1
|
De Domenico P, Gagliardi F, Roncelli F, Snider S, Mortini P. Tumor-infiltrating and circulating B cells mediate local and systemic immunomodulatory mechanisms in Glioblastoma. J Neurooncol 2025; 172:527-548. [PMID: 40080248 DOI: 10.1007/s11060-025-04989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Glioblastoma (GBM) demonstrates extensive immunomodulatory mechanisms that challenge effective therapeutic interventions. These phenomena extend well beyond the tumor microenvironment (TME) and are reflected in the circulating immunophenotype. B lymphocytes (B cells) have received limited attention in GBM studies despite their emerging importance in mediating both local and systemic immune responses. Recent findings highlight the complex regulatory interactions between B cells and other immune cell populations, including tumor-infiltrating macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and other infiltrating lymphocytes (TILs). B cells are believed to hinder the efficacy of modern immunotherapy strategies focusing on T cells. METHODS This is a focused review of available evidence regarding B cells in GBM through January 2025. RESULTS Peripheral blood reflects a systemically dampened immune response, with sustained lymphopenia, increased plasma cells, and dysfunctional memory B cells. The tumor immune landscape is enriched in cells of B-lineage. Subsets of poorly characterized B regulatory cells (Bregs) populate the TME, developing their phenotype due to their proximity to MDSCs, TAMs, and tumoral cells. The Bregs inhibit CD8+ T activity and may have potential prognostic significance. CONCLUSION Understanding the role of B cells, how they are recruited, and their differentiation shifted towards an immunomodulatory role could inform better therapeutic strategies and unleash their full antitumoral potential in GBM.
Collapse
Affiliation(s)
- Pierfrancesco De Domenico
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy.
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Roncelli
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Snider
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
2
|
Tomar MS, Mohit, Kumar A, Shrivastava A. Circadian immunometabolism: A future insight for targeted therapy in cancer. Sleep Med Rev 2025; 80:102031. [PMID: 39603026 DOI: 10.1016/j.smrv.2024.102031] [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: 04/15/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Circadian rhythms send messages to regulate the sleep-wake cycle in living beings, which, regulate various biological activities. It is well known that altered sleep-wake cycles affect host metabolism and significantly deregulate the host immunity. The dysregulation of circadian-related genes is critical for various malignancies. One of the hallmarks of cancer is altered metabolism, the effects of which spill into surrounding microenvironments. Here, we review the emerging literature linking the circadian immunometabolic axis to cancer. Small metabolites are the products of various metabolic pathways, that are usually perturbed in cancer. Genes that regulate circadian rhythms also regulate host metabolism and control metabolite content in the tumor microenvironment. Immune cell infiltration into the tumor site is critical to perform anticancer functions, and altered metabolite content affects their trafficking to the tumor site. A compromised immune response in the tumor microenvironment aids cancer cell proliferation and immune evasion, resulting in metastases. The role of circadian rhythms in these processes is largely overlooked and demands renewed attention in the search for targets against cancer growth and spread. The precision medicine approach requires targeting the circadian immune metabolism in cancer.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Mohit
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India; Department of Prosthodontics, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India.
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India.
| |
Collapse
|
3
|
You L, Wu Q. Cellular senescence in tumor immune escape: Mechanisms, implications, and therapeutic potential. Crit Rev Oncol Hematol 2025; 208:104628. [PMID: 39864532 DOI: 10.1016/j.critrevonc.2025.104628] [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: 12/23/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Cellular senescence, a hallmark of aging, has emerged as a captivating area of research in tumor immunology with profound implications for cancer prevention and treatment. In the tumor microenvironment, senescent cells exhibit a dual role, simultaneously hindering tumor development through collaboration with immune cells and evading immune cell attacks by upregulating immunoinhibitory proteins. However, the intricate immune escape mechanism of cellular senescence in the tumor microenvironment remains a subject of intense investigation. Chronic inflammation is exacerbated by cellular senescence through the upregulation of pro-inflammatory factors such as interleukin-1β, thereby augmenting the risk of tumorigenesis. Additionally, the interplay between autophagy and cellular senescence adds another layer of complexity. Autophagy, known to slow down the aging process by reducing p53/p21 levels, may be downregulated by cellular senescence. To harness the therapeutic potential of cellular senescence, targeting its immunological aspects has gained significant attention. Strategies such as immune checkpoint inhibitors and T-cell senescence inhibition are being explored in the context of cellular senescence immunotherapy. In this comprehensive review, we provide a compelling overview of the regulation of cellular senescence and delve into the influencing factors, including chronic inflammation, autophagy, and circadian rhythms, associated with senescence in the tumor microenvironment. We specifically focus on unraveling the enigmatic dual role of cellular senescence in tumor immune escape. By deciphering the intricate nature of cellular senescence in the tumor microenvironment, this review aims to advance our understanding and pave the way for leveraging senescence as a promising target for tumor immunotherapy applications.
Collapse
Affiliation(s)
- Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing 401520, China; College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou 434025, China.
| |
Collapse
|
4
|
Geng F, Zhao N, Ren Q. Circadian rhythm, microglia-mediated neuroinflammation, and Alzheimer's disease. Neurosci Biobehav Rev 2025; 170:106044. [PMID: 39914702 DOI: 10.1016/j.neubiorev.2025.106044] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Microglia, the brain's resident macrophages, are key mediators of neuroinflammation, responding to immune pathogens and toxins. They play a crucial role in clearing cellular debris, regulating synaptic plasticity, and phagocytosing amyloid-β (Aβ) plaques in Alzheimer's disease (AD). Recent studies indicate that microglia not only exhibit intrinsic circadian rhythms but are also regulated by circadian clock genes, influencing specific functions such as phagocytosis and the modulation of neuroinflammation. Disruption of the circadian rhythm is closely associated with AD pathology. In this review, we will provide an overview of how circadian rhythms regulate microglia-mediated neuroinflammation in the progression of AD, focusing on the pathway from the central nervous system (CNS) and the peripheral immune system. We also discuss potential therapeutic targets, including hormone modulation, lifestyle interventions, and anti-inflammatory therapies, aimed at maintaining brain health in AD. This will shed light on the involvement of circadian rhythm in AD and explore new avenues for AD treatment.
Collapse
Affiliation(s)
- Fan Geng
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Na Zhao
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China
| | - Qingguo Ren
- Department of Neurology, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
5
|
Shen Z, Zhao Y, Xu X, Yang H, He S, Ma J, Zhang S, Hou P, Sui F. Single-cell RNA sequencing integrated with bulk RNA sequencing analysis of clock circadian regulator with prognostic and immune microenvironment in thyroid cancer. Transl Oncol 2025; 53:102299. [PMID: 39892222 PMCID: PMC11833347 DOI: 10.1016/j.tranon.2025.102299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Disruption of circadian rhythm was found to be associated with immune infiltration and thyroid cancer. However, the role of clock circadian regulator (CLOCK) in the progression of thyroid cancer and its immune microenvironment remains largely unexplored. Therefore, our aim was to explore the role and potential mechanism of CLOCK in thyroid cancer. METHODS Single cell sequencing analysis and bulk RNA sequencing analysis was used for LASSO regression and Kaplan-Meier survival estimates. Potential mechanism analysis were gained through KEGG/GO analysis, GSEA analysis and PPI network. In vivo and in vitro experiment was used for further validation. RESULTS The result showed CLOCK protein was overexpressed in thyroid cancer compared with normal tissue in both thyroid specific mouse model and human sample. A prognostic model incorporating CLOCK and other related genes (FAT4, OR6K2, STK40, TMEM63A, HRCT1, SUPT5H, and OR2C3) was developed using LASSO regression. Functional assay and bioinformatics analysis indicated that CLOCK knockdown hindered tumor growth and the activity of MAPK signaling. Besides, analyses of gene enrichment, signaling pathways, and immune checkpoints suggested that CLOCK might inhibit immune infiltration within the tumor microenvironment. Confirmatory in vitro experiments and immunohistochemical assays in human samples further linked high CLOCK expression to reduced T cell cytotoxicity and infiltration. CONCLUSION These findings underscore the pivotal role of CLOCK in thyroid cancer prognosis and immune suppression, highlighting its potential as a target for therapeutic intervention and prognostic assessment in thyroid cancer management.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Yuelei Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Xinxin Xu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Huini Yang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Shuting He
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Junchi Ma
- School of Information Engineering, Chang'an University, Shaanxi Province, PR China
| | - Shaoqiang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, Xi'an 710061, Shaanxi Province, PR China
| | - Fang Sui
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| |
Collapse
|
6
|
Emir SM, Karaoğlan BS, Kaşmer R, Şirin HB, Sarıyıldız B, Karakaş N. Hunting glioblastoma recurrence: glioma stem cells as retrospective targets. Am J Physiol Cell Physiol 2025; 328:C1045-C1061. [PMID: 39818986 DOI: 10.1152/ajpcell.00344.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/11/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain malignancies in adults. Standard approaches, including surgical resection followed by adjuvant radio- and chemotherapy with temozolomide (TMZ), provide only transient control, as GBM frequently recurs due to its infiltrative nature and the presence of therapy-resistant subpopulations such as glioma stem cells (GSCs). GSCs, with their quiescent state and robust resistance mechanisms, evade conventional therapies, contributing significantly to relapse. Consequently, current treatment methods for GBM face significant limitations in effectively targeting GSCs. In this review, we emphasize the relationship between GBM recurrence and GSCs, discuss the current limitations, and provide future perspectives to overwhelm the challenges associated with targeting GSCs. Eliminating GSCs may suppress recurrence, achieve durable responses, and improve therapeutic outcomes for patients with GBM.
