1
|
Maire K, Chamy L, Ghazali S, Carratala-Lasserre M, Zahm M, Bouisset C, Métais A, Combes-Soia L, de la Fuente-Vizuete L, Trad H, Chaubet A, Savignac M, Gonzalez de Peredo A, Subramaniam A, Joffre O, Lutz PG, Lamsoul I. Fine-tuning levels of filamins a and b as a specific mechanism sustaining Th2 lymphocyte functions. Nat Commun 2024; 15:10574. [PMID: 39639023 PMCID: PMC11621393 DOI: 10.1038/s41467-024-53768-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Augmenting the portfolio of therapeutics for type 2-driven diseases is crucial to address unmet clinical needs and to design personalized treatment schemes. An attractive therapy for such diseases would consist in targeting the recruitment of T helper 2 (Th2) lymphocytes to inflammatory sites. Herein, we show the degradation of filamins (FLN) a and b by the ASB2α E3 ubiquitin ligase as a mechanism sustaining Th2 lymphocyte functions. Low levels of FLNa and FLNb confer an elongated shape to Th2 lymphocytes associated with efficient αVβ3 integrin-dependent cell migration. Genes encoding the αVβ3 integrin and ASB2α belong to the core of Th2-specific genes. Using genetically modified mice, we find that increasing the levels of FLNa and FLNb in Th2 lymphocytes reduces airway inflammation through diminished Th2 lymphocyte recruitment in inflamed lungs. Collectively, our results highlight ASB2α and its substrates FLNa and FLNb to alter Th2 lymphocyte-mediated responses.
Collapse
Affiliation(s)
- Kilian Maire
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Léa Chamy
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Samira Ghazali
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | | | - Margot Zahm
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Clément Bouisset
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Arnaud Métais
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lucie Combes-Soia
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Hussein Trad
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Adeline Chaubet
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Magali Savignac
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Arun Subramaniam
- Sanofi Immunology and Inflammation Research Therapeutic Area, Cambridge, MA, USA
| | - Olivier Joffre
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France
| | - Pierre G Lutz
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France.
| | - Isabelle Lamsoul
- Infinity, University of Toulouse, CNRS, Inserm, UPS, Toulouse, France.
| |
Collapse
|
2
|
Caillier A, Oleksyn D, Fowell DJ, Miller J, Oakes PW. T cells use focal adhesions to pull themselves through confined environments. J Cell Biol 2024; 223:e202310067. [PMID: 38889096 PMCID: PMC11187980 DOI: 10.1083/jcb.202310067] [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: 10/16/2023] [Revised: 05/16/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Immune cells are highly dynamic and able to migrate through environments with diverse biochemical and mechanical compositions. Their migration has classically been defined as amoeboid under the assumption that it is integrin independent. Here, we show that activated primary Th1 T cells require both confinement and extracellular matrix proteins to migrate efficiently. This migration is mediated through small and dynamic focal adhesions that are composed of the same proteins associated with canonical mesenchymal cell focal adhesions, such as integrins, talin, and vinculin. These focal adhesions, furthermore, localize to sites of contractile traction stresses, enabling T cells to pull themselves through confined spaces. Finally, we show that Th1 T cells preferentially follow tracks of other T cells, suggesting that these adhesions modify the extracellular matrix to provide additional environmental guidance cues. These results demonstrate not only that the boundaries between amoeboid and mesenchymal migration modes are ambiguous, but that integrin-mediated focal adhesions play a key role in T cell motility.
Collapse
Affiliation(s)
- Alexia Caillier
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - David Oleksyn
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Deborah J. Fowell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jim Miller
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Patrick W. Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
3
|
Ende K, Santos F, Guasch J, Kemkemer R. Migration of human T cells can be differentially directed by electric fields depending on the extracellular microenvironment. iScience 2024; 27:109746. [PMID: 38706849 PMCID: PMC11067362 DOI: 10.1016/j.isci.2024.109746] [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: 09/29/2023] [Revised: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
T cell migration plays an essential role in the immune response and T cell-based therapies. It can be modulated by chemical and physical cues such as electric fields (EFs). The mechanisms underlying electrotaxis (cell migration manipulated by EFs) are not fully understood and systematic studies with immune cells are rare. In this in vitro study, we show that direct current EFs with strengths of physiologically occurring EFs (25-200 mV/mm) can guide the migration of primary human CD4+ and CD8+ T cells on 2D substrates toward the anode and in a 3D environment differentially (CD4+ T cells show cathodal and CD8+ T cells show anodal electrotaxis). Overall, we find that EFs present a potent stimulus to direct T cell migration in different microenvironments in a cell-type-, substrate-, and voltage-dependent manner, while not significantly influencing T cell differentiation or viability.
Collapse
Affiliation(s)
- Karen Ende
- Reutlingen Research Institute and School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
| | - Fabião Santos
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain
- Dynamic Biomimetics for Cancer Immunotherapy, Max Planck Partner Group, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Judith Guasch
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain
- Dynamic Biomimetics for Cancer Immunotherapy, Max Planck Partner Group, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Ralf Kemkemer
- Reutlingen Research Institute and School of Life Sciences, Reutlingen University, 72762 Reutlingen, Germany
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Lyons-Cohen MR, Shamskhou EA, Gerner MY. Site-specific regulation of Th2 differentiation within lymph node microenvironments. J Exp Med 2024; 221:e20231282. [PMID: 38442268 PMCID: PMC10912907 DOI: 10.1084/jem.20231282] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
T helper 2 (Th2) responses protect against pathogens while also driving allergic inflammation, yet how large-scale Th2 responses are generated in tissue context remains unclear. Here, we used quantitative imaging to investigate early Th2 differentiation within lymph nodes (LNs) following cutaneous allergen administration. Contrary to current models, we observed extensive activation and "macro-clustering" of early Th2 cells with migratory type-2 dendritic cells (cDC2s), generating specialized Th2-promoting microenvironments. Macro-clustering was integrin-mediated and promoted localized cytokine exchange among T cells to reinforce differentiation, which contrasted the behavior during Th1 responses. Unexpectedly, formation of Th2 macro-clusters was dependent on the site of skin sensitization. Differences between sites were driven by divergent activation states of migratory cDC2 from different dermal tissues, with enhanced costimulatory molecule expression by cDC2 in Th2-generating LNs promoting prolonged T cell activation, macro-clustering, and cytokine sensing. Thus, the generation of dedicated Th2 priming microenvironments through enhanced costimulatory molecule signaling initiates Th2 responses in vivo and occurs in a skin site-specific manner.
