1
|
Li J, Long S, Yang Z, Wei W, Yu S, Liu Q, Hui X, Li X, Wang Y. Single-cell transcriptomics reveals IRF7 regulation of the tumor microenvironment in isocitrate dehydrogenase wild-type glioma. MedComm (Beijing) 2024; 5:e754. [PMID: 39492838 PMCID: PMC11531655 DOI: 10.1002/mco2.754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 11/05/2024] Open
Abstract
Mutations in isocitrate dehydrogenase (IDH) are important markers of glioma prognosis. However, few studies have examined the gene expression regulatory network (GRN) in IDH-mutant and wild-type gliomas. In this study, single-cell RNA sequencing and spatial transcriptome sequencing were used to analyze the GRN of cell subsets in patients with IDH-mutant and wild-type gliomas. Through gene transcriptional regulation analysis, we identified the M4 module, whose transcription factor activity is highly expressed in IDH wild-type gliomas compared to IDH-mutants. Enrichment analysis revealed that these genes were predominantly expressed in microglia and macrophages, with significant enrichment in interferon-related signaling pathways. Interferon regulatory factor 7 (IRF7), a transcription factor within this pathway, showed the highest percentage of enrichment and was primarily localized in the core region of wild-type IDH tumors. A machine-learning prognostic model identified novel subgroups within the wild-type IDH population. Additionally, IRF7 was shown to promote the proliferation and migration of T98G and U251 cells in vitro, and its knockdown affected glioma cell proliferation in vivo. This study systematically established the regulatory mechanism of IDH transcriptional activity in gliomas at the single-cell level and drew a corresponding cell map. The study presents a transcriptional regulatory activity map for IDH wild-type gliomas, involving single-cell RNA sequencing and spatial transcriptomics to identify gene regulatory networks, machine learning models for IDH subtyping, and experimental validation, highlighting the role of IRF7 in glioma progression.
Collapse
Affiliation(s)
- Jinwei Li
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Shengrong Long
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Brain Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zhang Yang
- Department of Vascular SurgeryFuwai Yunnan Cardiovascular HospitalAffiliated Cardiovascular Hospital of Kunming Medical UniversityKunmingChina
| | - Wei Wei
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Brain Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Shuangqi Yu
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Brain Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Quan Liu
- Department of NeurosurgeryThe Fourth Affiliated Hospital of Guangxi Medical UniversityLiuzhouChina
| | - Xuhui Hui
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Xiang Li
- Department of NeurosurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Brain Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Yinyan Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| |
Collapse
|
2
|
Motomura K, Sasaki K, Sugii N, Yamaguchi S, Inoue H, Oshima A, Tanaka K, Otani Y, Shirahata M, Shibahara I, Nagane M, Tsuzuki S, Matsutani T, Tsukamoto Y, Kijima N, Asano K, Ohno M, Inoue A, Mineharu Y, Miyake K, Mitobe Y, Hanihara M, Kawanishi Y, Deguchi S, Saito M, Matsuda R, Ujifuku K, Arita H, Sato Y, Yamashita S, Yonezawa U, Yamaguchi J, Momii Y, Ogawa T, Kambe A, Ohba S, Fukai J, Saito N, Kinoshita M, Sumi K, Otani R, Uzuka T, Takebe N, Koizumi S, Saito R, Arakawa Y, Narita Y. Cost of medical care for malignant brain tumors at hospitals in the Japan Clinical Oncology Group brain-tumor study group. Jpn J Clin Oncol 2024; 54:1123-1131. [PMID: 39223700 PMCID: PMC11456849 DOI: 10.1093/jjco/hyae116] [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: 04/27/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study aimed to investigate what treatment are selected for malignant brain tumors, particularly glioblastoma (GBM) and primary central nervous system lymphoma (PCNSL), in real-world Japan and the costs involved. METHODS We conducted a questionnaire survey regarding treatment selections for newly diagnosed GBM and PCNSL treated between July 2021 and June 2022 among 47 institutions in the Japan Clinical Oncology Group-Brain Tumor Study Group. We calculated the total cost and cost per month of the initial therapy for newly diagnosed GBM or PCNSL. RESULTS The most used regimen (46.8%) for GBM in patients aged ≤74 years was 'Surgery + radiotherapy concomitant with temozolomide'. This regimen's total cost was 7.50 million JPY (Japanese yen). Adding carmustine wafer implantation (used in 15.0%), TTFields (used in 14.1%), and bevacizumab (BEV) (used in 14.5%) to the standard treatment of GBM increased the cost by 1.24 million JPY for initial treatment, and 1.44 and 0.22 million JPY per month, respectively. Regarding PCNSL, 'Surgery (biopsy) + rituximab, methotrexate, procarbazine, and vincristine (R-MPV) therapy' was the most used regimen (42.5%) for patients of all ages. This regimen incurred 1.07 million JPY per month. The three PCNSL regimens based on R-MPV therapy were in ultra-high-cost medical care (exceeding 1 million JPY per month). CONCLUSIONS Treatment of malignant brain tumors is generally expensive, and cost-ineffective treatments such as BEV are frequently used. We believe that the results of this study can be used to design future economic health studies examining the cost-effectiveness of malignant brain tumors.
Collapse
Affiliation(s)
- Kazuya Motomura
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Keita Sasaki
- JCOG Data Center/Operations Office, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Narushi Sugii
- Department of Neurosurgery, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Japan
| | - Shigeru Yamaguchi
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, Japan
| | - Hirotaka Inoue
- Department of Neurosurgery, Graduate School of Medical Sciences, 1-1-1, Honjo, Chuo-ku, Kumamoto University, Kumamoto, Japan
| | - Akito Oshima
- Department of Neurosurgery, Yokohama City University, 3-9, Fukuura, Kanazawa, Yokohama, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Hospital, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, kita-ku, Okayama, Japan
| | - Mitsuaki Shirahata
- Department of Neurosurgery/Neuro-oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, Japan
| | - Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara, Japan
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, Japan
| | - Shunsuke Tsuzuki
- Department of Neurosurgery, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Tomoo Matsutani
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba, Japan
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Kenichiro Asano
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, Zaifu-cho 5, Hirosaki, Japan
| | - Makoto Ohno
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime university school of medicine, 454, Shitsukawa, Toon, Ehime, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, Japan
| | - Keisuke Miyake
- Department of Neurological Surgery, Kagawa University Faculty of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, Japan
| | - Yuta Mitobe
- Department of Neurosurgery, Yamagata University Hospital, 2-2-2, Iida-Nishi, Yamagata, Japan
| | - Mitsuto Hanihara
- Department of Neurosurgery, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, Japan
| | - Yu Kawanishi
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi, Japan
| | - Shoichi Deguchi
- Department of Neurosurgery, Shizuoka Cancer Center, Shimonagakubo 1007, Nagaizumi, Sunto-gun, Shizuoka, Japan
| | - Masato Saito
- Department of Neurosurgery, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Shijo-cho 840, Kashihara, Nara, Japan
| | - Kenta Ujifuku
- Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, Japan
| | - Hideyuki Arita
- Department of Neurosurgery, Osaka International Cancer Institute, 3-1-69, Otemae, Chuo-ku, Osaka, Japan
| | - Yuichi Sato
- Department of Neurosurgery, Iwate Medical University School of Medicine, 2-1-1 Idai-dori, Yahaba, Shiwa, Iwate, Japan
| | - Shinji Yamashita
- Department of Neurosurgery, Miyazaki University, 5200 Kihara Kiyotake, Miyazaki, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Hiroshima University Hospital, 1-2-3 Minamiku Kasumi, Hiroshima, Japan
| | - Junya Yamaguchi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Yasutomo Momii
- Department of Neurosurgery, Oita University Faculty of Medicine, 1-1 Idaigaoka Hasama-machi Yufu, Oita, Japan
| | - Takahiro Ogawa
- Department of Neurosurgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Atsushi Kambe
- Department of Brain and Neurosciences, Division of Neurosurgery, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, Japan
| | - Shigeo Ohba
- Department of Neurosurgery, Fujita Health University, 1-98Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, Japan
| | - Junya Fukai
- Department of Neurological Surgery, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, Japan
| | - Norihiko Saito
- Department of Neurosurgery, Toho University Ohashi Medical Center, 2-22-36, Ohashi, Meguro, Tokyo, Japan
| | - Masashi Kinoshita
- Department of Neurosurgery, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Koichiro Sumi
- Department of Neurological Surgery, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Ryohei Otani
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, Japan
| | - Takeo Uzuka
- Department of Neurosurgery, Dokkyo Medical University Hospital, 880 Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, Japan
| | - Noriyoshi Takebe
- Department of Neurosurgery, Medical Research Institute Kitano Hospital, PIIF Tazuke-kofukai, 2-4-20 Ohgimachi, Kita-ku, Osaka, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka, Japan
| | - Ryuta Saito
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | | |
Collapse
|
3
|
Sousa F, Lee H, Almeida M, Bazzoni A, Rothen-Rutishauser B, Petri-Fink A. Immunostimulatory nanoparticles delivering cytokines as a novel cancer nanoadjuvant to empower glioblastoma immunotherapy. Drug Deliv Transl Res 2024; 14:2655-2667. [PMID: 38161192 PMCID: PMC11385014 DOI: 10.1007/s13346-023-01509-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Glioblastoma (GBM) stands as a highly aggressive and deadly malignant primary brain tumor with a median survival time of under 15 months upon disease diagnosis. While immunotherapies have shown promising results in solid cancers, brain cancers are still unresponsive to immunotherapy due to immunological dysfunction and the presence of a blood-brain barrier. Interleukin-12 (IL-12) emerges as a potent cytokine in fostering anti-tumor immunity by triggering interferon-gamma production in T and natural killer cells and changing macrophages to a tumoricidal phenotype. However, systemic administration of IL-12 toxicity in clinical trials often leads to significant toxicity, posing a critical hurdle. To overcome this major drawback, we have formulated a novel nanoadjuvant composed of immunostimulatory nanoparticles (ISN) loaded with IL-12 to decrease IL-12 toxicity and enhance the immune response by macrophages and GBM cancer cells. Our in vitro results reveal that ISN substantially increase the production of pro-inflammatory cytokines in GBM cancer cells (e.g. 2.6 × increase in IL-8 expression compared to free IL-12) and macrophages (e.g. 2 × increase in TNF-α expression and 6 × increase in IL-6 expression compared to the free IL-12). These findings suggest a potential modulation of the tumor microenvironment. Additionally, our study demonstrates the effective intracellular delivery of IL-12 by ISN, triggering alterations in the levels of pro-inflammatory cytokines at both transcriptional and protein expression levels. These results highlight the promise of the nanoadjuvant as a prospective platform for resharing the GBM microenvironment and empowering immunotherapy.
Collapse
Affiliation(s)
- Flávia Sousa
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland.
- National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland.
| | - Henry Lee
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Mauro Almeida
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Amelie Bazzoni
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
- Chemistry Department, University of Fribourg, Chemin du Musée 9, 1700, Fribourg, Switzerland
| |
Collapse
|
4
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [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: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
5
|
Li D, Zhang Z, Wang L. Emerging role of tumor microenvironmental nutrients and metabolic molecules in ferroptosis: Mechanisms and clinical implications. Biomed Pharmacother 2024; 179:117406. [PMID: 39255738 DOI: 10.1016/j.biopha.2024.117406] [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: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
In recent years, ferroptosis has gradually attracted increasing attention because of its important role in tumors. Ferroptosis resistance is an important cause of tumor metastasis, recurrence and drug resistance. Exploring the initiating factors and specific mechanisms of ferroptosis has become a key strategy to block tumor progression and improve drug sensitivity. As the external space in direct contact with tumor cells, the tumor microenvironment has a great impact on the biological function of tumor cells. The relationships between abnormal environmental characteristics (hypoxia, lactic acid accumulation, etc.) in the microenvironment and ferroptosis of tumor cells has not been fully characterized. This review focuses on the characteristics of the tumor microenvironment and summarizes the mechanisms of ferroptosis under different environmental factors, aiming to provide new insights for subsequent targeted therapy. Moreover, considering the presence of anticancer drugs in the microenvironment, we further summarize the mechanisms of ferroptosis to provide new strategies for the sensitization of tumor cells to drugs.
