1
|
Mao W, Zhang H, Wang K, Geng J, Wu J. Research progress of MUC1 in genitourinary cancers. Cell Mol Biol Lett 2024; 29:135. [PMID: 39491020 PMCID: PMC11533421 DOI: 10.1186/s11658-024-00654-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: 06/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
MUC1 is a highly glycosylated transmembrane protein with a high molecular weight. It plays a role in lubricating and protecting mucosal epithelium, participates in epithelial cell renewal and differentiation, and regulates cell adhesion, signal transduction, and immune response. MUC1 is expressed in both normal and malignant epithelial cells, and plays an important role in the diagnosis, prognosis prediction and clinical monitoring of a variety of tumors and is expected to be a new therapeutic target. This article reviews the structural features, expression regulation mechanism, and research progress of MUC1 in the development of genitourinary cancers and its clinical applications.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| | - Houliang Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Keyi Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Jiang Geng
- Department of Urology, Bengbu First People's Hospital, Bengbu, People's Republic of China.
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Jianping Wu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
2
|
Galassi C, Esteller M, Vitale I, Galluzzi L. Epigenetic control of immunoevasion in cancer stem cells. Trends Cancer 2024; 10:1052-1071. [PMID: 39244477 DOI: 10.1016/j.trecan.2024.08.004] [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: 03/30/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
Cancer stem cells (CSCs) are a poorly differentiated population of malignant cells that (at least in some neoplasms) is responsible for tumor progression, resistance to therapy, and disease relapse. According to a widely accepted model, all stages of cancer progression involve the ability of neoplastic cells to evade recognition or elimination by the host immune system. In line with this notion, CSCs are not only able to cope with environmental and therapy-elicited stress better than their more differentiated counterparts but also appear to better evade tumor-targeting immune responses. We summarize epigenetic modifications of DNA and histones through which CSCs evade immune recognition or elimination, and propose that such alterations constitute promising therapeutic targets to increase the sensitivity of some malignancies to immunotherapy.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Ning B, Huang T, Liu Y, Wei Y. A depression-related lncRNA signature predicts the clinical outcome and immune characteristics of gastric cancer. Heliyon 2024; 10:e34399. [PMID: 39144985 PMCID: PMC11320146 DOI: 10.1016/j.heliyon.2024.e34399] [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: 07/29/2023] [Revised: 06/29/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
Background Depression and long non-coding RNA (lncRNA) have been reported to be associated with tumor progression and prognosis in gastric cancer (GC). This study aims to build a GC risk classification and prognosis model based on depression-related lncRNAs (DRLs). Methods To develop a risk model, we performed univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses using RNA sequencing data of GC from The Cancer Genome Atlas (TCGA) and depression-related genes (DRGs) from previous studies. Kaplan-Meier analysis, receiver operating characteristic (ROC) curve analysis, nomogram construction, pathway enrichment analysis, assessment of immunological features, and drug sensitivity testing were conducted using a series of bioinformatics methods. Results Seven DRLs were identified to build a prognostic model, whose robustness was verified in an internal cohort. Subsequent prognostic analyses found that high risk scores were associated with worse overall survival (OS). Univariate and multivariate analyses revealed that the risk score could be used as an independent prognostic factor. Furthermore, the ROC curve indicated that the risk score had higher diagnostic efficiency than traditional clinicopathological features. The calibration curve confirmed the accuracy and reliability of the nomogram. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that there were differences in digestive system and nervous system-related pathways between the high- and low-risk groups. Results of tumor mutational burden (TMB) and tumor immune dysfunction and exclusion (TIDE) analyses indicated that patients in the low-risk group had a better response rate to immunotherapy. Finally, the results of drug sensitivity analysis showed that risk score could influence sensitivity to EHT 1864 in GC. Conclusion We have successfully developed and verified a 7-DRL risk model which can assess the prognosis and immunological features and guide individualized therapy of GC patients.
Collapse
Affiliation(s)
- Biao Ning
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Cancer Clinical Study Center Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
| | - Tianhe Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Cancer Clinical Study Center Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
| | - Yixin Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Cancer Clinical Study Center Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
- Hubei Cancer Clinical Study Center Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei Province, 430071, China
| |
Collapse
|
4
|
Bhattacharya A, Wang K, Penailillo J, Chan CN, Fushimi A, Yamashita N, Daimon T, Haratake N, Ozawa H, Nakashoji A, Shigeta K, Morimoto Y, Miyo M, Kufe DW. MUC1-C regulates NEAT1 lncRNA expression and paraspeckle formation in cancer progression. Oncogene 2024; 43:2199-2214. [PMID: 38802648 PMCID: PMC11226401 DOI: 10.1038/s41388-024-03068-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
The MUC1 gene evolved in mammals for adaptation of barrier tissues in response to infections and damage. Paraspeckles are nuclear bodies formed on the NEAT1 lncRNA in response to loss of homeostasis. There is no known intersection of MUC1 with NEAT1 or paraspeckles. Here, we demonstrate that the MUC1-C subunit plays an essential role in regulating NEAT1 expression. MUC1-C activates the NEAT1 gene with induction of the NEAT1_1 and NEAT1_2 isoforms by NF-κB- and MYC-mediated mechanisms. MUC1-C/MYC signaling also induces expression of the SFPQ, NONO and FUS RNA binding proteins (RBPs) that associate with NEAT1_2 and are necessary for paraspeckle formation. MUC1-C integrates activation of NEAT1 and RBP-encoding genes by recruiting the PBAF chromatin remodeling complex and increasing chromatin accessibility of their respective regulatory regions. We further demonstrate that MUC1-C and NEAT1 form an auto-inductive pathway that drives common sets of genes conferring responses to inflammation and loss of homeostasis. Of functional significance, we find that the MUC1-C/NEAT1 pathway is of importance for the cancer stem cell (CSC) state and anti-cancer drug resistance. These findings identify a previously unrecognized role for MUC1-C in the regulation of NEAT1, RBPs, and paraspeckles that has been co-opted in promoting cancer progression.