Collapse
Affiliation(s)
- Miranda R. Lyons-Cohen
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Elya A. Shamskhou
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Michael Y. Gerner
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Fierro Morales JC, Redfearn C, Titus MA, Roh-Johnson M. Reduced PaxillinB localization to cell-substrate adhesions promotes cell migration in Dictyostelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585764. [PMID: 38562712 PMCID: PMC10983970 DOI: 10.1101/2024.03.19.585764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Many cells adhere to extracellular matrix for efficient cell migration. This adhesion is mediated by focal adhesions, a protein complex linking the extracellular matrix to the intracellular cytoskeleton. Focal adhesions have been studied extensively in mesenchymal cells, but recent research in physiological contexts and amoeboid cells suggest focal adhesion regulation differs from the mesenchymal focal adhesion paradigm. We used Dictyostelium discoideum to uncover new mechanisms of focal adhesion regulation, as Dictyostelium are amoeboid cells that form focal adhesion-like structures for migration. We show that PaxillinB, the Dictyostelium homologue of Paxillin, localizes to dynamic focal adhesion-like structures during Dictyostelium migration. Unexpectedly, reduced PaxillinB recruitment to these structures increases Dictyostelium cell migration. Quantitative analysis of focal adhesion size and dynamics show that lack of PaxillinB recruitment to focal adhesions does not alter focal adhesion size, but rather increases focal adhesion turnover. These findings are in direct contrast to Paxillin function at focal adhesions during mesenchymal migration, challenging the established focal adhesion model.
Collapse
Affiliation(s)
| | - Chandler Redfearn
- Department of Kinesiology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Minna Roh-Johnson
- Department of Biochemistry, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Kinesiology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Zhou M, Feng Y, Zhang X, Chen J, Yao N, Fu S, Ni T, Chen Y, Xie F, Roy S, Liu J, Yang Y, He Y, Zhao Y, Yang N. Platelet-derived microparticles adoptively transfer integrin β3 to promote antitumor effect of tumor-infiltrating T cells. Oncoimmunology 2024; 13:2304963. [PMID: 38235317 PMCID: PMC10793703 DOI: 10.1080/2162402x.2024.2304963] [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: 08/23/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
Approximately two-thirds of hepatocellular carcinoma (HCC) is considered a "cold tumor" characterized by few tumor-infiltrating T cells and an abundance of immunosuppressive cells. Cilengitide, an integrin αvβ3 inhibitor, has failed in clinical trials as a potential anticancer drug. This failure implies that integrin αvβ3 may play an important role in immune cells. However, the expression and potential role of integrin αvβ3 in T cells of HCC patients remain unknown. Here, we established two HCC models and found that cilengitide had a dual effect on the HCC microenvironment by exerting both antitumor effect and immunosuppressive effect on T cells. This may partly explain the failure of cilengitide in clinical trials. In clinical specimens, HCC-infiltrating T cells exhibited deficient expression and activation of integrin β3, which was associated with poor T-cell infiltration into tumors. Additionally, integrin β3 functioned as a positive immunomodulatory molecule to facilitate T-cell infiltration and T helper 1-type immune response in vitro. Furthermore, T cells and platelet-derived microparticles (PMPs) co-culture assay revealed that PMPs adoptively transferred integrin β3 to T cells and positively regulated T cell immune response. This process was mediated by clathrin-dependent endocytosis and macropinocytosis. Our data demonstrate that integrin β3 deficiency on HCC-infiltrating T cells may be involved in shaping the immunosuppressive tumor microenvironment. PMPs transfer integrin β3 to T cells and positively regulate T cell immune response, which may provide a new insight into immune therapy of HCC.
Collapse
Affiliation(s)
- Mimi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yali Feng
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaoli Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jianguo Chen
- School of Software Engineering, Sun Yat-Sen University, Zhuhai, China
| | - Naijuan Yao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shan Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Tianzhi Ni
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yi Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fei Xie
- Health Science Center, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Sahasrabda Roy
- School of International Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jinfeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuan Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yingli He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yingren Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Nan Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institution of Hepatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Clinical Research Center for Infectious Diseases, Xi’an, Shaanxi, China
- Clinical Research Center for Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
7
|
Caillier A, Oleksyn D, Fowell DJ, Miller J, Oakes PW. T cells Use Focal Adhesions to Pull Themselves Through Confined Environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562587. [PMID: 37904911 PMCID: PMC10614902 DOI: 10.1101/2023.10.16.562587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Immune cells are highly dynamic and able to migrate through environments with diverse biochemical and mechanical composition. Their migration has classically been defined as amoeboid under the assumption that it is integrin-independent. Here we show that activated primary Th1 T cells require both confinement and extracellular matrix protein to migrate efficiently. This migration is mediated through small and dynamic focal adhesions that are composed of the same proteins associated with canonical mesenchymal focal adhesions, such as integrins, talin, and vinculin. These focal adhesions, furthermore, localize to sites of contractile traction stresses, enabling T cells to pull themselves through confined spaces. Finally, we show that Th1 T cell preferentially follows tracks of other T cells, suggesting that these adhesions are modifying the extracellular matrix to provide additional environmental guidance cues. These results demonstrate not only that the boundaries between amoeboid and mesenchymal migration modes are ambiguous, but that integrin-mediated adhesions play a key role in T cell motility.
Collapse
Affiliation(s)
- Alexia Caillier
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - David Oleksyn
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah J Fowell
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Jim Miller
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Patrick W Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| |
Collapse
|
8
|
Lyons-Cohen MR, Shamskhou EA, Gerner MY. Prolonged T cell - DC macro-clustering within lymph node microenvironments initiates Th2 cell differentiation in a site-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.547554. [PMID: 37461439 PMCID: PMC10350056 DOI: 10.1101/2023.07.07.547554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Formation of T helper 2 (Th2) responses has been attributed to low-grade T cell stimulation, yet how large-scale polyclonal Th2 responses are generated in vivo remains unclear. Here, we used quantitative imaging to investigate early Th2 differentiation within lymph nodes (LNs) following cutaneous allergen administration. Contrary to current models, Th2 differentiation was associated with enhanced T cell activation and extensive integrin-dependent 'macro-clustering' at the T-B border, which also contrasted clustering behavior seen during Th1 differentiation. Unexpectedly, formation of Th2 macro-clusters within LNs was highly dependent on the site of skin sensitization. Differences between sites were driven by divergent activation states of migratory cDC2 from different dermal tissues, with enhanced costimulatory molecule expression by cDC2 in Th2-generating LNs promoting T cell macro-clustering and cytokine sensing. Thus, generation of dedicated priming micro-environments through enhanced costimulatory molecule signaling initiates the generation of Th2 responses in vivo and occurs in a skin site-specific manner.
