1
|
Mastrogiovanni M, Donnadieu E, Pathak R, Di Bartolo V. Subverting Attachment to Prevent Attacking: Alteration of Effector Immune Cell Migration and Adhesion as a Key Mechanism of Tumor Immune Evasion. BIOLOGY 2024; 13:860. [PMID: 39596815 PMCID: PMC11591779 DOI: 10.3390/biology13110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Cell adhesion regulates specific migratory patterns, location, communication with other cells, physical interactions with the extracellular matrix, and the establishment of effector programs. Proper immune control of cancer strongly depends on all these events occurring in a highly accurate spatiotemporal sequence. In response to cancer-associated inflammatory signals, effector immune cells navigating the bloodstream shift from their patrolling exploratory migration mode to establish adhesive interactions with vascular endothelial cells. This interaction enables them to extravasate through the blood vessel walls and access the cancer site. Further adhesive interactions within the tumor microenvironment (TME) are crucial for coordinating their distribution in situ and for mounting an effective anti-tumor immune response. In this review, we examine how alterations of adhesion cues in the tumor context favor tumor escape by affecting effector immune cell infiltration and trafficking within the TME. We discuss the mechanisms by which tumors directly modulate immune cell adhesion and migration patterns to affect anti-tumor immunity and favor tumor evasion. We also explore indirect immune escape mechanisms that involve modifications of TME characteristics, such as vascularization, immunogenicity, and structural topography. Finally, we highlight the significance of these aspects in designing more effective drug treatments and cellular immunotherapies.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emmanuel Donnadieu
- Equipe Labellisée Ligue Contre le Cancer, CNRS, INSERM, Institut Cochin, Université Paris Cité, F-75014 Paris, France;
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Vincenzo Di Bartolo
- Immunoregulation Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
2
|
De Luca C, Virtuoso A, Papa M, Cirillo G, La Rocca G, Corvino S, Barbarisi M, Altieri R. The Three Pillars of Glioblastoma: A Systematic Review and Novel Analysis of Multi-Omics and Clinical Data. Cells 2024; 13:1754. [PMID: 39513861 PMCID: PMC11544881 DOI: 10.3390/cells13211754] [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: 09/14/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma is the most fatal and common malignant brain tumor, excluding metastasis and with a median survival of approximately one year. While solid tumors benefit from newly approved drugs, immunotherapy, and prevention, none of these scenarios are opening for glioblastoma. The key to unlocking the peculiar features of glioblastoma is observing its molecular and anatomical features tightly entangled with the host's central nervous system (CNS). In June 2024, we searched the PUBMED electronic database. Data collection and analysis were conducted independently by two reviewers. Results: A total of 215 articles were identified, and 192 were excluded based on inclusion and exclusion criteria. The remaining 23 were used for collecting divergent molecular pathways and anatomical features of glioblastoma. The analysis of the selected papers revealed a multifaced tumor with extreme variability and cellular reprogramming that are observable within the same patient. All the variability of glioblastoma could be clustered into three pillars to dissect the physiology of the tumor: 1. necrotic core; 2. vascular proliferation; 3. CNS infiltration. These three pillars support glioblastoma survival, with a pivotal role of the neurovascular unit, as supported by the most recent paper published by experts in the field.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
- ISBE Italy, SYSBIO Centre of Systems Biology, 20126 Milan, Italy
| | - Giovanni Cirillo
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (M.P.); (G.C.)
| | - Giuseppe La Rocca
- Department of Neurosurgery, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Catholic University of Rome School of Medicine, 00153 Rome, Italy;
| | - Sergio Corvino
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Neurosurgical Clinic, University “Federico II” of Naples, 80131 Naples, Italy;
| | - Manlio Barbarisi
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| | - Roberto Altieri
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy (R.A.)
| |
Collapse
|
3
|
Qi T, Liao X, Cao Y. Development of bispecific T cell engagers: harnessing quantitative systems pharmacology. Trends Pharmacol Sci 2023; 44:880-890. [PMID: 37852906 PMCID: PMC10843027 DOI: 10.1016/j.tips.2023.09.009] [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: 09/10/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Bispecific T cell engagers (bsTCEs) have emerged as a promising class of cancer immunotherapy. Several bsTCEs have achieved marketing approval; dozens more are under clinical investigation. However, the clinical development of bsTCEs remains rife with challenges, including nuanced pharmacology, limited translatability of preclinical findings, frequent on-target toxicity, and convoluted dosing regimens. In this opinion article we present a distinct perspective on how quantitative systems pharmacology (QSP) can serve as a powerful tool for overcoming these obstacles. Recent advances in QSP modeling have empowered developers of bsTCEs to gain a deeper understanding of their context-dependent pharmacology, bridge gaps in experimental data, guide first-in-human (FIH) dose selection, design dosing regimens with expanded therapeutic windows, and improve long-term treatment outcomes. We use recent case studies to exemplify the potential of QSP techniques to support future bsTCE development.
