1
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Giammello F, Biella C, Priori EC, Filippo MADS, Leone R, D'Ambrosio F, Paterno' M, Cassioli G, Minetti A, Macchi F, Spalletti C, Morella I, Ruberti C, Tremonti B, Barbieri F, Lombardi G, Brambilla R, Florio T, Galli R, Rossi P, Brandalise F. Modulating voltage-gated sodium channels to enhance differentiation and sensitize glioblastoma cells to chemotherapy. Cell Commun Signal 2024; 22:434. [PMID: 39251990 PMCID: PMC11382371 DOI: 10.1186/s12964-024-01819-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: 04/05/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle. METHODS We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo. RESULTS We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav. CONCLUSIONS This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide treatment.
Collapse
Affiliation(s)
- Francesca Giammello
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
- PhD Program in Genetics, Molecular and Cellular Biology, University of Pavia, Pavia, Italy
| | - Chiara Biella
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Roberta Leone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Martina Paterno'
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Giulia Cassioli
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Antea Minetti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Francesca Macchi
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Cristina Spalletti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Ilaria Morella
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Cristina Ruberti
- Advanced Technology Platform, Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Beatrice Tremonti
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Federica Barbieri
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Giuseppe Lombardi
- Department of Oncology 1, Oncology, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, Padua, 35128, Italy
| | - Riccardo Brambilla
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Tullio Florio
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Rossella Galli
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | |
Collapse
|
3
|
Yi L, Lin X, She X, Gao W, Wu M. Chronic stress as an emerging risk factor for the development and progression of glioma. Chin Med J (Engl) 2024; 137:394-407. [PMID: 38238191 PMCID: PMC10876262 DOI: 10.1097/cm9.0000000000002976] [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: 05/13/2023] [Indexed: 02/21/2024] Open
Abstract
ABSTRACT Gliomas tend to have a poor prognosis and are the most common primary malignant tumors of the central nervous system. Compared with patients with other cancers, glioma patients often suffer from increased levels of psychological stress, such as anxiety and fear. Chronic stress (CS) is thought to impact glioma profoundly. However, because of the complex mechanisms underlying CS and variability in individual tolerance, the role of CS in glioma remains unclear. This review suggests a new proposal to redivide the stress system into two parts. Neuronal activity is dominant upstream. Stress-signaling molecules produced by the neuroendocrine system are dominant downstream. We discuss the underlying molecular mechanisms by which CS impacts glioma. Potential pharmacological treatments are also summarized from the therapeutic perspective of CS.
Collapse
Affiliation(s)
- Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| | - Xiaoling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
4
|
Gondarenko E, Mazur D, Masliakova M, Ryabukha Y, Kasheverov I, Utkin Y, Tsetlin V, Shahparonov M, Kudryavtsev D, Antipova N. Subtype-Selective Peptide and Protein Neurotoxic Inhibitors of Nicotinic Acetylcholine Receptors Enhance Proliferation of Patient-Derived Glioblastoma Cell Lines. Toxins (Basel) 2024; 16:80. [PMID: 38393158 PMCID: PMC10891657 DOI: 10.3390/toxins16020080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain cancer, with a poor prognosis. GBM cells, which develop in the environment of neural tissue, often exploit neurotransmitters and their receptors to promote their own growth and invasion. Nicotinic acetylcholine receptors (nAChRs), which play a crucial role in central nervous system signal transmission, are widely represented in the brain, and GBM cells express several subtypes of nAChRs that are suggested to transmit signals from neurons, promoting tumor invasion and growth. Analysis of published GBM transcriptomes revealed spatial heterogeneity in nAChR subtype expression, and functional nAChRs of α1*, α7, and α9 subtypes are demonstrated in our work on several patient-derived GBM microsphere cultures and on the U87MG GBM cell line using subtype-selective neurotoxins and fluorescent calcium mobilization assay. The U87MG cell line shows reactions to nicotinic agonists similar to those of GBM patient-derived culture. Selective α1*, α7, and α9 nAChR neurotoxins stimulated cell growth in the presence of nicotinic agonists. Several cultivating conditions with varying growth factor content have been proposed and tested. The use of selective neurotoxins confirmed that cell cultures obtained from patients are representative GBM models, but the use of media containing fetal bovine serum can lead to alterations in nAChR expression and functioning.
Collapse
Affiliation(s)
- Elena Gondarenko
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Diana Mazur
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Marina Masliakova
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Yana Ryabukha
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Igor Kasheverov
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Yuri Utkin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Victor Tsetlin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
| | - Mikhail Shahparonov
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| | - Denis Kudryavtsev
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (E.G.); (I.K.); (V.T.); (D.K.)
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia
| | - Nadine Antipova
- Department of Functioning of Living Systems, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of Russian Academy of Sciences, 117997 Moscow, Russia; (D.M.); (M.M.); (Y.R.); (M.S.); (N.A.)
| |
Collapse
|
5
|
Yu H, Yu J, Wang M, Jiang X. Characterization of Prognostic Apoptosis-Related Gene Signature to Evaluate Glioma Immune Microenvironment and Experimental Verification. Genet Test Mol Biomarkers 2024; 28:22-32. [PMID: 38294358 DOI: 10.1089/gtmb.2023.0483] [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: 02/01/2024] Open
Abstract
Purpose: Recently, apoptosis-related genes were shown to modulate cancer immunity. However, the role of apoptosis-related genes in the glioma immune microenvironment (GIME) remains unknown. This study aimed to explore the prognostic value of apoptosis-related genes in glioma. Methods: Doxorubicin was used to induce glioma cell apoptosis, and four differentially expressed apoptosis-related genes were identified: CREM, TNFSF12, PEA15, and PRKCD. Kaplan-Meier analyses, receiver operating characteristic curve analyses, and nomograms were established to determine the relationship between risk markers and the prognosis of patients with glioma. Results: Risk biomarkers were significantly associated with overall survival, immune cell infiltration, and immune checkpoints in patients with glioma. Somatic mutations and anti-PD-1/L1 immunotherapy were associated with worse prognosis in the high-risk group receiving anti-PD-1/L1 therapy. The expression of these four apoptosis-related genes was verified using quantitative polymerase chain reaction and immunohistochemistry, and the relationship between these four genes and apoptosis was examined using flow cytometry. Conclusions: This study suggests that apoptosis-related genes play a critical role in shaping the GIME. Assessing the apoptotic patterns of individual tumors will enhance our understanding of GIME infiltration features and lead to improved strategies for immunotherapy.