Collapse
Affiliation(s)
| | - Elya A. Shamskhou
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael Y. Gerner
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
9
|
Su H, Jin Y, Tao C, Yang H, Yang E, Zhang WG, Feng F. Th2 cells infiltrating high-grade serous ovarian cancer: a feature that may account for the poor prognosis. J Gynecol Oncol 2023:34.e48. [PMID: 36998223 DOI: 10.3802/jgo.2023.34.e48] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 12/17/2022] [Accepted: 01/18/2023] [Indexed: 04/01/2023] Open
Abstract
OBJECTIVE We aimed to investigate the differences of transcriptome profile between 2 groups of high-grade serous ovarian cancer (HGSOC) patients with distinct outcomes and identify potential biomarkers for recurrence. METHODS RNA sequencing was performed in 2 groups of HGSOC patients with similar demographic characteristics but exhibiting distinct progression-free survival (PFS). Transcriptome data of poor response (PR; PFS ≤6 months) and good response (GR; PFS ≥12 months) group were compared. We employed xCell to evaluate the abundance of 63 cells in tumor microenvironment. The predictive value of recurrence-related tumor infiltration cells was validated in cohort data from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) dataset. The weighted correlation network analysis was performed to identify the genes related to cell infiltration. RESULTS PR patients exhibited a distinct tumor infiltration immune cells-related transcriptional profile compared to GR patients, such as lower signatures of leukocyte differentiation, activation and chemotaxis. The fraction of T-helper 2 (Th2) cells infiltration was significantly higher in PR group than in GR group. High infiltration of Th2 was significantly associated with unfavorable prognosis in the GEO cohort (area under the curve=0.84 at 6 months recurrence) and TCGA cohort (p=0.008). Genes enriched to extracellular matrix organization and integrin binding were relevant to Th2 infiltration. CONCLUSION Patients with HGSOC having shorter PFS exhibited a distinct gene signature that related to tumor-infiltrating immune cells. The level of Th2 infiltration could facilitate patient recurrence risk stratification and may be a promising biomarker for prognosis prediction and immune-related treatment.
Collapse
Affiliation(s)
- Hao Su
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yueqi Jin
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Changyu Tao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hua Yang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ence Yang
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wei-Guang Zhang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| | - Fengzhi Feng
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Recent Emerging Immunological Treatments for Primary Brain Tumors: Focus on Chemokine-Targeting Immunotherapies. Cells 2023; 12:cells12060841. [PMID: 36980182 PMCID: PMC10046911 DOI: 10.3390/cells12060841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Primary brain tumors are a leading cause of death worldwide and are characterized by extraordinary heterogeneity and high invasiveness. Current drug and radiotherapy therapies combined with surgical approaches tend to increase the five-year survival of affected patients, however, the overall mortality rate remains high, thus constituting a clinical challenge for which the discovery of new therapeutic strategies is needed. In this field, novel immunotherapy approaches, aimed at overcoming the complex immunosuppressive microenvironment, could represent a new method of treatment for central nervous system (CNS) tumors. Chemokines especially are a well-defined group of proteins that were so named due to their chemotactic properties of binding their receptors. Chemokines regulate the recruitment and/or tissue retention of immune cells as well as the mobilization of tumor cells that have undergone epithelial–mesenchymal transition, promoting tumor growth. On this basis, this review focuses on the function and involvement of chemokines and their receptors in primary brain tumors, specifically examining chemokine-targeting immunotherapies as one of the most promising strategies in neuro-oncology.
Collapse
|
11
|
Li Z, Cheng L, Zhan H, Li Y. Chemokines and chemokine receptors in Behçet's disease. Front Immunol 2023; 14:1109147. [PMID: 36742301 PMCID: PMC9889923 DOI: 10.3389/fimmu.2023.1109147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Behçet's disease (BD), a chronic vascular inflammatory disease, is characterized by the symptoms of ocular lesions, recurrent genital and oral ulcers, skin symptoms and arthritis in addition to neurological, intestinal and vascular involvement. The pathogenesis of BD is poorly understood, and there are no effective laboratory markers for the diagnosis of BD. In addition, BD is presently incurable. Chemokines, a family of small secreted chemotactic cytokines, interact with chemokine receptors and mediate the migration, localization and cellular interactions of inflammatory cells. Several studies have suggested that chemokines and their receptors play an important role in the occurrence and development of BD and that these chemokines along with their receptors can be utilized as biomarkers and therapeutic targets. In the present review, chemokines and chemokine receptors involved in BD and their potential application in diagnosis and therapy have been discussed.
Collapse
Affiliation(s)
- Zhan Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Szeto ACH, Ferreira ACF, Mannion J, Clark PA, Sivasubramaniam M, Heycock MWD, Crisp A, Jolin HE, Kozik P, Knolle MD, McKenzie ANJ. An αvβ3 integrin checkpoint is critical for efficient T H2 cell cytokine polarization and potentiation of antigen-specific immunity. Nat Immunol 2023; 24:123-135. [PMID: 36550322 DOI: 10.1038/s41590-022-01378-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/03/2022] [Indexed: 12/24/2022]
Abstract
Naive CD4+ T lymphocytes initially undergo antigen-specific activation to promote a broad-spectrum response before adopting bespoke cytokine expression profiles shaped by intercellular microenvironmental cues, resulting in pathogen-focused modular cytokine responses. Interleukin (IL)-4-induced Gata3 upregulation is important for the helper type 2 T cell (TH2 cell) polarization associated with anti-helminth immunity and misdirected allergic inflammation. Whether additional microenvironmental factors participate is unclear. Using whole mouse-genome CRISPR-Cas9 screens, we discovered a previously unappreciated role for αvβ3 integrin in TH2 cell differentiation. Low-level αvβ3 expression by naive CD4+ T cells contributed to pan-T cell activation by promoting T-T cell clustering and IL-2/CD25/STAT5 signaling. Subsequently, IL-4/Gata3-induced selective upregulation of αvβ3 licensed intercellular αvβ3-Thy1 interactions among TH2 cells, enhanced mammalian target of rapamycin (mTOR) signaling, supported differentiation and promoted IL-5/IL-13 production. In mice, αvβ3 was required for efficient, allergen-driven, antigen-specific lung TH2 cell responses. Thus, αvβ3-expressing TH2 cells form multicellular factories to propagate and amplify TH2 cell responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Martin D Knolle
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Cambridge University Hospitals, Cambridge, UK
| | | |
Collapse
|
13
|
Thompson SB, Waldman MM, Jacobelli J. Polymerization power: effectors of actin polymerization as regulators of T lymphocyte migration through complex environments. FEBS J 2022; 289:6154-6171. [PMID: 34273243 PMCID: PMC8761786 DOI: 10.1111/febs.16130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/24/2021] [Accepted: 07/16/2021] [Indexed: 11/26/2022]
Abstract
During their life span, T cells are tasked with patrolling the body for potential pathogens. To do so, T cells migrate through numerous distinct anatomical sites and tissue environments with different biophysical characteristics. To migrate through these different environments, T cells use various motility strategies that rely on actin network remodeling to generate shape changes and mechanical forces. In this review, we initially discuss the migratory journey of T cells and then cover the actin polymerization effectors at play in T cells, and finally, we focus on the function of these effectors of actin cytoskeleton remodeling in mediating T-cell migration through diverse tissue environments. Specifically, we will discuss the current state of the field pertaining to our understanding of the roles in T-cell migration played by members of the three main families of actin polymerization machinery: the Arp2/3 complex; formin proteins; and Ena/VASP proteins.
