1
|
Pan BS, Lin CY, Lee GA, Lin HK. Targeting SETDB1 in cancer and immune regulation: Potential therapeutic strategies in cancer. Kaohsiung J Med Sci 2025; 41:e12933. [PMID: 39764697 DOI: 10.1002/kjm2.12933] [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: 12/05/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 03/12/2025] Open
Abstract
SET domain bifurcated histone lysine methyltransferase 1 (SETDB1/ESET), a pivotal H3K9 methyltransferase, has been extensively studied since its discovery over two decades ago. SETDB1 plays critical roles in immune regulation, including B cell maturation, T-cell activity modulation, and endogenous retrovirus (ERV) silencing. While essential for normal immune cell function, SETDB1 overexpression in cancer cells disrupts immune responses by suppressing tumor immunogenicity and facilitating immune evasion. This is achieved through the repression of anti-tumor immune cell production, ERV silencing, and interference with the type I interferon pathway leading to inhibiting immune checkpoint blockade (ICB) efficacy. Beyond its immunological implications, SETDB1 overexpression fosters tumor growth and metastasis via transcriptional silencing of tumor suppressor genes through histone regulation and activating oncogenic signaling by non-histone regulation. These multifaceted roles make SETDB1 an attractive epigenetic target for novel cancer therapies. This review explores SETDB1's dual function in immune regulation and tumor progression, emphasizing its potential in the development of innovative cancer treatments targeting epigenetic dysregulation and oncogenic signaling.
Collapse
Affiliation(s)
- Bo-Syong Pan
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Cheng-Yu Lin
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Gilbert Aaron Lee
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
2
|
Igarashi Y, Akiyama Y, Shimada S, Watanabe S, Hatano M, Kodera K, Okazaki K, Tanji Y, Tsukihara S, Taniai T, Nara A, Yamane M, Kamachi A, Umemura K, Yasukawa K, Ono H, Akahoshi K, Tanabe M, Haruki K, Furukawa K, Ikegami T, Tanaka S. Identification and clinical implications of endogenous retrovirus elements suppressed by SETDB1 in hepatocellular carcinoma. JHEP Rep 2025; 7:101307. [DOI: 10.1016/j.jhepr.2024.101307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2025] Open
|
3
|
Verdier E, Gaspar N, Marques Da Costa ME, Marchais A. SETDB1 amplification in osteosarcomas: Insights from its role in healthy tissues and other cancer types. Oncotarget 2025; 16:51-62. [PMID: 39945463 PMCID: PMC11823473 DOI: 10.18632/oncotarget.28688] [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: 08/02/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Epigenetic modifications, which reversibly regulate gene expression without altering the DNA sequence, are increasingly described in the literature as essential elements in the processes leading to cancer development. SETDB1 regulates histone 3 (H3) K9 di- and trimethylation, promoting heterochromatin formation, and plays a key role in gene silencing. Epigenetic deregulation of SETDB1 expression appears to be involved in different cancers types, particularly in aggressive, relapsing or treatment-resistant subtypes. Despite advances in research, the full range of mechanisms through which this protein acts remains unclear; however, it is evident that SETDB1 has a pivotal role, particularly in the mesenchymal stem cells differentiation, tumor evasion and treatment resistance. Its role in genetically complex sarcomas, such as osteosarcoma, has not been fully explored, although recent Omics analyses suggest its presence and amplification in osteosarcoma. Given its involvement in osteoblastogenesis and adipogenesis, we discuss the potential of SETDB1 as a key target for new therapeutic strategies in osteosarcoma.
Collapse
Affiliation(s)
- Elodie Verdier
- UMR 1015 Tumour Immunology and anti-cancer immunotherapy Unit, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Nathalie Gaspar
- UMR 1015 Tumour Immunology and anti-cancer immunotherapy Unit, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Maria Eugenia Marques Da Costa
- UMR 1015 Tumour Immunology and anti-cancer immunotherapy Unit, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| | - Antonin Marchais
- UMR 1015 Tumour Immunology and anti-cancer immunotherapy Unit, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Department of Oncology for Child and Adolescent, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif 94805, France
| |
Collapse
|
4
|
Wang R, Dong X, Zhang X, Liao J, Cui W, Li W. Exploring viral mimicry combined with epigenetics and tumor immunity: new perspectives in cancer therapy. Int J Biol Sci 2025; 21:958-973. [PMID: 39897033 PMCID: PMC11781167 DOI: 10.7150/ijbs.103877] [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: 09/19/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Viral mimicry refers to an active antiviral response triggered by the activation of endogenous retroviruses (ERVs), usually manifested by the formation of double-stranded RNA (dsRNA) and activation of the cellular interferon response, which activates the immune system and produces anti-tumor effects. Epigenetic studies have shown that epigenetic modifications (e.g. DNA methylation, histone modifications, etc.) play a crucial role in tumorigenesis, progression, and treatment resistance. Particularly, alterations in DNA methylation may be closely associated with the suppression of ERVs expression, and treatment by demethylation may restore ERVs activity and thus strengthen the tumor immune response. Therefore, we propose that viral mimicry can induce immune responses in the tumor microenvironment by activating the expression of ERVs, and that epigenetic alterations may play a key regulatory role in this process. In this paper, we review the intersection of viral mimicry, epigenetics and tumor immunotherapy, and explore the possible interactions and synergistic effects among the three, aiming to provide a new theoretical basis and potential strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Xin Dong