Collapse
Affiliation(s)
- Sümeyra Mengüç Emir
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Birnur Sinem Karaoğlan
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Ramazan Kaşmer
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Hilal Buse Şirin
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Batuhan Sarıyıldız
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Nihal Karakaş
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
- Department of Medical Biology, International School of Medicine, İstanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
7
|
Yu Q, Mei H, Gu Q, Zeng R, Li Y, Zhang J, Gao C, Fang H, Qu J, Liu J. OLFML3 Promotes IRG1 Mitochondrial Localization and Modulates Mitochondrial Function in Macrophages. Int J Biol Sci 2025; 21:2275-2295. [PMID: 40083707 PMCID: PMC11900800 DOI: 10.7150/ijbs.103859] [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: 02/15/2025] [Indexed: 03/16/2025] Open
Abstract
Olfactomedin-like protein 3 (OLFML3), belonging to olfactomedin (OLF) protein family, has poorly defined functions. Recent studies have reported the functions of OLFML3 in anti-viral immunity and tumorigenesis. In this study, we investigated the roles of OLFML3 in macrophages. In LPS- or Pseudomonas aeruginosa-induced acute lung injury (ALI) mouse model, OLFML3 depletion exacerbated inflammatory response, leading to reduced survival. OLFML3 achieved the in vivo activity by regulating macrophage phagocytosis and migration. Mass spectrometry analysis revealed immunoresponsive gene 1 (IRG1) as an OLFML3-interacting protein. IRG1 is a mitochondrial decarboxylase that catalyzes the conversion of cis-aconitate to itaconate, a myeloid-borne mitochondrial metabolite with immunomodulatory activities. Further investigation showed that OLFML3 could prevent LPS-induced mitochondrial dysfunction in macrophages by maintaining the homeostasis of mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (mtROS) and itaconate-related metabolites. In-depth protein-protein interaction studies showed that OLFML3 could promote IRG1 mitochondrial localization via a mitochondrial transport protein, apoptosis inducing factor mitochondria associated 1 (AIFM1). In summary, our study showed that OLFML3 could facilitate IRG1 mitochondrial localization and prevent LPS-induced mitochondrial dysfunction in macrophages.
Collapse
Affiliation(s)
- Qijun Yu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qian Gu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ran Zeng
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Yanan Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Junjie Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chenxu Gao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Guangzhou Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China
| |
Collapse
|
8
|
Vinel C, Boot J, Jin W, Pomella N, Hadaway A, Mein C, Zabet NR, Marino S. Mapping chromatin remodelling in glioblastoma identifies epigenetic regulation of key molecular pathways and novel druggable targets. BMC Biol 2025; 23:26. [PMID: 39915814 PMCID: PMC11804007 DOI: 10.1186/s12915-025-02127-9] [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: 06/24/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Glioblastoma is the most common and aggressive malignant brain tumour in the adult population and its prognosis is dismal. The heterogeneous nature of the tumour, to which epigenetic dysregulation significantly contributes, is among the main therapeutic challenges of the disease. RESULTS We have leveraged SYNGN, an experimental pipeline enabling the syngeneic comparison of glioblastoma stem cells and expanded potential stem cell (EPSC)-derived neural stem cells to identify regulatory features driven by chromatin remodelling specifically in glioblastoma stem cells. CONCLUSIONS We show epigenetic regulation of the expression of genes and related signalling pathways contributing to glioblastoma development. We also identify novel epigenetically regulated druggable target genes on a patient-specific level, including SMOX and GABBR2.
Collapse
Affiliation(s)
- Claire Vinel
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - James Boot
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
- Genome Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Weiwei Jin
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Nicola Pomella
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Alexandra Hadaway
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Charles Mein
- Genome Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Nicolae Radu Zabet
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK
| | - Silvia Marino
- Brain Tumour Research Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK.
- Barts Brain Tumour Centre, Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University London, London, UK.
| |
Collapse
|
9
|
Liao J, Duan Y, Xu X, Liu Y, Zhan C, Xiao G. Circadian rhythm related genes signature in glioma for drug resistance prediction: a comprehensive analysis integrating transcriptomics and machine learning. Discov Oncol 2025; 16:119. [PMID: 39909964 PMCID: PMC11799505 DOI: 10.1007/s12672-025-01863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Gliomas, 24% of all primary brain tumors, have diverse histology and poor survival rates, with about 70% recurring due to acquired or de novo resistance. Insomnia in patients is correlated strongly with circadian rhythm disruptions. The correlation between circadian rhythm disorders and drug resistance of some tumors has been proved. However, the precise mechanism underlying the relationship between glioma and circadian rhythm disorders has not been elucidated. METHODS Circadian rhythm-related genes (CRRGs) were identified using the least absolute shrinkage and selection operator (LASSO) regression, and stochastic gradient descent (SGD) was performed to form a circadian rhythm-related score (CRRS) model. The studies of immune cell infiltration, genetic variations, differential gene expression pattern, and single cell analysis were performed for exploring the mechanisms of chemotherapy resistance in glioma. The relationship between CRRGs and chemosensitivity was also confirmed by IC 50 (half maximal inhibitory concentration) analysis. RESULT Signatures of 16 CRRGs were screened out and identified. Based on the CRRS model, an optimal comprehensive nomogram was created, exhibiting a favorable potential for predicting drug resistance in samples. Immune infiltration, cell-cell communication, and single cell analysis all indicated that high CRRS group was closely related to innate immune cells. IC50 analysis showed that CRRG knockdown enhanced the chemosensitivity of glioma. CONCLUSION A significant correlation between CRRGs, drug resistance of glioma, and innate immune cells was found, which might hold a significant role in the drug resistance of glioma.
Collapse
Affiliation(s)
- Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxing Duan
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xiangwang Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaxue Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chaohong Zhan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
10
|
Wang X, Yang K, Yang B, Wang R, Zhu Y, Pan T. ANKRD22 participates in the proinflammatory activities of macrophages in the colon cancer tumor microenvironment. Cancer Immunol Immunother 2025; 74:86. [PMID: 39891675 PMCID: PMC11787111 DOI: 10.1007/s00262-024-03930-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/21/2024] [Indexed: 02/03/2025]
Abstract
Tumor-associated macrophages (TAMs) are among the most common types of immune cells in the colon cancer microenvironment. Reprogramming M2-type TAMs with immunosuppressive functions into M1-type TAMs with proinflammatory functions is a novel strategy for reshaping the tumor microenvironment (TME) and enhancing the efficacy of immunotherapy in colon cancer. However, the key molecules and mechanisms underlying TAM polarization require further clarification. Our previous study suggested that ANKRD22 may play a role in regulating the functional state transition of macrophages. However, the expression levels of ANKRD22 in colon TAMs and its specific effects on tumor proliferation remain unclear. In the present study, we observed elevated ANKRD22 expression in M1-type TAMs. The expression level of ANKRD22 was positively correlated with the survival period of patients with colon cancer and with the infiltration abundance of M1-type TAMs, and ANKRD22 expression was negatively correlated with the infiltration abundance of M2-type TAMs. A significant decrease in ANKRD22 expression in macrophages cocultured with colon cancer cell culture supernatant as well as in macrophages directly derived from colorectal cancer tissues was observed. Single-cell RNA sequencing, spatial transcriptomic studies, and subcutaneous xenograft experiments in mice revealed that Ankrd22 silencing altered the subtype distribution of macrophages, attenuated their proinflammatory activity, and enhanced their protumor activity. Additionally, we identified a small-molecule ANKRD22 upregulator that could aid in the development of novel therapeutics targeting TAM remodeling.
Collapse
Affiliation(s)
- Xiaoying Wang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Keqing Yang
- Internal Medicine, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, People's Republic of China
| | - Bin Yang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Rui Wang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China.
| | - Tianhui Pan
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China.
| |
Collapse
|
11
|
Licón-Muñoz Y, Avalos V, Subramanian S, Granger B, Martinez F, García-Montaño LA, Varela S, Moore D, Perkins E, Kogan M, Berto S, Chohan MO, Bowers CA, Piccirillo SGM. Single-nucleus and spatial landscape of the sub-ventricular zone in human glioblastoma. Cell Rep 2025; 44:115149. [PMID: 39752252 DOI: 10.1016/j.celrep.2024.115149] [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: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The sub-ventricular zone (SVZ) is the most well-characterized neurogenic area in the mammalian brain. We previously showed that in 65% of patients with glioblastoma (GBM), the SVZ is a reservoir of cancer stem-like cells that contribute to treatment resistance and the emergence of recurrence. Here, we build a single-nucleus RNA-sequencing-based microenvironment landscape of the tumor mass and the SVZ of 15 patients and two histologically normal SVZ samples as controls. We identify a ZEB1-centered mesenchymal signature in the tumor cells of the SVZ. Moreover, the SVZ microenvironment is characterized by tumor-supportive microglia, which spatially coexist and establish crosstalks with tumor cells. Last, differential gene expression analyses, predictions of ligand-receptor and incoming/outgoing interactions, and functional assays reveal that the interleukin (IL)-1β/IL-1RAcP and Wnt-5a/Frizzled-3 pathways represent potential therapeutic targets in the SVZ. Our data provide insights into the biology of the SVZ in patients with GBM and identify potential targets of this microenvironment.
Collapse
Affiliation(s)
- Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Vanessa Avalos
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Suganya Subramanian
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bryan Granger
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Frank Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Leopoldo A García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Samantha Varela
- University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Drew Moore
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Stefano Berto
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Muhammad O Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Christian A Bowers
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Sara G M Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
12
|
Shao F, Wang Z, Ye L, Wu R, Wang J, Yu QX, Wusiman D, Tuo Z, Yoo KH, Shu Z, Wei W, Li D, Cho WC, Liu Z, Feng D. Basic helix-loop-helix ARNT like 1 regulates the function of immune cells and participates in the development of immune-related diseases. BURNS & TRAUMA 2025; 13:tkae075. [PMID: 39830193 PMCID: PMC11741524 DOI: 10.1093/burnst/tkae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 01/22/2025]
Abstract
The circadian clock is an internal timekeeper system that regulates biological processes through a central circadian clock and peripheral clocks controlling various genes. Basic helix-loop-helix ARNT-like 1 (BMAL1), also known as aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL1), is a key component of the circadian clock. The deletion of BMAL1 alone can abolish the circadian rhythms of the human body. BMAL1 plays a critical role in immune cell function. Dysregulation of BMAL1 is linked to immune-related diseases such as autoimmune diseases, infectious diseases, and cancer, and vice versa. This review highlights the significant role of BMAL1 in governing immune cells, including their development, differentiation, migration, homing, metabolism, and effector functions. This study also explores how dysregulation of BMAL1 can have far-reaching implications and potentially contribute to the onset of immune-related diseases such as autoimmune diseases, infectious diseases, cancer, sepsis, and trauma. Furthermore, this review discusses treatments for immune-related diseases that target BMAL1 disorders. Understanding the impact of BMAL1 on immune function can provide insights into the pathogenesis of immune-related diseases and help in the development of more effective treatment strategies. Targeting BMAL1 has been demonstrated to achieve good efficacy in immune-related diseases, indicating its promising potential as a targetable therapeutic target in these diseases.