Collapse
Affiliation(s)
- Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaozhi Liao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Shetab Boushehri S, Essig K, Chlis NK, Herter S, Bacac M, Theis FJ, Glasmacher E, Marr C, Schmich F. Explainable machine learning for profiling the immunological synapse and functional characterization of therapeutic antibodies. Nat Commun 2023; 14:7888. [PMID: 38036503 PMCID: PMC10689847 DOI: 10.1038/s41467-023-43429-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/09/2023] [Indexed: 12/02/2023] Open
Abstract
Therapeutic antibodies are widely used to treat severe diseases. Most of them alter immune cells and act within the immunological synapse; an essential cell-to-cell interaction to direct the humoral immune response. Although many antibody designs are generated and evaluated, a high-throughput tool for systematic antibody characterization and prediction of function is lacking. Here, we introduce the first comprehensive open-source framework, scifAI (single-cell imaging flow cytometry AI), for preprocessing, feature engineering, and explainable, predictive machine learning on imaging flow cytometry (IFC) data. Additionally, we generate the largest publicly available IFC dataset of the human immunological synapse containing over 2.8 million images. Using scifAI, we analyze class frequency and morphological changes under different immune stimulation. T cell cytokine production across multiple donors and therapeutic antibodies is quantitatively predicted in vitro, linking morphological features with function and demonstrating the potential to significantly impact antibody design. scifAI is universally applicable to IFC data. Given its modular architecture, it is straightforward to incorporate into existing workflows and analysis pipelines, e.g., for rapid antibody screening and functional characterization.
Collapse
Affiliation(s)
- Sayedali Shetab Boushehri
- Institute of AI for Health, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich, Department of Mathematics, Munich, Germany
- Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany
| | - Katharina Essig
- Large Molecule Research (LMR), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany
| | - Nikolaos-Kosmas Chlis
- Large Molecule Research (LMR), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany
| | - Sylvia Herter
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Zurich, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zurich, Roche Pharma Research and Early Development (pRED), Zurich, Switzerland
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich, Department of Mathematics, Munich, Germany
| | - Elke Glasmacher
- Research and Early Development (RED), Roche Diagnostics Solutions, Roche Innovation Center Munich, Munich, Germany.
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Fabian Schmich
- Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Munich, Germany.
| |
Collapse
|
5
|
Li WS, Zhang QQ, Li Q, Liu SY, Yuan GQ, Pan YW. Innate immune response restarts adaptive immune response in tumors. Front Immunol 2023; 14:1260705. [PMID: 37781382 PMCID: PMC10538570 DOI: 10.3389/fimmu.2023.1260705] [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: 07/20/2023] [Accepted: 08/25/2023] [Indexed: 10/03/2023] Open
Abstract
The imbalance of immune response plays a crucial role in the development of diseases, including glioblastoma. It is essential to comprehend how the innate immune system detects tumors and pathogens. Endosomal and cytoplasmic sensors can identify diverse cancer cell antigens, triggering the production of type I interferon and pro-inflammatory cytokines. This, in turn, stimulates interferon stimulating genes, enhancing the presentation of cancer antigens, and promoting T cell recognition and destruction of cancer cells. While RNA and DNA sensing of tumors and pathogens typically involve different receptors and adapters, their interaction can activate adaptive immune response mechanisms. This review highlights the similarity in RNA and DNA sensing mechanisms in the innate immunity of both tumors and pathogens. The aim is to enhance the anti-tumor innate immune response, identify regions of the tumor that are not responsive to treatment, and explore new targets to improve the response to conventional tumor therapy and immunotherapy.