Collapse
Affiliation(s)
- Scott B. Thompson
- Department of Immunology and Microbiology, University of Colorado School of Medicine
| | - Monique M. Waldman
- Department of Immunology and Microbiology, University of Colorado School of Medicine
- Barbara Davis Research Center, University of Colorado School of Medicine
| | - Jordan Jacobelli
- Department of Immunology and Microbiology, University of Colorado School of Medicine
- Barbara Davis Research Center, University of Colorado School of Medicine
| |
Collapse
|
14
|
Yun L, Li W, Wu T, Zhang M. Effect of sea cucumber peptides on the immune response and gut microbiota composition in ovalbumin-induced allergic mice. Food Funct 2022; 13:6338-6349. [PMID: 35612003 DOI: 10.1039/d2fo00536k] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The prevalence of food allergies has increased in Asian countries. The aim of this study was to determine the potential value of sea cucumber peptide (SCP) for anti-allergic therapeutics in terms of their effect on immune response and gut microbiota composition. Results exhibited that SCP could significantly improve the allergy symptoms caused by ovalbumin and could reduce the risk of IgE mediated allergic disorders, as well as repair the morphological damage in the colon. Flow cytometry analysis indicated that SCP could improve the ratio of CD4+/CD8+ T lymphocytes. 16S rRNA results indicated that SCP could differently impact the composition of microbiota. The relative abundances of Bacteroidetes and Firmicutes and the Bacteroidetes/Firmicutes ratio were altered in normal mice. When compared with the OVA treated group, the SCP treated groups showed an increase in the relative abundance of Lachnospiraceae, Muribaculaceae and Ruminococcaceae, and a decrease in Bacteroidaceae, Prevotellaceae, and Lactobacillaceae. These results demonstrate that SCP exhibits potential antiallergic activities in a mouse model of ovalbumin allergy by regulating intestinal microbiota diversity and upregulating the immune response of T lymphocyte subpopulations, which might provide important evidence that SCP can be developed into a novel functional food for inhibiting ovalbumin allergy.
Collapse
Affiliation(s)
- Liyuan Yun
- China-Russia Agricultural Processing Joint Laboratory, Tianjin Agricultural University, Tianjin 300392, P. R. China.
| | - Wen Li
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Min Zhang
- China-Russia Agricultural Processing Joint Laboratory, Tianjin Agricultural University, Tianjin 300392, P. R. China. .,State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
15
|
Goncalves P, Doisne JM, Eri T, Charbit B, Bondet V, Posseme C, Llibre A, Casrouge A, Lenoir C, Neven B, Duffy D, Fischer A, Di Santo JP. Defects in mucosal immunity and nasopharyngeal dysbiosis in HSC-transplanted SCID patients with IL2RG/JAK3 deficiency. Blood 2022; 139:2585-2600. [PMID: 35157765 PMCID: PMC11022929 DOI: 10.1182/blood.2021014654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Both innate and adaptive lymphocytes have critical roles in mucosal defense that contain commensal microbial communities and protect against pathogen invasion. Here we characterize mucosal immunity in patients with severe combined immunodeficiency (SCID) receiving hematopoietic stem cell transplantation (HSCT) with or without myeloablation. We confirmed that pretransplant conditioning had an impact on innate (natural killer and innate lymphoid cells) and adaptive (B and T cells) lymphocyte reconstitution in these patients with SCID and now show that this further extends to generation of T helper 2 and type 2 cytotoxic T cells. Using an integrated approach to assess nasopharyngeal immunity, we identified a local mucosal defect in type 2 cytokines, mucus production, and a selective local immunoglobulin A (IgA) deficiency in HSCT-treated SCID patients with genetic defects in IL2RG/GC or JAK3. These patients have a reduction in IgA-coated nasopharyngeal bacteria and exhibit microbial dysbiosis with increased pathobiont carriage. Interestingly, intravenous immunoglobulin replacement therapy can partially normalize nasopharyngeal immunoglobulin profiles and restore microbial communities in GC/JAK3 patients. Together, our results suggest a potential nonredundant role for type 2 immunity and/or of local IgA antibody production in the maintenance of nasopharyngeal microbial homeostasis and mucosal barrier function.
Collapse
Affiliation(s)
- Pedro Goncalves
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Toshiki Eri
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Bruno Charbit
- Institut Pasteur, Université de Paris Cité, Center for Translational Science, Paris, France
| | - Vincent Bondet
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Celine Posseme
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Alba Llibre
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Armanda Casrouge
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Christelle Lenoir
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Imagine Institut, Université de Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Bénédicte Neven
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Darragh Duffy
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Alain Fischer
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Collège de France, Paris, France
| | - James P. Di Santo
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - The Milieu Intérieur Consortium
- Institut Pasteur, Université de Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
- Institut Pasteur, Université de Paris Cité, Center for Translational Science, Paris, France
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
- Inserm Unité Mixte de Recherche 1163, Paris, France
- Imagine Institut, Université de Paris Descartes Sorbonne Paris Cité, Paris, France
- Department of Pediatric Immunology, Hematology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Collège de France, Paris, France
| |
Collapse
|
16
|
van Steijn L, Wortel IMN, Sire C, Dupré L, Theraulaz G, Merks RMH. Computational modelling of cell motility modes emerging from cell-matrix adhesion dynamics. PLoS Comput Biol 2022; 18:e1009156. [PMID: 35157694 PMCID: PMC8880896 DOI: 10.1371/journal.pcbi.1009156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/25/2022] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
Lymphocytes have been described to perform different motility patterns such as Brownian random walks, persistent random walks, and Lévy walks. Depending on the conditions, such as confinement or the distribution of target cells, either Brownian or Lévy walks lead to more efficient interaction with the targets. The diversity of these motility patterns may be explained by an adaptive response to the surrounding extracellular matrix (ECM). Indeed, depending on the ECM composition, lymphocytes either display a floating motility without attaching to the ECM, or sliding and stepping motility with respectively continuous or discontinuous attachment to the ECM, or pivoting behaviour with sustained attachment to the ECM. Moreover, on the long term, lymphocytes either perform a persistent random walk or a Brownian-like movement depending on the ECM composition. How the ECM affects cell motility is still incompletely understood. Here, we integrate essential mechanistic details of the lymphocyte-matrix adhesions and lymphocyte intrinsic cytoskeletal induced cell propulsion into a Cellular Potts model (CPM). We show that the combination of de novo cell-matrix adhesion formation, adhesion growth and shrinkage, adhesion rupture, and feedback of adhesions onto cell propulsion recapitulates multiple lymphocyte behaviours, for different lymphocyte subsets and various substrates. With an increasing attachment area and increased adhesion strength, the cells’ speed and persistence decreases. Additionally, the model predicts random walks with short-term persistent but long-term subdiffusive properties resulting in a pivoting type of motility. For small adhesion areas, the spatial distribution of adhesions emerges as a key factor influencing cell motility. Small adhesions at the front allow for more persistent motility than larger clusters at the back, despite a similar total adhesion area. In conclusion, we present an integrated framework to simulate the effects of ECM proteins on cell-matrix adhesion dynamics. The model reveals a sufficient set of principles explaining the plasticity of lymphocyte motility. During immunosurveillance, lymphocytes patrol through tissues to interact with cancer cells, other immune cells, and pathogens. The efficiency of this process depends on the kinds of trajectories taken, ranging from simple Brownian walks to Lévy walks. The composition of the extracellular matrix (ECM), a network of macromolecules, affects the formation of cell-matrix adhesions, thus strongly influencing the way lymphocytes move. Here, we present a model of lymphocyte motility driven by adhesions that grow, shrink and rupture in response to the ECM and cellular forces. Compared to other models, our model is computationally light making it suitable for generating long term cell track data, while still capturing actin dynamics and adhesion turnover. Our model suggests that cell motility is affected by the force required to break adhesions and the rate at which new adhesions form. Adhesions can promote cell protrusion by inhibiting retrograde actin flow. After introducing this effect into the model, we found that it reduces the cellular diffusivity and that it promotes stick-slip behaviour. Furthermore, location and size of adhesion clusters determined cell persistence. Overall, our model explains the plasticity of lymphocyte behaviour in response to the ECM.