Collapse
Affiliation(s)
- Fanglin Shao
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, First Ring Road, Qingyang District, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, West Gate Street, Linhai City 317000, Zhejiang Province, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Qing-Xin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Huancheng North Road, Jiangbei District, Ningbo, Zhejiang Province, 315211, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, 615 W. State Street, West Lafayette, IN 47907, USA
| | - Zhouting Tuo
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Jinzhai South Road, Shushan District, Hefei, Anhui 230032, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, South Korea
| | - Ziyu Shu
- Department of Earth Science and Engineering, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, UK
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Shapingba Street, Shapingba District, Chongqing 400044, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Gascoigne Road, Yau Ma Tei, Kowloon, Hong Kong SAR, China
| | - Zhihong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
- Division of Surgery & Interventional Science, University College London, Gower Street, London W1T 6JF, London W1W 7TS, UK
| |
Collapse
|
13
|
Xu W, Li L, Cao Z, Ye J, Gu X. Circadian Rhythms and Lung Cancer in the Context of Aging: A Review of Current Evidence. Aging Dis 2025:AD.2024.1188. [PMID: 39812541 DOI: 10.14336/ad.2024.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Circadian rhythm is the internal homeostatic physiological clock that regulates the 24-hour sleep/wake cycle. This biological clock helps to adapt to environmental changes such as light, dark, temperature, and behaviors. Aging, on the other hand, is a process of physiological changes that results in a progressive decline in cells, tissues, and other vital systems of the body. Both aging and the circadian clock are highly interlinked phenomena with a bidirectional relationship. The process of aging leads to circadian disruptions while dysfunctional circadian rhythms promote age-related complications. Both processes involve diverse physiological, molecular, and cellular changes such as modifications in the DNA repair mechanisms, mechanisms, ROS generation, apoptosis, and cell proliferation. This review aims to examine the role of aging and circadian rhythms in the context of lung cancer. This will also review the existing literature on the role of circadian disruptions in the process of aging and vice versa. Various molecular pathways and genes such as BMAL1, SIRT1, HLF, and PER1 and their implications in aging, circadian rhythms, and lung cancer will also be discussed.
Collapse
Affiliation(s)
- Wenhui Xu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Jinghong Ye
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, China
| | - Xuyu Gu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
14
|
Zhu X, Zhang Z, Zhang J, Xiao Y, Wang H, Wang M, Jiang M, Xu Y. Single-cell and Bulk Transcriptomic Analyses Reveal a Stemness and Circadian Rhythm Disturbance-related Signature Predicting Clinical Outcome and Immunotherapy Response in Hepatocellular Carcinoma. Curr Gene Ther 2025; 25:178-193. [PMID: 38847249 DOI: 10.2174/0115665232298240240529131358] [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: 01/22/2024] [Revised: 04/30/2024] [Accepted: 05/19/2024] [Indexed: 11/30/2024]
Abstract
AIMS Investigating the impact of stemness-related circadian rhythm disruption (SCRD) on hepatocellular carcinoma (HCC) prognosis and its potential as a predictor for immunotherapy response. BACKGROUND Circadian disruption has been linked to tumor progression through its effect on the stemness of cancer cells. OBJECTIVE Develop a novel signature for SCRD to accurately predict clinical outcomes and immune therapy response in patients with HCC. METHODS The stemness degree of patients with HCC was assessed based on the stemness index (mRNAsi). The co-expression circadian genes significantly correlated with mRNAsi were identified and defined as stemness- and circadian-related genes (SCRGs). The SCRD scores of samples and cells were calculated based on the SCRGs. Differentially expressed genes with a prognostic value between distinct SCRD groups were identified in bulk and single-cell datasets to develop an SCRD signature. RESULTS A higher SCRD score indicates a worse patient survival rate. Analysis of the tumor microenvironment revealed a significant correlation between SCRD and infiltrating immune cells. Heterogeneous expression patterns, functional states, genomic variants, and cell-cell interactions between two SCRD populations were revealed by transcriptomic, genomic, and interaction analyses. The robust SCRD signature for predicting immunotherapy response and prognosis in patients with HCC was developed and validated in multiple independent cohorts. CONCLUSIONS In summary, distinct tumor immune microenvironment patterns were confirmed under SCRD in bulk and single-cell transcriptomic, and SCRD signature associated with clinical outcomes and immunotherapy response was developed and validated in HCC.
Collapse
Affiliation(s)
- Xiaojing Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zixin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jiaxing Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanqi Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingwei Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Minghui Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Zhang Y, Zhang Q, Liu R, Zhang D, Hu G, Chen X. Circadian disruption in cancer and regulation of cancer stem cells by circadian clock genes: An updated review. Cancer Lett 2024; 611:217391. [PMID: 39672457 DOI: 10.1016/j.canlet.2024.217391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Circadian rhythm, regulated by a time keeping system termed as the circadian clock, is important for many biological processes in eukaryotes. Disordered circadian rhythm is implicated in different human diseases, including cardiovascular disease, neurologic disease, metabolic disorders, and cancer. The stem like-cancer cells (or cancer stem cells, CSCs) are proposed to stand at the top of the heterogeneous hierarchy in different solid tumors, which are responsible for tumor initiation, development, therapy resistance and metastasis. Emerging evidence has shown that circadian clock genes potentially regulate the stemness and features of CSCs in several malignant systems, including leukemia, glioblastoma, breast cancer, colorectal cancer and prostate cancer. The chronotherapies targeting CSCs are therefore of therapeutic potentials in treating malignancies. In this review, we have summarized our current knowledge of circadian clock gene regulation in normal stem/progenitor cells. Moreover, we have provided evidence linking dysregulations of circadian clock genes and cancer development. Importantly, we have listed the potential mechanisms underlying circadian clock gene regulation of CSCs. Finally, we have offered the current attempts of chronotherapy targeting CSCs. Elucidating the molecular regulation of circadian clock gene in CSCs will provide us a novel direction for the development of therapeutics to target CSCs.
Collapse
Affiliation(s)
- Yiling Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rundong Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dingxiao Zhang
- Provincial Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Elguindy MM, Young JS, Ho WS, Lu RO. Co-evolution of glioma and immune microenvironment. J Immunother Cancer 2024; 12:e009175. [PMID: 39631850 PMCID: PMC11624716 DOI: 10.1136/jitc-2024-009175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/03/2024] [Indexed: 12/07/2024] Open
Abstract
Glioma evolution is governed by a multitude of dynamic interactions between tumor cells and heterogenous neighboring, non-cancerous cells. This complex ecosystem, termed the tumor microenvironment (TME), includes diverse immune cell types that have gained increasing attention for their critical and paradoxical roles in tumor control and tumorigenesis. Recent work has revealed that the cellular composition and functional state of immune cells in the TME can evolve extensively depending on the tumor stage and intrinsic features of surrounding glioma cells. Concurrently, adaptations to the glioma cellular phenotype, including activation of various cellular states, occur in the context of these immune cell alterations. In this review, we summarize important features of the immune TME that play key roles during each stage of glioma progression, from initiation to immune escape, invasion and recurrence. Understanding the complex interplay between tumor and immune cells is critical for the development of effective immunotherapies for glioma treatment.
Collapse
Affiliation(s)
- Mahmoud M Elguindy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Winson S Ho
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Rongze O Lu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
Liu Y, Zhou F, Ali H, Lathia JD, Chen P. Immunotherapy for glioblastoma: current state, challenges, and future perspectives. Cell Mol Immunol 2024; 21:1354-1375. [PMID: 39406966 PMCID: PMC11607068 DOI: 10.1038/s41423-024-01226-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: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive and lethal type of brain tumor in human adults. The standard of care offers minimal clinical benefit, and most GBM patients experience tumor recurrence after treatment. In recent years, significant advancements have been made in the development of novel immunotherapies or other therapeutic strategies that can overcome immunotherapy resistance in many advanced cancers. However, the benefit of immune-based treatments in GBM is limited because of the unique brain immune profiles, GBM cell heterogeneity, and immunosuppressive tumor microenvironment. In this review, we present a detailed overview of current immunotherapeutic strategies and discuss the challenges and potential molecular mechanisms underlying immunotherapy resistance in GBM. Furthermore, we provide an in-depth discussion regarding the strategies that can overcome immunotherapy resistance in GBM, which will likely require combination therapies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Fei Zhou
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Heba Ali
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA
| | - Peiwen Chen
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.
| |
Collapse
|
18
|
Kricha A, Bouchmaa N, Ben Mkaddem S, Abbaoui A, Ben Mrid R, El Fatimy R. Glioblastoma-associated macrophages: A key target in overcoming glioblastoma therapeutic resistance. Cytokine Growth Factor Rev 2024; 80:97-108. [PMID: 39510901 DOI: 10.1016/j.cytogfr.2024.10.009] [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: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Glioblastoma multiforme (GBM) is recognized as the most aggressive and malignant form of brain cancer, characterized by a highly heterogeneous phenotype, poor prognosis, and a median survival time of less than 16 months. Recent studies have highlighted the critical role of glioblastoma-associated macrophages (GAMs) in promoting tumor progression and resistance not only to immunotherapy but also to radiotherapy and chemotherapy. GAMs actively suppress immune responses, promote angiogenesis, facilitate tumor metastasis, and induce therapy resistance, through various mechanisms such as cytokines production, signaling pathways regulation, and angiogenesis. In this context, understanding these regulatory mechanisms is essential for developing efficient therapies. Preclinical studies have investigated diverse approaches to target these cells, both as monotherapies or in combination with other treatments. While these approaches have shown promise in strengthening antitumor immune responses in animal models, their clinical success remains to be fully determined. Herein, we provide a comprehensive overview of GAMs's role in GBM therapeutic resistance and summarizes existing approaches to target GAMs in overcoming tumor resistance.