Collapse
Affiliation(s)
- Wen-shan Li
- The Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Department of Neurosurgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Qing-qing Zhang
- Department of Respiratory and Critical Care Medicine, Qinghai University Affiliated Hospital, Xining, Qinghai, China
| | - Qiao Li
- The Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Shang-yu Liu
- The Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Guo-qiang Yuan
- The Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Ya-wen Pan
- The Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Ide H, Aoshi T, Saito M, Espulgar WV, Briones JC, Hosokawa M, Matsunaga H, Arikawa K, Takeyama H, Koyama S, Takamatsu H, Tamiya E. Linking antigen specific T-cell dynamics in a microfluidic chip to single cell transcription patterns. Biochem Biophys Res Commun 2023; 657:8-15. [PMID: 36963175 DOI: 10.1016/j.bbrc.2023.03.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
A new non-invasive screening profile has been realized that can aid in determining T-cell activation state at single-cell level. Production of activated T-cells with good specificity and stable proliferation is greatly beneficial for advancing adoptive immunotherapy as innate immunological cells are not effective in recognizing and eliminating cancer as expected. The screening method is realized by relating intracellular Ca2+ intensity and motility of T-cells interacting with APC (Antigen Presenting Cells) in a microfluidic chip. The system is tested using APC pulsed with OVA257-264 peptide and its modified affinities (N4, Q4, T4 and V4), and the T-cells from OT-1 mice. In addition, single cell RNA sequencing reveals the activation states of the cells and the clusters from the derived profiles can be indicative of the T-cell activation state. The presented system here can be versatile for a comprehensive application to proceed with T-cell-based immunotherapy and screen the antigen-specific T-cells with excellent efficiency and high proliferation.
Collapse
Affiliation(s)
- Hiroki Ide
- Graduate School of Engineering Osaka Univ, Japan; PhotoBIO Lab, AIST-Osaka Univ, Japan
| | - Taiki Aoshi
- Research Institute for Microbial Diseases, Osaka Univ, Japan
| | - Masato Saito
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | | | - Jonathan Campos Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | | | | | - Eiichi Tamiya
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Institute of Scientific and Industrial Research, Osaka University, Japan
| |
Collapse
|
7
|
Carmena Moratalla A, Carpentier Solorio Y, Lemaître F, Farzam-Kia N, Da Cal S, Guimond JV, Haddad E, Duquette P, Girard JM, Prat A, Larochelle C, Arbour N. Specific alterations in NKG2D + T lymphocytes in relapsing-remitting and progressive multiple sclerosis patients. Mult Scler Relat Disord 2023; 71:104542. [PMID: 36716577 DOI: 10.1016/j.msard.2023.104542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND T lymphocytes exhibit numerous alterations in relapsing-remitting (RRMS), secondary progressive (SPMS), and primary progressive multiple sclerosis (PPMS). The NKG2D pathway has been involved in MS pathology. NKG2D is a co-activating receptor on subsets of CD4+ and most CD8+ T lymphocytes. The ligands of NKG2D are expressed at low levels in normal tissues but are elevated in MS postmortem brain tissues compared with controls. Whether the NKG2D pathway shows specific changes in different forms of MS remains unclear. METHODS We performed unsupervised and supervised flow cytometry analysis to characterize peripheral blood T lymphocytes from RRMS, SPMS, and PPMS patients and healthy controls (HC). We used an in vitro microscopy approach to assess the role of NKG2D in the interactions between human CD8+T lymphocytes and human astrocytes. RESULTS Specific CD8+, CD4+, and CD4-CD8- T cell populations exhibited altered frequency in MS patients' subgroups. The proportion of NKG2D+ T lymphocytes declined with age in PPMS patients but not in RRMS and HC. This reduced percentage of NKG2D+ cells was due to lower abundance of γδ and αβ CD4-CD8- T lymphocytes in PPMS patients. NKG2D+ T lymphocytes were significantly less abundant in RRMS than in HC; this was caused by a decreased frequency of CD4-CD8- and CD8+ T lymphocytes and was not linked to age. Blocking NKG2D increased the motility of CD8+ T lymphocytes co-cultured with astrocytes expressing NKG2D ligand. Moreover, preventing NKG2D from interacting with its ligands increased the proportion of CD8+ T lymphocytes exhibiting a kinapse-like behavior characterized by short-term interaction while reducing those displaying a long-lasting synapse-like behavior. These results support that NKG2D participates in the establishment of long-term interactions between activated CD8+ T lymphocytes and astrocytes. CONCLUSION Our data demonstrate specific alterations in NKG2D+ T lymphocytes in MS patients' subgroups and suggest that NKG2D contributes to the interactions between human CD8+ T lymphocytes and human astrocytes.