Collapse
Affiliation(s)
| | - Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Johany Penailillo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chi Ngai Chan
- Tissue Technologies Unit, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nami Yamashita
- Breast Surgical Oncology, Breast Oncology Center, The Cancer Institute Hospital of the JFCR, Tokyo, Japan
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Keisuke Shigeta
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yoshihiro Morimoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masaaki Miyo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Huang H, Mu Y, Li S. The biological function of Serpinb9 and Serpinb9-based therapy. Front Immunol 2024; 15:1422113. [PMID: 38966643 PMCID: PMC11222584 DOI: 10.3389/fimmu.2024.1422113] [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: 04/23/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024] Open
Abstract
Recent breakthroughs in discovering novel immune signaling pathways have revolutionized different disease treatments. SERPINB9 (Sb9), also known as Proteinase Inhibitor 9 (PI-9), is a well-known endogenous inhibitor of Granzyme B (GzmB). GzmB is a potent cytotoxic molecule secreted by cytotoxic T lymphocytes and natural killer cells, which plays a crucial role in inducing apoptosis in target cells during immune responses. Sb9 acts as a protective mechanism against the potentially harmful effects of GzmB within the cells of the immune system itself. On the other hand, overexpression of Sb9 is an important mechanism of immune evasion in diseases like cancers and viral infections. The intricate functions of Sb9 in different cell types represent a fine-tuned regulatory mechanism for preventing immunopathology, protection against autoimmune diseases, and the regulation of cell death, all of which are essential for maintaining health and responding effectively to disease challenges. Dysregulation of the Sb9 will disrupt human normal physiological condition, potentially leading to a range of diseases, including cancers, inflammatory conditions, viral infections or other pathological disorders. Deepening our understanding of the role of Sb9 will aid in the discovery of innovative and effective treatments for various medical conditions. Therefore, the objective of this review is to consolidate current knowledge regarding the biological role of Sb9. It aims to offer insights into its discovery, structure, functions, distribution, its association with various diseases, and the potential of nanoparticle-based therapies targeting Sb9.
Collapse
Affiliation(s)
- Haozhe Huang
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yiqing Mu
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Song Li
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
6
|
Wang K, Bhattacharya A, Haratake N, Daimon T, Nakashoji A, Ozawa H, Peng B, Li W, Kufe D. XIST and MUC1-C form an auto-regulatory pathway in driving cancer progression. Cell Death Dis 2024; 15:330. [PMID: 38740827 PMCID: PMC11091074 DOI: 10.1038/s41419-024-06684-9] [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: 10/10/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
The long non-coding RNA X-inactive specific transcript (lncRNA XIST) and MUC1 gene are dysregulated in chronic inflammation and cancer; however, there is no known interaction of their functions. The present studies demonstrate that MUC1-C regulates XIST lncRNA levels by suppressing the RBM15/B, WTAP and METTL3/14 components of the m6A methylation complex that associate with XIST A repeats. MUC1-C also suppresses the YTHDF2-CNOT1 deadenylase complex that recognizes m6A sites and contributes to XIST decay with increases in XIST stability and expression. In support of an auto-regulatory pathway, we show that XIST regulates MUC1-C expression by promoting NF-κB-mediated activation of the MUC1 gene. Of significance, MUC1-C and XIST regulate common genes associated with inflammation and stemness, including (i) miR-21 which is upregulated across pan-cancers, and (ii) TDP-43 which associates with the XIST E repeats. Our results further demonstrate that the MUC1-C/XIST pathway (i) is regulated by TDP-43, (ii) drives stemness-associated genes, and (iii) is necessary for self-renewal capacity. These findings indicate that the MUC1-C/XIST auto-regulatory axis is of importance in cancer progression.