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiongjian Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| |
Collapse
|
5
|
Yin CQ, Song CQ. Tumor Intrinsic Immunogenicity Suppressor SETDB1 Worsens the Prognosis of Patients with Hepatocellular Carcinoma. Cells 2024; 13:2102. [PMID: 39768193 PMCID: PMC11675013 DOI: 10.3390/cells13242102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is clinically distinguished by its covert onset, rapid progression, high recurrence rate, and poor prognosis. Studies have revealed that SETDB1 (SET Domain Bifurcated 1) is a histone H3 methyltransferase located on chromosome 1 and plays a crucial role in carcinogenesis. Therefore, we aimed to evaluate the clinical significance of SETDB1 expression in HCC. In patients with HCC, elevated levels of SETDB1 correlated with a poorer overall survival (OS) rate, marking it as an independent prognostic factor for HCC, as revealed by both univariate and multivariate Cox analyses. Furthermore, we utilized the SangerBox and TISIDB databases to profile the tumor immune microenvironment in HCC, including scoring the tumor microenvironment and assessing immune cell infiltration. The TIDE algorithm was employed to examine the association between SETDB1 expression and immune responses. Our findings indicated that SETDB1 expression negatively correlated with the majority of immune cells, a wide range of immune cell marker genes, and numerous immune pathways, thereby leading to the reduced effectiveness of immune checkpoint inhibitors. Lastly, both in vivo and ex vivo experiments were conducted to substantiate the role of SETDB1 in HCC tumorigenesis. In conclusion, the upregulation of SETDB1 is associated with a poorer prognosis in HCC patients and inversely correlates with immune cell infiltration, potentially serving as a predictive marker for immunotherapy response.
Collapse
Affiliation(s)
- Chang-Qing Yin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Laboratory of Gene Therapeutic Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Chun-Qing Song
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Laboratory of Gene Therapeutic Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
6
|
Yang Y, Dong S, You B, Zhou C. Dual roles of human endogenous retroviruses in cancer progression and antitumor immune response. Biochim Biophys Acta Rev Cancer 2024; 1879:189201. [PMID: 39427821 DOI: 10.1016/j.bbcan.2024.189201] [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: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Human endogenous retroviruses (HERVs) are a class of transposable elements formed by the integration of ancient retroviruses into the germline genome. They are inherited in a Mendelian manner and approximately constitute 8 % of the human genome. HERVs were considered as "junk DNA" for decades, but increasing evidence suggests that they play significant roles in pathological inflammation, neural differentiation, and oncogenesis. Specifically, HERVs expression has been implicated in several oncogenic processes and the formation of the tumor microenvironment. Indeed, the dual roles of HERVs in cancer, serving as both promoters of oncogenesis and forerunners of the innate antitumor immune response, remain a subject of debate. In this review, we will discuss how HERVs participate in cancer progression and how they are regulated. Our aim is to provide a comprehensive understanding of the fundamental properties and potential function of HERVs in propagating oncogenesis and activating the antitumor immune response. We hope that updated knowledge will reshape our understanding of the critical roles played by HERVs in human evolution and cancer progression.
Collapse
Affiliation(s)
- Yang Yang
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| | - Surong Dong
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Benshuai You
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| | - Chenglin Zhou
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| |
Collapse
|
7
|
Gold S, Shilatifard A. Epigenetic therapies targeting histone lysine methylation: complex mechanisms and clinical challenges. J Clin Invest 2024; 134:e183391. [PMID: 39403928 PMCID: PMC11473148 DOI: 10.1172/jci183391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
As epigenetic therapies continue to gain ground as potential treatment strategies for cancer and other diseases, compounds that target histone lysine methylation and the enzyme complexes represent a major frontier for therapeutic development. Clinically viable therapies targeting the activities of histone lysine methyltransferases (HKMT) and demethylases (HKDMs) have only recently begun to emerge following FDA approval of the EZH2 inhibitor tazemetostat in 2020 and remain limited to compounds targeting the well-studied SET domain-containing HKMTs and their opposing HKDMs. These include the H3K27 methyltransferases EZH2/EZH1, the singular H3K79 methyltransferase DOT1L, and the H3K4 methyltransferase MLL1/COMPASS as well as H3K9 and H3K36 methyltransferases. They additionally include the H3K4/9-preferential demethylase LSD1 and the H3K4-, H3K27-, and H3K36-preferential KDM5, KDM6, and KDM2 demethylase subfamilies, respectively. This Review discusses the results of recent clinical and preclinical studies relevant to all of these existing and potential therapies. It provides an update on advancements in therapeutic development, as well as more basic molecular understanding, within the past 5 years approximately. It also offers a perspective on histone lysine methylation that departs from the long-predominant "histone code" metaphor, emphasizing complex-disrupting inhibitors and proximity-based approaches rather than catalytic domain inhibitors in the outlook for future therapeutic development.