Collapse
Affiliation(s)
- Hao Yu
- Department of Neurosurgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jiapeng Yu
- Department of Neurosurgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Minjie Wang
- Department of Neurosurgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Genoud V, Kinnersley B, Brown NF, Ottaviani D, Mulholland P. Therapeutic Targeting of Glioblastoma and the Interactions with Its Microenvironment. Cancers (Basel) 2023; 15:5790. [PMID: 38136335 PMCID: PMC10741850 DOI: 10.3390/cancers15245790] [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: 10/17/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumour, and it confers a dismal prognosis despite intensive multimodal treatments. Whilst historically, research has focussed on the evolution of GBM tumour cells themselves, there is growing recognition of the importance of studying the tumour microenvironment (TME). Improved characterisation of the interaction between GBM cells and the TME has led to a better understanding of therapeutic resistance and the identification of potential targets to block these escape mechanisms. This review describes the network of cells within the TME and proposes treatment strategies for simultaneously targeting GBM cells, the surrounding immune cells, and the crosstalk between them.
Collapse
Affiliation(s)
- Vassilis Genoud
- Glioblastoma Research Group, University College London, London WC1E 6DD, UK (B.K.)
- Department of Oncology, University College London Hospitals, London NW1 2PB, UK
- Department of Oncology, University Hospitals of Geneva, 1205 Geneva, Switzerland
- Centre for Translational Research in Onco-Haematology, University of Geneva, 1205 Geneva, Switzerland
| | - Ben Kinnersley
- Glioblastoma Research Group, University College London, London WC1E 6DD, UK (B.K.)
- Department of Oncology, University College London Hospitals, London NW1 2PB, UK
| | - Nicholas F. Brown
- Glioblastoma Research Group, University College London, London WC1E 6DD, UK (B.K.)
- Guy’s Cancer, Guy’s & St Thomas’ NHS Foundation Trust, London SE1 3SS, UK
| | - Diego Ottaviani
- Glioblastoma Research Group, University College London, London WC1E 6DD, UK (B.K.)
- Department of Oncology, University College London Hospitals, London NW1 2PB, UK
| | - Paul Mulholland
- Glioblastoma Research Group, University College London, London WC1E 6DD, UK (B.K.)
- Department of Oncology, University College London Hospitals, London NW1 2PB, UK
| |
Collapse
|
7
|
Park G, Jin Z, Ge Q, Pan Y, Du J. Neuronal acid-sensing ion channel 1a regulates neuron-to-glioma synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555794. [PMID: 37693494 PMCID: PMC10491214 DOI: 10.1101/2023.08.31.555794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Neuronal activity promotes high-grade glioma progression via secreted proteins and neuron-to-glioma synapses, and glioma cells boost neuronal activity to further reinforce the malignant cycle. Whereas strong evidence supports that the activity of neuron-to-glioma synapses accelerates tumor progression, the molecular mechanisms that modulate the formation and function of neuron-to-glioma synapses remain largely unknown. Our recent findings suggest that a proton (H + ) signaling pathway actively mediates neuron-to-glioma synaptic communications by activating neuronal acid-sensing ion channel 1a (Asic1a), a predominant H + receptor in the central nervous system (CNS). Supporting this idea, our preliminary data revealed that local acid puff on neurons in high-grade glioma-bearing brain slices induces postsynaptic currents of glioma cells. Stimulating Asic1a knockout (Asic1a -/- ) neurons results in lower AMPA receptor-dependent excitatory postsynaptic currents (EPSCs) in glioma cells than stimulating wild-type (WT) neurons. Moreover, glioma-bearing Asic1a -/- mice exhibited reduced tumor size and survived longer than the glioma-bearing WT mice. Finally, pharmacologically targeting brain Asic1a inhibited high-grade glioma progression. In conclusion, our findings suggest that the neuronal H + -Asic1a axis plays a key role in regulating the neuron-glioma synapse. The outcomes of this study will greatly expand our understanding of how this deadly tumor integrates into the neuronal microenvironment.
Collapse
|
8
|
The Role of Hyperexcitability in Gliomagenesis. Int J Mol Sci 2023; 24:ijms24010749. [PMID: 36614191 PMCID: PMC9820922 DOI: 10.3390/ijms24010749] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor. Recent studies have demonstrated that excitatory or activity-dependent signaling-both synaptic and non-synaptic-contribute to the progression of glioblastoma. Glutamatergic receptors may be stimulated via neuron-tumor synapses or release of glutamate by the tumor itself. Ion currents generated by these receptors directly alter the structure of membrane adhesion molecules and cytoskeletal proteins to promote migratory behavior. Additionally, the hyperexcitable milieu surrounding glioma increases the rate at which tumor cells proliferate and drive recurrent disease. Inhibition of excitatory signaling has shown to effectively reduce its pro-migratory and -proliferative effects.
Collapse
|