Collapse
Affiliation(s)
- Aymane Kricha
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Najat Bouchmaa
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Sanae Ben Mkaddem
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Abdellatif Abbaoui
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Reda Ben Mrid
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| | - Rachid El Fatimy
- Institute of Biological Sciences (IBS), Faculty of Medical Sciences, Mohammed VI Polytechnic University (FMS-UM6P), Benguerir, Morocco.
| |
Collapse
|
19
|
Liu X, Shi Z, Liu X, Cao Y, Yang X, Liu J, Xu T, Yang W, Chen L, Zou Z, Jia Q, Li M. The role of PDCD6 in stemness maintenance of Glioblastoma. Pathol Res Pract 2024; 264:155727. [PMID: 39561536 DOI: 10.1016/j.prp.2024.155727] [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: 11/11/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Glioblastoma (GBM) poses formidable challenges due to its high malignancy and therapeutic resistance and still exhibits dismal 5-year survival rates, high recurrence propensity, and limited treatment modalities. There is an acute need for innovative treatments for recurrent glioblastoma due to the lack of established protocols. This necessity is driving research into the cellular underpinnings that initiate and drive the disease forward, aiming to discover groundbreaking targets for therapy that could enhance the efficacy of medical interventions. METHODS Patient-derived glioblastoma stem cells (GSCs) were harvested and isolated. Subsequently, PDCD6 expression was quantified through both western blotting (WB) and real-time PCR (RT-PCR) techniques. The stem-like properties of the GSCs were evaluated using sphere-forming assays. All gathered data, inclusive of TCGA datasets, were analyzed using SPSS (IBM) version 23.0. RESULTS Elevated PDCD6 expression characterized classical GBM tumor tissues. PDCD6 overexpression significantly correlated with diminished overall survival in GBM patients, emerging as an independent prognostic indicator. Notably, primary GBM cells exhibited heightened PDCD6 levels in GSCs compared to NSTCs. Moreover, alterations in stemness markers paralleled PDCD6 modulation, where PDCD6 knockdown attenuated tumor size in GSCs. CONCLUSION Our findings illuminate PDCD6's role in fostering stemness within classical GBM, hinting at its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xiyu Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zuolin Shi
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Xuantong Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuan Cao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinyu Yang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiaming Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Weiyi Yang
- Department of Neurology, Xi'an Daxing Hospital, Xi'an, China
| | - Ligang Chen
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zheng Zou
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
20
|
Pathikonda S, Amirmahani F, Mathew D, Muthukrishnan SD. Histone acetyltransferases as promising therapeutic targets in glioblastoma resistance. Cancer Lett 2024; 604:217269. [PMID: 39326554 DOI: 10.1016/j.canlet.2024.217269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Glioblastoma (GBM) is a fatal adult brain tumor with an extremely poor prognosis. GBM poses significant challenges for targeted therapies due to its intra- and inter-tumoral heterogeneity, a highly immunosuppressive microenvironment, diffuse infiltration into normal brain parenchyma, protection by the blood-brain barrier and acquisition of therapeutic resistance. Recent studies have implicated epigenetic modifiers as key players driving tumorigenesis, resistance, and progression of GBM. While the vast majority of GBM research on epigenetic modifiers thus far has focused predominantly on elucidating the functional roles and targeting of DNA methyltransferases and histone deacetylases, emerging evidence indicates that histone acetyltransferases (HATs) also play a key role in mediating plasticity and therapeutic resistance in GBM. Here, we will provide an overview of HATs, their dual roles and functions in cancer as both tumor suppressors and oncogenes and focus specifically on their implications in GBM resistance. We also discuss the technical challenges in developing selective HAT inhibitors and highlight their promise as potential anti-cancer therapeutics for treating intractable cancers such as GBM.
Collapse
Affiliation(s)
- Spoorthy Pathikonda
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Farzaneh Amirmahani
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Diya Mathew
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Sree Deepthi Muthukrishnan
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| |
Collapse
|
21
|
Hui Y, Zhong Y, Kuang L, Xu J, Hao Y, Cao J, Zheng T. O-GlcNAcylation of circadian clock protein Bmal1 impairs cognitive function in diabetic mice. EMBO J 2024; 43:5667-5689. [PMID: 39375536 PMCID: PMC11574178 DOI: 10.1038/s44318-024-00263-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Neuronal damage in the hippocampus induced by high glucose has been shown to promote the onset and development of cognitive impairment in diabetes, but the underlying molecular mechanism remains unclear. Guided by single-cell RNA sequencing, we here report that high glucose increases O-GlcNAcylation of Bmal1 in hippocampal neurons. This glycosylation promotes the binding of Clock to Bmal1, resulting in the expression of transcription factor Bhlhe41 and its target Dnajb4. Upregulated Dnajb4 in turn leads to ubiquitination and degradation of the mitochondrial Na + /Ca2+ exchanger NCLX, thereby inducing mitochondrial calcium overload that causes neuronal damage and cognitive impairment in mice. Notably, Bhlhe41 downregulation or treatment with a short peptide that specifically blocks O-GlcNAcylation of Bmal1 on Ser424 mitigated these adverse effects in diabetic mouse models. These data highlight the crucial role of O-GlcNAcylation in circadian clock gene expression and may facilitate the design of targeted therapies for diabetes-associated cognitive impairment.
Collapse
Affiliation(s)
- Ya Hui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuanmei Zhong
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Liuyu Kuang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxi Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuqi Hao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxue Cao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Tianpeng Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
| |
Collapse
|
22
|
Quist M, van Os M, van Laake LW, Bovenschen N, Crnko S. Integration of circadian rhythms and immunotherapy for enhanced precision in brain cancer treatment. EBioMedicine 2024; 109:105395. [PMID: 39413708 PMCID: PMC11530607 DOI: 10.1016/j.ebiom.2024.105395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
Circadian rhythms significantly impact (patho)physiological processes, with disruptions linked to neurodegenerative diseases and heightened cancer vulnerability. While immunotherapy has shown promise in treating various cancers, its efficacy in brain malignancies remains limited. This review explores the nexus of circadian rhythms and immunotherapy in brain cancer treatment, emphasising precision through alignment with the body's internal clock. We evaluate circadian regulation of immune responses, including cell localisation and functional phenotype, and discuss how circadian dysregulation affects anti-cancer immunity. Additionally, we analyse and assess the effectiveness of current immunotherapeutic approaches for brain cancer including immune checkpoint blockades, adoptive cellular therapies, and other novel strategies. Future directions, such as chronotherapy and personalised treatment schedules, are proposed to optimise immunotherapy precision against brain cancers. Overall, this review provides an understanding of the often-overlooked role of circadian rhythms in brain cancer and suggests avenues for improving immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Matthias Quist
- Department of Pathology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maas van Os
- Department of Pathology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Linda W van Laake
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Centre Utrecht, Utrecht, the Netherlands; Regenerative Medicine Centre and Circulatory Health Research Centre, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands; Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sandra Crnko
- Department of Pathology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
23
|
Yang Y, Zhang X, Cai D, Zheng X, Zhao X, Zou JX, Zhang J, Borowsky AD, Dall’Era MA, Corey E, Mitsiades N, Kung HJ, Chen X, Li JJ, Downes M, Evans RM, Chen HW. Functional inversion of circadian regulator REV-ERBα leads to tumorigenic gene reprogramming. Proc Natl Acad Sci U S A 2024; 121:e2411321121. [PMID: 39383000 PMCID: PMC11494309 DOI: 10.1073/pnas.2411321121] [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: 06/05/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024] Open
Abstract
Profound functional switch of key regulatory factors may play a major role in homeostasis and disease. Dysregulation of circadian rhythm (CR) is strongly implicated in cancer with mechanisms poorly understood. We report here that the function of REV-ERBα, a major CR regulator of the orphan nuclear receptor subfamily, is dramatically altered in tumors in both its genome binding and functional mode. Loss of CR is linked to a functional inversion of REV-ERBα from a repressor in control of CR and metabolic gene programs in normal tissues to a strong activator in different cancers. Through changing its association from NCoR/HDAC3 corepressor complex to BRD4/p300 coactivators, REV-ERBα directly activates thousands of genes including tumorigenic programs such as MAPK and PI3K-Akt signaling. Functioning as a master transcriptional activator, REV-ERBα partners with pioneer factor FOXA1 and directly stimulates a large number of signaling genes, including multiple growth factors, receptor tyrosine kinases, RASs, AKTs, and MAPKs. Moreover, elevated REV-ERBα reprograms FOXA1 to bind new targets through a BRD4-mediated increase in local chromatin accessibility. Pharmacological targeting with SR8278 diminishes the function of both REV-ERBα and FOXA1 and synergizes with BRD4 inhibitor in effective suppression of tumorigenic programs and tumor growth. Thus, our study revealed a functional inversion by a CR regulator in driving gene reprogramming as an unexpected paradigm of tumorigenesis mechanism and demonstrated a high effectiveness of therapeutic targeting such switch.
Collapse
Affiliation(s)
- Yatian Yang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Xiong Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Demin Cai
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Xingling Zheng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Xuan Zhao
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - June X. Zou
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Jin Zhang
- Department of Surgical & Radiological Sciences, University of California-Davis, Davis, CA95616
| | - Alexander D. Borowsky
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Marc A. Dall’Era
- Department of Urologic Surgery, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA98195
| | - Nicholas Mitsiades
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California Davis, Sacramento, CA95817
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA95817
| | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Xinbin Chen
- Department of Surgical & Radiological Sciences, University of California-Davis, Davis, CA95616
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA95817
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA95817
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA95817
- Veterans Affairs Northern California Health Care System-Mather, Mather, CA95655
| |
Collapse
|
24
|
Pan Y, Chiu TP, Zhou L, Chan P, Kuo TT, Battaglin F, Soni S, Jayachandran P, Li JJ, Lenz HJ, Mumenthaler SM, Rohs R, Torres ER, Kay SA. Targeting circadian transcriptional programs through a cis-regulatory mechanism in triple negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590360. [PMID: 38746115 PMCID: PMC11092448 DOI: 10.1101/2024.04.26.590360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Circadian clock genes are emerging targets in many types of cancer, but their mechanistic contributions to tumor progression are still largely unknown. This makes it challenging to stratify patient populations and develop corresponding treatments. In this work, we show that in breast cancer, the disrupted expression of circadian genes has the potential to serve as biomarkers. We also show that the master circadian transcription factors (TFs) BMAL1 and CLOCK are required for the proliferation of metastatic mesenchymal stem-like (mMSL) triple-negative breast cancer (TNBC) cells. Using currently available small molecule modulators, we found that a stabilizer of cryptochrome 2 (CRY2), the direct repressor of BMAL1 and CLOCK transcriptional activity, synergizes with inhibitors of proteasome, which is required for BMAL1 and CLOCK function, to repress a transcriptional program comprising circadian cycling genes in mMSL TNBC cells. Omics analyses on drug-treated cells implied that this repression of transcription is mediated by the transcription factor binding sites (TFBSs) features in the cis-regulatory elements (CRE) of clock-controlled genes. Through a massive parallel reporter assay, we defined a set of CRE features that are potentially repressed by the specific drug combination. The identification of cis -element enrichment might serve as a new concept of defining and targeting tumor types through the modulation of cis -regulatory programs, and ultimately provide a new paradigm of therapy design for cancer types with unclear drivers like TNBC.