Collapse
|
8
|
Zheng J, Feng H, Lin J, Zhou J, Xi Z, Zhang Y, Ling F, Liu Y, Wang J, Hou T, Xing F, Li Y. KDM3A Ablation Activates Endogenous Retrovirus Expression to Stimulate Antitumor Immunity in Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309983. [PMID: 39031630 PMCID: PMC11515915 DOI: 10.1002/advs.202309983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/04/2024] [Indexed: 07/22/2024]
Abstract
The success of immunotherapy for cancer treatment is limited by the presence of an immunosuppressive tumor microenvironment (TME); Therefore, identifying novel targets to that can reverse this immunosuppressive TME and enhance immunotherapy efficacy is essential. In this study, enrichment analysis based on publicly available single-cell and bulk RNA sequencing data from gastric cancer patients are conducted, and found that tumor-intrinsic interferon (IFN) plays a central role in TME regulation. The results shows that KDM3A over-expression suppresses the tumor-intrinsic IFN response and inhibits KDM3A, either genomically or pharmacologically, which effectively promotes IFN responses by activating endogenous retroviruses (ERVs). KDM3A ablation reconfigures the dsRNA-MAVS-IFN axis by modulating H3K4me2, enhancing the infiltration and function of CD8 T cells, and simultaneously reducing the presence of regulatory T cells, resulting in a reshaped TME in vivo. In addition, combining anti-PD1 therapy with KDM3A inhibition effectively inhibited tumor growth. In conclusions, this study highlights KDM3A as a potential target for TME remodeling and the enhancement of antitumor immunity in gastric cancer through the regulation of the ERV-MAVS-IFN axis.
Collapse
Affiliation(s)
- Jiabin Zheng
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Huolun Feng
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong510006China
| | - Jiatong Lin
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong510006China
| | - Jianlong Zhou
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhihui Xi
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong510006China
| | - Yucheng Zhang
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Fa Ling
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yongfeng Liu
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Junjiang Wang
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Tieying Hou
- Medical Experimental CenterShenzhen Nanshan People's HospitalShenzhenGuangdong518052China
- Shenzhen University Medical SchoolShenzhenGuangdong518073China
| | - Fan Xing
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouGuangdong510080China
| | - Yong Li
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong510006China
| |
Collapse
|
9
|
Rødland GE, Temelie M, Eek Mariampillai A, Hauge S, Gilbert A, Chevalier F, Savu DI, Syljuåsen RG. Potential Benefits of Combining Proton or Carbon Ion Therapy with DNA Damage Repair Inhibitors. Cells 2024; 13:1058. [PMID: 38920686 PMCID: PMC11201490 DOI: 10.3390/cells13121058] [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: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The use of charged particle radiotherapy is currently increasing, but combination therapy with DNA repair inhibitors remains to be exploited in the clinic. The high-linear energy transfer (LET) radiation delivered by charged particles causes clustered DNA damage, which is particularly effective in destroying cancer cells. Whether the DNA damage response to this type of damage is different from that elicited in response to low-LET radiation, and if and how it can be targeted to increase treatment efficacy, is not fully understood. Although several preclinical studies have reported radiosensitizing effects when proton or carbon ion irradiation is combined with inhibitors of, e.g., PARP, ATR, ATM, or DNA-PKcs, further exploration is required to determine the most effective treatments. Here, we examine what is known about repair pathway choice in response to high- versus low-LET irradiation, and we discuss the effects of inhibitors of these pathways when combined with protons and carbon ions. Additionally, we explore the potential effects of DNA repair inhibitors on antitumor immune signaling upon proton and carbon ion irradiation. Due to the reduced effect on healthy tissue and better immune preservation, particle therapy may be particularly well suited for combination with DNA repair inhibitors.
Collapse
Affiliation(s)
- Gro Elise Rødland
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Mihaela Temelie
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Adrian Eek Mariampillai
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France (F.C.)
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France (F.C.)
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
10
|
Fang Y, Barrows D, Dabas Y, Carroll T, Singer S, Tap W, Nacev B. ATRX guards against aberrant differentiation in mesenchymal progenitor cells. Nucleic Acids Res 2024; 52:4950-4968. [PMID: 38477352 PMCID: PMC11109985 DOI: 10.1093/nar/gkae160] [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: 01/19/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Alterations in the tumor suppressor ATRX are recurrently observed in mesenchymal neoplasms. ATRX has multiple epigenetic functions including heterochromatin formation and maintenance and regulation of transcription through modulation of chromatin accessibility. Here, we show in murine mesenchymal progenitor cells (MPCs) that Atrx deficiency aberrantly activated mesenchymal differentiation programs. This includes adipogenic pathways where ATRX loss induced expression of adipogenic transcription factors and enhanced adipogenic differentiation in response to differentiation stimuli. These changes are linked to loss of heterochromatin near mesenchymal lineage genes together with increased chromatin accessibility and gains of active chromatin marks. We additionally observed depletion of H3K9me3 at transposable elements, which are derepressed including near mesenchymal genes where they could serve as regulatory elements. Finally, we demonstrated that loss of ATRX in a mesenchymal malignancy, undifferentiated pleomorphic sarcoma, results in similar epigenetic disruption and de-repression of transposable elements. Together, our results reveal a role for ATRX in maintaining epigenetic states and transcriptional repression in mesenchymal progenitors and tumor cells and in preventing aberrant differentiation in the progenitor context.