Collapse
Affiliation(s)
- Ana Carmena Moratalla
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Yves Carpentier Solorio
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Florent Lemaître
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Negar Farzam-Kia
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Sandra Da Cal
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Jean Victor Guimond
- CLSC des Faubourgs, CIUSSS du Centre-Sud-de-l'Ile-de-Montréal, Montreal, QC, Canada
| | - Elie Haddad
- Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics, Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Pierre Duquette
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - J Marc Girard
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Alexandre Prat
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Catherine Larochelle
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Nathalie Arbour
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9.
| |
Collapse
|
8
|
Anikeeva N, Steblyanko M, Kuri-Cervantes L, Buggert M, Betts MR, Sykulev Y. The immune synapses reveal aberrant functions of CD8 T cells during chronic HIV infection. Nat Commun 2022; 13:6436. [PMID: 36307445 PMCID: PMC9616955 DOI: 10.1038/s41467-022-34157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 10/14/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic HIV infection causes persistent low-grade inflammation that induces premature aging of the immune system including senescence of memory and effector CD8 T cells. To uncover the reasons of gradually diminished potency of CD8 T cells from people living with HIV, here we expose the T cells to planar lipid bilayers containing ligands for T-cell receptor and a T-cell integrins and analyze the cellular morphology, dynamics of synaptic interface formation and patterns of the cellular degranulation. We find a large fraction of phenotypically naive T cells from chronically infected people are capable to form mature synapse with focused degranulation, a signature of a differentiated T cells. Further, differentiation of aberrant naive T cells may lead to the development of anomalous effector T cells undermining their capacity to control HIV and other pathogens that could be contained otherwise.
Collapse
Affiliation(s)
- Nadia Anikeeva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Steblyanko
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcus Buggert
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Michael R Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuri Sykulev
- Departments of Immunology and Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sydney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Idoate Gastearena MA, López-Janeiro Á, Lecumberri Aznarez A, Arana-Iñiguez I, Guillén-Grima F. A Quantitative Digital Analysis of Tissue Immune Components Reveals an Immunosuppressive and Anergic Immune Response with Relevant Prognostic Significance in Glioblastoma. Biomedicines 2022; 10:biomedicines10071753. [PMID: 35885058 PMCID: PMC9313250 DOI: 10.3390/biomedicines10071753] [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: 05/17/2022] [Revised: 07/10/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives: Immunostimulatory therapies using immune checkpoint blockers show clinical activity in a subset of glioblastoma (GBM) patients. Several inhibitory mechanisms play a relevant role in the immune response to GBM. With the objective of analyzing the tumor immune microenvironment and its clinical significance, we quantified several relevant immune biomarkers. Design: We studied 76 primary (non-recurrent) GBMs with sufficient clinical follow-up, including a subgroup of patients treated with a dendritic cell vaccine. The IDH-mutation, EGFR-amplification, and MGMT methylation statuses were determined. Several relevant immune biomarkers, including CD163, CD8, PD1, and PDL1, were quantified in representative selected areas by digital image analysis and semiquantitative evaluation. The percentage of each immune expression was calculated with respect to the total number of tumor cells. Results: All GBMs were wild-type IDH, with a subgroup of classical GBMs according to the EGFR amplification (44%). Morphologically, CD163 immunostained microglia and intratumor clusters of macrophages were observed. A significant direct correlation was found between the expression of CD8 and the mechanisms of lymphocyte immunosuppression, in such a way that higher values of CD8 were directly associated with higher values of CD163 (p < 0.001), PDL1 (0.026), and PD1 (0.007). In a multivariate analysis, high expressions of CD8+ (HR = 2.05, 95%CI (1.02−4.13), p = 0.034) and CD163+ cells (HR 2.50, 95%CI (1.29−4.85), p = 0.007), were associated with shorter survival durations. The expression of immune biomarkers was higher in the non-classical (non-EGFR amplified tumors) GBMs. Other relevant prognostic factors were age, receipt of the dendritic cell vaccine, and MGMT methylation status. Conclusions: In accordance with the inverse correlation between CD8 and survival and the direct correlation between effector cells and CD163 macrophages and immune-checkpoint expression, we postulate that CD8 infiltration could be placed in a state of anergy or lymphocytic inefficient activity. Furthermore, the significant inverse correlation between CD163 tissue concentration and survival explains the relevance of this type of immune cell when creating a strong immunosuppressive environment. This information may potentially be used to support the selection of patients for immunotherapy.
Collapse
Affiliation(s)
- Miguel A. Idoate Gastearena
- Pathology Department, Clinica Universidad de Navarra and School of Medicine, University of Navarra, 31008 Pamplona, Spain; (Á.L.-J.); (A.L.A.); (I.A.-I.)