Collapse
|
25
|
Liu Y, Wu J, Najem H, Lin Y, Pang L, Khan F, Zhou F, Ali H, Heimberger AB, Chen P. Dual targeting macrophages and microglia is a therapeutic vulnerability in models of PTEN-deficient glioblastoma. J Clin Invest 2024; 134:e178628. [PMID: 39352749 PMCID: PMC11563674 DOI: 10.1172/jci178628] [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: 12/18/2023] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
Tumor-associated macrophages and microglia (TAMs) are critical for tumor progression and therapy resistance in glioblastoma (GBM), a type of incurable brain cancer. We previously identified lysyl oxidase (LOX) and olfactomedin like-3 (OLFML3) as essential macrophage and microglia chemokines, respectively, in GBM. Here, single-cell transcriptomics and multiplex sequential immunofluorescence followed by functional studies demonstrate that macrophages negatively correlate with microglia in the GBM tumor microenvironment. LOX inhibition in PTEN-deficient GBM cells upregulates OLFML3 expression via the NF-κB-PATZ1 signaling pathway, inducing a compensatory increase of microglia infiltration. Dual targeting macrophages and microglia via inhibition of LOX and the CLOCK-OLFML3 axis generates potent antitumor effects and offers a complete tumor regression in more than 60% of animals when combined with anti-PD1 therapy in PTEN-deficient GBM mouse models. Thus, our findings provide a translational triple therapeutic strategy for this lethal disease.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Junyan Wu
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yiyun Lin
- Department of Genetics and
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lizhi Pang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fatima Khan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fei Zhou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Heba Ali
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
27
|
Li W, Yang LJ, Xiong YY, Li ZS, Li X, Wen Y. 4,5-Dimethoxycanthin-6-one Inhibits Glioblastoma Stem Cell and Tumor Growth by Inhibiting TSPAN1 Interaction with TM4SF1. Neurochem Res 2024; 49:2897-2909. [PMID: 39060768 DOI: 10.1007/s11064-024-04211-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024]
Abstract
Glioblastoma stem cells (GSCs) have been implicated in the self-renewal and treatment resistance of glioblastoma (GBM). Our previous study found that 4,5-dimethoxycanthin-6-one has the potential to inhibit GBM cell proliferation. This current study aims to elucidate the molecular mechanism underlying the effects of 4,5-dimethoxycanthin-6-one in GBM development. The effect of 4,5-dimethoxycanthin-6-one on GSC formation and differentiation was explored in human GBM cell lines U251 and U87. Subsequently, 4,5-dimethoxycanthin-6-one binding to tetraspanin 1 (TSPAN1) / transmembrane 4 L six family member 1 (TM4SF1) was analyzed by molecular simulation docking. Co-immunoprecipitation (Co-IP) and immunofluorescence (IF) were used to assess the interactions between TSPAN1 and TM4SF1 in GSCs. Cell proliferation was detected by cell counting kit-8 (CCK-8) and colony formation assay. To evaluate cell migration, invasion and apoptosis, we employed wound healing assay, transwell and flow cytometry, respectively. Furthermore, subcutaneous xenograft tumor models in nude mice were constructed to evaluate the impact of 4,5-dimethoxycanthin-6-one on GSCs in vivo by examining tumor growth and histological characteristics. 4,5-Dimethoxycanthin-6-one inhibited GSC formation and promoted stem cell differentiation in a concentration-dependent manner. Molecular docking models of 4,5-dimethoxycanthin-6-one with TM4SF1 and TSPAN1 were constructed. Then, the interaction between TSPAN1 and TM4SF1 in GSC was clarified. Moreover, 4,5-dimethoxycanthin-6-one significantly inhibited the expressions of TM4SF1 and TSPAN1 in vitro and in vivo. Overexpression of TSPAN1 partially reversed the inhibitory effects of 4,5-dimethoxycanthin-6-one on GSC formation, proliferation, migration and invasion. 4,5-Dimethoxycanthin-6-one inhibited GBM progression by inhibiting TSPAN1/TM4SF1 axis. 4,5-Dimethoxycanthin-6-one might be a novel and effective drug for the treatment of GBM.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Li-Jian Yang
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yuan-Yuan Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zeng-Shi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xi Li
- Department of Neurosurgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi Wen
- Department of Imaging, Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China.
- Department of Imaging, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
28
|
Rashed N, Liu W, Zhou X, Bode AM, Luo X. The role of circadian gene CLOCK in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119782. [PMID: 38871225 DOI: 10.1016/j.bbamcr.2024.119782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
Circadian Locomotor Output Cycles Kaput (CLOCK) is one of the circadian clock genes and is considered to be a fundamental regulatory gene in the circadian rhythm, responsible for mediating several biological processes. Therefore, abnormal expression of CLOCK affects its role in the circadian clock and its more general function as a direct regulator of gene expression. This dysfunction can lead to severe pathological effects, including cancer. To better understand the role of CLOCK in cancer, we compiled this review to describe the biological function of CLOCK, and especially highlighted its function in cancer development, progression, tumor microenvironment, cancer cell metabolism, and drug resistance.
Collapse
Affiliation(s)
- Nasot Rashed
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Wenbin Liu
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Xinran Zhou
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
29
|
Chan P, Nagai Y, Wu Q, Hovsepyan A, Mkhitaryan S, Wang J, Karapetyan G, Kamenecka T, Solt LA, Cope J, Moats RA, Hirota T, Rich JN, Kay SA. Advancing Clinical Response Against Glioblastoma: Evaluating SHP1705 CRY2 Activator Efficacy in Preclinical Models and Safety in Phase I Trials. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613520. [PMID: 39345648 PMCID: PMC11429762 DOI: 10.1101/2024.09.17.613520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background It has been reported that circadian clock components, Brain and Muscle ARNT-Like 1 (BMAL1) and Circadian Locomotor Output Cycles Kaput (CLOCK), are uniquely essential for glioblastoma (GBM) stem cell (GSC) biology and survival. Consequently, we developed a novel Cryptochrome (CRY) activator SHP1705, which inhibits BMAL1-CLOCK transcriptional activity. Methods We analyzed buffy coats isolated from Phase 1 clinical trial subjects' blood to assess any changes to circadian, housekeeping, and blood transcriptome-based biomarkers following SHP1705 treatment. We utilized GlioVis to determine which circadian genes are differentially expressed in non-tumor versus GBM tissues. We employed in vitro and in vivo methods to test the efficacy of SHP1705 against patient-derived GSCs and xenografts in comparison to earlier CRY activator scaffolds. Additionally, we applied a novel-REV-ERB agonist SR29065, which inhibits BMAL1 transcription, to determine whether targeting both negative limbs of the circadian transcription-translation feedback loop (TTFL) would yield synergistic effects against various GBM cells. Results SHP1705 is safe and well-tolerated in Phase I clinical trials. SHP1705 has increased selectivity for the CRY2 isoform and potency against GSC viability compared to previously published CRY activators. SHP1705 prolonged survival in mice bearing GBM tumors established with GSCs. When combined with the novel REV-ERB agonist SR29065, SHP1705 displayed synergy against multiple GSC lines and differentiated GSCs (DGCs). Conclusions These demonstrate the efficacy of SHP1705 against GSCs, which pose for GBM patient outcomes. They highlight the potential of novel circadian clock compounds in targeting GBM as single agents or in combination with each other or current standard-of-care. KEY POINTS SHP1705 is a novel CRY2 activator that has shown success in Phase 1 safety trialsSHP1705 has a significantly improved efficacy against GSCs and GBM PDX tumorsNovel REV-ERB agonist SR29065 and SHP1705 display synergistic effects against GSCs. IMPORTANCE OF THE STUDY CRY2 is decreased in GBM tissues compared to CRY1 suggesting that promoting CRY2 activity will be an efficacious GBM treatment paradigm. SHP1705, a CRY2 activator that has shown success in Phase 1 safety trials, has significantly improved preclinical efficacy. Novel REV-ERB agonist SR29065 displays synergistic effects against diverse GBM cells.
Collapse
|
30
|
Yadav H, Bakshi A, Anamika, Singh V, Paul P, Murugan NA, Maurya SK. Co-localization and co-expression of Olfml3 with Iba1 in brain of mice. J Neuroimmunol 2024; 394:578411. [PMID: 39079458 DOI: 10.1016/j.jneuroim.2024.578411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/30/2024]
Abstract
Olfml3 is a microglia-specific protein whose role in neuroinflammation is elusive. In silico analysis was conducted to characterize the Olfml3 protein, followed by molecular docking and MD simulation to check possible interaction with Iba1. Further, expression and co-localization analysis was performed in the LPS-induced neuroinflammatory mice brains. Results suggest that Olfml3 physically interacts with Iba1. Olfml3 and Iba1 expression increases during neuroinflammation in mice brains. Olfml3 was observed to co-localize with Iba1, and the number of Olfml3 and Iba1 dual-positive cells increased in the brain of the neuroinflammatory mice model. Thus, Olfml3 could potentially participate in microglia functions by interacting with Iba1.
Collapse
Affiliation(s)
- Himanshi Yadav
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Amrita Bakshi
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Anamika
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Vishal Singh
- Electron Microscope Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prateek Paul
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - N Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Industrial Estate, Delhi, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India.
| |
Collapse
|
31
|
Le MK, Tran NQV, Nguyen PT, Nguyen TA, Nakao A, Kondo T. Comprehensive analysis of distinct circadian clock subtypes of adult diffuse glioma and their associations with clinicopathological, genetic, and epigenetic profiles. J Neuropathol Exp Neurol 2024; 83:736-744. [PMID: 38964366 DOI: 10.1093/jnen/nlae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
The circadian clock (CC) has biological and clinical implications in gliomas. Most studies focused on CC effects on the tumor microenvironment and the application of chronotherapy. The present study focused on CC gene expression patterns and intracellular oncogenic activities. Glioma gene expression data were collected from The Human Cancer Genome Atlas (TCGA) project. After applying inclusion and exclusion criteria, we selected 666 patients from TCGA-GBM and TCGA-LGG projects and included important clinicopathological variables. The entire cohort was subjected to clustering analysis and divided into CC1 and CC2 subtypes based on statistical, biological, and clinical criteria. CC2 gliomas showed higher expression of BMAL1 and CRY1 and lower expression of CRY2 and PER2 (adjusted P < .001). CC2 gliomas had q higher activity of cell proliferation, metabolic reprogramming, angiogenesis, hypoxia, and many oncogenic signals (P < .001). The CC2 subtype contained a higher proportion of glioblastomas (P < .001) and had a worse prognosis (P < .001). Stratified Kaplan-Meier and multivariable Cox analyses illustrated that the CC subtype is an independent prognostic factor to clinicopathological characteristics (P < .001), genetic aberrations (P = .006), and biological processes (P < .001). Thus, this study shows statistical evidence of CC subtypes and their biological, and clinicopathological significance in adult gliomas.