Collapse
Affiliation(s)
- Yan Fang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Douglas Barrows
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065, USA
| | - Yakshi Dabas
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065, USA
| | - Sam Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065, USA
| | - Benjamin A Nacev
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Hassanie H, Penteado AB, de Almeida LC, Calil RL, da Silva Emery F, Costa-Lotufo LV, Trossini GHG. SETDB1 as a cancer target: challenges and perspectives in drug design. RSC Med Chem 2024; 15:1424-1451. [PMID: 38799223 PMCID: PMC11113007 DOI: 10.1039/d3md00366c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/16/2024] [Indexed: 05/29/2024] Open
Abstract
Genome stability is governed by chromatin structural dynamics, which modify DNA accessibility under the influence of intra- and inter-nucleosomal contacts, histone post-translational modifications (PTMs) and variations, besides the activity of ATP-dependent chromatin remodelers. These are the main ways by which chromatin dynamics are regulated and connected to nuclear processes, which when dysregulated can frequently be associated with most malignancies. Recently, functional crosstalk between histone modifications and chromatin remodeling has emerged as a critical regulatory method of transcriptional regulation during cell destiny choice. Therefore, improving therapeutic outcomes for patients by focusing on epigenetic targets dysregulated in malignancies should help prevent cancer cells from developing resistance to anticancer treatments. For this reason, SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) has gained a lot of attention recently as a cancer target. SETDB1 is a histone lysine methyltransferase that plays an important role in marking euchromatic and heterochromatic regions. Hence, it promotes the silencing of tumor suppressor genes and contributes to carcinogenesis. Some studies revealed that SETDB1 was overexpressed in various human cancer types, which enhanced tumor growth and metastasis. Thus, SETDB1 appears to be an attractive epigenetic target for new cancer treatments. In this review, we have discussed the effects of its overexpression on the progression of tumors and the development of inhibitor drugs that specifically target this enzyme.
Collapse
Affiliation(s)
- Haifa Hassanie
- School of Pharmaceutical Sciences, University of São Paulo Brazil
| | | | | | | | - Flávio da Silva Emery
- School of Pharmaceutical Sciences of the Ribeirão Preto, University of São Paulo Brazil
| | | | | |
Collapse
|
12
|
Huang KCY, Chiang SF, Chang HY, Hong WZ, Chen JY, Lee PC, Liang JA, Ke TW, Peng SL, Shiau AC, Chen TW, Yang PC, Chen WTL, Chao KSC. Colorectal cancer-specific IFNβ delivery overcomes dysfunctional dsRNA-mediated type I interferon signaling to increase the abscopal effect of radiotherapy. J Immunother Cancer 2024; 12:e008515. [PMID: 38749537 PMCID: PMC11097864 DOI: 10.1136/jitc-2023-008515] [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] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Cancer-intrinsic type I interferon (IFN-I) production triggered by radiotherapy (RT) is mainly dependent on cytosolic double-stranded DNA (dsDNA)-mediated cGAS/STING signaling and increases cancer immunogenicity and enhances the antitumor immune response to increase therapeutic efficacy. However, cGAS/STING deficiency in colorectal cancer (CRC) may suppress the RT-induced antitumor immunity. Therefore, we aimed to evaluate the importance of the dsRNA-mediated antitumor immune response induced by RT in patients with CRC. METHODS Cytosolic dsRNA level and its sensors were evaluated via cell-based assays (co-culture assay, confocal microscopy, pharmacological inhibition and immunofluorescent staining) and in vivo experiments. Biopsies and surgical tissues from patients with CRC who received preoperative chemoradiotherapy (neoCRT) were collected for multiplex cytokine assays, immunohistochemical analysis and SNP genotyping. We also generated a cancer-specific adenovirus-associated virus (AAV)-IFNβ1 construct to evaluate its therapeutic efficacy in combination with RT, and the immune profiles were analyzed by flow cytometry and RNA-seq. RESULTS Our studies revealed that RT stimulates the autonomous release of dsRNA from cancer cells to activate TLR3-mediated IFN-I signatures to facilitate antitumor immune responses. Patients harboring a dysfunctional TLR3 variant had reduced serum levels of IFN-I-related cytokines and intratumoral CD8+ immune cells and shorter disease-free survival following neoCRT treatment. The engineered cancer-targeted construct AAV-IFNβ1 significantly improved the response to RT, leading to systematic eradication of distant tumors and prolonged survival in defective TLR3 preclinical models. CONCLUSION Our results support that increasing cancer-intrinsic IFNβ1 expression is an immunotherapeutic strategy that enhances the RT-induced antitumor immune response in locally patients with advanced CRC with dysfunctional TLR3.