Collapse
Affiliation(s)
- Dongyu Li
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
6
|
Zhang Z, Yin W, Wang S, Zheng X, Dong S. MBFusion: Multi-modal balanced fusion and multi-task learning for cancer diagnosis and prognosis. Comput Biol Med 2024; 181:109042. [PMID: 39180856 DOI: 10.1016/j.compbiomed.2024.109042] [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: 01/27/2024] [Revised: 07/11/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Pathological images and molecular omics are important information for predicting diagnosis and prognosis. The two kinds of heterogeneous modal data contain complementary information, and the effective fusion of the two modals can better reveal the complex mechanisms of cancer. However, due to the different representation learning methods, the expression strength of different modals in different tasks varies greatly, so that many multimodal fusions do not achieve the best results. In this paper, MBFusion is proposed, to achieve multiple tasks such as prediction of diagnosis and prognosis through multi-modal balanced fusion. The MBFusion framework uses two kinds of specially constructed graph convolutional network to extract the features of molecular omics data, and uses ResNet to extract the features of pathological image data and retain important deep features by using attention and clustering, which effectively improves both kinds of the features representation, making their expressive ability balanced and comparable. The features of these two modal data are then fused through cross-attention Transformer, and the fused features are used to learn both tasks of cancer subtype classification and survival analysis by using multi-task learning. In this paper, MBFusion and other state of the art methods are compared on two public cancer datasets, and MBFusion shows an improvement of up to 10.1% by three kinds of evaluation metrics. In the ablation experiment, MBFusion explores the contribution of each modal data and each framework module to the performance. Furthermore, the interpretability of MBFusion is explained in detail to show the value of application.
Collapse
Affiliation(s)
- Ziye Zhang
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Wendong Yin
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Shijin Wang
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Xiaorou Zheng
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Shoubin Dong
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China.
| |
Collapse
|
7
|
Zhao T, Yang X, Duan G, Chen J, He K, Chen Y, Luo S. Phosphorylation-regulated phase separation of syndecan-4 and syntenin promotes the biogenesis of exosomes. Cell Prolif 2024; 57:e13645. [PMID: 38601993 PMCID: PMC11471451 DOI: 10.1111/cpr.13645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/08/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
The biogenesis of exosomes that mediate cell-to-cell communication by transporting numerous biomolecules to neighbouring cells is an essential cellular process. The interaction between the transmembrane protein syndecan-4 (SDC4) and cytosolic protein syntenin plays a key role in the biogenesis of exosomes. However, how the relatively weak binding of syntenin to SDC4 efficiently enables syntenin sorting for packaging into exosomes remains unclear. Here, we demonstrate for the first time that SDC4 can undergo liquid-liquid phase separation (LLPS) to form condensates both in vitro and in the cell membrane and that, the SDC4 cytoplasmic domain (SDC4-CD) is a key contributor to this process. The phase separation of SDC4 greatly enhances the recruitment of syntenin to the plasma membrane (PM) despite the weak SDC4-syntenin interaction, facilitating syntenin sorting for inclusion in exosomes. Interestingly, phosphorylation at the only serine (179) in the SDC4-CD (Ser179) disrupts SDC4 LLPS, and inhibited phosphorylation or dephosphorylation restores the SDC4 LLPS to promote its recruitment of syntenin to the PM and syntenin inclusion into exosomes. This research reveals a novel phosphorylation-regulated phase separation property of SDC4 in the PM through which SDC4 efficiently recruits cytosolic syntenin and facilitates the biogenesis of exosomes, providing potential intervention targets for exosome-mediated biomedical events.
Collapse
Affiliation(s)
- Tian Zhao
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Xiaolan Yang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Guangfei Duan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Jialin Chen
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Kefeng He
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Yong‐Xiang Chen
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of ChemistryTsinghua UniversityBeijingChina
| | - Shi‐Zhong Luo
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| |
Collapse
|
8
|
Manescu (Paltanea) V, Antoniac I, Paltanea G, Nemoianu IV, Mohan AG, Antoniac A, Rau JV, Laptoiu SA, Mihai P, Gavrila H, Al-Moushaly AR, Bodog AD. Magnetic Hyperthermia in Glioblastoma Multiforme Treatment. Int J Mol Sci 2024; 25:10065. [PMID: 39337552 PMCID: PMC11432100 DOI: 10.3390/ijms251810065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Glioblastoma multiforme (GBM) represents one of the most critical oncological diseases in neurological practice, being considered highly aggressive with a dismal prognosis. At a worldwide level, new therapeutic methods are continuously being researched. Magnetic hyperthermia (MHT) has been investigated for more than 30 years as a solution used as a single therapy or combined with others for glioma tumor assessment in preclinical and clinical studies. It is based on magnetic nanoparticles (MNPs) that are injected into the tumor, and, under the effect of an external alternating magnetic field, they produce heat with temperatures higher than 42 °C, which determines cancer cell death. It is well known that iron oxide nanoparticles have received FDA approval for anemia treatment and to be used as contrast substances in the medical imagining domain. Today, energetic, efficient MNPs are developed that are especially dedicated to MHT treatments. In this review, the subject's importance will be emphasized by specifying the number of patients with cancer worldwide, presenting the main features of GBM, and detailing the physical theory accompanying the MHT treatment. Then, synthesis routes for thermally efficient MNP manufacturing, strategies adopted in practice for increasing MHT heat performance, and significant in vitro and in vivo studies are presented. This review paper also includes combined cancer therapies, the main reasons for using these approaches with MHT, and important clinical studies on human subjects found in the literature. This review ends by describing the most critical challenges associated with MHT and future perspectives. It is concluded that MHT can be successfully and regularly applied as a treatment for GBM if specific improvements are made.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Iulian Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, RO-050094 Bucharest, Romania
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Iosif Vasile Nemoianu
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
| | - Aurel George Mohan
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania
- Department of Neurosurgery, Clinical Emergency Hospital Oradea, 65 Gheorghe Doja Street, RO-410169 Oradea, Romania
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy;
- Institute of Pharmacy, Department of Analytical, Physical and Colloid Chemistry, I.M. Sechenov First Moscow State Medical University, Trubetskaya St. 8, Build.2, 119048 Moscow, Russia
| | - Stefan Alexandru Laptoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (I.A.); (A.A.)
| | - Petruta Mihai
- Faculty of Entrepreneurship, Business Engineering and Management, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Horia Gavrila
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (I.V.N.)
- Technical Sciences Academy of Romania, 26 Bulevardul Dacia, RO-030167 Bucharest, Romania
| | | | - Alin Danut Bodog
- Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania
| |
Collapse
|
9
|
Wen J, Liu D, Zhu H, Shu K. Microenvironmental regulation of tumor-associated neutrophils in malignant glioma: from mechanism to therapy. J Neuroinflammation 2024; 21:226. [PMID: 39285276 PMCID: PMC11406851 DOI: 10.1186/s12974-024-03222-4] [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: 05/17/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates. Tumor-associated neutrophils (TANs) are essential components of the tumor microenvironment (TME) in glioma and play a crucial role in glioma cell proliferation, invasion and proneural-mesenchymal transition. Besides the interactions between TANs and tumor cells, the multi-dimensional crosstalk between TANs and other components within TME have been reported to participate in glioma progression. More importantly, several therapies targeting TANs have been developed and relevant preclinical and clinical studies have been conducted in cancer therapy. In this review, we introduce the origin of TANs and the functions of TANs in malignant behaviors of glioma, highlighting the microenvironmental regulation of TANs. Moreover, we focus on summarizing the TANs-targeted methods in cancer therapy, aiming to provide insights into the mechanisms and therapeutic opportunities of TANs in the malignant glioma microenvironment.
Collapse
Affiliation(s)
- Jiayi Wen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Dan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
10
|
Inocencio JF, Mitrasinovic S, Asad M, Parney IF, Zang X, Himes BT. Immune checkpoint pathways in glioblastoma: a diverse and evolving landscape. Front Immunol 2024; 15:1424396. [PMID: 39346924 PMCID: PMC11427296 DOI: 10.3389/fimmu.2024.1424396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Immune checkpoint (IC) inhibition in glioblastoma (GBM) has not shown promising results in the last decade compared to other solid tumors. Several factors contributing to the lack of immunotherapy response include the profound immunosuppressive nature of GBM, highly redundant signaling pathways underlying immune checkpoints, and the negative immunogenic impact of current standard of care on the tumor microenvironment. In this review, we will discuss various ICs in the context of GBM, their interplay with the tumor immune microenvironment, relevant pre-clinical and clinical studies, and the impact of current treatment modalities on GBM IC blockade therapy. Understanding the molecular mechanisms that drive ICs, and how they contribute to an immunosuppressive tumor microenvironment is critical in advancing IC inhibition therapy in GBM. Furthermore, revisiting current treatment modalities and their impact on the immune landscape is instrumental in designing future combinatorial therapies that may overcome treatment resistance.
Collapse
Affiliation(s)
- Julio F Inocencio
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Stefan Mitrasinovic
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mohammad Asad
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Benjamin T Himes
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
11
|
Feng F, Shen J, Qi Q, Zhang Y, Ni S. Empowering brain tumor management: chimeric antigen receptor macrophage therapy. Theranostics 2024; 14:5725-5742. [PMID: 39310093 PMCID: PMC11413779 DOI: 10.7150/thno.98290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Brain tumors pose formidable challenges in oncology due to the intricate biology and the scarcity of effective treatment modalities. The emergence of immunotherapy has opened new avenues for innovative therapeutic strategies. Chimeric antigen receptor, originally investigated in T cell-based therapy, has now expanded to encompass macrophages, presenting a compelling avenue for augmenting anti-tumor immune surveillance. This emerging frontier holds promise for advancing the repertoire of therapeutic options against brain tumors, offering potential breakthroughs in combating the formidable malignancies of the central nervous system. Tumor-associated macrophages constitute a substantial portion, ranging from 30% to 50%, of the tumor tissue and exhibit tumor-promoting phenotypes within the immune-compromised microenvironment. Constructing CAR-macrophages can effectively repolarize M2-type macrophages towards an M1-type phenotype, thereby eliciting potent anti-tumor effects. CAR-macrophages can recruit T cells to the brain tumor site, thereby orchestrating a remodeling of the immune niche to effectively inhibit tumor growth. In this review, we explore the potential limitations as well as strategies for optimizing CAR-M therapy, offering insights into the future direction of this innovative therapeutic approach.