Collapse
Affiliation(s)
- Minh-Khang Le
- Department of Human Pathology, University of Yamanashi, Yamanashi, Japan
| | | | - Phuc-Tan Nguyen
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Thuy-An Nguyen
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Atsuhito Nakao
- Department of Immunology, University of Yamanashi, Yamanashi, Japan
| | - Tetsuo Kondo
- Department of Human Pathology, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
32
|
Kabir AU, Zeng C, Subramanian M, Wu J, Kim M, Krchma K, Wang X, Halabi CM, Pan H, Wickline SA, Fremont DH, Artyomov MN, Choi K. ZBTB46 coordinates angiogenesis and immunity to control tumor outcome. Nat Immunol 2024; 25:1546-1554. [PMID: 39134750 DOI: 10.1038/s41590-024-01936-4] [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: 03/11/2023] [Accepted: 07/16/2024] [Indexed: 09/01/2024]
Abstract
Tumor angiogenesis and immunity show an inverse correlation in cancer progression and outcome1. Here, we report that ZBTB46, a repressive transcription factor and a widely accepted marker for classical dendritic cells (DCs)2,3, controls both tumor angiogenesis and immunity. Zbtb46 was downregulated in both DCs and endothelial cells by tumor-derived factors to facilitate robust tumor growth. Zbtb46 downregulation led to a hallmark pro-tumor microenvironment (TME), including dysfunctional vasculature and immunosuppressive conditions. Analysis of human cancer data revealed a similar association of low ZBTB46 expression with an immunosuppressive TME and a worse prognosis. In contrast, enforced Zbtb46 expression led to TME changes to restrict tumor growth. Mechanistically, Zbtb46-deficient endothelial cells were highly angiogenic, and Zbtb46-deficient bone marrow progenitors upregulated Cebpb and diverted the DC program to immunosuppressive myeloid lineage output, potentially explaining the myeloid lineage skewing phenomenon in cancer4. Conversely, enforced Zbtb46 expression normalized tumor vessels and, by suppressing Cebpb, skewed bone marrow precursors toward immunostimulatory myeloid lineage output, leading to an immune-hot TME. Remarkably, Zbtb46 mRNA treatment synergized with anti-PD1 immunotherapy to improve tumor management in preclinical models. These findings identify ZBTB46 as a critical factor for angiogenesis and for myeloid lineage skewing in cancer and suggest that maintaining its expression could have therapeutic benefits.
Collapse
Affiliation(s)
- Ashraf Ul Kabir
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carisa Zeng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Madhav Subramanian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Minseo Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen M Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Hua Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel A Wickline
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
33
|
Wang Z, Ma L, Meng Y, Fang J, Xu D, Lu Z. The interplay of the circadian clock and metabolic tumorigenesis. Trends Cell Biol 2024; 34:742-755. [PMID: 38061936 DOI: 10.1016/j.tcb.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 09/08/2024]
Abstract
The circadian clock and cell metabolism are both dysregulated in cancer cells through intrinsic cell-autonomous mechanisms and external influences from the tumor microenvironment. The intricate interplay between the circadian clock and cancer cell metabolism exerts control over various metabolic processes, including aerobic glycolysis, de novo nucleotide synthesis, glutamine and protein metabolism, lipid metabolism, mitochondrial metabolism, and redox homeostasis in cancer cells. Importantly, oncogenic signaling can confer a moonlighting function on core clock genes, effectively reshaping cellular metabolism to fuel cancer cell proliferation and drive tumor growth. These interwoven regulatory mechanisms constitute a distinctive feature of cancer cell metabolism.
Collapse
Affiliation(s)
- Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China
| | - Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Jing Fang
- Department of Oncology, The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, Shandong 266003, China.
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| |
Collapse
|
34
|
Yin H, Liu Y, Dong Q, Wang H, Yan Y, Wang X, Wan X, Yuan G, Pan Y. The mechanism of extracellular CypB promotes glioblastoma adaptation to glutamine deprivation microenvironment. Cancer Lett 2024; 597:216862. [PMID: 38582396 DOI: 10.1016/j.canlet.2024.216862] [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: 02/13/2024] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Glioblastoma, previously known as glioblastoma multiform (GBM), is a type of glioma with a high degree of malignancy and rapid growth rate. It is highly dependent on glutamine (Gln) metabolism during proliferation and lags in neoangiogenesis, leading to extensive Gln depletion in the core region of GBM. Gln-derived glutamate is used to synthesize the antioxidant Glutathione (GSH). We demonstrated that GSH levels are also reduced in Gln deficiency, leading to increased reactive oxygen species (ROS) levels. The ROS production induces endoplasmic reticulum (ER) stress, and the proteins in the ER are secreted into the extracellular medium. We collected GBM cell supernatants cultured with or without Gln medium; the core and peripheral regions of human GBM tumor tissues. Proteomic analysis was used to screen out the target-secreted protein CypB. We demonstrated that the extracellular CypB expression is associated with Gln deprivation. Then, we verified that GBM can promote the glycolytic pathway by activating HIF-1α to upregulate the expression of GLUT1 and LDHA. Meanwhile, the DRP1 was activated, increasing mitochondrial fission, thus inhibiting mitochondrial function. To explore the specific mechanism of its regulation, we constructed a si-CD147 knockout model and added human recombinant CypB protein to verify that extracellular CypB influenced the expression of downstream p-AKT through its cell membrane receptor CD147 binding. Moreover, we confirmed that p-AKT could upregulate HIF-1α and DRP1. Finally, we observed that extracellular CypB can bind to the CD147 receptor, activate p-AKT, upregulate HIF-1α and DRP1 in order to promote glycolysis while inhibiting mitochondrial function to adapt to the Gln-deprived microenvironment.
Collapse
Affiliation(s)
- Hang Yin
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Yang Liu
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China
| | - Qiang Dong
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hongyu Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Yunji Yan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoqing Wang
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China
| | - Xiaoyu Wan
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore, Singapore; School of Basic Medicine, Henan University, Kaifeng, China
| | - Guoqiang Yuan
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China.
| | - Yawen Pan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
35
|
Trebucq LL, Salvatore N, Wagner PM, Golombek DA, Chiesa JJ. Circadian Clock Gene bmal1 Acts as a Tumor Suppressor Gene in a Mice Model of Human Glioblastoma. Mol Neurobiol 2024; 61:5216-5229. [PMID: 38180613 DOI: 10.1007/s12035-023-03895-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
Glioblastomas derived from malignant astrocytes are the most common primary tumors of the central nervous system in humans, exhibiting very bad prognosis. Treatment with surgery, radiotherapy, and chemotherapy (mainly using temozolomide), generates as much one-year survival. The circadian clock controls different aspects of tumor development, and its role in GBM is beginning to be explored. Here, the role of the canonic circadian clock gene bmal1 was studied in vivo in a nude mice model bearing human GBMs from LN229 cells xenografted orthotopically in the dorsal striatum. For that aim, a bmal1 knock-down was generated in LN229 cells by CRISPR/Cas9 gene editing tool, and tumor progression was followed in male mice by measuring survival, tumor growth, cell proliferation and prognosis with CD44 marker, as well as astrocyte activation in the tumor microenvironment with GFAP and nestin markers. Disruption of bmal1 in the tumor decreased survival, increased tumor growth and CD44 expression, worsened motor performance, as well as increased GFAP expression in astrocytes at tumor microenvironment. In addition, survival and tumor progression was not affected in mice bearing LN229 wild type GBM that underwent circadian disruption by constant light, as compared to mice synchronized to 12:12 light-dark cycles. These results consistently demonstrate in an in vivo orthotopic model of human GBM, that bmal1 has a key role as a tumor suppressor gene regulating GBM progression.
Collapse
Affiliation(s)
- Laura L Trebucq
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Roque S. Peña 352, B1876BXD, Bernal, Buenos Aires, Argentina
| | - Nicolas Salvatore
- Laboratorio de Biotransformaciones y Química de Ácidos Nucleicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Roque S. Peña 352, B1876BXD, Bernal, Buenos Aires, Argentina
| | - Paula M Wagner
- CIQUIBIC-CONICET y Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Diego A Golombek
- Laboratorio Interdisciplinario del Tiempo (LITERA), Universidad de San Andrés, B1644BID, Victoria, Buenos Aires, Argentina
| | - Juan J Chiesa
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Roque S. Peña 352, B1876BXD, Bernal, Buenos Aires, Argentina.
| |
Collapse
|
36
|
Lu J, Zhang X, Su K, Luo H, Liu C, Yang Y, He B, Wang C, Zhao Z, Liu X, Wang X, Meng P, Lv D, Wang C, Kelley KW, Wang L, Cui B, Liu Q, Peng F. Olanzapine suppresses mPFC activity-norepinephrine releasing to alleviate CLOCK-enhanced cancer stemness under chronic stress. Cell Commun Signal 2024; 22:375. [PMID: 39054537 PMCID: PMC11270788 DOI: 10.1186/s12964-024-01747-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Olanzapine (OLZ) reverses chronic stress-induced anxiety. Chronic stress promotes cancer development via abnormal neuro-endocrine activation. However, how intervention of brain-body interaction reverses chronic stress-induced tumorigenesis remains elusive. METHODS KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model were used to study the effect of OLZ on cancer stemness and anxiety-like behaviors. Cancer stemness was evaluated by qPCR, western-blotting, immunohistology staining and flow-cytometry analysis of stemness markers, and cancer stem-like function was assessed by serial dilution tumorigenesis in mice and extreme limiting dilution analysis in primary tumor cells. Anxiety-like behaviors in mice were detected by elevated plus maze and open field test. Depression-like behaviors in mice were detected by tail suspension test. Anxiety and depression states in human were assessed by Hospital Anxiety and Depression Scale (HADS). Chemo-sensitivity of lung cancer was assessed by in vivo syngeneic tumor model and in vitro CCK-8 assay in lung cancer cell lines. RESULTS In this study, we found that OLZ reversed chronic stress-enhanced lung tumorigenesis in both KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model. OLZ relieved anxiety and depression-like behaviors by suppressing neuro-activity in the mPFC and reducing norepinephrine (NE) releasing under chronic stress. NE activated ADRB2-cAMP-PKA-CREB pathway to promote CLOCK transcription, leading to cancer stem-like traits. As such, CLOCK-deficiency or OLZ reverses NE/chronic stress-induced gemcitabine (GEM) resistance in lung cancer. Of note, tumoral CLOCK expression is positively associated with stress status, serum NE level and poor prognosis in lung cancer patients. CONCLUSION We identify a new mechanism by which OLZ ameliorates chronic stress-enhanced tumorigenesis and chemoresistance. OLZ suppresses mPFC-NE-CLOCK axis to reverse chronic stress-induced anxiety-like behaviors and lung cancer stemness. Decreased NE-releasing prevents activation of ADRB2-cAMP-PKA-CREB pathway to inhibit CLOCK transcription, thus reversing lung cancer stem-like traits and chemoresistance under chronic stress.