Collapse
Affiliation(s)
- Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
- Translation Research Core, China Medical University Hospital, Taichung, Taiwan
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital Ministry of Health and Welfare, Taichung, Taiwan
| | - Hsin-Yu Chang
- Translation Research Core, China Medical University Hospital, Taichung, Taiwan
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Ze Hong
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
| | - Jhen-Yu Chen
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
- Translation Research Core, China Medical University Hospital, Taichung, Taiwan
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Chih Lee
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Tao-Wei Ke
- Department of Colorectal Cancer, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - An-Cheng Shiau
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Tsung-Wei Chen
- Department of Pathology, Asia University, Taichung, Taiwan
| | - Pei-Chen Yang
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
| | - William Tzu-Liang Chen
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Colorectal Cancer, China Medical University Hospital, Taichung, Taiwan
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University Hospital, HsinChu, Taiwan
| | - K S Clifford Chao
- Proton Cancer, China Medical University Hospital, Taichung, Taiwan
- Department of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
13
|
Prashanth S, Radha Maniswami R, Rajajeyabalachandran G, Jegatheesan SK. SETDB1, an H3K9-specific methyltransferase: An attractive epigenetic target to combat cancer. Drug Discov Today 2024; 29:103982. [PMID: 38614159 DOI: 10.1016/j.drudis.2024.103982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) is an important epigenetic regulator catalyzing histone H3 lysine 9 (H3K9) methylation, specifically di-/tri-methylation. This regulation promotes gene silencing through heterochromatin formation. Aberrant SETDB1 expression, and its oncogenic role is evident in many cancers. Thus, SETDB1 is a valid target with novel therapeutic benefits. In this review, we explore the structural and biochemical features of SETDB1, its regulatory mechanisms, and its role in various cancers. We also discuss recent discoveries in small molecules targeting SETDB1 and provide suggestions for future research.
Collapse
Affiliation(s)
- Seema Prashanth
- Informatics, AI & ML, Jubilant Biosys Ltd., Bangalore, India
| | | | | | | |
Collapse
|
14
|
Ma T, Xu F, Hou Y, Shu Y, Zhao Z, Zhang Y, Bai L, Feng L, Zhong L. SETDB1: Progress and prospects in cancer treatment potential and inhibitor research. Bioorg Chem 2024; 145:107219. [PMID: 38377821 DOI: 10.1016/j.bioorg.2024.107219] [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: 12/27/2023] [Revised: 02/03/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024]
Abstract
SET domain bifurcated methyltransferase 1 (SETDB1) serves as a histone lysine methyltransferase, catalyzing the di- and tri-methylation of histone H3K9. Mounting evidence indicates that the abnormal expression or activity of SETDB1, either through amplification or mutation, plays a crucial role in tumorigenesis and progression. This is particularly evident in the context of tumor immune evasion and resistance to immune checkpoint blockade therapy. Furthermore, there is a robust association between SETDB1 dysregulation and an unfavorable prognosis across various types of tumors. The oncogenic role of SETDB1 primarily arises from its methyltransferase function, which contributes to the establishment of a condensed and transcriptionally inactive heterochromatin state. This results in the inactivation of genes that typically hinder cancer development and silencing of retrotransposons that could potentially trigger an immune response. These findings underscore the substantial potential for SETDB1 as an anti-tumor therapeutic target. Nevertheless, despite significant strides in recent years in tumor biology research, challenges persist in SETDB1-targeted therapy. To better facilitate the development of anti-tumor therapy targeting SETDB1, we have conducted a comprehensive review of SETDB1 in this account. We present the structure and function of SETDB1, its role in various tumors and immune regulation, as well as the advancements made in SETDB1 antagonists. Furthermore, we discuss the challenges encountered and provide perspectives for the development of SETDB1-targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Tingnan Ma
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Feifei Xu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China; State Key Laboratory of Southwestern Chinese Medicine Resources; Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry of Education, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yingying Hou
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Yongquan Shu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Zhipeng Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Yaru Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Lu Feng
- Department of Emergency, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610041, China.
| | - Lei Zhong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China.
| |
Collapse
|
15
|
Yang H, Sui L, Cai C, Chu H, Diao Y. SETDB1 promotes progression through upregulation of SF3B4 expression and regulates the immunity in ovarian cancer. J Ovarian Res 2024; 17:34. [PMID: 38317200 PMCID: PMC10840244 DOI: 10.1186/s13048-024-01358-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecologic malignant tumour. The mechanism promoting OC initiation and progression remains unclear. SET domain bifurcated histone lysine methyltransferase 1(SETDB1) acts as an oncogene in a variety of tumours. This study aims to explore the role of SETDB1 in OC. METHODS GEO, TCGA, CSIOVDB and CPTAC databases jointly analysed SETDB1 mRNA and protein expression. Effect of SETDB1 expression on the clinical prognosis of OC patients was analysed through online Kaplan‒Meier plotter and CSIOVDB database. Then, the effect of SETDB1 in OC cells progression and mobility was examined using MTT, EdU, colony formation and transwell assay. Additionally, Cistrome DB database was used to visualize the binding of SETDB1 protein and splicing factor 3b subunit 4 (SF3B4) promoter, and dual-luciferase reporter gene assay was performed to confirm the interaction. Finally, bioinformatics analysis was employed to reveal the relationship between SETDB1 and the microenvironment of OC. RESULTS In the present study, we found that SETDB1 was obviously upregulated in OC and its overexpression predicted poor prognosis of OC patients. Then, we verified that SETDB1 promoted the progression and motility of OC cells in vitro. Knockdown of SETDB1 had the opposite effect. Further research showed that SETDB1 acted as a transcription factor to activate SF3B4 expression. SF3B4 knockdown impaired the effect of SETDB1 to promote the proliferative capacity and motility of OC cells. Finally, the results of bioinformatics analysis confirmed that SETDB1 regulated the immune microenvironment of ovarian cancer. CONCLUSION SETDB1 promoted ovarian cancer progression by upregulating the expression of SF3B4 and inhibiting the tumour immunity. SETDB1 may be a promising prognostic and therapeutic marker for OC.