Collapse
Affiliation(s)
| | - Inge M. N. Wortel
- Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | - Clément Sire
- Laboratoire de Physique Théorique, Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
| | - Loïc Dupré
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Université de Toulouse, Toulouse, France
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Guy Theraulaz
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
- Centre for Ecological Sciences, Indian Institute of Science, Bengaluru, India
| | - Roeland M. H. Merks
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
17
|
Frattolin J, Watson DJ, Bonneuil WV, Russell MJ, Fasanella Masci F, Bandara M, Brook BS, Nibbs RJB, Moore JE. The Critical Importance of Spatial and Temporal Scales in Designing and Interpreting Immune Cell Migration Assays. Cells 2021; 10:3439. [PMID: 34943947 PMCID: PMC8700135 DOI: 10.3390/cells10123439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Intravital microscopy and other direct-imaging techniques have allowed for a characterisation of leukocyte migration that has revolutionised the field of immunology, resulting in an unprecedented understanding of the mechanisms of immune response and adaptive immunity. However, there is an assumption within the field that modern imaging techniques permit imaging parameters where the resulting cell track accurately captures a cell's motion. This notion is almost entirely untested, and the relationship between what could be observed at a given scale and the underlying cell behaviour is undefined. Insufficient spatial and temporal resolutions within migration assays can result in misrepresentation of important physiologic processes or cause subtle changes in critical cell behaviour to be missed. In this review, we contextualise how scale can affect the perceived migratory behaviour of cells, summarise the limited approaches to mitigate this effect, and establish the need for a widely implemented framework to account for scale and correct observations of cell motion. We then extend the concept of scale to new approaches that seek to bridge the current "black box" between single-cell behaviour and systemic response.
Collapse
Affiliation(s)
- Jennifer Frattolin
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Daniel J. Watson
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Willy V. Bonneuil
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| | - Matthew J. Russell
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Francesca Fasanella Masci
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Mikaila Bandara
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - Bindi S. Brook
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (M.J.R.); (B.S.B.)
| | - Robert J. B. Nibbs
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK; (F.F.M.); (M.B.); (R.J.B.N.)
| | - James E. Moore
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK; (J.F.); (D.J.W.); (W.V.B.)
| |
Collapse
|
18
|
Krüppel-like Factor 2 (KLF2) in Immune Cell Migration. Vaccines (Basel) 2021; 9:vaccines9101171. [PMID: 34696279 PMCID: PMC8539188 DOI: 10.3390/vaccines9101171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 01/30/2023] Open
Abstract
Krüppel-like factor 2 (KLF2), a transcription factor of the krüppel-like family, is a key regulator of activation, differentiation, and migration processes in various cell types. In this review, we focus on the functional relevance of KLF2 in immune cell migration and homing. We summarize the key functions of KLF2 in the regulation of chemokine receptors and adhesion molecules and discuss the relevance of the KLF2-mediated control of immune cell migration in the context of immune responses, infections, and diseases.
Collapse
|
19
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Fowell DJ, Kim M. The spatio-temporal control of effector T cell migration. Nat Rev Immunol 2021; 21:582-596. [PMID: 33627851 PMCID: PMC9380693 DOI: 10.1038/s41577-021-00507-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Effector T cells leave the lymph nodes armed with specialized functional attributes. Their antigenic targets may be located anywhere in the body, posing the ultimate challenge: how to efficiently identify the target tissue, navigate through a complex tissue matrix and, ultimately, locate the immunological insult. Recent advances in real-time in situ imaging of effector T cell migratory behaviour have revealed a great degree of mechanistic plasticity that enables effector T cells to push and squeeze their way through inflamed tissues. This process is shaped by an array of 'stop' and 'go' guidance signals including target antigens, chemokines, integrin ligands and the mechanical cues of the inflamed microenvironment. Effector T cells must sense and interpret these competing signals to correctly position themselves to mediate their effector functions for complete and durable responses in infectious disease and malignancy. Tuning T cell migration therapeutically will require a new understanding of this complex decision-making process.
Collapse
Affiliation(s)
- Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY.,Department of Microbiology and Immunology, Cornell University, Ithaca, NY
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
21
|
Abstract
T cells experience complex temporal patterns of stimulus via receptor-ligand-binding interactions with surrounding cells. From these temporal patterns, T cells are able to pick out antigenic signals while establishing self-tolerance. Although features such as duration of antigen binding have been examined, our understanding of how T cells interpret signals with different frequencies or temporal stimulation patterns is relatively unexplored. We engineered T cells to respond to light as a stimulus by building an optogenetically controlled chimeric antigen receptor (optoCAR). We discovered that T cells respond to minute-scale oscillations of activation signal by stimulating optoCAR T cells with tunable pulse trains of light. Systematically scanning signal oscillation period from 1 to 150 min revealed that expression of CD69, a T cell activation marker, reached a local minimum at a period of ∼25 min (corresponding to 5 to 15 min pulse widths). A combination of inhibitors and genetic knockouts suggest that this frequency filtering mechanism lies downstream of the Erk signaling branch of the T cell response network and may involve a negative feedback loop that diminishes Erk activity. The timescale of CD69 filtering corresponds with the duration of T cell encounters with self-peptide-presenting APCs observed via intravital imaging in mice, indicating a potential functional role for temporal filtering in vivo. This study illustrates that the T cell signaling machinery is tuned to temporally filter and interpret time-variant input signals in discriminatory ways.
Collapse
|
22
|
CXCL10 + peripheral activation niches couple preferred sites of Th1 entry with optimal APC encounter. Cell Rep 2021; 36:109523. [PMID: 34380032 PMCID: PMC9218982 DOI: 10.1016/j.celrep.2021.109523] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/02/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Correct positioning of T cells within infected tissues is critical for T cell activation and pathogen control. Upon tissue entry, effector T cells must efficiently locate antigen-presenting cells (APC) for peripheral activation. We reveal that tissue entry and initial peripheral activation of Th1 effector T cells are tightly linked to perivascular positioning of chemokine-expressing APCs. Dermal inflammation induces tissue-wide de novo generation of discrete perivascular CXCL10+ cell clusters, enriched for CD11c+MHC-II+ monocyte-derived dendritic cells. These chemokine clusters are "hotspots" for both Th1 extravasation and activation in the inflamed skin. CXCR3-dependent Th1 localization to the cluster micro-environment prolongs T-APC interactions and boosts function. Both the frequency and range of these clusters are enhanced via a T helper 1 (Th1)-intrinsic, interferon-gamma (IFNγ)-dependent positive-feedback loop. Thus, the perivascular CXCL10+ clusters act as initial peripheral activation niches, optimizing controlled activation broadly throughout the tissue by coupling Th1 tissue entry with enhanced opportunities for Th1-APC encounter.