Collapse
Affiliation(s)
- Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Department of Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Congcong Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuqing Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhuoran Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xianxian Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Peixuan Meng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chunli Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine, Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ling Wang
- Department of Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
37
|
Fekry B, Ugartemendia L, Esnaola NF, Goetzl L. Extracellular Vesicles, Circadian Rhythms, and Cancer: A Comprehensive Review with Emphasis on Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:2552. [PMID: 39061191 PMCID: PMC11274441 DOI: 10.3390/cancers16142552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
This review comprehensively explores the complex interplay between extracellular vesicles (ECVs)/exosomes and circadian rhythms, with a focus on the role of this interaction in hepatocellular carcinoma (HCC). Exosomes are nanovesicles derived from cells that facilitate intercellular communication by transporting bioactive molecules such as proteins, lipids, and RNA/DNA species. ECVs are implicated in a range of diseases, where they play crucial roles in signaling between cells and their surrounding environment. In the setting of cancer, ECVs are known to influence cancer initiation and progression. The scope of this review extends to all cancer types, synthesizing existing knowledge on the various roles of ECVs. A unique aspect of this review is the emphasis on the circadian-controlled release and composition of exosomes, highlighting their potential as biomarkers for early cancer detection and monitoring metastasis. We also discuss how circadian rhythms affect multiple cancer-related pathways, proposing that disruptions in the circadian clock can alter tumor development and treatment response. Additionally, this review delves into the influence of circadian clock components on ECV biogenesis and their impact on reshaping the tumor microenvironment, a key component driving HCC progression. Finally, we address the potential clinical applications of ECVs, particularly their use as diagnostic tools and drug delivery vehicles, while considering the challenges associated with clinical implementation.
Collapse
Affiliation(s)
- Baharan Fekry
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| | - Lierni Ugartemendia
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| | - Nestor F. Esnaola
- Division of Surgical Oncology and Gastrointestinal Surgery, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA;
| | - Laura Goetzl
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| |
Collapse
|
38
|
Peng F, Lu J, Su K, Liu X, Luo H, He B, Wang C, Zhang X, An F, Lv D, Luo Y, Su Q, Jiang T, Deng Z, He B, Xu L, Guo T, Xiang J, Gu C, Wang L, Xu G, Xu Y, Li M, Kelley KW, Cui B, Liu Q. Oncogenic fatty acid oxidation senses circadian disruption in sleep-deficiency-enhanced tumorigenesis. Cell Metab 2024; 36:1598-1618.e11. [PMID: 38772364 DOI: 10.1016/j.cmet.2024.04.018] [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: 11/02/2022] [Revised: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Circadian disruption predicts poor cancer prognosis, yet how circadian disruption is sensed in sleep-deficiency (SD)-enhanced tumorigenesis remains obscure. Here, we show fatty acid oxidation (FAO) as a circadian sensor relaying from clock disruption to oncogenic metabolic signal in SD-enhanced lung tumorigenesis. Both unbiased transcriptomic and metabolomic analyses reveal that FAO senses SD-induced circadian disruption, as illustrated by continuously increased palmitoyl-coenzyme A (PA-CoA) catalyzed by long-chain fatty acyl-CoA synthetase 1 (ACSL1). Mechanistically, SD-dysregulated CLOCK hypertransactivates ACSL1 to produce PA-CoA, which facilitates CLOCK-Cys194 S-palmitoylation in a ZDHHC5-dependent manner. This positive transcription-palmitoylation feedback loop prevents ubiquitin-proteasomal degradation of CLOCK, causing FAO-sensed circadian disruption to maintain SD-enhanced cancer stemness. Intriguingly, timed β-endorphin resets rhythmic Clock and Acsl1 expression to alleviate SD-enhanced tumorigenesis. Sleep quality and serum β-endorphin are negatively associated with both cancer development and CLOCK/ACSL1 expression in patients with cancer, suggesting dawn-supplemented β-endorphin as a potential chronotherapeutic strategy for SD-related cancer.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Jinxin Lu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Keyu Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinyu Liu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Huandong Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Fan An
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Dekang Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Yuanyuan Luo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Qitong Su
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Tonghui Jiang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Ziqian Deng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lingzhi Xu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jin Xiang
- State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ling Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guowang Xu
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning, China
| | - Ying Xu
- Cambridge-Soochow University Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Mindian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Keith W Kelley
- Department of Pathology, College of Medicine and Department of Animal Sciences, College of ACES, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China; State Key Laboratory of Oncology in South China, Psychobehavioral Cancer Research Center, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
39
|
Niu Z, Yang Z, Sun S, Zeng Z, Han Q, Wu L, Bai J, Li H, Xia H. Clinical analysis of the efficacy of radiation therapy for primary high-grade gliomas guided by biological rhythms. Transl Oncol 2024; 45:101973. [PMID: 38705052 PMCID: PMC11089398 DOI: 10.1016/j.tranon.2024.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/05/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE High-grade glioma (HGG) patients frequently encounter treatment resistance and relapse, despite numerous interventions seeking enhanced survival outcomes yielding limited success. Consequently, this study, rooted in our prior research, aimed to ascertain whether leveraging circadian rhythm phase attributes could optimize radiotherapy results. METHODS In this retrospective analysis, we meticulously selected 121 HGG cases with synchronized rhythms through Cosinor analysis. Post-surgery, all subjects underwent standard radiotherapy alongside Temozolomide chemotherapy. Random allocation ensued, dividing patients into morning (N = 69) and afternoon (N = 52) radiotherapy cohorts, enabling a comparison of survival and toxicity disparities. RESULTS The afternoon radiotherapy group exhibited improved overall survival (OS) and progression-free survival (PFS) relative to the morning cohort. Notably, median OS extended to 25.6 months versus 18.5 months, with P = 0.014, with median PFS at 20.6 months versus 13.3 months, with P = 0.022, post-standardized radiotherapy. Additionally, lymphocyte expression levels in the afternoon radiation group 32.90(26.10, 39.10) significantly exceeded those in the morning group 31.30(26.50, 39.20), with P = 0.032. CONCLUSIONS This study underscores the markedly prolonged average survival within the afternoon radiotherapy group. Moreover, lymphocyte proportion demonstrated a notable elevation in the afternoon group. Timely and strategic adjustments of therapeutic interventions show the potential to improve therapeutic efficacy, while maintaining vigilant systemic immune surveillance. A comprehensive grasp of physiological rhythms governing both the human body and tumor microenvironment can refine treatment efficacy, concurrently curtailing immune-related damage-a crucial facet of precision medicine.
Collapse
Affiliation(s)
- Zhanfeng Niu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Zhihua Yang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Shengyu Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Zhong Zeng
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China
| | - Qian Han
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China
| | - Liang Wu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Jinbo Bai
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Hailiang Li
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, PR China; Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, PR China.
| |
Collapse
|
40
|
Fortin BM, Pfeiffer SM, Insua-Rodríguez J, Alshetaiwi H, Moshensky A, Song WA, Mahieu AL, Chun SK, Lewis AN, Hsu A, Adam I, Eng OS, Pannunzio NR, Seldin MM, Marazzi I, Marangoni F, Lawson DA, Kessenbrock K, Masri S. Circadian control of tumor immunosuppression affects efficacy of immune checkpoint blockade. Nat Immunol 2024; 25:1257-1269. [PMID: 38806707 PMCID: PMC11374317 DOI: 10.1038/s41590-024-01859-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Bridget M Fortin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Shannon M Pfeiffer
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Jacob Insua-Rodríguez
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Hamad Alshetaiwi
- Department of Pathology, University of Hail, Hail, Saudi Arabia
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Alexander Moshensky
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Wei A Song
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alisa L Mahieu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Sung Kook Chun
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Amber N Lewis
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alex Hsu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Isam Adam
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Oliver S Eng
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Surgery, Division of Surgical Oncology, University of California Irvine, Orange, CA, USA
| | - Nicholas R Pannunzio
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Francesco Marangoni
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Devon A Lawson
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA
| | - Selma Masri
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
41
|
Feldman L. Hypoxia within the glioblastoma tumor microenvironment: a master saboteur of novel treatments. Front Immunol 2024; 15:1384249. [PMID: 38994360 PMCID: PMC11238147 DOI: 10.3389/fimmu.2024.1384249] [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: 02/09/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Glioblastoma (GBM) tumors are the most aggressive primary brain tumors in adults that, despite maximum treatment, carry a dismal prognosis. GBM tumors exhibit tissue hypoxia, which promotes tumor aggressiveness and maintenance of glioma stem cells and creates an overall immunosuppressive landscape. This article reviews how hypoxic conditions overlap with inflammatory responses, favoring the proliferation of immunosuppressive cells and inhibiting cytotoxic T cell development. Immunotherapies, including vaccines, immune checkpoint inhibitors, and CAR-T cell therapy, represent promising avenues for GBM treatment. However, challenges such as tumor heterogeneity, immunosuppressive TME, and BBB restrictiveness hinder their effectiveness. Strategies to address these challenges, including combination therapies and targeting hypoxia, are actively being explored to improve outcomes for GBM patients. Targeting hypoxia in combination with immunotherapy represents a potential strategy to enhance treatment efficacy.
Collapse
Affiliation(s)
- Lisa Feldman
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
42
|
Pang L, Zhou F, Liu Y, Ali H, Khan F, Heimberger AB, Chen P. Epigenetic regulation of tumor immunity. J Clin Invest 2024; 134:e178540. [PMID: 39133578 PMCID: PMC11178542 DOI: 10.1172/jci178540] [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: 08/16/2024] Open
Abstract
Although cancer has long been considered a genetic disease, increasing evidence shows that epigenetic aberrations play a crucial role in affecting tumor biology and therapeutic response. The dysregulated epigenome in cancer cells reprograms the immune landscape within the tumor microenvironment, thereby hindering antitumor immunity, promoting tumor progression, and inducing immunotherapy resistance. Targeting epigenetically mediated tumor-immune crosstalk is an emerging strategy to inhibit tumor progression and circumvent the limitations of current immunotherapies, including immune checkpoint inhibitors. In this Review, we discuss the mechanisms by which epigenetic aberrations regulate tumor-immune interactions and how epigenetically targeted therapies inhibit tumor progression and synergize with immunotherapy.