Collapse
Affiliation(s)
- Hongjuan Yang
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong Province, China
| | - Lei Sui
- Department of Gynecological Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266000, Shandong, China
| | - Cuicui Cai
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong Province, China
| | - Huijun Chu
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong Province, China
| | - Yuchao Diao
- Department of Obstetrics and Gynecology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
16
|
Jackson-Jones KA, McKnight Á, Sloan RD. The innate immune factor RPRD2/REAF and its role in the Lv2 restriction of HIV. mBio 2023; 14:e0257221. [PMID: 37882563 PMCID: PMC10746242 DOI: 10.1128/mbio.02572-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
Abstract
Intracellular innate immunity involves co-evolved antiviral restriction factors that specifically inhibit infecting viruses. Studying these restrictions has increased our understanding of viral replication, host-pathogen interactions, and pathogenesis, and represent potential targets for novel antiviral therapies. Lentiviral restriction 2 (Lv2) was identified as an unmapped early-phase restriction of HIV-2 and later shown to also restrict HIV-1 and simian immunodeficiency virus. The viral determinants of Lv2 susceptibility have been mapped to the envelope and capsid proteins in both HIV-1 and HIV-2, and also viral protein R (Vpr) in HIV-1, and appears dependent on cellular entry mechanism. A genome-wide screen identified several likely contributing host factors including members of the polymerase-associated factor 1 (PAF1) and human silencing hub (HUSH) complexes, and the newly characterized regulation of nuclear pre-mRNA domain containing 2 (RPRD2). Subsequently, RPRD2 (or RNA-associated early-stage antiviral factor) has been shown to be upregulated upon T cell activation, is highly expressed in myeloid cells, binds viral reverse transcripts, and potently restricts HIV-1 infection. RPRD2 is also bound by HIV-1 Vpr and targeted for degradation by the proteasome upon reverse transcription, suggesting RPRD2 impedes reverse transcription and Vpr targeting overcomes this block. RPRD2 is mainly localized to the nucleus and binds RNA, DNA, and DNA:RNA hybrids. More recently, RPRD2 has been shown to negatively regulate genome-wide transcription and interact with the HUSH and PAF1 complexes which repress HIV transcription and are implicated in maintenance of HIV latency. In this review, we examine Lv2 restriction and the antiviral role of RPRD2 and consider potential mechanism(s) of action.
Collapse
Affiliation(s)
- Kathryn A. Jackson-Jones
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
- Division of Infectious Diseases & Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Áine McKnight
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Richard D. Sloan
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
- ZJU-UoE Institute, Zhejiang University, Haining, China
| |
Collapse
|
17
|
Luo H, Wu X, Zhu XH, Yi X, Du D, Jiang DS. The functions of SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) in biological process and disease. Epigenetics Chromatin 2023; 16:47. [PMID: 38057834 DOI: 10.1186/s13072-023-00519-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/27/2023] [Indexed: 12/08/2023] Open
Abstract
Histone methyltransferase SETDB1 (SET domain bifurcated histone lysine methyltransferase 1, also known as ESET or KMT1E) is known to be involved in the deposition of the di- and tri-methyl marks on H3K9 (H3K9me2 and H3K9me3), which are associated with transcription repression. SETDB1 exerts an essential role in the silencing of endogenous retroviruses (ERVs) in embryonic stem cells (mESCs) by tri-methylating H3K9 (H3K9me3) and interacting with DNA methyltransferases (DNMTs). Additionally, SETDB1 is engaged in regulating multiple biological processes and diseases, such as ageing, tumors, and inflammatory bowel disease (IBD), by methylating both histones and non-histone proteins. In this review, we provide an overview of the complex biology of SETDB1, review the upstream regulatory mechanisms of SETDB1 and its partners, discuss the functions and molecular mechanisms of SETDB1 in cell fate determination and stem cell, as well as in tumors and other diseases. Finally, we discuss the current challenges and prospects of targeting SETDB1 for the treatment of different diseases, and we also suggest some future research directions in the field of SETDB1 research.
Collapse
Affiliation(s)
- Hanshen Luo
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Xingliang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue-Hai Zhu
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dunfeng Du
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Ferlita AL, Nigita G, Tsyba L, Palamarchuk A, Alaimo S, Pulvirenti A, Balatti V, Rassenti L, Tsichlis PN, Kipps T, Pekarsky Y, Croce CM. Expression signature of human endogenous retroviruses in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2023; 120:e2307593120. [PMID: 37871223 PMCID: PMC10622969 DOI: 10.1073/pnas.2307593120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is one of the most diagnosed forms of leukemia worldwide and it is usually classified into two forms: indolent and aggressive. These two forms are characterized by distinct molecular features that drive different responses to treatment and clinical outcomes. In this context, a better understanding of the molecular landscape of the CLL forms may potentially lead to the development of new drugs or the identification of novel biomarkers. Human endogenous retroviruses (HERVs) are a class of transposable elements that have been associated with the development of different human cancers, including different forms of leukemias. However, no studies about HERVs in CLL have ever been reported so far. Here, we present the first locus-specific profiling of HERV expression in both the aggressive and indolent forms of CLL. Our analyses revealed several dysregulations in HERV expression occurring in CLL and some of them were specific for either the aggressive or indolent form of CLL. Such results were also validated by analyzing an external cohort of CLL patients and by RT-qPCR. Moreover, in silico analyses have shown relevant signaling pathways associated with them suggesting a potential involvement of the dysregulated HERVs in these pathways and consequently in CLL development.