Collapse
|
23
|
Moreau JM, Gouirand V, Rosenblum MD. T-Cell Adhesion in Healthy and Inflamed Skin. JID INNOVATIONS 2021; 1:100014. [PMID: 35024681 PMCID: PMC8669513 DOI: 10.1016/j.xjidi.2021.100014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
The diverse populations of tissue-resident and transitory T cells present in the skin share a common functional need to enter, traverse, and interact with their environment. These processes are largely dependent on the regulated expression of adhesion molecules, such as selectins and integrins, which mediate bidirectional interactions between immune cells and skin stroma. Dysregulation and engagement of adhesion pathways contribute to ectopic T-cell activity in tissues, leading to the initiation and/or exacerbation of chronic inflammation. In this paper, we review how the molecular interactions supported by adhesion pathways contribute to T-cell dynamics and function in the skin. A comprehensive understanding of the molecular mechanisms underpinning T-cell adhesion in inflammatory skin disorders will facilitate the development of novel tissue-specific therapeutic strategies.
Collapse
Key Words
- AD, atopic dermatitis
- BM, basement membrane
- DC, dendritic cell
- DETC, dendritic epidermal γδ T cell
- ECM, extracellular matrix
- HF, hair follicle
- JC, John Cunningham
- LAD, leukocyte adhesion deficiency
- PML, progressive multifocal leukoencephalopathy
- Th, T helper
- Treg, regulatory T cell
- Trm, tissue-resident memory
Collapse
Affiliation(s)
- Joshua M. Moreau
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Victoire Gouirand
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Michael D. Rosenblum
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
Zayats R, Uzonna JE, Murooka TT. Visualizing the In Vivo Dynamics of Anti- Leishmania Immunity: Discoveries and Challenges. Front Immunol 2021; 12:671582. [PMID: 34093571 PMCID: PMC8172142 DOI: 10.3389/fimmu.2021.671582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/07/2021] [Indexed: 11/20/2022] Open
Abstract
Intravital microscopy, such as 2-photon microscopy, is now a mainstay in immunological research to visually characterize immune cell dynamics during homeostasis and pathogen infections. This approach has been especially beneficial in describing the complex process of host immune responses to parasitic infections in vivo, such as Leishmania. Human-parasite co-evolution has endowed parasites with multiple strategies to subvert host immunity in order to establish chronic infections and ensure human-to-human transmission. While much focus has been placed on viral and bacterial infections, intravital microscopy studies during parasitic infections have been comparatively sparse. In this review, we will discuss how in vivo microscopy has provided important insights into the generation of innate and adaptive immunity in various organs during parasitic infections, with a primary focus on Leishmania. We highlight how microscopy-based approaches may be key to providing mechanistic insights into Leishmania persistence in vivo and to devise strategies for better parasite control.
Collapse
Affiliation(s)
- Romaniya Zayats
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jude E. Uzonna
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas T. Murooka
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
25
|
Norman MU, Chow Z, Snelgrove SL, Prakongtham P, Hickey MJ. Dynamic Regulation of the Molecular Mechanisms of Regulatory T Cell Migration in Inflamed Skin. Front Immunol 2021; 12:655499. [PMID: 34040606 PMCID: PMC8143438 DOI: 10.3389/fimmu.2021.655499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022] Open
Abstract
The presence of regulatory T cells (Tregs) in skin is important in controlling inflammatory responses in this peripheral tissue. Uninflamed skin contains a population of relatively immotile Tregs often located in clusters around hair follicles. Inflammation induces a significant increase both in the abundance of Tregs within the dermis, and in the proportion of Tregs that are highly migratory. The molecular mechanisms underpinning Treg migration in the dermis are unclear. In this study we used multiphoton intravital microscopy to examine the role of RGD-binding integrins and signalling through phosphoinositide 3-kinase P110δ (PI3K p110δ) in intradermal Treg migration in resting and inflamed skin. We found that inflammation induced Treg migration was dependent on RGD-binding integrins in a context-dependent manner. αv integrin was important for Treg migration 24 hours after induction of inflammation, but contributed to Treg retention at 48 hours, while β1 integrin played a role in Treg retention at the later time point but not during the peak of inflammation. In contrast, inhibition of signalling through PI3K p110δ reduced Treg migration throughout the entire inflammatory response, and also in the absence of inflammation. Together these observations demonstrate that the molecular mechanisms controlling intradermal Treg migration vary markedly according to the phase of the inflammatory response.
Collapse
Affiliation(s)
- M Ursula Norman
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Zachary Chow
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Sarah L Snelgrove
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Peemapat Prakongtham
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| |
Collapse
|
26
|
Miao Y, Zhao Y, Han L, Ma X, Deng J, Yang J, Lü S, Shao F, Kong W, Wang W, Xu Q, Wang X, Feng J. NSun2 regulates aneurysm formation by promoting autotaxin expression and T cell recruitment. Cell Mol Life Sci 2021; 78:1709-1727. [PMID: 32734582 PMCID: PMC11073013 DOI: 10.1007/s00018-020-03607-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/09/2020] [Accepted: 07/22/2020] [Indexed: 01/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration and aggravated by hyperhomocysteinemia (HHcy). It is unknown whether the homocysteine (Hcy)-activated RNA methyltransferase NOP2/Sun domain family member 2 (NSun2) is associated with AAA. Here, we found that NSun2 deficiency significantly attenuated elastase-induced and HHcy-aggravated murine AAA with decreased T cell infiltration in the vessel walls. T cell labeling and adoptive transfer experiments confirmed that NSun2 deficiency inhibited the chemotaxis of vessels to T cells. RNA sequencing of endothelial cells showed that Hcy induced the accumulation of various metabolic enzymes of the phospholipid PC-LPC-LPA metabolic pathway, especially autotaxin (ATX). In the elastase-induced mouse model of AAA, ATX was specifically expressed in the endothelium and the plasma ATX concentration was upregulated and even higher in the HHcy group, which were decreased dramatically by NSun2 knockdown. In vitro Transwell experiments showed that ATX dose-dependently promoted T cell migration. HHcy may upregulate endothelial ATX expression and secretion and in turn recruit T cells into the vessel walls to induce vascular inflammation and consequently accelerate the pathogenesis of AAA. Mechanistically, secreted ATX interacted with T cells by binding to integrin α4, which subsequently activated downstream FAK/Src-RhoA signaling pathways and then induced T cell chemokinesis and adhesion. ATX overexpression in the vessel walls reversed the inhibited development of AAA in the NSun2-deficient mice. Therefore, NSun2 mediates the development of HHcy-aggravated AAA primarily by increasing endothelial ATX expression, secretion and T cell migration, which is a novel mechanism for HHcy-aggravated vascular inflammation and pathogenesis of AAA.
Collapse
Affiliation(s)
- Yutong Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | - Lulu Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Xiaolong Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Jiacheng Deng
- Cardiovascular Division, BHF Center for Vascular Regeneration, King's College London, London, UK
| | - Juan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Silin Lü
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Fangyu Shao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Qingbo Xu
- Cardiovascular Division, BHF Center for Vascular Regeneration, King's College London, London, UK
| | - Xian Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China.