Collapse
Affiliation(s)
| | | | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| |
Collapse
|
12
|
Zhang S, Zhang Y, Sun X. Targeting GPR133 via miR-106a-5p inhibits the proliferation, invasion, migration and epithelial-mesenchymal transition (EMT) of glioma cells. Int J Neurosci 2024; 134:991-1002. [PMID: 37036013 DOI: 10.1080/00207454.2023.2201873] [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: 01/21/2023] [Revised: 03/07/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Background: Glioma is the most common malignant brain tumor. GPR133 is a key factor in the progression of glioma. However, the role of GPR133 in glioma invasion and EMT and the microRNAs (miRNAs) associated with this pathway are still poorly understood.Objective: This study aims to elucidate the biological function of miR-106a-5p and GPR133 in glioma as well as the molecular mechanism of their interaction.Methods: The mRNA expression of miR-106a-5p and GPR133 in glioma specimens and cells was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). The protein level of GPR133 and the levels of invasion- and EMT-related proteins were measured by western blotting. miR-106a-5p and GPR133 function in glioma cells was determined through cell counting kit-8 (CCK-8), transwell, wound healing, colony formation assays in vitro and xenograft assays in vivo. To determine the targeting relationship between miR-106a-5p and GPR133, a dual-luciferase reporter assay was conducted.Results: A marked reduction in miR-106a-5p expression was observed in glioma cells and specimens. Patients with high expression of miR-106a-5p had a good prognosis, while patients with high expression of GPR133 had a shorter OS. Additionally, overexpression of miR-106a-5p or downregulation of GPR133 inhibited the progression of glioma cells. Furthermore, miR-106a-5p negatively regulated GPR133 expression by binding to its 3'-UTR, and restrained the invasion, migration, proliferation and EMT of glioma cells by targeting GPR133.Conclusions: miR-106a-5p is a tumor suppressor that negatively regulates GPR133. The miR-106a-5p/GPR133 axis could potentially serve as a therapeutic target for glioma.
Collapse
Affiliation(s)
- Shiyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PRChina
- Department of Neurosurgery, Suining Central Hospital, Suining, PR China
| | - Yuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PRChina
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PRChina
| |
Collapse
|
13
|
Wang Y, Zhang Y, Chen Y, Wang S, Liu W, Liu Z, Hu M. [ 18F]AlF-NOTA-PCP2: a novel PET/CT tracer for enhanced PD-L1 heterogeneity imaging and comparative analysis with [ 18F]AlF-NOTA-WL12 in glioblastoma xenografts. Eur J Nucl Med Mol Imaging 2024; 51:3161-3175. [PMID: 38713298 DOI: 10.1007/s00259-024-06743-5] [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: 01/04/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE The unsatisfactory efficacy of PD-L1 antibodies in glioblastoma (GBM) is largely due to the temporal and spatial heterogeneity of PD-L1 expression. Molecular imaging can enhance understanding of the tumor immune microenvironment and guide immunotherapy. However, highly sensitive imaging agents capable of effectively visualizing PD-L1 heterogeneity are limited. This study introduces a novel PET tracer, offering improved imaging of PD-L1 heterogeneity in GBM xenografts, with a comparative analysis to [18F]AlF-NOTA-WL12. METHODS [18F]AlF-NOTA-PCP2 was synthesized with high purity and its affinity for PD-L1 was characterized using surface plasmon resonance (SPR) and cell binding assays. Its specificity for PD-L1 was evaluated both in vitro using various cell lines and in vivo with GBM xenograft models in NOD/SCID mice. PET/CT imaging was conducted to evaluate the tracer's biodistribution, pharmacokinetics, and ability to quantify tumoral spatial heterogeneity of PD-L1 expression. A focused comparative analysis between [18F]AlF-NOTA-PCP2 and [18F]AlF-NOTA-WL12 was conducted, examining binding affinity, biodistribution, pharmacokinetics, and imaging effectiveness in GBM xenografts. Additionally, human radiation dosimetry estimates compared the safety profiles of both tracers. RESULTS [18F]AlF-NOTA-PCP2 demonstrated high radiochemical purity (> 95%) and a strong affinity for PD-L1, comparable to [18F]AlF-NOTA-WL12. In vitro and in vivo studies confirmed its specificity for PD-L1, with increased uptake in PD-L1 expressing cells and tumors. Toxicological profiles indicated no significant abnormalities in serum biochemical indicators or major organ tissues. MicroPET/CT imaging showed [18F]AlF-NOTA-PCP2's effectiveness in visualizing PD-L1 expression levels and spatial heterogeneity in GBM xenografts. Comparative studies revealed [18F]AlF-NOTA-PCP2's improved pharmacokinetic properties, including higher tumor-to-blood ratios and lower nonspecific liver uptake, as well as reduced radiation exposure compared to [18F]AlF-NOTA-WL12. CONCLUSION [18F]AlF-NOTA-PCP2 distinguishes itself as an exceptionally sensitive PET/CT tracer, adept at non-invasively and accurately quantifying PD-L1 expression and its spatial heterogeneity in tumors, especially in GBM. Its favorable pharmacokinetic properties, safety profile, and high affinity for PD-L1 highlight its potential for enhancing the precision of cancer immunotherapy and guiding individualized treatment strategies. While promising, its clinical translation, especially in brain imaging, necessitates further validation in clinical trials.
Collapse
Affiliation(s)
- Yong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Yunhao Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shijie Wang
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wei Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Zhiguo Liu
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
14
|
Ghazi B, Harmak Z, Rghioui M, Kone AS, El Ghanmi A, Badou A. Decoding the secret of extracellular vesicles in the immune tumor microenvironment of the glioblastoma: on the border of kingdoms. Front Immunol 2024; 15:1423232. [PMID: 39267734 PMCID: PMC11390556 DOI: 10.3389/fimmu.2024.1423232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
Over the last decades, extracellular vesicles (EVs) have become increasingly popular for their roles in various pathologies, including cancer and neurological and immunological disorders. EVs have been considered for a long time as a means for normal cells to get rid of molecules it no longer needs. It is now well established that EVs play their biological roles also following uptake or by the interaction of EV surface proteins with cellular receptors and membranes. In this review, we summarize the current status of EV production and secretion in glioblastoma, the most aggressive type of glioma associated with high mortality. The main purpose is to shed light on the EVs as a universal mediator of interkingdom and intrakingdom communication in the context of tumor microenvironment heterogeneity. We focus on the immunomodulatory EV functions in glioblastoma-immune cross-talk to enhance immune escape and reprogram tumor-infiltrating immune cells. We critically examine the evidence that GBM-, immune cell-, and microbiome-derived EVs impact local tumor microenvironment and host immune responses, and can enter the circulatory system to disseminate and drive premetastatic niche formation in distant organs. Taking into account the current state of the art in intratumoral microbiome studies, we discuss the emerging role of bacterial EV in glioblastoma and its response to current and future therapies including immunotherapies.
Collapse
Affiliation(s)
- Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Zakia Harmak
- Immuno-genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mounir Rghioui
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Abdou-Samad Kone
- Immuno-genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Abdallah Badou
- Immuno-genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
| |
Collapse
|
15
|
Fu X, Ren C, Dai K, Ren M, Yan C. Epithelial-Mesenchymal Transition Related Score Functions as a Predictive Tool for Immunotherapy and Candidate Drugs in Glioma. J Chem Inf Model 2024; 64:6648-6661. [PMID: 39116318 DOI: 10.1021/acs.jcim.4c00620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Gliomas are aggressive CNS tumors where the epithelial-mesenchymal transition (EMT) is crucial for prognosis. We developed an EMT-based score predicting overall survival (OS) and conducted pathway analyses, revealing functions such as cell proliferation and immune response in glioma progression. The EMT score, correlated with immune functions and cell infiltration, shows potential as an immune response indicator. We identified two promising compounds, BIX02189 and QL-XI-92, as potential glioma treatments based on candidate gene analysis.
Collapse
Affiliation(s)
- Xiaojun Fu
- Sanbo Brain Hospital, Capital Medical University No. 50, Yikesong Road, Xiangshan, Haidian District, Beijing 100070, China
- Laboratory for Clinical Medicine, Capital Medical University, No. 10, You'anmenwai, Fengtai District, Beijing 100070, China
| | - Changyuan Ren
- Sanbo Brain Hospital, Capital Medical University No. 50, Yikesong Road, Xiangshan, Haidian District, Beijing 100070, China
- Beijing Neurosurgical Institute, Capital Medical University, No. 119 South Fourth Ring Road West, Fengtai District, Beijing 100070, China
| | - Kaining Dai
- Sanbo Brain Hospital, Capital Medical University No. 50, Yikesong Road, Xiangshan, Haidian District, Beijing 100070, China
- Laboratory for Clinical Medicine, Capital Medical University, No. 10, You'anmenwai, Fengtai District, Beijing 100070, China
| | - Ming Ren
- Sanbo Brain Hospital, Capital Medical University No. 50, Yikesong Road, Xiangshan, Haidian District, Beijing 100070, China
| | - Changxiang Yan
- Sanbo Brain Hospital, Capital Medical University No. 50, Yikesong Road, Xiangshan, Haidian District, Beijing 100070, China
| |
Collapse
|
16
|
Vasudevan MT, Rangaraj K, Ramesh R, Muthusami S, Govindasamy C, Khan MI, Arulselvan P, Muruganantham B. Inhibitory effects of Gracilaria edulis and Gracilaria salicornia against the MGMT and VEGFA biomarkers involved in the onset and advancement of glioblastoma using in silico and in vitro approaches. Biotechnol Appl Biochem 2024. [PMID: 39168850 DOI: 10.1002/bab.2657] [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: 06/04/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024]
Abstract
Glioblastoma (GBM), an aggressive primary brain tumor originating from glial cells, poses significant treatment challenges due to its rapid growth and invasiveness. The exact mechanisms of GBM's brain damage remain unclear. This study examines primary molecular markers commonly assessed in GBM patients, including brain-derived neurotrophic factor (BDNF), platelet-derived growth factor receptor A (PDGFRA), O6-methylguanine DNA methyltransferase (MGMT), epidermal growth factor receptor (EGFR), and vascular endothelial growth factor A (VEGFA) using computational approaches. The study revealed significant differences (p ≤ 0.05) in PDGFRA, EGFR, and VEGFA expression rates, which are particularly interesting. Additionally, MGMT and VEGFA showed higher hazard ratios. Expression levels of MGMT and VEGFA were visualized in immune and malignant cells using single-cell RNA datasets GSE103224 and GSE148842. From a total of 48 compounds in Gracilaria edulis and 86 in Gracilaria salicornia, we identified 15 compounds capable of crossing the blood-brain barrier. Notably, 2-tridecanone (binding affinity [BA] = -4.2 kcal/mol; root mean square deviation [RMSD] = 1.479 Å) and decanoic acid, ethyl ester (BA = -4.2 kcal/mol; RMSD = 1.702 Å) from G. edulis interacted with MGMT via hydrogen bonds. The compound alpha-terpineol interacted with MGMT (BA = -5.7 kcal/mol; RMSD = 0.501 Å) and VEGFA (BA = -4.7 kcal/mol; RMSD = 2.483 Å). Ethanolic and methanolic extracts from G. edulis and G. salicornia demonstrated mild anti-cell proliferation properties in the GBM LN-229 cell line, suggesting potential therapeutic benefits. This study highlights the significance of molecular markers and natural compounds in understanding and potentially treating GBM.