Collapse
Affiliation(s)
- Alessandro La Ferlita
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Liudmyla Tsyba
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Alexey Palamarchuk
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Salvatore Alaimo
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania95123, Italy
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania95123, Italy
| | - Veronica Balatti
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Laura Rassenti
- Department of Medicine, University of California San Diego, La JollaCA92093
| | - Philip N. Tsichlis
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Thomas Kipps
- Department of Medicine, University of California San Diego, La JollaCA92093
| | - Yuri Pekarsky
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH43210
| |
Collapse
|
19
|
Musella M, Manduca N, Maccafeo E, Sistigu A. Epigenetics behind tumor immunology: a mini review. Oncogene 2023; 42:2932-2938. [PMID: 37604925 DOI: 10.1038/s41388-023-02791-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/23/2023]
Abstract
Immunogenic- and immune-therapies have become hot spots in the treatment of cancer. Although promising, these strategies are frequently associated with innate or acquired resistance, calling for combined targeting of immune inhibitory signals. Epigenetic therapy is attracting considerable attention as a combination partner for immune-based therapies due to its role in molding the state and fate of cancer and immune cells in the tumor microenvironment. Here, we describe epigenetic dysregulations in cancer, with a particular focus on those related to innate immune signaling and Type I interferons, and emphasize opportunities and current efforts to translate this knowledge into treatment regimens with improved clinical benefit.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
- Fondazione Policlinico Universitario 'A. Gemelli' - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168, Rome, Italy.
| |
Collapse
|
20
|
Fang Y, Barrows D, Dabas Y, Carroll TS, Tap WD, Nacev BA. ATRX guards against aberrant differentiation in mesenchymal progenitor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552433. [PMID: 37609273 PMCID: PMC10441338 DOI: 10.1101/2023.08.08.552433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Alterations in the tumor suppressor ATRX are recurrently observed in several cancer types including sarcomas, which are mesenchymal neoplasms. ATRX has multiple epigenetic functions including heterochromatin formation and maintenance and regulation of transcription through modulation of chromatin accessibility. Here, we show in murine mesenchymal progenitor cells (MPCs) that Atrx deficiency aberrantly activated mesenchymal differentiation programs. This includes adipogenic pathways where ATRX loss induced expression of adipogenic transcription factors (Pparγ and Cebpα) and enhanced adipogenic differentiation in response to differentiation stimuli. These changes are linked to loss of heterochromatin near mesenchymal lineage genes together with increased chromatin accessibility and gains of active chromatin marks at putative enhancer elements and promoters. Finally, we observed depletion of H3K9me3 at transposable elements, which are derepressed including near mesenchymal genes where they could serve as regulatory elements. Our results demonstrate that ATRX functions to buffer against differentiation in mesenchymal progenitor cells, which has implications for understanding ATRX loss of function in sarcomas.
Collapse
Affiliation(s)
- Yan Fang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY10065
| | - Douglas Barrows
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065
| | - Yakshi Dabas
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY10065
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller University, New York, NY10065
| | - William D. Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY10065
| | - Benjamin A. Nacev
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213
| |
Collapse
|
21
|
Carelock ME, Master RP, Kim MC, Jin Z, Wang L, Maharjan CK, Hua N, De U, Kolb R, Xiao Y, Liao D, Zheng G, Zhang W. Targeting intracellular proteins with cell type-specific functions for cancer immunotherapy. LIFE MEDICINE 2023; 2:lnad019. [PMID: 39872303 PMCID: PMC11749652 DOI: 10.1093/lifemedi/lnad019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/24/2023] [Indexed: 01/30/2025]
Abstract
Immune checkpoint inhibitors (ICIs) use antibodies that block cell surface immune checkpoint proteins with great efficacy in treating immunogenic or "immune hot" tumors such as melanoma, kidney, and lung adenocarcinoma. ICIs have limited response rates to other non-immunogenic cancers. The tumor microenvironment (TME) consists of many cell types that collectively promote tumor progression. Cancer therapeutics are commonly designed to target one molecule in one defined cell type. There is growing evidence that long-term therapeutic responses require the targeting of cancer cells and tumor-promoting populations within the TME. The question remains whether we can identify targetable molecules/pathways that are critical for multiple cell types. Here, we will discuss several molecular targets that may fit a "two or multiple birds, one stone" model, including the B-cell lymphoma-2 (BCL-2) family pro-survival factors, transcriptional factors including signal transducer and activator of transcription 3, the nuclear receptor 4A family (NR4A1, NR4A2, and NR4A3), as well as epigenetic regulators such as bromodomain and extra-terminal (BET) family proteins, histone deacetylase family, SET domain bifurcated histone lysine methyltransferase 1 (SETDB1), and lysine-specific demethylase 1 (LSD1/KDM1A). We will focus on the rationale of these targets in immune modulation, as well as the strategies for targeting these important proteins for cancer therapy.