- Pathology Department, Virgen Macarena University Hospital and School of Medicine, University of Seville, 41009 Seville, Spain
- Correspondence: ; Tel.: +34-660460714
| | - Álvaro López-Janeiro
- Pathology Department, Clinica Universidad de Navarra and School of Medicine, University of Navarra, 31008 Pamplona, Spain; (Á.L.-J.); (A.L.A.); (I.A.-I.)
| | - Arturo Lecumberri Aznarez
- Pathology Department, Clinica Universidad de Navarra and School of Medicine, University of Navarra, 31008 Pamplona, Spain; (Á.L.-J.); (A.L.A.); (I.A.-I.)
| | - Iñigo Arana-Iñiguez
- Pathology Department, Clinica Universidad de Navarra and School of Medicine, University of Navarra, 31008 Pamplona, Spain; (Á.L.-J.); (A.L.A.); (I.A.-I.)
| | - Francisco Guillén-Grima
- Department of Preventive Medicine, Clinica Universidad de Navarra, University of Navarra, 31008 Pamplona, Spain;
| |
Collapse
|
10
|
Abrams RE, Pierre K, El-Murr N, Seung E, Wu L, Luna E, Mehta R, Li J, Larabi K, Ahmed M, Pelekanou V, Yang ZY, van de Velde H, Stamatelos SK. Quantitative systems pharmacology modeling sheds light into the dose response relationship of a trispecific T cell engager in multiple myeloma. Sci Rep 2022; 12:10976. [PMID: 35768621 PMCID: PMC9243109 DOI: 10.1038/s41598-022-14726-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 06/10/2022] [Indexed: 02/08/2023] Open
Abstract
In relapsed and refractory multiple myeloma (RRMM), there are few treatment options once patients progress from the established standard of care. Several bispecific T-cell engagers (TCE) are in clinical development for multiple myeloma (MM), designed to promote T-cell activation and tumor killing by binding a T-cell receptor and a myeloma target. In this study we employ both computational and experimental tools to investigate how a novel trispecific TCE improves activation, proliferation, and cytolytic activity of T-cells against MM cells. In addition to binding CD3 on T-cells and CD38 on tumor cells, the trispecific binds CD28, which serves as both co-stimulation for T-cell activation and an additional tumor target. We have established a robust rule-based quantitative systems pharmacology (QSP) model trained against T-cell activation, cytotoxicity, and cytokine data, and used it to gain insight into the complex dose response of this drug. We predict that CD3-CD28-CD38 killing capacity increases rapidly in low dose levels, and with higher doses, killing plateaus rather than following the bell-shaped curve typical of bispecific TCEs. We further predict that dose–response curves are driven by the ability of tumor cells to form synapses with activated T-cells. When competition between cells limits tumor engagement with active T-cells, response to therapy may be diminished. We finally suggest a metric related to drug efficacy in our analysis—“effective” receptor occupancy, or the proportion of receptors engaged in synapses. Overall, this study predicts that the CD28 arm on the trispecific antibody improves efficacy, and identifies metrics to inform potency of novel TCEs.
Collapse
Affiliation(s)
- R E Abrams
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.,Daichi Sankyo, 211 Mt. Airy Rd., Basking Ridge, NJ, 07920, USA
| | - K Pierre
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.
| | - N El-Murr
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - E Seung
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | - L Wu
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | | | - J Li
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA
| | - K Larabi
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - M Ahmed
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA
| | - V Pelekanou
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA.,Bayer Pharmaceuticals, Cambridge, MA, 02142, USA
| | - Z-Y Yang
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | - S K Stamatelos
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA. .,Bayer Pharmaceuticals, PH100 Bayer Boulevard, Whippany, NJ, 07981, USA.
| |
Collapse
|
11
|
Braun M, Aguilera AR, Sundarrajan A, Corvino D, Stannard K, Krumeich S, Das I, Lima LG, Meza Guzman LG, Li K, Li R, Salim N, Jorge MV, Ham S, Kelly G, Vari F, Lepletier A, Raghavendra A, Pearson S, Madore J, Jacquelin S, Effern M, Quine B, Koufariotis LT, Casey M, Nakamura K, Seo EY, Hölzel M, Geyer M, Kristiansen G, Taheri T, Ahern E, Hughes BGM, Wilmott JS, Long GV, Scolyer RA, Batstone MD, Landsberg J, Dietrich D, Pop OT, Flatz L, Dougall WC, Veillette A, Nicholson SE, Möller A, Johnston RJ, Martinet L, Smyth MJ, Bald T. CD155 on Tumor Cells Drives Resistance to Immunotherapy by Inducing the Degradation of the Activating Receptor CD226 in CD8 + T Cells. Immunity 2021; 53:805-823.e15. [PMID: 33053330 DOI: 10.1016/j.immuni.2020.09.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/21/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
The activating receptor CD226 is expressed on lymphocytes, monocytes, and platelets and promotes anti-tumor immunity in pre-clinical models. Here, we examined the role of CD226 in the function of tumor-infiltrating lymphocytes (TILs) and resistance to immunotherapy. In murine tumors, a large proportion of CD8+ TILs had decreased surface expression of CD226 and exhibited features of dysfunction, whereas CD226hi TILs were highly functional. This correlation was seen also in TILs isolated from HNSCC patients. Mutation of CD226 at tyrosine 319 (Y319) led to increased CD226 surface expression, enhanced anti-tumor immunity and improved efficacy of immune checkpoint blockade (ICB). Mechanistically, tumor-derived CD155, the ligand for CD226, initiated phosphorylation of Y319 by Src kinases, thereby enabling ubiquitination of CD226 by CBL-B, internalization, and proteasomal degradation. In pre-treatment samples from melanoma patients, CD226+CD8+ T cells correlated with improved progression-free survival following ICB. Our findings argue for the development of therapies aimed at maintaining the expression of CD226.