Collapse
Affiliation(s)
- Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Ashrafizadeh M, Zhang W, Tian Y, Sethi G, Zhang X, Qiu A. Molecular panorama of therapy resistance in prostate cancer: a pre-clinical and bioinformatics analysis for clinical translation. Cancer Metastasis Rev 2024; 43:229-260. [PMID: 38374496 DOI: 10.1007/s10555-024-10168-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 01/04/2024] [Indexed: 02/21/2024]
Abstract
Prostate cancer (PCa) is a malignant disorder of prostate gland being asymptomatic in early stages and high metastatic potential in advanced stages. The chemotherapy and surgical resection have provided favourable prognosis of PCa patients, but advanced and aggressive forms of PCa including CRPC and AVPC lack response to therapy properly, and therefore, prognosis of patients is deteriorated. At the advanced stages, PCa cells do not respond to chemotherapy and radiotherapy in a satisfactory level, and therefore, therapy resistance is emerged. Molecular profile analysis of PCa cells reveals the apoptosis suppression, pro-survival autophagy induction, and EMT induction as factors in escalating malignant of cancer cells and development of therapy resistance. The dysregulation in molecular profile of PCa including upregulation of STAT3 and PI3K/Akt, downregulation of STAT3, and aberrant expression of non-coding RNAs are determining factor for response of cancer cells to chemotherapy. Because of prevalence of drug resistance in PCa, combination therapy including co-utilization of anti-cancer drugs and nanotherapeutic approaches has been suggested in PCa therapy. As a result of increase in DNA damage repair, PCa cells induce radioresistance and RelB overexpression prevents irradiation-mediated cell death. Similar to chemotherapy, nanomaterials are promising for promoting radiosensitivity through delivery of cargo, improving accumulation in PCa cells, and targeting survival-related pathways. In respect to emergence of immunotherapy as a new tool in PCa suppression, tumour cells are able to increase PD-L1 expression and inactivate NK cells in mediating immune evasion. The bioinformatics analysis for evaluation of drug resistance-related genes has been performed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Yu Tian
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China.
| | - Aiming Qiu
- Department of Geriatrics, the Fifth People's Hospital of Wujiang District, Suzhou, China.
| |
Collapse
|
8
|
Tecalco-Cruz AC, Medina-Abreu KH, Oropeza-Martínez E, Zepeda-Cervantes J, Vázquez-Macías A, Macías-Silva M. Deregulation of interferon-gamma receptor 1 expression and its implications for lung adenocarcinoma progression. World J Clin Oncol 2024; 15:195-207. [PMID: 38455133 PMCID: PMC10915940 DOI: 10.5306/wjco.v15.i2.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Interferon-gamma (IFN-γ) plays a dual role in cancer; it is both a pro- and an antitumorigenic cytokine, depending on the type of cancer. The deregulation of the IFN-γ canonic pathway is associated with several disorders, including vulnerability to viral infections, inflammation, and cancer progression. In particular, the interplay between lung adenocarcinoma (LUAD) and viral infections appears to exist in association with the deregulation of IFN-γ signaling. In this mini-review, we investigated the status of the IFN-γ signaling pathway and the expression level of its components in LUAD. Interestingly, a reduction in IFNGR1 expression seems to be associated with LUAD progression, affecting defenses against viruses such as severe acute respiratory syndrome coronavirus 2. In addition, alterations in the expression of IFNGR1 may inhibit the antiproliferative action of IFN-γ signaling in LUAD.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | - Karen H Medina-Abreu
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | | | - Jesus Zepeda-Cervantes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | - Aleida Vázquez-Macías
- Colegio de Ciencias y Humanidades, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | - Marina Macías-Silva
- Instituo de Fisiología Celular, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| |
Collapse
|
9
|
Perevalova AM, Gulyaeva LF, Pustylnyak VO. Roles of Interferon Regulatory Factor 1 in Tumor Progression and Regression: Two Sides of a Coin. Int J Mol Sci 2024; 25:2153. [PMID: 38396830 PMCID: PMC10889282 DOI: 10.3390/ijms25042153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
IRF1 is a transcription factor well known for its role in IFN signaling. Although IRF1 was initially identified for its involvement in inflammatory processes, there is now evidence that it provides a function in carcinogenesis as well. IRF1 has been shown to affect several important antitumor mechanisms, such as induction of apoptosis, cell cycle arrest, remodeling of tumor immune microenvironment, suppression of telomerase activity, suppression of angiogenesis and others. Nevertheless, the opposite effects of IRF1 on tumor growth have also been demonstrated. In particular, the "immune checkpoint" molecule PD-L1, which is responsible for tumor immune evasion, has IRF1 as a major transcriptional regulator. These and several other properties of IRF1, including its proposed association with response and resistance to immunotherapy and several chemotherapeutic drugs, make it a promising object for further research. Numerous mechanisms of IRF1 regulation in cancer have been identified, including genetic, epigenetic, transcriptional, post-transcriptional, and post-translational mechanisms, although their significance for tumor progression remains to be explored. This review will focus on the established tumor-suppressive and tumor-promoting functions of IRF1, as well as the molecular mechanisms of IRF1 regulation identified in various cancers.
Collapse
Affiliation(s)
- Alina M. Perevalova
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| | - Lyudmila F. Gulyaeva
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| | - Vladimir O. Pustylnyak
- Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Pirogova Street, 1, Novosibirsk 630090, Russia; (A.M.P.)