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, People's Republic of China.
| |
Collapse
|
27
|
Hazlewood JE, Dumenil T, Le TT, Slonchak A, Kazakoff SH, Patch AM, Gray LA, Howley PM, Liu L, Hayball JD, Yan K, Rawle DJ, Prow NA, Suhrbier A. Injection site vaccinology of a recombinant vaccinia-based vector reveals diverse innate immune signatures. PLoS Pathog 2021; 17:e1009215. [PMID: 33439897 PMCID: PMC7837487 DOI: 10.1371/journal.ppat.1009215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/26/2021] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Poxvirus systems have been extensively used as vaccine vectors. Herein a RNA-Seq analysis of intramuscular injection sites provided detailed insights into host innate immune responses, as well as expression of vector and recombinant immunogen genes, after vaccination with a new multiplication defective, vaccinia-based vector, Sementis Copenhagen Vector. Chikungunya and Zika virus immunogen mRNA and protein expression was associated with necrosing skeletal muscle cells surrounded by mixed cellular infiltrates. The multiple adjuvant signatures at 12 hours post-vaccination were dominated by TLR3, 4 and 9, STING, MAVS, PKR and the inflammasome. Th1 cytokine signatures were dominated by IFNγ, TNF and IL1β, and chemokine signatures by CCL5 and CXCL12. Multiple signatures associated with dendritic cell stimulation were evident. By day seven, vaccine transcripts were absent, and cell death, neutrophil, macrophage and inflammation annotations had abated. No compelling arthritis signatures were identified. Such injection site vaccinology approaches should inform refinements in poxvirus-based vector design. Poxvirus vector systems have been widely developed for vaccine applications. Despite considerable progress, so far only one recombinant poxvirus vectored vaccine has to date been licensed for human use, with ongoing efforts seeking to enhance immunogenicity whilst minimizing reactogenicity. The latter two characteristics are often determined by early post-vaccination events at the injection site. We therefore undertook an injection site vaccinology approach to analyzing gene expression at the vaccination site after intramuscular inoculation with a recombinant, multiplication defective, vaccinia-based vaccine. This provided detailed insights into inter alia expression of vector-encoded immunoregulatory genes, as well as host innate and adaptive immune responses. We propose that such injection site vaccinology can inform rational vaccine vector design, and we discuss how the information and approach elucidated herein might be used to improve immunogenicity and limit reactogenicity of poxvirus-based vaccine vector systems.
Collapse
Affiliation(s)
- Jessamine E. Hazlewood
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Thuy T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Stephen H. Kazakoff
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Ann-Marie Patch
- Clinical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Lesley-Ann Gray
- Australian Genome Research Facility Ltd., Melbourne, Australia
| | | | - Liang Liu
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - John D. Hayball
- Sementis Ltd., Hackney, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Daniel J. Rawle
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Natalie A. Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Australian Infectious Disease Research Centre, Brisbane, Australia
- * E-mail:
| |
Collapse
|
28
|
Bares AJ, Mejooli MA, Pender MA, Leddon SA, Tilley S, Lin K, Dong J, Kim M, Fowell DJ, Nishimura N, Schaffer CB. Hyperspectral multiphoton microscopy for in vivo visualization of multiple, spectrally overlapped fluorescent labels. OPTICA 2020; 7:1587-1601. [PMID: 33928182 PMCID: PMC8081374 DOI: 10.1364/optica.389982] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/30/2020] [Indexed: 05/17/2023]
Abstract
The insensitivity of multiphoton microscopy to optical scattering enables high-resolution, high-contrast imaging deep into tissue, including in live animals. Scattering does, however, severely limit the use of spectral dispersion techniques to improve spectral resolution. In practice, this limited spectral resolution together with the need for multiple excitation wavelengths to excite different fluorophores limits multiphoton microscopy to imaging a few, spectrally-distinct fluorescent labels at a time, restricting the complexity of biological processes that can be studied. Here, we demonstrate a hyperspectral multiphoton microscope that utilizes three different wavelength excitation sources together with multiplexed fluorescence emission detection using angle-tuned bandpass filters. This microscope maintains scattering insensitivity, while providing high enough spectral resolution on the emitted fluorescence and capitalizing on the wavelength-dependent nonlinear excitation of fluorescent dyes to enable clean separation of multiple, spectrally overlapping labels, in vivo. We demonstrated the utility of this instrument for spectral separation of closely-overlapped fluorophores in samples containing ten different colors of fluorescent beads, live cells expressing up to seven different fluorescent protein fusion constructs, and in multiple in vivo preparations in mouse cortex and inflamed skin with up to eight different cell types or tissue structures distinguished.
Collapse
Affiliation(s)
- Amanda J. Bares
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Menansili A. Mejooli
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mitchell A. Pender
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Scott A. Leddon
- Center for Vaccine Biology and Immunology, Dept. of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven Tilley
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Karen Lin
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jingyuan Dong
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Minsoo Kim
- Center for Vaccine Biology and Immunology, Dept. of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah J. Fowell
- Center for Vaccine Biology and Immunology, Dept. of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Nozomi Nishimura
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Chris B. Schaffer
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
29
|
Emo K, Reilly EC, Sportiello M, Yang H, Topham DJ. T cell and chemokine receptors differentially control CD8 T cell motility behavior in the infected airways immediately before and after virus clearance in a primary infection. PLoS One 2020; 15:e0227157. [PMID: 32817719 PMCID: PMC7444504 DOI: 10.1371/journal.pone.0227157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/21/2020] [Indexed: 11/19/2022] Open
Abstract
In mice, experimental influenza virus infection stimulates CD8 T cell infiltration of the airways. Virus is cleared by day 9, and between days 8 and 9 there is an abrupt change in CD8 T cell motility behavior transitioning from low velocity and high confinement on day 8, to high velocity with continued high confinement on day 9. We hypothesized that loss of virus and/or antigen signals in the context of high chemokine levels drives the T cells into a rapid surveillance mode. Virus infection induces chemokine production, which may change when the virus is cleared. We therefore sought to examine this period of rapid changes to the T cell environment in the tissue and seek evidence on the roles of peptide-MHC and chemokine receptor interactions. Experiments were performed to block G protein coupled receptor (GPCR) signaling with Pertussis toxin (Ptx). Ptx treatment generally reduced cell velocities and mildly increased confinement suggesting chemokine mediated arrest (velocity <2 μm/min) (Friedman RS, 2005), except on day 8 when velocity increased and confinement was relieved. Blocking specific peptide-MHC with monoclonal antibody unexpectedly decreased velocities on days 7 through 9, suggesting TCR/peptide-MHC interactions promote cell mobility in the tissue. Together, these results suggest the T cells are engaged with antigen bearing and chemokine producing cells that affect motility in ways that vary with the day after infection. The increase in velocities on day 9 were reversed by addition of specific peptide, consistent with the idea that antigen signals become limiting on day 9 compared to earlier time points. Thus, antigen and chemokine signals act to alternately promote and restrict CD8 T cell motility until the point of virus clearance, suggesting the switch in motility behavior on day 9 may be due to a combination of limiting antigen in the presence of high chemokine signals as the virus is cleared.