Collapse
Affiliation(s)
- Matthias Braun
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Amelia Roman Aguilera
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ashmitha Sundarrajan
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Dillon Corvino
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kimberley Stannard
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sophie Krumeich
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Indrajit Das
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Luize G Lima
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Lizeth G Meza Guzman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Kunlun Li
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Rui Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC, Canada
| | - Nazhifah Salim
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Maria Villancanas Jorge
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sunyoung Ham
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Gabrielle Kelly
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Frank Vari
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ailin Lepletier
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Ashwini Raghavendra
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sally Pearson
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Jason Madore
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Sebastien Jacquelin
- Gordon and Jessie Gilmour Leukemia Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Maike Effern
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany; Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC, Australia
| | - Brodie Quine
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Lambros T Koufariotis
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Mika Casey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Eun Y Seo
- Immuno-Oncology Discovery, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Touraj Taheri
- Pathology Queensland, Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Elizabeth Ahern
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; The University of Sydney, Central Clinical School, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; The University of Sydney, Central Clinical School, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Martin D Batstone
- Royal Brisbane and Women's Hospital, University of Queensland Herston, Herston, QLD, Australia
| | - Jennifer Landsberg
- Department of Dermatology and Allergy, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Dimo Dietrich
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Oltin T Pop
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, St.Gallen, Switzerland; Department of Dermatology, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - William C Dougall
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC, Canada; Department of Medicine, University of Montréal, Montréal, QC, Canada
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andreas Möller
- Tumor Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Robert J Johnston
- Immuno-Oncology Discovery, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Ludovic Martinet
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse F-31000, France
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.
| | - Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.
| |
Collapse
|
12
|
Friedman D, Simmonds P, Hale A, Bere L, Hodson NW, White MRH, Davis DM. Natural killer cell immune synapse formation and cytotoxicity are controlled by tension of the target interface. J Cell Sci 2021; 134:jcs258570. [PMID: 33712452 PMCID: PMC8077183 DOI: 10.1242/jcs.258570] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells can kill infected or transformed cells via a lytic immune synapse. Diseased cells may exhibit altered mechanical properties but how this impacts NK cell responsiveness is unknown. We report that human NK cells were stimulated more effectively to secrete granzymes A and B, FasL (also known as FasLG), granulysin and IFNγ, by stiff (142 kPa) compared to soft (1 kPa) planar substrates. To create surrogate spherical targets of defined stiffness, sodium alginate was used to synthesise soft (9 kPa), medium (34 kPa) or stiff (254 kPa) cell-sized beads, coated with antibodies against activating receptor NKp30 (also known as NCR3) and the integrin LFA-1 (also known as ITGAL). Against stiff beads, NK cells showed increased degranulation. Polarisation of the microtubule-organising centre and lytic granules were impaired against soft targets, which instead resulted in the formation of unstable kinapses. Thus, by varying target stiffness to characterise the mechanosensitivity of immune synapses, we identify soft targets as a blind spot in NK cell recognition. This article has an associated First Person interview with the co-first authors of the paper.