Collapse
Affiliation(s)
- Miji Thandaserry Vasudevan
- Department of Biochemistry, Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Kaviyaprabha Rangaraj
- Department of Biochemistry, Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Ragupathi Ramesh
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Ibrar Khan
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Palanisamy Arulselvan
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Bharathi Muruganantham
- Department of Biochemistry, Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| |
Collapse
|
17
|
Zheng G, Zeng G, Wei D. The role of NPC2 gene in glioma was investigated based on bioinformatics analysis. Sci Rep 2024; 14:19134. [PMID: 39160329 PMCID: PMC11333723 DOI: 10.1038/s41598-024-70221-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: 12/11/2023] [Accepted: 08/14/2024] [Indexed: 08/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most malignant primary brain tumors in adults. The NPC2 gene (Niemann-Pick type C intracellular cholesterol transporter 2) is a protein-coding gene with a lipid recognition domain. The NPC2 gene was found to be significantly increased in gliomas (LGG and GBM), and it is now thought to be a risk factor. COX analysis demonstrated that NPC2 was a significant risk factor for glioma. Functional enrichment analysis of genes that were differentially expressed between high and low expression groups revealed that genes were primarily enriched in the regulation of trans-synaptic signaling, Retrograde endocannabinoid signaling and other pathways. According to the findings of the immunoinfiltration investigation, the NPC2 gene and macrophage, DC, etc. have a strong positive association. In addition, patients with high NPC2 expression had higher levels of immune cell expression. Medication sensitivity research revealed that NPC2's differential expression had some bearing on patients' medication sensitivity. There was a strong correlation between the prognosis of glioma patients and the gene sets NUDT19 and NUME. In brief, the NPC2 gene was identified to be a possible biomarker of glioma, and preliminary analysis was done on the role of the NPC2 gene in immunological microenvironment of glioma.
Collapse
Affiliation(s)
- Guangwei Zheng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Guangming Zeng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - De Wei
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
18
|
Zhang S, Ji X, Liu Z, Xie Z, Wang Y, Wang H, Ni D. Bimetallic Nanoplatforms for Prostate Cancer Treatment by Interfering Cellular Communication. J Am Chem Soc 2024; 146:22530-22540. [PMID: 39082227 DOI: 10.1021/jacs.4c06267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Cellular communication mediated by messenger molecules plays an important role in the progression of cancer. Herein, pH-sensitive zeolitic imidazolate framework-8 (ZIF-8) loaded with PtCl2(OH)2(NH3)2 [i.e., Pt(IV)] bimetallic nanoplatforms were developed for prostate cancer therapy by interfering inositol-1, 4, 5-trisphosphate (IP3)-mediated cellular communication. As an important messenger in cells, the function of IP3 was found to be efficiently interfered with by the Pt(IV)-binding inositol unit. This finding effect of Pt(IV) is totally different from its traditional function as a prodrug of cis-platinum for chemotherapy. The decreased IP3 signal further downregulated the cytoplasmic Ca2+ concentration and downstream signal transduction to inhibit proliferation and invasion of tumor cells. Meanwhile, Zn2+ released from ZIF-8 under an acidic tumor microenvironment decreased adenosine triphosphate biosynthesis, which could further limit the cellular communication. Such a proposed strategy of interfering cellular communication has demonstrated its feasibility in this study, which may provide new perspectives for cancer therapy.
Collapse
Affiliation(s)
- Siyang Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiuru Ji
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zeyang Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiwen Xie
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yue Wang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
19
|
Hwang YK, Lee DH, Lee EC, Oh JS. Importance of Autophagy Regulation in Glioblastoma with Temozolomide Resistance. Cells 2024; 13:1332. [PMID: 39195222 DOI: 10.3390/cells13161332] [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: 06/20/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive and common malignant and CNS tumor, accounting for 47.7% of total cases. Glioblastoma has an incidence rate of 3.21 cases per 100,000 people. The regulation of autophagy, a conserved cellular process involved in the degradation and recycling of cellular components, has been found to play an important role in GBM pathogenesis and response to therapy. Autophagy plays a dual role in promoting tumor survival and apoptosis, and here we discuss the complex interplay between autophagy and GBM. We summarize the mechanisms underlying autophagy dysregulation in GBM, including PI3K/AKT/mTOR signaling, which is most active in brain tumors, and EGFR and mutant EGFRvIII. We also review potential therapeutic strategies that target autophagy for the treatment of GBM, such as autophagy inhibitors used in combination with the standard of care, TMZ. We discuss our current understanding of how autophagy is involved in TMZ resistance and its role in glioblastoma development and survival.
Collapse
Affiliation(s)
- Young Keun Hwang
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong-Hun Lee
- Industry-Academic Cooperation Foundation, The Catholic University of Korea, 222, Banpo-daro, Seocho-gu, Seoul 06591, Republic of Korea
| | - Eun Chae Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae Sang Oh
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
20
|
Mendanha D, Casanova MR, Gimondi S, Ferreira H, Neves NM. Microfluidic-Derived Docosahexaenoic Acid Liposomes for Targeting Glioblastoma and Its Inflammatory Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40543-40554. [PMID: 39042828 PMCID: PMC11310905 DOI: 10.1021/acsami.4c01368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, characterized by limited treatment options and a poor prognosis. Its aggressiveness is attributed not only to the uncontrolled proliferation and invasion of tumor cells but also to the complex interplay between these cells and the surrounding microenvironment. Within the tumor microenvironment, an intricate network of immune cells, stromal cells, and various signaling molecules creates a pro-inflammatory milieu that supports tumor growth and progression. Docosahexaenoic acid (DHA), an essential ω3 polyunsaturated fatty acid for brain function, is associated with anti-inflammatory and anticarcinogenic properties. Therefore, in this work, DHA liposomes were synthesized using a microfluidic platform to target and reduce the inflammatory environment of GBM. The liposomes were rapidly taken up by macrophages in a time-dependent manner without causing cytotoxicity. Moreover, DHA liposomes successfully downregulated the expression of inflammatory-associated genes (IL-6; IL-1β; TNFα; NF-κB, and STAT-1) and the secretion of key cytokines (IL-6 and TNFα) in stimulated macrophages and GBM cells. Conversely, no significant differences were observed in the expression of IL-10, an anti-inflammatory gene expressed in alternatively activated macrophages. Additionally, DHA liposomes were found to be more efficient in regulating the inflammatory profile of these cells compared with a free formulation of DHA. The nanomedicine platform established in this work opens new opportunities for developing liposomes incorporating DHA to target GBM and its inflammatory milieu.
Collapse
Affiliation(s)
- Daniel Mendanha
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Marta R. Casanova
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Sara Gimondi
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Helena Ferreira
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| |
Collapse
|
21
|
Mu Y, Zhang Z, Zhou H, Ma L, Wang DA. Applications of nanotechnology in remodeling the tumour microenvironment for glioblastoma treatment. Biomater Sci 2024; 12:4045-4064. [PMID: 38993162 DOI: 10.1039/d4bm00665h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
With the increasing research and deepening understanding of the glioblastoma (GBM) tumour microenvironment (TME), novel and more effective therapeutic strategies have been proposed. The GBM TME involves intricate interactions between tumour and non-tumour cells, promoting tumour progression. Key therapeutic goals for GBM treatment include improving the immunosuppressive microenvironment, enhancing the cytotoxicity of immune cells against tumours, and inhibiting tumour growth and proliferation. Consequently, remodeling the GBM TME using nanotechnology has emerged as a promising approach. Nanoparticle-based drug delivery enables targeted delivery, thereby improving treatment specificity, facilitating combination therapies, and optimizing drug metabolism. This review provides an overview of the GBM TME and discusses the methods of remodeling the GBM TME using nanotechnology. Specifically, it explores the application of nanotechnology in ameliorating immune cell immunosuppression, inducing immunogenic cell death, stimulating, and recruiting immune cells, regulating tumour metabolism, and modulating the crosstalk between tumours and other cells.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
22
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2024:S2090-1232(24)00315-1. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [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: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
23
|
Li J, Zhang Y, Liang C, Yan X, Hui X, Liu Q. Advancing precision medicine in gliomas through single-cell sequencing: unveiling the complex tumor microenvironment. Front Cell Dev Biol 2024; 12:1396836. [PMID: 39156969 PMCID: PMC11327033 DOI: 10.3389/fcell.2024.1396836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma (GBM) displays an infiltrative growth characteristic that recruits neighboring normal cells to facilitate tumor growth, maintenance, and invasion into the brain. While the blood-brain barrier serves as a critical natural defense mechanism for the central nervous system, GBM disrupts this barrier, resulting in the infiltration of macrophages from the peripheral bone marrow and the activation of resident microglia. Recent advancements in single-cell transcriptomics and spatial transcriptomics have refined the categorization of cells within the tumor microenvironment for precise identification. The intricate interactions and influences on cell growth within the tumor microenvironment under multi-omics conditions are succinctly outlined. The factors and mechanisms involving microglia, macrophages, endothelial cells, and T cells that impact the growth of GBM are individually examined. The collaborative mechanisms of tumor cell-immune cell interactions within the tumor microenvironment synergistically promote the growth, infiltration, and metastasis of gliomas, while also influencing the immune status and therapeutic response of the tumor microenvironment. As immunotherapy continues to progress, targeting the cells within the inter-tumor microenvironment emerges as a promising novel therapeutic approach for GBM. By comprehensively understanding and intervening in the intricate cellular interactions within the tumor microenvironment, novel therapeutic modalities may be developed to enhance treatment outcomes for patients with GBM.
Collapse
Affiliation(s)
- Jinwei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Yang Zhang
- Graduate School of Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Cong Liang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Xianlei Yan
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quan Liu
- Department of Neurosurgery, Liuzhou Workers Hospital, Liuzhou, Guangxi, China
| |
Collapse
|
24
|
Zhao Y, Pan J, Han B, Hou W, Li B, Wang J, Wang G, He Y, Ma M, Zhou J, Yu C, Sun SK. Ultrahigh-Resolution Visualization of Vascular Heterogeneity in Brain Tumors via Magnetic Nanoparticles-Enhanced Susceptibility-Weighted Imaging. ACS NANO 2024. [PMID: 39094075 DOI: 10.1021/acsnano.4c02611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The precise assessment of vascular heterogeneity in brain tumors is vital for diagnosing, grading, predicting progression, and guiding treatment decisions. However, currently, there is a significant shortage of high-resolution imaging approaches. Herein, we propose a contrast-enhanced susceptibility-weighted imaging (CE-SWI) utilizing the minimalist dextran-modified Fe3O4 nanoparticles (Dextran@Fe3O4 NPs) for ultrahigh-resolution mapping of vasculature in brain tumors. The Dextran@Fe3O4 NPs are prepared via a facile coprecipitation method under room temperature, and exhibit small hydrodynamic size (28 nm), good solubility, excellent biocompatibility, and high transverse relaxivity (r2*, 159.7 mM-1 s-1) under 9.4 T magnetic field. The Dextran@Fe3O4 NPs-enhanced SWI can increase the contrast-to-noise ratio (CNR) of cerebral vessels to 2.5 times that before injection and achieves ultrahigh-spatial-resolution visualization of microvessels as small as 0.1 mm in diameter. This advanced imaging capability not only allows for the detailed mapping of both enlarged peritumoral drainage vessels and the intratumoral microvessels, but also facilitates the sensitive imaging detection of vascular permeability deterioration in a C6 cells-bearing rat glioblastoma model. Our proposed Dextran@Fe3O4 NPs-enhanced SWI provides a powerful imaging technique with great clinical translation potential for the precise theranostics of brain tumors.