Collapse
Affiliation(s)
- Kris Emo
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Emma C. Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
| | - Hongmei Yang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
30
|
Fernandes NRJ, Reilly NS, Schrock DC, Hocking DC, Oakes PW, Fowell DJ. CD4 + T Cell Interstitial Migration Controlled by Fibronectin in the Inflamed Skin. Front Immunol 2020; 11:1501. [PMID: 32793204 PMCID: PMC7393769 DOI: 10.3389/fimmu.2020.01501] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 01/21/2023] Open
Abstract
The extracellular matrix (ECM) is extensively remodeled during inflammation providing essential guidance cues for immune cell migration and signals for cell activation and survival. There is increasing interest in the therapeutic targeting of ECM to mitigate chronic inflammatory diseases and enhance access to the tumor microenvironment. T cells utilize the ECM as a scaffold for interstitial migration, dependent on T cell expression of matrix-binding integrins αVβ1/αVβ3 and tissue display of the respective RGD-containing ligands. The specific ECM components that control T cell migration are unclear. Fibronectin (FN), a canonical RGD-containing matrix component, is heavily upregulated in inflamed tissues and in vitro can serve as a substrate for leukocyte migration. However, limited by lack of tools to intravitally visualize and manipulate FN, the specific role of FN in effector T cell migration in vivo is unknown. Here, we utilize fluorescently-tagged FN to probe for FN deposition, and intravital multiphoton microscopy to visualize T cell migration relative to FN in the inflamed ear dermis. Th1 cells were found to migrate along FN fibers, with T cells appearing to actively push or pull against flexible FN fibers. To determine the importance of T cell interactions with FN, we used a specific inhibitor of FN polymerization, pUR4. Intradermal delivery of pUR4 (but not the control peptide) to the inflamed skin resulted in a local reduction in FN deposition. We also saw a striking attenuation of Th1 effector T cell movement at the pUR4 injection site, suggesting FN plays a key role in T cell interstitial migration. In mechanistic studies, pUR4 incubation with FN in vitro resulted in enhanced tethering of T cells to FN matrix, limiting productive migration. In vivo, such tethering led to increased Th1 accumulation in the inflamed dermis. Enhanced Th1 accumulation exacerbated inflammation with increased Th1 activation and IFNγ cytokine production. Thus, our studies highlight the importance of ECM FN fibrils for T cell migration in inflamed tissues and suggest that manipulating local levels of ECM FN may prove beneficial in promoting T cell accumulation in tissues and enhancing local immunity to infection or cancer.
Collapse
Affiliation(s)
- Ninoshka R. J. Fernandes
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Nicholas S. Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States
| | - Dillon C. Schrock
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Denise C. Hocking
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States
| | - Patrick W. Oakes
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
31
|
Donnadieu E, Reisinger KB, Scharf S, Michel Y, Bein J, Hansen S, Loth AG, Flinner N, Hartmann S, Hansmann ML. Landscape of T Follicular Helper Cell Dynamics in Human Germinal Centers. THE JOURNAL OF IMMUNOLOGY 2020; 205:1248-1255. [PMID: 32699157 DOI: 10.4049/jimmunol.1901475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/28/2020] [Indexed: 11/19/2022]
Abstract
T follicular helper (Tfh) cells play a very important role in mounting a humoral response. Studies conducted in mouse models have revealed with good kinetic and spatial resolution the dynamics of these cells in germinal centers (GC) and their cross-talk with B cells upon an immune response. However, whether a similar migratory behavior is performed by human Tfh cells is unclear, as technology to track them in situ has been lacking. In this study, we combined traditional immunohistochemistry and real-time fluorescent imaging approaches on fresh human adenoid slices to provide static and dynamic information on Tfh cells. Our data indicate that GC light zones are composed of two distinct areas in terms of Tfh cell distribution and migration. In the outer GC light zones, Tfh cells migrate actively and with a high ability to form dynamic clusters showing intense and rapid reorganization. In these outer regions, Tfh cells demonstrate multiple interactions between each other. Conversely, in central regions of GC light zones, Tfh cells are much more static, forming long-lasting conjugates. These findings reveal for the first time, to our knowledge, the dynamic behavior whereby Tfh cells migrate in human GC and highlight the heterogeneity of GC for Tfh cell motility.
Collapse
Affiliation(s)
- Emmanuel Donnadieu
- Département Immunologie, Inflammation, et Infection, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Université de Paris, F-75014 Paris, France;
| | | | - Sonja Scharf
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Yvonne Michel
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Julia Bein
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany
| | - Susanne Hansen
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Andreas G Loth
- Department of Otolaryngology, Head and Neck Surgery, University Hospital, 60590 Frankfurt am Main, Germany; and
| | - Nadine Flinner
- Frankfurt Institute for Advanced Studies, 60438 Frankfurt, Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany
| | - Martin-Leo Hansmann
- Dr. Senckenberg Institute of Pathology, Goethe University, 60590 Frankfurt am Main, Germany.,Reference and Consultant Center for Lymph Node and Lymphoma Diagnostics, 60590 Frankfurt, Germany.,Frankfurt Institute for Advanced Studies, 60438 Frankfurt, Germany
| |
Collapse
|
32
|
Allan-Rahill NH, Lamont MRE, Chilian WM, Nishimura N, Small DM. Intravital Microscopy of the Beating Murine Heart to Understand Cardiac Leukocyte Dynamics. Front Immunol 2020; 11:92. [PMID: 32117249 PMCID: PMC7010807 DOI: 10.3389/fimmu.2020.00092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of worldwide mortality. Intravital microscopy has provided unprecedented insight into leukocyte biology by enabling the visualization of dynamic responses within living organ systems at the cell-scale. The heart presents a uniquely dynamic microenvironment driven by periodic, synchronous electrical conduction leading to rhythmic contractions of cardiomyocytes, and phasic coronary blood flow. In addition to functions shared throughout the body, immune cells have specific functions in the heart including tissue-resident macrophage-facilitated electrical conduction and rapid monocyte infiltration upon injury. Leukocyte responses to cardiac pathologies, including myocardial infarction and heart failure, have been well-studied using standard techniques, however, certain questions related to spatiotemporal relationships remain unanswered. Intravital imaging techniques could greatly benefit our understanding of the complexities of in vivo leukocyte behavior within cardiac tissue, but these techniques have been challenging to apply. Different approaches have been developed including high frame rate imaging of the beating heart, explantation models, micro-endoscopy, and mechanical stabilization coupled with various acquisition schemes to overcome challenges specific to the heart. The field of cardiac science has only begun to benefit from intravital microscopy techniques. The current focused review presents an overview of leukocyte responses in the heart, recent developments in intravital microscopy for the murine heart, and a discussion of future developments and applications for cardiovascular immunology.
Collapse
Affiliation(s)
- Nathaniel H Allan-Rahill
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Michael R E Lamont
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Nozomi Nishimura
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - David M Small
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| |
Collapse
|