- Federal Research Center of Fundamental and Translational Medicine, Timakova Street, 2/12, Novosibirsk 630117, Russia
| |
Collapse
|
10
|
Chen X, Sandrine IK, Yang M, Tu J, Yuan X. MUC1 and MUC16: critical for immune modulation in cancer therapeutics. Front Immunol 2024; 15:1356913. [PMID: 38361923 PMCID: PMC10867145 DOI: 10.3389/fimmu.2024.1356913] [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: 12/16/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
The Mucin (MUC) family, a range of highly glycosylated macromolecules, is ubiquitously expressed in mammalian epithelial cells. Such molecules are pivotal in establishing protective mucosal barriers, serving as defenses against pathogenic assaults. Intriguingly, the aberrant expression of specific MUC proteins, notably Mucin 1 (MUC1) and Mucin 16 (MUC16), within tumor cells, is intimately associated with oncogenesis, proliferation, and metastasis. This association involves various mechanisms, including cellular proliferation, viability, apoptosis resistance, chemotherapeutic resilience, metabolic shifts, and immune surveillance evasion. Due to their distinctive biological roles and structural features in oncology, MUC proteins have attracted considerable attention as prospective targets and biomarkers in cancer therapy. The current review offers an exhaustive exploration of the roles of MUC1 and MUC16 in the context of cancer biomarkers, elucidating their critical contributions to the mechanisms of cellular signal transduction, regulation of immune responses, and the modulation of the tumor microenvironment. Additionally, the article evaluates the latest advances in therapeutic strategies targeting these mucins, focusing on innovations in immunotherapies and targeted drugs, aiming to enhance customization and accuracy in cancer treatments.
Collapse
Affiliation(s)
| | | | | | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Li BE, Li GY, Cai W, Zhu Q, Seruggia D, Fujiwara Y, Vakoc CR, Orkin SH. In vivo CRISPR/Cas9 screening identifies Pbrm1 as a regulator of myeloid leukemia development in mice. Blood Adv 2023; 7:5281-5293. [PMID: 37428871 PMCID: PMC10506108 DOI: 10.1182/bloodadvances.2022009455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023] Open
Abstract
CRISPR/Cas9 screening approaches are powerful tool for identifying in vivo cancer dependencies. Hematopoietic malignancies are genetically complex disorders in which the sequential acquisition of somatic mutations generates clonal diversity. Over time, additional cooperating mutations may drive disease progression. Using an in vivo pooled gene editing screen of epigenetic factors in primary murine hematopoietic stem and progenitor cells (HSPCs), we sought to uncover unrecognized genes that contribute to leukemia progression. We, first, modeled myeloid leukemia in mice by functionally abrogating both Tet2 and Tet3 in HSPCs, followed by transplantation. We, then, performed pooled CRISPR/Cas9 editing of genes encoding epigenetic factors and identified Pbrm1/Baf180, a subunit of the polybromo BRG1/BRM-associated factor SWItch/Sucrose Non-Fermenting chromatin-remodeling complex, as a negative driver of disease progression. We found that Pbrm1 loss promoted leukemogenesis with a significantly shortened latency. Pbrm1-deficient leukemia cells were less immunogenic and were characterized by attenuated interferon signaling and reduced major histocompatibility complex class II (MHC II) expression. We explored the potential relevance to human leukemia by assessing the involvement of PBRM1 in the control of interferon pathway components and found that PBRM1 binds to the promoters of a subset of these genes, most notably IRF1, which in turn regulates MHC II expression. Our findings revealed a novel role for Pbrm1 in leukemia progression. More generally, CRISPR/Cas9 screening coupled with phenotypic readouts in vivo has helped identify a pathway by which transcriptional control of interferon signaling influences leukemia cell interactions with the immune system.
Collapse
Affiliation(s)
- Bin E. Li
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Grace Y. Li
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Wenqing Cai
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Qian Zhu
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | - Davide Seruggia
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Yuko Fujiwara
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| | | | - Stuart H. Orkin
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
12
|
Huang Z, Bu D, Yang N, Huang W, Zhang L, Li X, Ding BS. Integrated analyses of single-cell transcriptomics identify metastasis-associated myeloid subpopulations in breast cancer lung metastasis. Front Immunol 2023; 14:1180402. [PMID: 37483625 PMCID: PMC10361816 DOI: 10.3389/fimmu.2023.1180402] [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: 03/06/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Lung metastasis of breast cancer is closely associated with patient morbidity and mortality, which correlates with myeloid cells in the lung microenvironment. However, the heterogeneity and specificity of metastasis-associated myeloid cells have not been fully established in lung metastasis. Here, by integrating and analyzing single-cell transcriptomics, we found that myeloid subpopulations (Tppp3 + monocytes, Isg15 + macrophages, Ifit3 + neutrophils, and Il12b + DCs) play critical roles in the formation and development of the metastatic niche. Gene enrichment analyses indicate that several tumor-promoting pathways should be responsible for the process, including angiogenesis (Anxa1 and Anxa2 by Tppp3 + monocytes), immunosuppression (Isg15 and Cxcl10 by Isg15 + macrophages; Il12b and Ccl22 by Il12b + DCs), and tumor growth and metastasis (Isg15 and Isg20 by Ifit3 + neutrophils). Furthermore, we have validated these subpopulations in lung microenvironment of MMTV-PyVT transgenic mice and verified their association with poor progression of human breast cancer. Also, our results elucidated a crosstalk network among four myeloid subpopulations by cell-cell communication analysis. This study, therefore, highlights the crucial role of myeloid cells in lung metastasis and provides insights into underlying molecular mechanisms, which pave the way for therapeutic interventions in breast cancer metastasis to lung.