Collapse
Affiliation(s)
- Daniel Friedman
- The Lydia Becker Institute, Faculty of Biology, Medicine and Health, University of Manchester, Core Technology Facility building, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| | - Poppy Simmonds
- The Lydia Becker Institute, Faculty of Biology, Medicine and Health, University of Manchester, Core Technology Facility building, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| | - Alexander Hale
- The Lydia Becker Institute, Faculty of Biology, Medicine and Health, University of Manchester, Core Technology Facility building, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| | - Leoma Bere
- The Lydia Becker Institute, Faculty of Biology, Medicine and Health, University of Manchester, Core Technology Facility building, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| | - Nigel W. Hodson
- BioAFM Facility, Faculty of Biology, Medicine and Health, Stopford Building, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Michael R. H. White
- Michael Smith Building, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Daniel M. Davis
- The Lydia Becker Institute, Faculty of Biology, Medicine and Health, University of Manchester, Core Technology Facility building, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| |
Collapse
|
13
|
Cribaro GP, Saavedra-López E, Romarate L, Mitxitorena I, Díaz LR, Casanova PV, Roig-Martínez M, Gallego JM, Perez-Vallés A, Barcia C. Three-dimensional vascular microenvironment landscape in human glioblastoma. Acta Neuropathol Commun 2021; 9:24. [PMID: 33579378 PMCID: PMC7879533 DOI: 10.1186/s40478-020-01115-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
The cellular complexity of glioblastoma microenvironments is still poorly understood. In-depth, cell-resolution tissue analyses of human material are rare but highly necessary to understand the biology of this deadly tumor. Here we present a unique 3D visualization revealing the cellular composition of human GBM in detail and considering its critical association with the neo-vascular niche. Our images show a complex vascular map of human 3D biopsies with increased vascular heterogeneity and altered spatial relationship with astrocytes or glioma-cell counterparts. High-resolution analysis of the structural layers of the blood brain barrier showed a multilayered fenestration of endothelium and basement membrane. Careful examination of T cell position and migration relative to vascular walls revealed increased infiltration corresponding with tumor proliferation. In addition, the analysis of the myeloid landscape not only showed a volumetric increase in glioma-associated microglia and macrophages relative to GBM proliferation but also revealed distinct phenotypes in tumor nest and stroma. Images and data sets are available on demand as a resource for public access.
Collapse
|
14
|
Hwang SH, Yeom H, Han BI, Ham BJ, Lee YM, Han MR, Lee M. Predicting Carcinogenic Mechanisms of Non-Genotoxic Carcinogens via Combined Analysis of Global DNA Methylation and In Vitro Cell Transformation. Int J Mol Sci 2020; 21:ijms21155387. [PMID: 32751172 PMCID: PMC7432388 DOI: 10.3390/ijms21155387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/17/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
An in vitro cell transformation assay (CTA) is useful for the detection of non-genotoxic carcinogens (NGTXCs); however, it does not provide information on their modes of action. In this study, to pursue a mechanism-based approach in the risk assessment of NGTXCs, we aimed to develop an integrated strategy comprising an in vitro Bhas 42 CTA and global DNA methylation analysis. For this purpose, 10 NGTXCs, which were also predicted to be negative through Derek/Sarah structure-activity relationship analysis, were first tested for transforming activity in Bhas 42 cells. Methylation profiles using reduced representation bisulfite sequencing were generated for seven NGTXCs that were positive in CTAs. In general, the differentially methylated regions (DMRs) within promoter regions showed slightly more bias toward hypermethylation than the DMRs across the whole genome. We also identified 13 genes associated with overlapping DMRs within the promoter regions in four NGTXCs, of which seven were hypermethylated and six were hypomethylated. Using ingenuity pathway analysis, the genes with DMRs at the CpG sites were found to be enriched in cancer-related categories, including "cell-to-cell signaling and interaction" as well as "cell death and survival". Moreover, the networks related to "cell death and survival", which were considered to be associated with carcinogenesis, were identified in six NGTXCs. These results suggest that epigenetic changes supporting cell transformation processes occur during non-genotoxic carcinogenesis. Taken together, our combined system can become an attractive component for an integrated approach for the testing and assessment of NGTXCs.
Collapse
Affiliation(s)
- Sung-Hee Hwang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (S.-H.H.); (H.Y.)
| | - Hojin Yeom
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (S.-H.H.); (H.Y.)
| | - Byeal-I Han
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea;
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea;
| | - Yong-Moon Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheoungju-si, Chungcheongbuk-do 28160, Korea;
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (S.-H.H.); (H.Y.)
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea;
- INU Human Genome Center, Incheon National University, Incheon 22012, Korea
- Correspondence: (M.-R.H.); (M.L.); Tel.: +82-32-835-8247 (M.L.)
| | - Michael Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (S.-H.H.); (H.Y.)