Collapse
|
43
|
Wu L, Zhao Z, Shin YJ, Yin Y, Raju A, Vaiyapuri TS, Idzham K, Son M, Lee Y, Sa JK, Chua JYH, Unal B, Zhai Y, Fan W, Huang L, Hu H, Gunaratne J, Nam DH, Jiang T, Tergaonkar V. Tumour microenvironment programming by an RNA-RNA-binding protein complex creates a druggable vulnerability in IDH-wild-type glioblastoma. Nat Cell Biol 2024; 26:1003-1018. [PMID: 38858501 PMCID: PMC11178504 DOI: 10.1038/s41556-024-01428-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Patients with IDH-wild-type glioblastomas have a poor five-year survival rate along with limited treatment efficacy due to immune cell (glioma-associated microglia and macrophages) infiltration promoting tumour growth and resistance. To enhance therapeutic options, our study investigated the unique RNA-RNA-binding protein complex LOC-DHX15. This complex plays a crucial role in driving immune cell infiltration and tumour growth by establishing a feedback loop between cancer and immune cells, intensifying cancer aggressiveness. Targeting this complex with blood-brain barrier-permeable small molecules improved treatment efficacy, disrupting cell communication and impeding cancer cell survival and stem-like properties. Focusing on RNA-RNA-binding protein interactions emerges as a promising approach not only for glioblastomas without the IDH mutation but also for potential applications beyond cancer, offering new avenues for developing therapies that address intricate cellular relationships in the body.
Collapse
Affiliation(s)
- Lele Wu
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Zheng Zhao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yong Jae Shin
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Yiyun Yin
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Anandhkumar Raju
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Thamil Selvan Vaiyapuri
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Khaireen Idzham
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Miseol Son
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Yeri Lee
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joelle Yi Heng Chua
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Bilal Unal
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - You Zhai
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wenhua Fan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lijie Huang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Huimin Hu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jayantha Gunaratne
- Laboratory of Translational Biomedical Proteomics, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Do-Hyun Nam
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurosurgery, Samsung Medical Center, Seoul, Republic of Korea
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Republic of Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Republic of Singapore.
| |
Collapse
|
44
|
Chen K, Wang Y, Li D, Wu R, Wang J, Wei W, Zhu W, Xie W, Feng D, He Y. Biological clock regulation by the PER gene family: a new perspective on tumor development. Front Cell Dev Biol 2024; 12:1332506. [PMID: 38813085 PMCID: PMC11133573 DOI: 10.3389/fcell.2024.1332506] [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: 11/03/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
The Period (PER) gene family is one of the core components of the circadian clock, with substantial correlations between the PER genes and cancers identified in extensive researches. Abnormal mutations in PER genes can influence cell function, metabolic activity, immunity, and therapy responses, thereby promoting the initiation and development of cancers. This ultimately results in unequal cancers progression and prognosis in patients. This leads to variable cancer progression and prognosis among patients. In-depth studies on the interactions between the PER genes and cancers can reveal novel strategies for cancer detection and treatment. In this review, we aim to provide a comprehensive overview of the latest research on the role of the PER gene family in cancer.
Collapse
Affiliation(s)
- Kai Chen
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Wang
- Department of Urology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhu
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| | - Wenhua Xie
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Yi He
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jia Xing, China
| |
Collapse
|
45
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
46
|
Loh JJ, Ma S. Hallmarks of cancer stemness. Cell Stem Cell 2024; 31:617-639. [PMID: 38701757 DOI: 10.1016/j.stem.2024.04.004] [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: 12/31/2023] [Revised: 03/11/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024]
Abstract
Cancer stemness is recognized as a key component of tumor development. Previously coined "cancer stem cells" (CSCs) and believed to be a rare population with rigid hierarchical organization, there is good evidence to suggest that these cells exhibit a plastic cellular state influenced by dynamic CSC-niche interplay. This revelation underscores the need to reevaluate the hallmarks of cancer stemness. Herein, we summarize the techniques used to identify and characterize the state of these cells and discuss their defining and emerging hallmarks, along with their enabling and associated features. We also highlight potential future directions in this field of research.
Collapse
Affiliation(s)
- Jia-Jian Loh
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong SAR, China; Laboratory of Synthetic Chemistry and Chemical Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China; Centre for Translational and Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China.
| |
Collapse
|
47
|
Liu Y, Ali H, Khan F, Pang L, Chen P. Epigenetic regulation of tumor-immune symbiosis in glioma. Trends Mol Med 2024; 30:429-442. [PMID: 38453529 PMCID: PMC11081824 DOI: 10.1016/j.molmed.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
Glioma is a type of aggressive and incurable brain tumor. Patients with glioma are highly resistant to all types of therapies, including immunotherapies. Epigenetic reprogramming is a key molecular hallmark in tumors across cancer types, including glioma. Mounting evidence highlights a pivotal role of epigenetic regulation in shaping tumor biology and therapeutic responses through mechanisms involving both glioma cells and immune cells, as well as their symbiotic interactions in the tumor microenvironment (TME). In this review, we discuss the molecular mechanisms of epigenetic regulation that impacts glioma cell biology and tumor immunity in both a cell-autonomous and non-cell-autonomous manner. Moreover, we provide an overview of potential therapeutic approaches that can disrupt epigenetic-regulated tumor-immune symbiosis in the glioma TME.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heba Ali
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lizhi Pang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
48
|
Zeng Y, Guo Z, Wu M, Chen F, Chen L. Circadian rhythm regulates the function of immune cells and participates in the development of tumors. Cell Death Discov 2024; 10:199. [PMID: 38678017 PMCID: PMC11055927 DOI: 10.1038/s41420-024-01960-1] [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: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
Circadian rhythms are present in almost all cells and play a crucial role in regulating various biological processes. Maintaining a stable circadian rhythm is essential for overall health. Disruption of this rhythm can alter the expression of clock genes and cancer-related genes, and affect many metabolic pathways and factors, thereby affecting the function of the immune system and contributing to the occurrence and progression of tumors. This paper aims to elucidate the regulatory effects of BMAL1, clock and other clock genes on immune cells, and reveal the molecular mechanism of circadian rhythm's involvement in tumor and its microenvironment regulation. A deeper understanding of circadian rhythms has the potential to provide new strategies for the treatment of cancer and other immune-related diseases.
Collapse
Affiliation(s)
- Yuen Zeng
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China
| | - Zichan Guo
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Mengqi Wu
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China
| | - Fulin Chen
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medical Sciences, Air Force Medical University, Xi'an, China.
| |
Collapse
|
49
|
Licón-Muñoz Y, Avalos V, Subramanian S, Granger B, Martinez F, Varela S, Moore D, Perkins E, Kogan M, Berto S, Chohan M, Bowers C, Piccirillo S. Single-nucleus and spatial landscape of the sub-ventricular zone in human glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590852. [PMID: 38712234 PMCID: PMC11071523 DOI: 10.1101/2024.04.24.590852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The sub-ventricular zone (SVZ) is the most well-characterized neurogenic area in the mammalian brain. We previously showed that in 65% of patients with glioblastoma (GBM), the SVZ is a reservoir of cancer stem-like cells that contribute to treatment resistance and emergence of recurrence. Here, we built a single-nucleus RNA-sequencing-based microenvironment landscape of the tumor mass (T_Mass) and the SVZ (T_SVZ) of 15 GBM patients and 2 histologically normal SVZ (N_SVZ) samples as controls. We identified a mesenchymal signature in the T_SVZ of GBM patients: tumor cells from the T_SVZ relied on the ZEB1 regulatory network, whereas tumor cells in the T_Mass relied on the TEAD1 regulatory network. Moreover, the T_SVZ microenvironment was predominantly characterized by tumor-supportive microglia, which spatially co-exist and establish heterotypic interactions with tumor cells. Lastly, differential gene expression analyses, predictions of ligand-receptor and incoming/outgoing interactions, and functional assays revealed that the IL-1β/IL-1RAcP and Wnt-5a/Frizzled-3 pathways are therapeutic targets in the T_SVZ microenvironment. Our data provide insights into the biology of the SVZ in GBM patients and identify specific targets of this microenvironment.
Collapse
Affiliation(s)
- Y. Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - V. Avalos
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - S. Subramanian
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - B. Granger
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - F. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - S. Varela
- University of New Mexico School of Medicine, Albuquerque, NM
| | - D. Moore
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - E. Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS
| | - M. Kogan
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM
| | - S. Berto
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - M.O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS
| | - C.A. Bowers
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM
| | - S.G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| |
Collapse
|
50
|
Zhang Z, Zhang Y. Transcriptional regulation of cancer stem cell: regulatory factors elucidation and cancer treatment strategies. J Exp Clin Cancer Res 2024; 43:99. [PMID: 38561775 PMCID: PMC10986082 DOI: 10.1186/s13046-024-03021-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer stem cells (CSCs) were first discovered in the 1990s, revealing the mysteries of cancer origin, migration, recurrence and drug-resistance from a new perspective. The expression of pluripotent genes and complex signal regulatory networks are significant features of CSC, also act as core factors to affect the characteristics of CSC. Transcription is a necessary link to regulate the phenotype and potential of CSC, involving chromatin environment, nucleosome occupancy, histone modification, transcription factor (TF) availability and cis-regulatory elements, which suffer from ambient pressure. Especially, the expression and activity of pluripotent TFs are deeply affected by both internal and external factors, which is the foundation of CSC transcriptional regulation in the current research framework. Growing evidence indicates that regulating epigenetic modifications to alter cancer stemness is effective, and some special promoters and enhancers can serve as targets to influence the properties of CSC. Clarifying the factors that regulate CSC transcription will assist us directly target key stem genes and TFs, or hinder CSC transcription through environmental and other related factors, in order to achieve the goal of inhibiting CSC and tumors. This paper comprehensively reviews the traditional aspects of transcriptional regulation, and explores the progress and insights of the impact on CSC transcription and status through tumor microenvironment (TME), hypoxia, metabolism and new meaningful regulatory factors in conjunction with the latest research. Finally, we present opinions on omnidirectional targeting CSCs transcription to eliminate CSCs and address tumor resistance.
Collapse
Affiliation(s)
- Zhengyue Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, People's Republic of China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, People's Republic of China
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, People's Republic of China.
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, People's Republic of China.
| |
Collapse
|