Collapse
|
13
|
Kufe D. Dependence on MUC1-C in Progression of Neuroendocrine Prostate Cancer. Int J Mol Sci 2023; 24:3719. [PMID: 36835130 PMCID: PMC9967814 DOI: 10.3390/ijms24043719] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Castration resistant prostate cancer (CRPC) is responsive to androgen receptor (AR) axis targeted agents; however, patients invariably relapse with resistant disease that often progresses to neuroendocrine prostate cancer (NEPC). Treatment-related NEPC (t-NEPC) is highly aggressive with limited therapeutic options and poor survival outcomes. The molecular basis for NEPC progression remains incompletely understood. The MUC1 gene evolved in mammals to protect barrier tissues from loss of homeostasis. MUC1 encodes the transmembrane MUC1-C subunit, which is activated by inflammation and contributes to wound repair. However, chronic activation of MUC1-C contributes to lineage plasticity and carcinogenesis. Studies in human NEPC cell models have demonstrated that MUC1-C suppresses the AR axis and induces the Yamanaka OSKM pluripotency factors. MUC1-C interacts directly with MYC and activates the expression of the BRN2 neural transcription factor (TF) and other effectors, such as ASCL1, of the NE phenotype. MUC1-C also induces the NOTCH1 stemness TF in promoting the NEPC cancer stem cell (CSC) state. These MUC1-C-driven pathways are coupled with activation of the SWI/SNF embryonic stem BAF (esBAF) and polybromo-BAF (PBAF) chromatin remodeling complexes and global changes in chromatin architecture. The effects of MUC1-C on chromatin accessibility integrate the CSC state with the control of redox balance and induction of self-renewal capacity. Importantly, targeting MUC1-C inhibits NEPC self-renewal, tumorigenicity and therapeutic resistance. This dependence on MUC1-C extends to other NE carcinomas, such as SCLC and MCC, and identify MUC1-C as a target for the treatment of these aggressive malignancies with the anti-MUC1 agents now under clinical and preclinical development.
Collapse
Affiliation(s)
- Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
14
|
Morimoto Y, Yamashita N, Daimon T, Hirose H, Yamano S, Haratake N, Ishikawa S, Bhattacharya A, Fushimi A, Ahmad R, Takahashi H, Dashevsky O, Mitsiades C, Kufe D. MUC1-C is a master regulator of MICA/B NKG2D ligand and exosome secretion in human cancer cells. J Immunother Cancer 2023; 11:e006238. [PMID: 36754452 PMCID: PMC9923360 DOI: 10.1136/jitc-2022-006238] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND The MUC1-C protein evolved in mammals to protect barrier tissues from loss of homeostasis; however, MUC1-C promotes oncogenesis in association with chronic inflammation. Aberrant expression of MUC1-C in cancers has been linked to depletion and dysfunction of T cells in the tumor microenvironment. In contrast, there is no known involvement of MUC1-C in the regulation of natural killer (NK) cell function. METHODS Targeting MUC1-C genetically and pharmacologically in cancer cells was performed to assess effects on intracellular and cell surface expression of the MHC class I chain-related polypeptide A (MICA) and MICB ligands. The MICA/B promoters were analyzed for H3K27 and DNA methylation. Shedding of MICA/B was determined by ELISA. MUC1-C interactions with ERp5 and RAB27A were assessed by coimmunoprecipitation and direct binding studies. Exosomes were isolated for analysis of secretion. Purified NK cells were assayed for killing of cancer cell targets. RESULTS Our studies demonstrate that MUC1-C represses expression of the MICA and MICB ligands that activate the NK group 2D receptor. We show that the inflammatory MUC1-C→NF-κB pathway drives enhancer of zeste homolog 2-mediated and DNMT-mediated methylation of the MICA and MICB promoter regions. Targeting MUC1-C genetically and pharmacologically with the GO-203 inhibitor induced intracellular and cell surface MICA/B expression but not MICA/B cleavage. Mechanistically, MUC1-C regulates the ERp5 thiol oxidoreductase that is necessary for MICA/B protease digestion and shedding. In addition, MUC1-C interacts with the RAB27A protein, which is required for exosome formation and secretion. As a result, targeting MUC1-C markedly inhibited secretion of exosomes expressing MICA/B. In concert with these results, we show that targeting MUC1-C promotes NK cell-mediated killing. CONCLUSIONS These findings uncover pleotropic mechanisms by which MUC1-C confers evasion of cancer cells to NK cell recognition and destruction.