Collapse
Affiliation(s)
- Yujie Zhao
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bing Han
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenjing Hou
- Department of Diagnostic and Therapeutic Ultrasonography, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Bingjie Li
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jiaojiao Wang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Guohe Wang
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300204, China
| | - Yujing He
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Min Ma
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junzi Zhou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin 300204, China
| |
Collapse
|
25
|
Ma Y, Yi J, Ruan J, Ma J, Yang Q, Zhang K, Zhang M, Zeng G, Jin L, Huang X, Li J, Yang H, Wu W, Sun D. Engineered Cell Membrane-Coated Nanoparticles: New Strategies in Glioma Targeted Therapy and Immune Modulation. Adv Healthc Mater 2024; 13:e2400514. [PMID: 38652681 DOI: 10.1002/adhm.202400514] [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: 02/08/2024] [Revised: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Gliomas, the most prevalent primary brain tumors, pose considerable challenges due to their heterogeneity, intricate tumor microenvironment (TME), and blood-brain barrier (BBB), which restrict the effectiveness of traditional treatments like surgery and chemotherapy. This review provides an overview of engineered cell membrane technologies in glioma therapy, with a specific emphasis on targeted drug delivery and modulation of the immune microenvironment. This study investigates the progress in engineered cell membranes, encompassing physical, chemical, and genetic alterations, to improve drug delivery across the BBB and effectively target gliomas. The examination focuses on the interaction of engineered cell membrane-coated nanoparticles (ECM-NPs) with the TME in gliomas, emphasizing their potential to modulate glioma cell behavior and TME to enhance therapeutic efficacy. The review further explores the involvement of ECM-NPs in immunomodulation techniques, highlighting their impact on immune reactions. While facing obstacles related to membrane stability and manufacturing scalability, the review outlines forthcoming research directions focused on enhancing membrane performance. This review underscores the promise of ECM-NPs in surpassing conventional therapeutic constraints, proposing novel approaches for efficacious glioma treatment.
Collapse
Affiliation(s)
- Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Maolan Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Guoming Zeng
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Haifeng Yang
- JinFeng Laboratory, Chongqing, 401329, China
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
26
|
Yang Y, Jin X, Xie Y, Ning C, Ai Y, Wei H, Xu X, Ge X, Yi T, Huang Q, Yang X, Jiang T, Wang X, Piao Y, Jin X. The CEBPB + glioblastoma subcluster specifically drives the formation of M2 tumor-associated macrophages to promote malignancy growth. Theranostics 2024; 14:4107-4126. [PMID: 38994023 PMCID: PMC11234274 DOI: 10.7150/thno.93473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
Rationale: The heterogeneity of tumor cells within the glioblastoma (GBM) microenvironment presents a complex challenge in curbing GBM progression. Understanding the specific mechanisms of interaction between different GBM cell subclusters and non-tumor cells is crucial. Methods: In this study, we utilized a comprehensive approach integrating glioma single-cell and spatial transcriptomics. This allowed us to examine the molecular interactions and spatial localization within GBM, focusing on a specific tumor cell subcluster, GBM subcluster 6, and M2-type tumor-associated macrophages (M2 TAMs). Results: Our analysis revealed a significant correlation between a specific tumor cell subcluster, GBM cluster 6, and M2-type TAMs. Further in vitro and in vivo experiments demonstrated the specific regulatory role of the CEBPB transcriptional network in GBM subcluster 6, which governs its tumorigenicity, recruitment of M2 TAMs, and polarization. This regulation involves molecules such as MCP1 for macrophage recruitment and the SPP1-Integrin αvβ1-Akt signaling pathway for M2 polarization. Conclusion: Our findings not only deepen our understanding of the formation of M2 TAMs, particularly highlighting the differential roles played by heterogeneous cells within GBM in this process, but also provided new insights for effectively controlling the malignant progression of GBM.
Collapse
Affiliation(s)
- Yongchang Yang
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Medical University, Tianjin 300060, China
| | - Xingyu Jin
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Medical University, Tianjin 300060, China
| | - Yang Xie
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Medical University, Tianjin 300060, China
| | - Chunlan Ning
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Medical University, Tianjin 300060, China
| | - Yiding Ai
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin Medical University, Tianjin 300060, China
| | - Haotian Wei
- Tianjin Medical University, Tianjin 300060, China
| | - Xing Xu
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xianglian Ge
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Tailong Yi
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xuejun Yang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People's Republic of China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaoguang Wang
- Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin China
| | - Yingzhe Piao
- Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin China
| | - Xun Jin
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
27
|
Lv X, Wang B, Liu K, Li MJ, Yi X, Wu X. Decoding heterogeneous and coordinated tissue architecture in glioblastoma using spatial transcriptomics. iScience 2024; 27:110064. [PMID: 38947514 PMCID: PMC11214485 DOI: 10.1016/j.isci.2024.110064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal brain tumors, characterized by profound heterogeneity. While single-cell transcriptomic studies have revealed extensive intra-tumor heterogeneity, shed light on intra-tumor diversity, spatial intricacies remain largely unexplored. Leveraging clinical GBM specimens, this study employs spatial transcriptomics technology to delve into gene expression heterogeneity. Our investigation unveils a significant enrichment of tissue stem cell signature in regions bordering necrosis and the peritumoral area, positively correlated with the mesenchymal subtype signature. Moreover, upregulated genes in these regions are linked with extracellular matrix (ECM)-receptor interaction, proteoglycans, as well as vascular endothelial growth factor (VEGF) and angiopoietin-Tie (ANGPT) signaling pathways. In contrast, signatures related to glycogen metabolism and oxidative phosphorylation show no relevance to pathological zoning, whereas creatine metabolism signature is notably exclusive to vascular-enriched areas. These spatial profiles not only offer valuable references but also pave the way for future in-depth functional and mechanistic investigations into GBM progression.
Collapse
Affiliation(s)
- Xuejiao Lv
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Bo Wang
- Department of Neurosurgery, Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, No. 6 Jizhao Road, Tianjin 300350, China
| | - Kunlun Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xianfu Yi
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xudong Wu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
28
|
Weller J, Potthoff AL, Zeyen T, Schaub C, Duffy C, Schneider M, Herrlinger U. Current status of precision oncology in adult glioblastoma. Mol Oncol 2024. [PMID: 38899374 DOI: 10.1002/1878-0261.13678] [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: 11/16/2023] [Revised: 04/05/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The concept of precision oncology, the application of targeted drugs based on comprehensive molecular profiling, has revolutionized treatment strategies in oncology. This review summarizes the current status of precision oncology in glioblastoma (GBM), the most common and aggressive primary brain tumor in adults with a median survival below 2 years. Targeted treatments without prior target verification have consistently failed. Patients with BRAF V600E-mutated GBM benefit from BRAF/MEK-inhibition, whereas targeting EGFR alterations was unsuccessful due to poor tumor penetration, tumor cell heterogeneity, and pathway redundancies. Systematic screening for actionable molecular alterations resulted in low rates (< 10%) of targeted treatments. Efficacy was observed in one-third and currently appears to be limited to BRAF-, VEGFR-, and mTOR-directed treatments. Advancing precision oncology for GBM requires consideration of pathways instead of single alterations, new trial concepts enabling rapid and adaptive drug evaluation, a focus on drugs with sufficient bioavailability in the CNS, and the extension of target discovery and validation to the tumor microenvironment, tumor cell networks, and their interaction with immune cells and neurons.
Collapse
Affiliation(s)
- Johannes Weller
- Department of Neurooncology, Center for Neurology, University Hospital Bonn, Germany
| | | | - Thomas Zeyen
- Department of Neurooncology, Center for Neurology, University Hospital Bonn, Germany
| | - Christina Schaub
- Department of Neurooncology, Center for Neurology, University Hospital Bonn, Germany
| | - Cathrina Duffy
- Department of Neurooncology, Center for Neurology, University Hospital Bonn, Germany
| | | | - Ulrich Herrlinger
- Department of Neurooncology, Center for Neurology, University Hospital Bonn, Germany
| |
Collapse
|
29
|
Janneh AH. Sphingolipid Signaling and Complement Activation in Glioblastoma: A Promising Avenue for Therapeutic Intervention. BIOCHEM 2024; 4:126-143. [PMID: 38894892 PMCID: PMC11185840 DOI: 10.3390/biochem4020007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Glioblastoma is the most common and aggressive type of malignant brain tumor with a poor prognosis due to the lack of effective treatment options. Therefore, new treatment options are required. Sphingolipids are essential components of the cell membrane, while complement components are integral to innate immunity, and both play a critical role in regulating glioblastoma survival signaling. This review focuses on recent studies investigating the functional roles of sphingolipid metabolism and complement activation signaling in glioblastoma. It also discusses how targeting these two systems together may emerge as a novel therapeutic approach.
Collapse
Affiliation(s)
- Alhaji H Janneh
- Hollings Cancer Center, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
30
|
Wu Q, Berglund AE, Macaulay RJ, Etame AB. The Role of Mesenchymal Reprogramming in Malignant Clonal Evolution and Intra-Tumoral Heterogeneity in Glioblastoma. Cells 2024; 13:942. [PMID: 38891074 PMCID: PMC11171993 DOI: 10.3390/cells13110942] [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: 04/30/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J. Macaulay
- Departments of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
31
|
Uthanaphun T, Manochantr S, Tantrawatpan C, Tantikanlayaporn D, Kheolamai P. PL-hMSC and CH-hMSC derived soluble factors inhibit proliferation but improve hGBM cell migration by activating TGF-β and inhibiting Wnt signaling. Biosci Rep 2024; 44:BSR20231964. [PMID: 38687607 PMCID: PMC11130542 DOI: 10.1042/bsr20231964] [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: 11/17/2023] [Revised: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive brain tumors. GBM resists most chemotherapeutic agents, resulting in a high mortality rate in patients. Human mesenchymal stem cells (hMSCs), which are parts of the cancer stroma, have been shown to be involved in the development and progression of GBM. However, different sources of hMSCs might affect GBM cells differently. In the present study, we established hMSCs from placenta (PL-hMSC) and chorion (CH-hMSC) to study the effects of their released soluble factors on the proliferation, migration, invasion, gene expression, and survival of human GBM cells, U251. We found that the soluble factors derived from CH-hMSCs and PL-hMSCs suppressed the proliferation of U251 cells in a dose-dependent manner. In contrast, soluble factors derived from both hMSC sources increased U251 migration without affecting their invasive property. The soluble factors derived from these hMSCs decreased the expression levels of CyclinD1, E2Fs and MYC genes that promote GBM cell proliferation but increased the expression level of TWIST gene, which promotes EMT and GBM cell migration. The functional study suggests that both hMSCs might exert their effects, at least in part, by activating TGF-β and suppressing Wnt/β-catenin signaling in U251 cells. Our study provides a better understanding of the interaction between GBM cells and gestational tissue-derived hMSCs. This knowledge might be used to develop safer and more effective stem cell therapy that improves the survival and quality of life of patients with GBM by manipulating the interaction between hMSCs and GBM cells.
