indicating the intimate link between tumor immune microenvironment and intratumoral heterogeneity.
Collapse
Affiliation(s)
- Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiyin Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jianfeng Yang
- Department of Surgery, ShangNan Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| |
Collapse
|
50
|
Yang L, Wang X, Liu J, Liu X, Li S, Zheng F, Dong Q, Xu S, Xiong J, Fu B. Prognostic and tumor microenvironmental feature of clear cell renal cell carcinoma revealed by m6A and lactylation modification-related genes. Front Immunol 2023; 14:1225023. [PMID: 37638005 PMCID: PMC10450969 DOI: 10.3389/fimmu.2023.1225023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Background Both lactylation and m6A modification have important implications for the development of clear cell renal cell carcinoma (ccRCC), and we aimed to use crosstalk genes of both to reveal the prognostic and immunological features of ccRCC. Methods Our first step was to look for lactylation-related genes that differed between normal and tumor tissues, and then by correlation analysis, we found the genes associated with M6A. Following that, ccRCC subtypes will be identified and risk models will be constructed to compare the prognosis and tumor microenvironment among different subgroups. A nomogram was constructed to predict the prognosis of ccRCC, and in vitro, experiments were conducted to validate the expression and function of key genes. Results We screened 100 crosstalk genes and identified 2 ccRCC subtypes. A total of 11 prognostic genes were screened for building a risk model. we observed higher immune scores, elevated tumor mutational burden, and microsatellite instability scores in the high-risk group. Therefore, individuals classified as high-risk would derive greater benefits from immunotherapy. The nomogram's ability to predict overall survival with a 1-year AUC of 0.863 demonstrates its significant practical utility. In addition, HIBCH was identified as a potential therapeutic target and its expression and function were verified by in vitro experiments. Conclusion In addition to developing a precise prognostic nomogram for patients with ccRCC, our study also discovered the potential of HIBCH as a biomarker for the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Songhui Xu
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Xiong
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Fu
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|