Collapse
Affiliation(s)
- Yoshihiro Morimoto
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nami Yamashita
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tatsuaki Daimon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Haruka Hirose
- Division of Systems Biology, Nagoya University Graduate School of Medicine Faculty of Medicine, Nagoya, Japan
| | - Shizuka Yamano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Naoki Haratake
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Satoshi Ishikawa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Atrayee Bhattacharya
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Atsushi Fushimi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rehan Ahmad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Osaka University, Suita, Japan
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Constantine Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Donald Kufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Shishodia S, Nuñez R, Strohmier BP, Bursch KL, Goetz CJ, Olp MD, Jensen DR, Fenske TG, Ordonez-Rubiano SC, Blau ME, Roach MK, Peterson FC, Volkman BF, Dykhuizen EC, Smith BC. Selective and Cell-Active PBRM1 Bromodomain Inhibitors Discovered through NMR Fragment Screening. J Med Chem 2022; 65:13714-13735. [PMID: 36227159 PMCID: PMC9630929 DOI: 10.1021/acs.jmedchem.2c00864] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PBRM1 is a subunit of the PBAF chromatin remodeling complex that uniquely contains six bromodomains. PBRM1 can operate as a tumor suppressor or tumor promoter. PBRM1 is a tumor promoter in prostate cancer, contributing to migratory and immunosuppressive phenotypes. Selective chemical probes targeting PBRM1 bromodomains are desired to elucidate the association between aberrant PBRM1 chromatin binding and cancer pathogenesis and the contributions of PBRM1 to immunotherapy. Previous PBRM1 inhibitors unselectively bind SMARCA2 and SMARCA4 bromodomains with nanomolar potency. We used our protein-detected NMR screening pipeline to screen 1968 fragments against the second PBRM1 bromodomain, identifying 17 hits with Kd values from 45 μM to >2 mM. Structure-activity relationship studies on the tightest-binding hit resulted in nanomolar inhibitors with selectivity for PBRM1 over SMARCA2 and SMARCA4. These chemical probes inhibit the association of full-length PBRM1 to acetylated histone peptides and selectively inhibit growth of a PBRM1-dependent prostate cancer cell line.
Collapse
Affiliation(s)
- Shifali Shishodia
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Raymundo Nuñez
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Brayden P Strohmier
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Karina L Bursch
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Christopher J Goetz
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Michael D Olp
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Davin R Jensen
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Tyler G Fenske
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Sandra C Ordonez-Rubiano
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Maya E Blau
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Mallory K Roach
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Francis C Peterson
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Brian F Volkman
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Brian C Smith
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
16
|
Fushimi A, Morimoto Y, Ishikawa S, Yamashita N, Bhattacharya A, Daimon T, Rajabi H, Jin C, Hagiwara M, Yasumizu Y, Luan Z, Suo W, Wong KK, Withers H, Liu S, Long MD, Kufe D. Dependence on the MUC1-C Oncoprotein in Classic, Variant, and Non-neuroendocrine Small Cell Lung Cancer. Mol Cancer Res 2022; 20:1379-1390. [PMID: 35612556 PMCID: PMC9437561 DOI: 10.1158/1541-7786.mcr-22-0165] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/27/2022] [Accepted: 05/20/2022] [Indexed: 01/07/2023]
Abstract
Small cell lung cancer (SCLC) is a recalcitrant malignancy defined by subtypes on the basis of differential expression of the ASCL1, NEUROD1, and POU2F3 transcription factors. The MUC1-C protein is activated in pulmonary epithelial cells by exposure to environmental carcinogens and promotes oncogenesis; however, there is no known association between MUC1-C and SCLC. We report that MUC1-C is expressed in classic neuroendocrine (NE) SCLC-A, variant NE SCLC-N and non-NE SCLC-P cells and activates the MYC pathway in these subtypes. In SCLC cells characterized by NE differentiation and DNA replication stress, we show that MUC1-C activates the MYC pathway in association with induction of E2F target genes and dysregulation of mitotic progression. Our studies further demonstrate that the MUC1-C→MYC pathway is necessary for induction of (i) NOTCH2, a marker of pulmonary NE stem cells that are the proposed cell of SCLC origin, and (ii) ASCL1 and NEUROD1. We also show that the MUC1-C→MYC→NOTCH2 network is necessary for self-renewal capacity and tumorigenicity of NE and non-NE SCLC cells. Analyses of datasets from SCLC tumors confirmed that MUC1 expression in single SCLC cells significantly associates with activation of the MYC pathway. These findings demonstrate that SCLC cells are addicted to MUC1-C and identify a potential new target for SCLC treatment. IMPLICATIONS This work uncovers addiction of SCLC cells to MUC1-C, which is a druggable target that could provide new opportunities for advancing SCLC treatment.
