1
|
Liu X, Gutierrez Jauregui R, Lueder Y, Halle S, Ospina-Quintero L, Ritter C, Schimrock A, Willenzon S, Janssen A, Wagner K, Messerle M, Bošnjak B, Förster R. Protective function of ex vivo-expanded CD8 T cells in a mouse model of adoptive therapy for cytomegalovirus infection depends on integrin beta 1 but not CXCR3, CTLA4, or PD-1 expression. J Leukoc Biol 2025; 117:qiae256. [PMID: 40276928 DOI: 10.1093/jleuko/qiae256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/24/2024] [Indexed: 04/26/2025] Open
Abstract
The adoptive transfer of virus-specific T cells (VSTs) represents a therapeutic option for viral infection treatment in immunocompromised patients. Before administration, ex vivo culture enables VST expansion. However, it is unclear how ex vivo expansion affects the circulation, homing, and intra-tissue migration of administered VSTs. We established a model of VST immunotherapy of acute cytomegalovirus infection using adoptive transfer of ex vivo-expanded OT-I CD8 T cells (recognizing SIINFEKL peptide) into Rag2-/- mice infected with murine cytomegalovirus (MCMV) encoding for the SIINFEKL peptide. Ex vivo expansion induced an effector T cell phenotype and affected the expression of integrins and chemokine receptors. CRISPR/Cas9-mediated gene deletions enabled us to address the role of selected genes in the homing of VSTs following intravenous administration. We found that deletion of Itgb1, encoding for integrin beta 1, prevented OT-I cells from entering infected organs and drastically reduced their number in blood, suggesting that adoptively transferred VSTs primarily expand in the infected tissues. By contrast, Cxcr3-/- OT-I cells provided equal protection as their Cxcr3+/+ counterparts, indicating that this chemokine receptor does not contribute to VST entry into infected organs. Further, Pdcd1 and Ctla4 deletion did not impair the transferred OT-I cells' ability to protect mice from MCMV, arguing against quick exhaustion of VSTs with an effector T cell phenotype. Together, these data indicate that ex vivo expansion affects migration and activation properties of VSTs and suggest that future clinical evaluation of adoptive T cell therapy efficacy should include homing molecule expression assessment.
Collapse
Affiliation(s)
- Xiaokun Liu
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | | | - Yvonne Lueder
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Stephan Halle
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Laura Ospina-Quintero
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Christiane Ritter
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Anja Schimrock
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Stefanie Willenzon
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Karen Wagner
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
| |
Collapse
|
2
|
Sun M, Angelillo J, Hugues S. Lymphatic transport in anti-tumor immunity and metastasis. J Exp Med 2025; 222:e20231954. [PMID: 39969537 PMCID: PMC11837853 DOI: 10.1084/jem.20231954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Although lymphatic vessels (LVs) are present in many tumors, their importance in cancer has long been underestimated. In contrast to the well-studied tumor-associated blood vessels, LVs were previously considered to function as passive conduits for tumor metastasis. However, emerging evidence over the last two decades has shed light on their critical role in locally shaping the tumor microenvironment (TME). Here we review the involvement of LVs in tumor progression, metastasis, and modulation of anti-tumor immune response.
Collapse
Affiliation(s)
- Mengzhu Sun
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Julien Angelillo
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
3
|
Kunisch M, Beutler S, Pilger C, Kiefer F, Huser T, Wirth B. Active axial motion compensation in multiphoton-excited fluorescence microscopy. OPTICS EXPRESS 2025; 33:3620-3636. [PMID: 39876479 DOI: 10.1364/oe.547244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025]
Abstract
In living organisms, the natural motion caused by heartbeat, breathing, or muscle movements leads to the deformation of tissue caused by translation and stretching of the tissue structure. This effect results in the displacement or deformation of the plane of observation for intravital microscopy and causes motion-induced aberrations of the resulting image data. This, in turn, places severe limitations on the time during which specific events can be observed in intravital imaging experiments. These limitations can be overcome if the tissue motion can be compensated such that the plane of observation remains steady. We have developed a mathematical shape space model that can predict the periodic motion of a cylindrical tissue phantom resembling blood vessels. This model is then used to rapidly calculate the future position of the plane of observation of a two-photon laser scanning fluorescence microscope. The focal plane is continuously adjusted to the calculated position with a piezo-actuated objective lens holder. We demonstrate active motion compensation for non-harmonic axial displacements of the vessel phantom with a field of view up to 400 µm × 400 µm, vertical amplitudes of more than 100 µm, and at a rate of 0.5 Hz.