Collapse
Affiliation(s)
- Tanawat Uthanaphun
- Master of Science Program in Stem Cell and Molecular Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Sirikul Manochantr
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Chairat Tantrawatpan
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| |
Collapse
|
32
|
Köktürk S, Doğan S, Yılmaz CE, Cetinkol Y, Mutlu O. Expression of brain-derived neurotrophic factor and formation of migrasome increases in the glioma cells induced by the adipokinetic hormone. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231337. [PMID: 38775506 PMCID: PMC11110965 DOI: 10.1590/1806-9282.20231337] [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: 01/13/2024] [Accepted: 01/28/2024] [Indexed: 05/24/2024]
Abstract
OBJECTIVE It has been previously shown that brain-derived neurotrophic factor is linked with various types of cancer. Brain-derived neurotrophic factor is found to be highly expressed in multiple human cancers and associated with tumor growth, invasion, and metastasis. Adipokinetic hormones are functionally related to the vertebrate glucagon, as they have similar functionalities that manage the nutrient-dependent secretion of these two hormones. Migrasomes are new organelles that contain numerous small vesicles, which aid in transmitting signals between the migrating cells. Therefore, the aim of this study was to investigate the effects of Anax imperator adipokinetic hormone on brain-derived neurotrophic factor expression and ultrastructure of cells in the C6 glioma cell line. METHODS The rat C6 glioma cells were treated with concentrations of 5 and 10 Anax imperator adipokinetic hormone for 24 h. The effects of the Anax imperator adipokinetic hormone on the migrasome formation and brain-derived neurotrophic factor expression were analyzed using immunocytochemistry and transmission electron microscope. RESULTS The rat C6 glioma cells of the 5 and 10 μM Anax imperator adipokinetic hormone groups showed significantly high expressions of brain-derived neurotrophic factor and migrasomes numbers, compared with the control group. CONCLUSION A positive correlation was found between the brain-derived neurotrophic factor expression level and the formation of migrasome, which indicates that the increased expression of brain-derived neurotrophic factor and the number of migrasomes may be involved to metastasis of the rat C6 glioma cell line induced by the Anax imperator adipokinetic hormone. Therefore, the expression of brain-derived neurotrophic factor and migrasome formation may be promising targets for preventing tumor proliferation, invasion, and metastasis in glioma.
Collapse
Affiliation(s)
- Sibel Köktürk
- Istanbul University, Faculty of Medicine, Department of Histology and Embryology – İstanbul, Turkey
| | - Sibel Doğan
- Istanbul University, Faculty of Medicine, Department of Histology and Embryology – İstanbul, Turkey
| | - Cansu Eda Yılmaz
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Pathology – İstanbul, Turkey
| | - Yeliz Cetinkol
- Afyonkarahisar Health Sciences University, Faculty of Medicine, Department of Medical Microbiology – Afyonkarahisar, Turkey
| | - Oğuz Mutlu
- Kocaeli University, Faculty of Medicine, Department of Pharmacology – İzmit, Turkey
| |
Collapse
|
33
|
Wei M, Zhou G, Chen L, Zhang Y, Ma W, Gao L, Gao G. The prognostic and immune significance of PLBD1 in pan-cancer and its roles in proliferation and invasion of glioma. J Cancer 2024; 15:3857-3872. [PMID: 38911364 PMCID: PMC11190780 DOI: 10.7150/jca.96365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024] Open
Abstract
Cancer is a destructive disease and is currently the leading cause of major threats to human health. PLBD1 is a transcription factor that regulates phospholipid metabolism, but its role in tumors is unknown. We assessed pan-cancer expression, methylation, and mutation data of PLBD1 by multiple databases to investigate its clinical prognostic value. In addition, we examined the pan-cancer immunological signature of PLBD1, particularly in gliomas. Furthermore, we assessed the impact of PLBD1 knockdown on the proliferation and invasive capacity of glioma cells by in vitro experiments. Our results suggest that PLBD1 is highly expressed in multiple types of cancers, and it can serve as an independent prognostic factor for gliomas. In addition, we found that the epigenetic alterations of PLBD1 were highly heterogeneous in a variety of cancers, including gliomas, and that its high methylation was associated with poor prognosis in a broad range of cancers. Immunological profiling demonstrated that PLBD1 was significantly associated with immune cell infiltration and multiple immune checkpoints in gliomas and is a potential biomarker for gliomas. Furthermore, cellular experiments showed that knockdown of PLBD1 significantly inhibited the proliferation and invasive ability of glioma cells. In conclusion, PLBD1 is a potential tumor prognostic biomarker and immunotherapeutic target that plays a crucial role in glioma cell proliferation, invasion and immunotherapy.
Collapse
Affiliation(s)
- Minghao Wei
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
- Department of Neurosurgery Ward II, the Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Gaoyang Zhou
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lian Chen
- Department of Neurosurgery, the Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001 China
| | - Yufu Zhang
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Wei Ma
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| |
Collapse
|
34
|
Indira Chandran V, Gopala S, Venkat EH, Kjolby M, Nejsum P. Extracellular vesicles in glioblastoma: a challenge and an opportunity. NPJ Precis Oncol 2024; 8:103. [PMID: 38760427 PMCID: PMC11101656 DOI: 10.1038/s41698-024-00600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
Glioblastoma is a highly heterogeneous tumor whose pathophysiological complexities dictate both the diagnosis of disease severity as well as response to therapy. Conventional diagnostic tools and standard treatment regimens have only managed to achieve limited success in the management of patients suspected of glioblastoma. Extracellular vesicles are an emerging liquid biopsy tool that has shown great promise in resolving the limitations presented by the heterogeneous nature of glioblastoma. Here we discuss the contrasting yet interdependent dual role of extracellular vesicles as communication agents that contribute to the progression of glioblastoma by creating a heterogeneous microenvironment and as a liquid biopsy tool providing an opportunity to accurately identify the disease severity and progression.
Collapse
Affiliation(s)
- Vineesh Indira Chandran
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Easwer Hariharan Venkat
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology and Steno Diabetes Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
35
|
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
|
36
|
Chen Y, Li R, Li Z, Yang B, He J, Li J, Li P, Zhou Z, Wu Y, Zhao Y, Guo G. Bulk and single cells transcriptomes with experimental validation identify USP18 as a novel glioma prognosis and proliferation indicator. Exp Ther Med 2024; 27:229. [PMID: 38596661 PMCID: PMC11002833 DOI: 10.3892/etm.2024.12517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/21/2024] [Indexed: 04/11/2024] Open
Abstract
The mechanism by which ubiquitin-specific protease 18 (USP18) (enzyme commission: 3.4.19.12) inhibition in cancer promotes cell pyroptosis via the induction of interferon (IFN)-stimulated genes has been recently demonstrated. It is also known that USP18 influences the epithelial-mesenchymal transition of glioma cells. In the present study, the upregulation of USP18 in glioma was revealed through bulk transcriptome analysis, which was associated with poor prognosis in patients with glioma. Furthermore, USP18 levels affected the response to immunotherapy in patients with glioma. Single-cell transcriptome and enrichment analyses demonstrated that USP18 was associated with type 1 IFN responses in glioma T cells. To demonstrate the effect of USP18 expression levels on glioma cells, USP18 expression was knocked down in U251 and U87MG ATCC cell lines. A subsequent Cell Counting Kit-8 assay revealed that glioma cell viability was significantly decreased 4 days after USP18 knockdown. In addition, the knockdown of USP18 expression significantly inhibited the clonogenicity of U251 and U87MG ATCC cells. In conclusion, the present study demonstrated that knockdown of USP18 expression inhibited the proliferation of glioma cells, which may be mediated by the effect of USP18 on the IFN-I response.
Collapse
Affiliation(s)
- Yang Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ren Li
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ziao Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Biao Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jianhang He
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jiayu Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Peize Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zihan Zhou
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yongqiang Wu
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Geng Guo
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
37
|
Yang G, Su F, Han BX, Su HX, Guo CH, Yu SH, Guan QL, Hou XM. HNF1A induces glioblastoma by upregulating EPS8 and activating PI3K/AKT signaling pathway. Biochem Pharmacol 2024; 223:116133. [PMID: 38494066 DOI: 10.1016/j.bcp.2024.116133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Despite the exact biological role of HNF1 homolog A (HNF1A) in the regulatory mechanism of glioblastoma (GBM), the molecular mechanism, especially the downstream regulation as a transcription factor, remains to be further elucidated. Immunohistochemistry was used to detect the expression and clinical relevance of HNF1A in GBM patients. CCK8, TUNEL, and subcutaneous tumor formation in nude mice were used to evaluate the effect of HNF1A on GBM in vitro and in vivo. The correction between HNF1A and epidermal growth factor receptor pathway substrate 8 (EPS8) was illustrated by bioinformatics analysis and luciferase assay. Further mechanism was explored that the transcription factor HNF1A regulated the expression of EPS8 and downstream signaling pathways by directly binding to the promoter region of EPS8. Our comprehensive analysis of clinical samples in this study showed that upregulated expression of HNF1A was associated with poor survival in GBM patients. Further, we found that knockdown of HNF1A markedly suppressed the malignant phenotype of GBM cells in vivo and in vitro as well as promoted apoptosis of tumor cells, which was reversed by upregulation of HNF1A. Mechanistically, HNF1A could significantly activate PI3K/AKT signaling pathway by specifically binding to the promoter regions of EPS8. Moreover, overexpression of EPS8 was able to reverse the apoptosis of tumor cells caused by HNF1A knockdown, thereby exacerbating the GBM progression. Correctively, our study has clarified the explicit mechanism by which HNF1A promotes GBM malignancy and provides a new therapeutic target for further clinical application.
Collapse
Affiliation(s)
- Gang Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Neurosurgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Fei Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Bin-Xiao Han
- Gansu Institute of Medical Information, Institute of Gansu Medical Science Research, Lanzhou, Gansu 730000, PR China
| | - Hong-Xin Su
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Chen-Hao Guo
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Shao-Hua Yu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Quan-Lin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, PR China; Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China.
| | - Xiao-Ming Hou
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China.
| |
Collapse
|
38
|
Guo W, Peng D, Liao Y, Lou L, Guo M, Li C, Yu W, Tian X, Wang G, Lv P, Zuo J, Shen H, Li Y. Upregulation of HLA-II related to LAG-3 +CD4 + T cell infiltration is associated with patient outcome in human glioblastoma. Cancer Sci 2024; 115:1388-1404. [PMID: 38480275 PMCID: PMC11093187 DOI: 10.1111/cas.16128] [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: 11/19/2022] [Revised: 02/01/2024] [Accepted: 02/17/2024] [Indexed: 05/15/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant diffuse glioma of the brain. Although immunotherapy with immune checkpoint inhibitors (ICIs), such as programmed cell death protein (PD)-1/PD ligand-1 inhibitors, has revolutionized the treatment of several cancers, the clinical benefit in GBM patients has been limited. Lymphocyte-activation gene 3 (LAG-3) binding to human leukocyte antigen-II (HLA-II) plays an essential role in triggering CD4+ T cell exhaustion and could interfere with the efficiency of anti-PD-1 treatment; however, the value of LAG-3-HLA-II interactions in ICI immunotherapy for GBM patients has not yet been analyzed. Therefore, we aimed to investigate the expression and regulation of HLA-II in human GBM samples and the correlation with LAG-3+CD4+ T cell infiltration. Human leukocyte antigen-II was highly expressed in GBM and correlated with increased LAG-3+CD4+ T cell infiltration in the stroma. Additionally, HLA-IIHighLAG-3High was associated with worse patient survival. Increased interleukin-10 (IL-10) expression was observed in GBM, which was correlated with high levels of HLA-II and LAG-3+ T cell infiltration in stroma. HLA-IIHighIL-10High GBM associated with LAG-3+ T cells infiltration synergistically showed shorter overall survival in patients. Combined anti-LAG-3 and anti-IL-10 treatment inhibited tumor growth in a mouse brain GL261 tumor model. In vitro, CD68+ macrophages upregulated HLA-II expression in GBM cells through tumor necrosis factor-α (TNF-α). Blocking TNF-α-dependent inflammation inhibited tumor growth in a mouse GBM model. In summary, T cell-tumor cell interactions, such as LAG-3-HLA-II, could confer an immunosuppressive environment in human GBM, leading to poor prognosis in patients. Therefore, targeting the LAG-3-HLA-II interaction could be beneficial in ICI immunotherapy to improve the clinical outcome of GBM patients.