Collapse
Affiliation(s)
- Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yoshihiro Morimoto
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Satoshi Ishikawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Caining Jin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masayuki Hagiwara
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yota Yasumizu
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Zhou Luan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wenhao Suo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Henry Withers
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.,Corresponding Authors: Donald Kufe, Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215. E-mail: ; and Mark D. Long, Roswell Park Comprehensive Cancer Center, Carlton & Elm Streets, Buffalo, NY 14263. E-mail:
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Corresponding Authors: Donald Kufe, Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215. E-mail: ; and Mark D. Long, Roswell Park Comprehensive Cancer Center, Carlton & Elm Streets, Buffalo, NY 14263. E-mail:
| |
Collapse
|
17
|
Xu Y, Liu R. Analysis of the role of m6A and lncRNAs in prognosis and immunotherapy of hepatocellular carcinoma. Heliyon 2022; 8:e10612. [PMID: 36158075 PMCID: PMC9489786 DOI: 10.1016/j.heliyon.2022.e10612] [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: 01/16/2022] [Revised: 05/14/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
Objective To study the role of m6A and lncRNAs in the prognosis and immunotherapy of hepatocellular carcinoma, construct the risk score of overall survival of hepatocellular carcinoma, and search for new therapeutic targets and drugs. Methods The data used in this study are obtained from The Cancer Genome Atlas (TCGA) database. A total of 424 HCC samples were included. The co-expression of lncRNAs and M6A-related genes in HCC was analyzed, COX regression analysis was conducted to construct the risk score for HCC prognosis, and the model's validity was further verified in different clinical trials subtypes and principal component analysis. GO enrichment analysis and immune function analysis were performed for the differential genes in the high-risk group and the low-risk group divided by risk score and analyzed the prognostic effect of TMB on the two groups. Based on the results, potential therapeutic agents for HCC were screened. Results The risk score can better predict the prognosis of HCC, the area under the ROC curve is 0.727. Differential genes were mainly located in the extracellular matrix and chromosomal regions and may play regulatory roles in binding sites and catalytic enzymes, thereby affecting the chromosome division and cell proliferation of cells. Type Ⅱ IFN response, type Ⅰ IFN response and MHC class Ⅰ were the three most different functions in terms of immune function between the high-risk group and the low-risk group. Type II IFN response, type I IFN response was significantly down-regulated in the high-risk group, while MHC class I was up-regulated. 14 potential therapeutic drugs were screened out. Conclusions The risk score constructed with NRAV and AL031985.3 had a good predictive effect on the prognosis of HCC. Differences in genes and immune function between high-risk and low-risk groups promoted the occurrence and progression of HCC.
Collapse
Affiliation(s)
- Yan Xu
- Department of Hepatopancreatobiliary Surgical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China
| | - Rong Liu
- Department of Hepatopancreatobiliary Surgical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China
| |
Collapse
|
18
|
Feng A, He L, Chen T, Xu M. A novel cuproptosis-related lncRNA nomogram to improve the prognosis prediction of gastric cancer. Front Oncol 2022; 12:957966. [PMID: 36106123 PMCID: PMC9465020 DOI: 10.3389/fonc.2022.957966] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/29/2022] [Indexed: 12/14/2022] Open
Abstract
Background Cuproptosis is a copper-triggered modality of mitochondrial cell death and cuproptosis process may play important roles in gastric cancer development. However, little is known about cuproptosis-related lncRNAs in gastric adenocarcinoma (STAD). This study is aimed to investigate the potential prognostic signatures of cuproptosis-related lncRNAs in STAD. Methods The Cancer Genome Atlas (TCGA) database were used to obtain gene expression profiles, clinicopathological, and OS information for STAD. Cuproptosis-related genes were collected based on previous studies and cuproptosis-related lncRNAs were screened out by co-expression analysis. The nomogram constructed by Cox regression analysis with the minimum absolute contraction and selection operator (lasso) algorithm. In addition, the potential response of ICB therapy and immune evasion incidence were estimated with Tumor Immune Dysfunction and Exclusion (TIDE) algorithm. Immune checkpoint expressions associated with risk scores were also analyzed. The correlation of immune checkpoint CD209 and HAVCR2 expressions associated with risk scores were experimentally testified by RT-qPCR, Western Blot, and IHC. Results Patients were classified into high-risk and low-risk groups based on the risk score calculated in this model. The Kaplan–Meier survival curve analysis revealed that the high-risk group was associated with poor prognosis. Multivariate Cox regression analysis suggested that this lncRNA prediction model was an independent risk factor affecting the OS rate. Furthermore, ROC curve indicates that the nomogram was superior to traditional clinicopathological features in predicting STAD prognosis. Finally, functional enrichment analysis and immune checkpoint investigation revealed that the nomogram is notably associated with cholesterol metabolism and immune functions, RT-qPCR and Western Blotting demonstrated the co-expression relationship of LINC01150 with CD209 and HAVCR2. Conclusion A novel cuproptosis-related lncRNAs signature impacts on the prognosis and immunological features of GC.