Collapse
|
4
|
Song R, Ye J, Chung S, Meyer CC, Déchelette C, Purcell AW, Braun A. A tactile approach to introduce the skin autoimmune disease psoriasis to the general public and the vision-impaired community. Immunol Cell Biol 2024; 102:336-340. [PMID: 37982599 DOI: 10.1111/imcb.12713] [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] [Indexed: 11/21/2023]
Abstract
Scientific outreach activities play an important role in disseminating knowledge, connecting the general public to research and breaking down scientific skepticism barriers. However, the vision-impaired community is often disadvantaged when the most common audio-visual approach of scientific communication is applied. Here we integrated tactile clues in the scientific communication of immune processes involved in the autoimmune skin disease psoriasis. We fostered the involvement of the vision-impaired community through interactive experiences, including tactile scientific origami art, a haptic poster and wood-carved molecular models. Readily accessible science communication that engages a number of senses is a critical step toward making science more inclusive and engaging for individuals with a wide range of sensory abilities. The approach of the 2023 Monash Sensory Science exhibition aligns with the principles of equity, diversity and inclusion and helps to empower a more informed and scientifically literate public.
Collapse
Affiliation(s)
- Runqiu Song
- Department of Biochemistry and Molecular Biology, Immunity and Infection Programs, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jingran Ye
- Department of Biochemistry and Molecular Biology, Immunity and Infection Programs, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Shanzou Chung
- Department of Biochemistry and Molecular Biology, Immunity and Infection Programs, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | | | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, Immunity and Infection Programs, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Asolina Braun
- Department of Biochemistry and Molecular Biology, Immunity and Infection Programs, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
5
|
Houbaert D, Nikolakopoulos AP, Jacobs KA, Meçe O, Roels J, Shankar G, Agrawal M, More S, Ganne M, Rillaerts K, Boon L, Swoboda M, Nobis M, Mourao L, Bosisio F, Vandamme N, Bergers G, Scheele CLGJ, Agostinis P. An autophagy program that promotes T cell egress from the lymph node controls responses to immune checkpoint blockade. Cell Rep 2024; 43:114020. [PMID: 38554280 DOI: 10.1016/j.celrep.2024.114020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 04/01/2024] Open
Abstract
Lymphatic endothelial cells (LECs) of the lymph node (LN) parenchyma orchestrate leukocyte trafficking and peripheral T cell dynamics. T cell responses to immunotherapy largely rely on peripheral T cell recruitment in tumors. Yet, a systematic and molecular understanding of how LECs within the LNs control T cell dynamics under steady-state and tumor-bearing conditions is lacking. Intravital imaging combined with immune phenotyping shows that LEC-specific deletion of the essential autophagy gene Atg5 alters intranodal positioning of lymphocytes and accrues their persistence in the LNs by increasing the availability of the main egress signal sphingosine-1-phosphate. Single-cell RNA sequencing of tumor-draining LNs shows that loss of ATG5 remodels niche-specific LEC phenotypes involved in molecular pathways regulating lymphocyte trafficking and LEC-T cell interactions. Functionally, loss of LEC autophagy prevents recruitment of tumor-infiltrating T and natural killer cells and abrogates response to immunotherapy. Thus, an LEC-autophagy program boosts immune-checkpoint responses by guiding systemic T cell dynamics.
Collapse
Affiliation(s)
- Diede Houbaert
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Apostolos Panagiotis Nikolakopoulos
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; Laboratory of Intravital Microscopy and Dynamics of Tumor Progression, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Kathryn A Jacobs
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Odeta Meçe
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Jana Roels
- VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; VIB Single Cell Core, Leuven, Belgium
| | - Gautam Shankar
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, KU Leuven and UZ Leuven, Leuven, Belgium
| | - Madhur Agrawal
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Sanket More
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Maarten Ganne
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Kristine Rillaerts
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | | | - Magdalena Swoboda
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium
| | - Max Nobis
- Intravital Imaging Expertise Center, VIB-CCB, Leuven, Belgium
| | - Larissa Mourao
- VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; Laboratory of Intravital Microscopy and Dynamics of Tumor Progression, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Francesca Bosisio
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, KU Leuven and UZ Leuven, Leuven, Belgium
| | - Niels Vandamme
- VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; VIB Single Cell Core, Leuven, Belgium
| | - Gabriele Bergers
- VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Colinda L G J Scheele
- VIB Center for Cancer Biology Research (CCB), Leuven, Belgium; Laboratory of Intravital Microscopy and Dynamics of Tumor Progression, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology Research (CCB), Leuven, Belgium.