Collapse
Affiliation(s)
- Madison E Carelock
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan P Master
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Diagnostic Laboratory Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Research Institute of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Immunology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Chandra K Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nan Hua
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Umasankar De
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Yufeng Xiao
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Daiqing Liao
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Guangrong Zheng
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Immunology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
22
|
Johnson E, Salari K, Yang S. SETDB1: A perspective into immune cell function and cancer immunotherapy. Immunology 2023; 169:3-12. [PMID: 36524435 PMCID: PMC10121739 DOI: 10.1111/imm.13619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Oncogene SET Domain Bifurcated 1 (SETDB1)/ESET, an H3K9 methyltransferase, was originally discovered over two decades ago; however, its function in the immune response was not first reported until 2011. SETDB1 immune functions include B cell maturation, T cell activity regulation, and immune escape in cancer cells. In B lymphocytes, SETDB1 mediates the transition from pro-B to pre-B cells and represses endogenous retroviruses (ERV) to encourage B cell lineage differentiation and maturation. SETDB1 alters T cell function by methylating IL-2 and IL-17 promoters and mediating T cell lineage commitment and development. In addition, SETDB1 plays a critical role in ERV silencing within a variety of immune cells, which can indirectly weaken the immune response. Although SETDB1 is critical for normal immune cell function, overexpression in cancer cells negatively impacts immune cell fights against cancer through decreased tumour immunogenicity. Within cancer cells, SETDB1 overexpression represses production and infiltration of antitumour immune cells, mediates immune escape through TE and ERV silencing, represses the type I interferon pathway, and interferes in immune checkpoint blockade (ICB) outcomes by regulation of PD-L1 expression and IFN signalling. In this review, we further discuss the immunological mechanisms of SETDB1 in normal and cancerous cells and its implications in cancer immunotherapy.
Collapse
Affiliation(s)
- Eleanor Johnson
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kiarash Salari
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Shujie Yang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
23
|
Zhao Z, Feng L, Peng X, Ma T, Tong R, Zhong L. Role of histone methyltransferase SETDB1 in regulation of tumourigenesis and immune response. Front Pharmacol 2022; 13:1073713. [PMID: 36582533 PMCID: PMC9793902 DOI: 10.3389/fphar.2022.1073713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Epigenetic alterations are implicated in tumour immune evasion and immune checkpoint blockade (ICB) resistance. SET domain bifurcated histone methyltransferase 1 (SETDB1) is a histone lysine methyltransferase that catalyses histone H3K9 di- and tri-methylation on euchromatin, and growing evidence indicates that SETDB1 amplification and abnormal activation are significantly correlated with the unfavourable prognosis of multiple malignant tumours and contribute to tumourigenesis and progression, immune evasion and ICB resistance. The main underlying mechanism is H3K9me3 deposition by SETDB1 on tumour-suppressive genes, retrotransposons, and immune genes. SETDB1 targeting is a promising approach to cancer therapy, particularly immunotherapy, because of its regulatory effects on endogenous retroviruses. However, SETDB1-targeted therapy remains challenging due to potential side effects and the lack of antagonists with high selectivity and potency. Here, we review the role of SETDB1 in tumourigenesis and immune regulation and present the current challenges and future perspectives of SETDB1 targeted therapy.
Collapse
Affiliation(s)
- Zhipeng Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Feng
- Department of Emergency, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
| | - Xuerun Peng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingnan Ma
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Zhong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China,*Correspondence: Lei Zhong,
| |
Collapse
|
24
|
Eek Mariampillai A, Hauge S, Øynebråten I, Rødland GE, Corthay A, Syljuåsen RG. Caspase activation counteracts interferon signaling after G2 checkpoint abrogation by ATR inhibition in irradiated human cancer cells. Front Oncol 2022; 12:981332. [PMID: 36387237 PMCID: PMC9650454 DOI: 10.3389/fonc.2022.981332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022] Open
Abstract
Recent studies suggest that inhibition of the ATR kinase can potentiate radiation-induced antitumor immune responses, but the extent and mechanisms of such responses in human cancers remain scarcely understood. We aimed to assess whether the ATR inhibitors VE822 and AZD6738, by abrogating the G2 checkpoint, increase cGAS-mediated type I IFN response after irradiation in human lung cancer and osteosarcoma cell lines. Supporting that the checkpoint may prevent IFN induction, radiation-induced IFN signaling declined when the G2 checkpoint arrest was prolonged at high radiation doses. G2 checkpoint abrogation after co-treatment with radiation and ATR inhibitors was accompanied by increased radiation-induced IFN signaling in four out of five cell lines tested. Consistent with the hypothesis that the cytosolic DNA sensor cGAS may detect DNA from ruptured micronuclei after G2 checkpoint abrogation, cGAS co-localized with micronuclei, and depletion of cGAS or STING abolished the IFN responses. Contrastingly, one lung cancer cell line showed no increase in IFN signaling despite irradiation and G2 checkpoint abrogation. This cell line showed a higher level of the exonuclease TREX1 than the other cell lines, but TREX1 depletion did not enhance IFN signaling. Rather, addition of a pan-caspase inhibitor restored the IFN response in this cell line and also increased the responses in the other cell lines. These results show that treatment-induced caspase activation can suppress the IFN response after co-treatment with radiation and ATR inhibitors. Caspase activation thus warrants further consideration as a possible predictive marker for lack of IFN signaling.
Collapse
Affiliation(s)
- Adrian Eek Mariampillai
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Inger Øynebråten
- Tumor Immunology Lab, Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Gro Elise Rødland
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub – Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- *Correspondence: Randi G. Syljuåsen,
| |
Collapse
|