Collapse
Affiliation(s)
- Wenli Guo
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
| | - Daijun Peng
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Yuee Liao
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Lei Lou
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Moran Guo
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Chen Li
- Department of NeurosurgerySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Wangyang Yu
- Department of NeurosurgerySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaoxi Tian
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Guohui Wang
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
| | - Ping Lv
- Department of PharmacologyHebei Medical UniversityShijiazhuangChina
| | - Jing Zuo
- Department of OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Haitao Shen
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
- Hebei Collaborative Innovation Center of Tumor Microecological Metabolism RegulationHebei UniversityBaodingChina
| | - Yuehong Li
- Department of PathologyThe Second Hospital, Hebei Medical UniversityShijiazhuangChina
- Laboratory of PathologyHebei Medical UniversityShijiazhuangChina
| |
Collapse
|
39
|
Zhu N, Chen S, Jin Y, Wang M, Fang L, Xue L, Hua D, Zhang Z, Jia M, Hao M, Zhang C. Enhancing Glioblastoma Immunotherapy with Integrated Chimeric Antigen Receptor T Cells through the Re-Education of Tumor-Associated Microglia and Macrophages. ACS NANO 2024; 18:11165-11182. [PMID: 38626338 DOI: 10.1021/acsnano.4c00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Glioblastoma (GBM) is an aggressive brain cancer that is highly resistant to treatment including chimeric antigen receptor (CAR)-T cells. Tumor-associated microglia and macrophages (TAMs) are major contributors to the immunosuppressive GBM microenvironment, which promotes tumor progression and treatment resistance. Hence, the modulation of TAMs is a promising strategy for improving the immunotherapeutic efficacy of CAR-T cells against GBM. Molecularly targeting drug pexidartinib (PLX) has been reported to re-educate TAMs toward the antitumorigenic M1-like phenotype. Here, we developed a cell-drug integrated technology to reversibly conjugate PLX-containing liposomes (PLX-Lip) to CAR-T cells and establish tumor-responsive integrated CAR-T cells (PLX-Lip/AZO-T cells) as a combination therapy for GBM. We used a mouse model of GBM to show that PLX-Lip was stably maintained on the surface of PLX-Lip/AZO-T cells in circulation and these cells could transmigrate across the blood-brain barrier and deposit PLX-Lip at the tumor site. The uptake of PLX-Lip by TAMs effectively re-educated them into the M1-like phenotype, which in turn boosted the antitumor function of CAR-T cells. GBM tumor growth was completely eradicated in 60% of the mice after receiving PLX-Lip/AZO-T cells and extended their overall survival time beyond 50 days; in comparison, the median survival time of mice in other treatment groups did not exceed 35 days. Overall, we demonstrated the successful fusion of CAR-T cells and small-molecule drugs with the cell-drug integrated technology. These integrated CAR-T cells provided a superior combination strategy for GBM treatment and presented a reference for the construction of integrated cell-based drugs.
Collapse
Affiliation(s)
- Nianci Zhu
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Sijia Chen
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Yu Jin
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Meng Wang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Luyao Fang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Dexiang Hua
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Ziyao Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Meng Jia
- School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Meixi Hao
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| |
Collapse
|
40
|
Scuderi SA, Ardizzone A, Salako AE, Pantò G, De Luca F, Esposito E, Capra AP. Pentraxin 3: A Main Driver of Inflammation and Immune System Dysfunction in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2024; 16:1637. [PMID: 38730589 PMCID: PMC11083335 DOI: 10.3390/cancers16091637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Brain tumors are a heterogeneous group of brain neoplasms that are highly prevalent in individuals of all ages worldwide. Within this pathological framework, the most prevalent and aggressive type of primary brain tumor is glioblastoma (GB), a subtype of glioma that falls within the IV-grade astrocytoma group. The death rate for patients with GB remains high, occurring within a few months after diagnosis, even with the gold-standard therapies now available, such as surgery, radiation, or a pharmaceutical approach with Temozolomide. For this reason, it is crucial to continue looking for cutting-edge therapeutic options to raise patients' survival chances. Pentraxin 3 (PTX3) is a multifunctional protein that has a variety of regulatory roles in inflammatory processes related to extracellular matrix (ECM). An increase in PTX3 blood levels is considered a trustworthy factor associated with the beginning of inflammation. Moreover, scientific evidence suggested that PTX3 is a sensitive and earlier inflammation-related marker compared to the short pentraxin C-reactive protein (CRP). In several tumoral subtypes, via regulating complement-dependent and macrophage-associated tumor-promoting inflammation, it has been demonstrated that PTX3 may function as a promoter of cancer metastasis, invasion, and stemness. Our review aims to deeply evaluate the function of PTX3 in the pathological context of GB, considering its pivotal biological activities and its possible role as a molecular target for future therapies.
Collapse
Affiliation(s)
- Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Ayomide Eniola Salako
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
- University of Florence, 50121 Florence, Italy
| | - Giuseppe Pantò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (S.A.S.); (A.A.); (A.E.S.); (F.D.L.); (A.P.C.)
| |
Collapse
|
41
|
Tang Y, Wang M, Yu J, Lv G, Wang Y, Yu B. The antitumor action of endocannabinoids in the tumor microenvironment of glioblastoma. Front Pharmacol 2024; 15:1395156. [PMID: 38720772 PMCID: PMC11076672 DOI: 10.3389/fphar.2024.1395156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Approximately 80% of all malignant brain tumors are gliomas, which are primary brain tumors. The most prevalent subtype of glioma, glioblastoma multiforme (GBM), is also the most deadly. Chemotherapy, immunotherapy, surgery, and conventional pharmacotherapy are currently available therapeutic options for GBM; unfortunately, these approaches only prolong the patient's life by 5 years at most. Despite numerous intensive therapeutic options, GBM is considered incurable. Accumulating preclinical data indicate that overt antitumoral effects can be induced by pharmacologically activating endocannabinoid receptors on glioma cells by modifying important intracellular signaling cascades. The complex mechanism underlying the endocannabinoid receptor-evoked antitumoral activity in experimental models of glioma may inhibit the ability of cancer cells to invade, proliferate, and exhibit stem cell-like characteristics, along with altering other aspects of the complex tumor microenvironment. The exact biological function of the endocannabinoid system in the development and spread of gliomas, however, is remains unclear and appears to rely heavily on context. Previous studies have revealed that endocannabinoid receptors are present in the tumor microenvironment, suggesting that these receptors could be novel targets for the treatment of GBM. Additionally, endocannabinoids have demonstrated anticancer effects through signaling pathways linked to the classic features of cancer. Thus, the pharmacology of endocannabinoids in the glioblastoma microenvironment is the main topic of this review, which may promote the development of future GBM therapies.
Collapse
Affiliation(s)
- Yi Tang
- Department of Pharmacy, Sichuan Cancer Center, Sichuan Cancer Hospital and Institute, Affiliate Cancer Hospital of University of Electronic Science and Technology of China (UESTC), Chengdu, China
| | - Maoru Wang
- Drug Dispensing Department, Sichuan Mental Health Center, The Third Hospital of Mianyang, Mianyang, China
| | - Jiangping Yu
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Guangyao Lv
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yu Wang
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Bin Yu
- Department of Pharmacy, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
42
|
Kappel AD, Jha R, Guggilapu S, Smith WJ, Feroze AH, Dmytriw AA, Vicenty-Padilla J, Alcedo Guardia RE, Gessler FA, Patel NJ, Du R, See AP, Peruzzi PP, Aziz-Sultan MA, Bernstock JD. Endovascular Applications for the Management of High-Grade Gliomas in the Modern Era. Cancers (Basel) 2024; 16:1594. [PMID: 38672676 PMCID: PMC11049132 DOI: 10.3390/cancers16081594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
High-grade gliomas (HGGs) have a poor prognosis and are difficult to treat. This review examines the evolving landscape of endovascular therapies for HGGs. Recent advances in endovascular catheter technology and delivery methods allow for super-selective intra-arterial cerebral infusion (SSIACI) with increasing precision. This treatment modality may offer the ability to deliver anti-tumoral therapies directly to tumor regions while minimizing systemic toxicity. However, challenges persist, including blood-brain barrier (BBB) penetration, hemodynamic complexities, and drug-tumor residence time. Innovative adjunct techniques, such as focused ultrasound (FUS) and hyperosmotic disruption, may facilitate BBB disruption and enhance drug penetration. However, hemodynamic factors that limit drug residence time remain a limitation. Expanding therapeutic options beyond chemotherapy, including radiotherapy and immunobiologics, may motivate future investigations. While preclinical and clinical studies demonstrate moderate efficacy, larger randomized trials are needed to validate the clinical benefits. Additionally, future directions may involve endovascular sampling for peri-tumoral surveillance; changes in drug formulations to prolong residence time; and the exploration of non-pharmaceutical therapies, like radioembolization and photodynamic therapy. Endovascular strategies hold immense potential in reshaping HGG treatment paradigms, offering targeted and minimally invasive approaches. However, overcoming technical challenges and validating clinical efficacy remain paramount for translating these advancements into clinical care.
Collapse
Affiliation(s)
- Ari D. Kappel
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Rohan Jha
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
| | - Saibaba Guggilapu
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
| | - William J. Smith
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Abdullah H. Feroze
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Adam A. Dmytriw
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Juan Vicenty-Padilla
- Neurosurgery Section, School of Medicine University of Puerto Rico, Medical Sciences Campus, San Juan P.O. Box 365067, Puerto Rico (R.E.A.G.)
| | - Rodolfo E. Alcedo Guardia
- Neurosurgery Section, School of Medicine University of Puerto Rico, Medical Sciences Campus, San Juan P.O. Box 365067, Puerto Rico (R.E.A.G.)
| | - Florian A. Gessler
- Department of Neurosurgery, Rostock University Hospital, 18057 Rostock, Germany
| | - Nirav J. Patel
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Rose Du
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Alfred P. See
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Pier Paolo Peruzzi
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Mohammad A. Aziz-Sultan
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Joshua D. Bernstock
- Harvard Medical School, Boston, MA 02115, USA; (A.D.K.); (S.G.); (R.D.); (A.P.S.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
43
|
Wang M, Graner AN, Knowles B, McRae C, Fringuello A, Paucek P, Gavrilovic M, Redwine M, Hanson C, Coughlan C, Metzger B, Bolus V, Kopper T, Smith M, Zhou W, Lenz M, Abosch A, Ojemann S, Lillehei KO, Yu X, Graner MW. A tale of two tumors: differential, but detrimental, effects of glioblastoma extracellular vesicles (EVs) on normal human brain cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588622. [PMID: 38645117 PMCID: PMC11030303 DOI: 10.1101/2024.04.08.588622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Glioblastomas (GBMs) are dreadful brain tumors with abysmal survival outcomes. GBM EVs dramatically affect normal brain cells (largely astrocytes) constituting the tumor microenvironment (TME). EVs from different patient-derived GBM spheroids induced differential transcriptomic, secretomic, and proteomic effects on cultured astrocytes/brain tissue slices as GBM EV recipients. The net outcome of brain cell differential changes nonetheless converges on increased tumorigenicity. GBM spheroids and brain slices were derived from neurosurgical patient tissues following informed consent. Astrocytes were commercially obtained. EVs were isolated from conditioned culture media by ultrafiltration, ultraconcentration, and ultracentrifugation. EVs were characterized by nanoparticle tracking analysis, electron microscopy, biochemical markers, and proteomics. Astrocytes/brain tissues were treated with GBM EVs before downstream analyses. EVs from different GBMs induced brain cells to alter secretomes with pro-inflammatory or TME-modifying (proteolytic) effects. Astrocyte responses ranged from anti-viral gene/protein expression and cytokine release to altered extracellular signal-regulated protein kinase (ERK1/2) signaling pathways, and conditioned media from EV-treated cells increased GBM cell proliferation. Thus, astrocytes/brain slices treated with different GBM EVs underwent non-identical changes in various 'omics readouts and other assays, indicating "personalized" tumor-specific GBM EV effects on the TME. This raises concern regarding reliance on "model" systems as a sole basis for translational direction. Nonetheless, net downstream impacts from differential cellular and TME effects still led to increased tumorigenic capacities for the different GBMs.