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea;
- INU Human Genome Center, Incheon National University, Incheon 22012, Korea
- Correspondence: (M.-R.H.); (M.L.); Tel.: +82-32-835-8247 (M.L.)
| |
Collapse
|
15
|
Chabaud M, Paillon N, Gaus K, Hivroz C. Mechanobiology of antigen‐induced T cell arrest. Biol Cell 2020; 112:196-212. [DOI: 10.1111/boc.201900093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/19/2020] [Accepted: 03/29/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Mélanie Chabaud
- Institut Curie‐PSL Research University INSERM U932 Paris France
- EMBL Australia Node in Single Molecule Science, School of Medical SciencesUniversity of New South Wales Sydney NSW Australia
- ARC Centre of Excellence in Advanced Molecular ImagingUniversity of New South Wales Sydney NSW Australia
| | - Noémie Paillon
- Institut Curie‐PSL Research University INSERM U932 Paris France
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical SciencesUniversity of New South Wales Sydney NSW Australia
- ARC Centre of Excellence in Advanced Molecular ImagingUniversity of New South Wales Sydney NSW Australia
| | - Claire Hivroz
- Institut Curie‐PSL Research University INSERM U932 Paris France
| |
Collapse
|
16
|
Saavedra-López E, Roig-Martínez M, Cribaro GP, Casanova PV, Gallego JM, Pérez-Vallés A, Barcia C. Phagocytic glioblastoma-associated microglia and macrophages populate invading pseudopalisades. Brain Commun 2019; 2:fcz043. [PMID: 32954312 PMCID: PMC7491442 DOI: 10.1093/braincomms/fcz043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 09/02/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxic pseudopalisades are a pathological hallmark of human glioblastoma, which is linked to tumour malignancy and aggressiveness. Yet, their function and role in the tumour development have scarcely been explored. It is thought that pseudopalisades are formed by malignant cells escaping from the hypoxic environment, although evidence of the immune component of pseudopalisades has been elusive. In the present work, we analyse the immunological constituent of hypoxic pseudopalisades using high-resolution three-dimensional confocal imaging in tissue blocks from excised tumours of glioblastoma patients and mimic the hypoxic gradient in microfluidic platforms in vitro to understand the cellular motility. We visualize that glioblastoma-associated microglia and macrophages abundantly populate pseudopalisades, displaying an elongated kinetic morphology across the pseudopalisades, and are oriented towards the necrotic focus. In vitro experiments demonstrate that under hypoxic gradient, microglia show a particular motile behaviour characterized by the increase of cellular persistence in contrast with glioma cells. Importantly, we show that glioblastoma-associated microglia and macrophages utilize fibres of glioma cells as a haptotactic cue to navigate along the anisotropic structure of the pseudopalisades and display a high phagocytic activity at the necrotic border of the pseudopalisades. In this study, we demonstrate that glioblastoma-associated microglia and macrophages are the main immune cells of pseudopalisades in glioblastoma, travelling to necrotic areas to clear the resulting components of the prothrombotic milieu, suggesting that the scavenging features of glioblastoma-associated microglia and macrophages at the pseudopalisades serve as an essential counterpart for glioma cell invasion.
Collapse
Affiliation(s)
- Elena Saavedra-López
- Neuroimmunity Research Group, Department of Biochemistry and Molecular Biology, School of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Meritxell Roig-Martínez
- Neuroimmunity Research Group, Department of Biochemistry and Molecular Biology, School of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - George P Cribaro
- Neuroimmunity Research Group, Department of Biochemistry and Molecular Biology, School of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Paola V Casanova
- Neuroimmunity Research Group, Department of Biochemistry and Molecular Biology, School of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - José M Gallego
- Department of Neurosurgery, Valencia General Hospital, Valencia 46014, Spain
| | - Ana Pérez-Vallés
- Department of Pathology, Valencia General Hospital, Valencia 46014, Spain
| | - Carlos Barcia
- Neuroimmunity Research Group, Department of Biochemistry and Molecular Biology, School of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| |
Collapse
|
17
|
Abstract
T cells effectively explore the tissue in search for antigens. When activated, they dedicate a big amount of energy and resources to arrange a complex structure called immunological synapse (IS), containing a particular distribution of molecules defined as supramolecular activation clusters (SMACs), and become polarized toward the target cell in a manner that channels the information specifically. This arrangement is symmetrical and requires the polarization of the MTOC and the Golgi to be operational, especially for the proper delivery of lytic granules and the recycling of molecules three dimensionally segregated at the clustered interface. Alternatively, after the productive encounter, T cells need to rearrange again to newly navigate through the tissue, changing back to a motile state called immunological kinapse (IK). In this IK state, the MTOC and the Golgi apparatus are repositioned and recruited at the back of the T cell to facilitate motility, while the established symmetry of the elements of the SMACs is broken and distributed in a different pattern. Both states, IS and IK, are interchangeable and are mainly orchestrated by the MTOC/Golgi complex, being critical for an effective immune response.
Collapse
|