Collapse
Affiliation(s)
| | | | - Tao Chen
- *Correspondence: Meidong Xu, ; Tao Chen,
| | - Meidong Xu
- *Correspondence: Meidong Xu, ; Tao Chen,
| |
Collapse
|
19
|
Yamashita N, Kufe D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. Int J Mol Sci 2022; 23:8219. [PMID: 35897789 PMCID: PMC9331006 DOI: 10.3390/ijms23158219] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with limited treatment options. TNBC progression is associated with expansion of cancer stem cells (CSCs). Few insights are available regarding druggable targets that drive the TNBC CSC state. This review summarizes the literature on TNBC CSCs and the compelling evidence that they are addicted to the MUC1-C transmembrane protein. In normal epithelia, MUC1-C is activated by loss of homeostasis and induces reversible wound-healing responses of inflammation and repair. However, in settings of chronic inflammation, MUC1-C promotes carcinogenesis. MUC1-C induces EMT, epigenetic reprogramming and chromatin remodeling in TNBC CSCs, which are dependent on MUC1-C for self-renewal and tumorigenicity. MUC1-C-induced lineage plasticity in TNBC CSCs confers DNA damage resistance and immune evasion by chronic activation of inflammatory pathways and global changes in chromatin architecture. Of therapeutic significance, an antibody generated against the MUC1-C extracellular domain has been advanced in a clinical trial of anti-MUC1-C CAR T cells and in IND-enabling studies for development as an antibody-drug conjugate (ADC). Agents targeting the MUC1-C cytoplasmic domain have also entered the clinic and are undergoing further development as candidates for advancing TNBC treatment. Eliminating TNBC CSCs will be necessary for curing this recalcitrant cancer and MUC1-C represents a promising druggable target for achieving that goal.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
20
|
Xing L, Xu L, Zhang Y, Che Y, Wang M, Shao Y, Qiu D, Yu H, Zhao F, Zhang J. Recent Insight on Regulations of FBXW7 and Its Role in Immunotherapy. Front Oncol 2022; 12:925041. [PMID: 35814468 PMCID: PMC9263569 DOI: 10.3389/fonc.2022.925041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
SCFFBXW7 E3 ubiquitin ligase complex is a crucial enzyme of the ubiquitin proteasome system that participates in variant activities of cell process, and its component FBXW7 (F-box and WD repeat domain–containing 7) is responsible for recognizing and binding to substrates. The expression of FBXW7 is controlled by multiple pathways at different levels. FBXW7 facilitates the maturity and function maintenance of immune cells via functioning as a mediator of ubiquitination-dependent degradation of substrate proteins. FBXW7 deficiency or mutation results in the growth disturbance and dysfunction of immune cell, leads to the resistance against immunotherapy, and participates in multiple illnesses. It is likely that FBXW7 coordinating with its regulators and substrates could offer potential targets to improve the sensitivity and effects of immunotherapy. Here, we review the mechanisms of the regulation on FBXW7 and its tumor suppression role in immune filed among various diseases (mostly cancers) to explore novel immune targets and treatments.
Collapse
Affiliation(s)
- Liangliang Xing
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Leidi Xu
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yong Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yinggang Che
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Min Wang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yongxiang Shao
- Department of Anus and Intestine Surgery, The 942th Hospital of Joint Logistics Support Force, Yinchuan, China
| | - Dan Qiu
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Honglian Yu
- Department of Hemato-Oncology, The 942th Hospital of Joint Logistics Support Force, Yinchuan, China
| | - Feng Zhao
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Jian Zhang, ; Feng Zhao,
| | - Jian Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Jian Zhang, ; Feng Zhao,
| |
Collapse
|
21
|
Kufe DW. Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells. JOURNAL OF CANCER METASTASIS AND TREATMENT 2022; 8. [PMID: 35539431 PMCID: PMC9083497 DOI: 10.20517/2394-4722.2022.03] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mucin 1 (MUC1) gene emerged in mammals to afford protection of barrier epithelial tissues from the external environment. MUC1 encodes a transmembrane C-terminal (MUC1-C) subunit that is activated by loss of homeostasis and induces inflammatory, proliferative, and remodeling pathways associated with wound repair. As a consequence, chronic activation of MUC1-C promotes lineage plasticity, epigenetic reprogramming, and carcinogenesis. In driving cancer progression, MUC1-C is imported into the nucleus, where it induces NF-κB inflammatory signaling and the epithelial-mesenchymal transition (EMT). MUC1-C represses gene expression by activating (i) DNA methyltransferase 1 (DNMT1) and DNMT3b, (ii) Polycomb Repressive Complex 1 (PRC1) and PRC2, and (iii) the nucleosome remodeling and deacetylase (NuRD) complex. PRC1/2-mediated gene repression is counteracted by the SWI/SNF chromatin remodeling complexes. MUC1-C activates the SWI/SNF BAF and PBAF complexes in cancer stem cell (CSC) models with the induction of genome-wide differentially accessible regions and expressed genes. MUC1-C regulates chromatin accessibility of enhancer-like signatures in association with the induction of the Yamanaka pluripotency factors and recruitment of JUN and BAF, which promote increases in histone activation marks and opening of chromatin. These and other findings described in this review have uncovered a pivotal role for MUC1-C in integrating lineage plasticity and epigenetic reprogramming, which are transient in wound repair and sustained in promoting CSC progression.
Collapse
Affiliation(s)
- Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|