Collapse
|
44
|
Spinelli C, Adnani L, Meehan B, Montermini L, Huang S, Kim M, Nishimura T, Croul SE, Nakano I, Riazalhosseini Y, Rak J. Mesenchymal glioma stem cells trigger vasectasia-distinct neovascularization process stimulated by extracellular vesicles carrying EGFR. Nat Commun 2024; 15:2865. [PMID: 38570528 PMCID: PMC10991552 DOI: 10.1038/s41467-024-46597-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Targeting neovascularization in glioblastoma (GBM) is hampered by poor understanding of the underlying mechanisms and unclear linkages to tumour molecular landscapes. Here we report that different molecular subtypes of human glioma stem cells (GSC) trigger distinct endothelial responses involving either angiogenic or circumferential vascular growth (vasectasia). The latter process is selectively triggered by mesenchymal (but not proneural) GSCs and is mediated by a subset of extracellular vesicles (EVs) able to transfer EGFR/EGFRvIII transcript to endothelial cells. Inhibition of the expression and phosphorylation of EGFR in endothelial cells, either pharmacologically (Dacomitinib) or genetically (gene editing), abolishes their EV responses in vitro and disrupts vasectasia in vivo. Therapeutic inhibition of EGFR markedly extends anticancer effects of VEGF blockade in mice, coupled with abrogation of vasectasia and prolonged survival. Thus, vasectasia driven by intercellular transfer of oncogenic EGFR may represent a new therapeutic target in a subset of GBMs.
Collapse
Affiliation(s)
- Cristiana Spinelli
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lata Adnani
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Brian Meehan
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Laura Montermini
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Sidong Huang
- Department of Biochemistry, McGill University, Montreal, QC, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Minjun Kim
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tamiko Nishimura
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Sidney E Croul
- Department of Pathology & Laboratory Medicine, Dalhousie University, Halifax, NS, Canada
| | - Ichiro Nakano
- Department of Neurosurgery, Hokuto Social Medical Corporation, Hokuto Hospital, Kisen-7-5 Inadacho, Obihiro, Hokkaido, 080-0833, Japan
| | | | - Janusz Rak
- McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
45
|
Fan Q, Wang H, Gu T, Liu H, Deng P, Li B, Yang H, Mao Y, Shao Z. Modeling the precise interaction of glioblastoma with human brain region-specific organoids. iScience 2024; 27:109111. [PMID: 38390494 PMCID: PMC10882168 DOI: 10.1016/j.isci.2024.109111] [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: 07/21/2023] [Revised: 09/18/2023] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Glioblastoma is a highly aggressive malignant tumor of the central nervous system, but the interaction between glioblastoma and different types of neurons remains unclear. Here, we established a co-culture model in vitro using 3D printed molds with microchannels, in which glioblastoma organoids (GB), dorsal forebrain organoids (DO, mainly composed of excitatory neurons), and ventral forebrain organoids (VO, mainly composed of inhibitory neurons) were assembled. Our results indicate that DO has a greater impact on altered gene expression profiles of GB, resulting in increased invasive potential. GB cells preferentially invaded DO along axons, whereas this phenomenon was not observed in VO. Furthermore, GB cells selectively inhibited neurite outgrowth in DOs and reduced the expression of the vesicular GABA transporter (VGAT), leading to neuronal hyperexcitability. By revealing how glioblastoma interacts with brain cells, our study provides a more comprehensive understanding of this disease.
Collapse
Affiliation(s)
- Qi Fan
- Institutes for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Hanze Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Tianyi Gu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 201203, China
| | - Huihui Liu
- Institutes for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Peng Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhicheng Shao
- Institutes for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Cui X, Wang Q, Liu X, Kang C. Levetiracetam: A Potent Sword against Microglia Polarization in Gliomas. Clin Cancer Res 2024; 30:1073-1075. [PMID: 38170191 DOI: 10.1158/1078-0432.ccr-23-3322] [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: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Crosstalk between tumor cells and peritumoral cells contributes to immunosuppressive microenvironment formation in glioblastomas (GBM). A recent study revealed that glioma stem cells activated neuronal activity to promote microglial M2 polarization, leading to GBM progression, which could be pharmacologically blocked by levetiracetam, providing a practical strategy for GBM immunotherapy. See related article by Guo et al., p. 1160.
Collapse
Affiliation(s)
- Xiaoteng Cui
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| | - Qixue Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Nankai University, Tianjin, P.R. China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, P.R China
- Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, P.R. China
| |
Collapse
|
47
|
Zhang L, Li X, Gao H, Li P. The Role of Circular RNA Variants Generated from the NFIX Gene in Different Diseases. Mol Pharm 2024; 21:1027-1037. [PMID: 38315004 DOI: 10.1021/acs.molpharmaceut.3c00933] [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: 02/07/2024]
Abstract
Circular RNAs (circRNAs) have been identified as important regulators in different developmental processes and disease pathogenesis. The loop structure of circRNAs makes them very stable in different conditions and microenvironments. circRNAs can affect microRNA (miRNA) and RNA binding protein (RBP) activity, encode functional proteins and regulate gene transcription. Recently, two circNFIX variants derived from the same gene, the Nuclear Factor I X (NFIX) gene, were determined as participants in the pathological processes of various diseases such as heart diseases and cancers. Both circNFIX variants are exonic circular RNAs and mainly function by sponging miRNAs. In this review, we summarize the current knowledge on circRNAs, elucidate the origins and properties of two circNFIX variants, explore the roles of two circNFIX variants in different diseases, and present clinical perspectives.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, No. 38 DengZhou Road, Qingdao 266021, China
| |
Collapse
|
48
|
Sanchez JC, Pierpont TM, Argueta-Zamora D, Wilson K, August A, Cerione RA. PTEN loss in glioma cell lines leads to increased extracellular vesicles biogenesis and PD-L1 cargo in a PI3K-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.26.550575. [PMID: 38464280 PMCID: PMC10925116 DOI: 10.1101/2023.07.26.550575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Phosphatase and Tensin Homologue (PTEN) is one of the most frequently lost tumor suppressors in cancer and the predominant negative regulator of the PI3K/AKT signaling axis. A growing body of evidence has highlighted the loss of PTEN with immuno-modulatory functions including the upregulation of the programmed death ligand-1 (PD-L1), an altered tumor derived secretome that drives an immunosuppressive tumor immune microenvironment (TIME), and resistance to certain immunotherapies. Given their roles in immunosuppression and tumor growth, we examined whether the loss of PTEN would impact the biogenesis, cargo, and function of extracellular vesicles (EVs) in the context of the anti-tumor associated cytokine interferon-γ (IFN-γ). Through genetic and pharmacological approaches, we show that PD-L1 expression is regulated by JAK/STAT signaling, not PI3K signaling. Instead, we observe that PTEN loss positively upregulates cell surface levels of PD-L1 and enhances the biogenesis of EVs enriched with PD-L1 in a PI3K-dependent manner. We demonstrate that because of these changes, EVs derived from glioma cells lacking PTEN have a greater ability to suppress T cell receptor (TCR) signaling. Taken together, these findings provide important new insights into how the loss of PTEN can contribute to an immunosuppressive TIME, facilitate immune evasion, and highlight a novel role for PI3K signaling in the regulation of EV biogenesis and the cargo they contain.
Collapse
Affiliation(s)
- Julio C Sanchez
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Timothy M Pierpont
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Dariana Argueta-Zamora
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Kristin Wilson
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Richard A Cerione
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
49
|
Ahluwalia MS, Khosla AA, Ozair A, Gouda MA, Subbiah V. Impact of tissue-agnostic approvals on management of primary brain tumors. Trends Cancer 2024; 10:256-274. [PMID: 38245379 DOI: 10.1016/j.trecan.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 01/22/2024]
Abstract
Novel tissue-agnostic therapeutics targeting driver mutations in tumor cells have been recently approved by FDA, driven by basket trials that have demonstrated their efficacy and safety across diverse tumor histology. However, the relative rarity of primary brain tumors (PBTs) has limited their representation in early trials of tissue-agnostic medications. Thus, consensus continues to evolve regarding utility of tissue-agnostic medications in routine practice for PBTs, a diverse group of neoplasms characterized by limited treatment options and unfavorable prognoses. We describe current and potential impact of tissue-agnostic approvals on management of PBTs. We discuss data from clinical trials for PBTs regarding tissue-agnostic targets, including BRAFV600E, neurotrophic tyrosine receptor kinase (NTRK) fusions, microsatellite instability-high (MSI-High), mismatch repair deficiency (dMMR), and high tumor mutational burden (TMB-H), in context of challenges in managing PBTs. Described are additional tissue-agnostic targets that hold promise for benefiting patients with PBTs, including RET fusion, fibroblast growth factor receptor (FGFR), ERBB2/HER2, and KRASG12C, and TP53Y220C.
Collapse
Affiliation(s)
- Manmeet S Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Atulya A Khosla
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Department of Internal Medicine, William Beaumont University Hospital, Royal Oak, MI, USA
| | - Ahmad Ozair
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Early Phase Drug Development Program, Sarah Cannon Research Institute, Nashville, TN, USA.
| |
Collapse
|
50
|
Li Z, Guo K, Gao Z, Chen J, Ye Z, Cao M, Wang SE, Yin Y, Zhong W. Colocalization of protein and microRNA markers reveals unique extracellular vesicle subpopulations for early cancer detection. SCIENCE ADVANCES 2024; 10:eadh8689. [PMID: 38416840 PMCID: PMC10901469 DOI: 10.1126/sciadv.adh8689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Extracellular vesicles (EVs) play important roles in cell-cell communication but are highly heterogeneous, and each vesicle has dimensions smaller than 200 nm with very limited amounts of cargos encapsulated. The technique of NanOstirBar (NOB)-EnabLed Single Particle Analysis (NOBEL-SPA) reported in the present work permits rapid inspection of single EV with high confidence by confocal fluorescence microscopy, thus enables colocalization assessment for selected protein and microRNA (miRNA) markers in the EVs produced by various cell lines, or present in clinical sera samples. EV subpopulations marked by the colocalization of unique protein and miRNA combinations were discovered to be able to detect early-stage (stage I or II) breast cancer (BC). NOBEL-SPA can be adapted to analyze other types of cargo molecules or other small submicron biological particles. Study of the sorting of specific cargos to heterogeneous vesicles under different physiological conditions can help discover distinct vesicle subpopulations valuable in clinical examination and therapeutics development and gain better understanding of their biogenesis.
Collapse
Affiliation(s)
- Zongbo Li
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Kaizhu Guo
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Ziting Gao
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Junyi Chen
- Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA 92521, USA
| | - Zuyang Ye
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Minghui Cao
- Department of Pathology, University of California–San Diego, La Jolla, CA 92093, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California–San Diego, La Jolla, CA 92093, USA
| | - Yadong Yin
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
| | - Wenwan Zhong
- Department of Chemistry, University of California-Riverside, Riverside, CA 92521, USA
- Environmental Toxicology Graduate Program, University of California-Riverside, Riverside, CA 92521, USA
| |
Collapse
|