| |
Collapse
|
6
|
Freen-van Heeren JJ. Posttranscriptional Events Orchestrate Immune Homeostasis of CD8 + T Cells. Methods Mol Biol 2024; 2782:65-80. [PMID: 38622392 DOI: 10.1007/978-1-0716-3754-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Maintaining immune homeostasis is instrumental for host health. Immune cells, such as T cells, are instrumental for the eradication of pathogenic bacteria, fungi and viruses. Furthermore, T cells also play a major role in the fight against cancer. Through the formation of immunological memory, a pool of antigen-experienced T cells remains in the body to rapidly protect the host upon reinfection or retransformation. In order to perform their protective function, T cells produce cytolytic molecules, such as granzymes and perforin, and cytokines such as interferon γ and tumor necrosis factor α. Recently, it has become evident that posttranscriptional regulatory events dictate the kinetics and magnitude of cytokine production by murine and human CD8+ T cells. Here, the recent literature regarding the role posttranscriptional regulation plays in maintaining immune homeostasis of antigen-experienced CD8+ T cells is reviewed.
Collapse
|
7
|
Lyu A, Humphrey RS, Nam SH, Durham TA, Hu Z, Arasappan D, Horton TM, Ehrlich LIR. Integrin signaling is critical for myeloid-mediated support of T-cell acute lymphoblastic leukemia. Nat Commun 2023; 14:6270. [PMID: 37805579 PMCID: PMC10560206 DOI: 10.1038/s41467-023-41925-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/21/2023] [Indexed: 10/09/2023] Open
Abstract
We previously found that T-cell acute lymphoblastic leukemia (T-ALL) requires support from tumor-associated myeloid cells, which activate Insulin Like Growth Factor 1 Receptor (IGF1R) signaling in leukemic blasts. However, IGF1 is not sufficient to sustain T-ALL in vitro, implicating additional myeloid-mediated signals in leukemia progression. Here, we find that T-ALL cells require close contact with myeloid cells to survive. Transcriptional profiling and in vitro assays demonstrate that integrin-mediated cell adhesion activates downstream focal adhesion kinase (FAK)/ proline-rich tyrosine kinase 2 (PYK2), which are required for myeloid-mediated T-ALL support, partly through activation of IGF1R. Blocking integrin ligands or inhibiting FAK/PYK2 signaling diminishes leukemia burden in multiple organs and confers a survival advantage in a mouse model of T-ALL. Inhibiting integrin-mediated adhesion or FAK/PYK2 also reduces survival of primary patient T-ALL cells co-cultured with myeloid cells. Furthermore, elevated integrin pathway gene signatures correlate with higher FAK signaling and myeloid gene signatures and are associated with an inferior prognosis in pediatric T-ALL patients. Together, these findings demonstrate that integrin activation and downstream FAK/PYK2 signaling are important mechanisms underlying myeloid-mediated support of T-ALL progression.
Collapse
Affiliation(s)
- Aram Lyu
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Ryan S Humphrey
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Seo Hee Nam
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Tyler A Durham
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Zicheng Hu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Dhivya Arasappan
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, USA
| | - Terzah M Horton
- Department of Pediatrics, Baylor College of Medicine/Dan L. Duncan Cancer Center and Texas Children's Cancer Center, Houston, TX, USA
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA.
- Department of Oncology, Livestrong Cancer Institutes, The University of Texas at Austin Dell Medical School, Austin, TX, USA.
| |
Collapse
|
8
|
Hallisey VM, Schwab SR. Get me out of here: Sphingosine 1-phosphate signaling and T cell exit from tissues during an immune response. Immunol Rev 2023; 317:8-19. [PMID: 37212181 DOI: 10.1111/imr.13219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
During an immune response, the duration of T cell residence in lymphoid and non-lymphoid tissues likely affects T cell activation, differentiation, and memory development. The factors that govern T cell transit through inflamed tissues remain incompletely understood, but one important determinant of T cell exit from tissues is sphingosine 1-phosphate (S1P) signaling. In homeostasis, S1P levels are high in blood and lymph compared to lymphoid organs, and lymphocytes follow S1P gradients out of tissues into circulation using varying combinations of five G-protein coupled S1P receptors. During an immune response, both the shape of S1P gradients and the expression of S1P receptors are dynamically regulated. Here we review what is known, and key questions that remain unanswered, about how S1P signaling is regulated in inflammation and in turn how S1P shapes immune responses.
Collapse
Affiliation(s)
- Victoria M Hallisey
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Susan R Schwab
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
9
|
Guo J, Xu Z, Gunderson RC, Xu B, Michie SA. LFA-1/ICAM-1 Adhesion Pathway Mediates the Homeostatic Migration of Lymphocytes from Peripheral Tissues into Lymph Nodes through Lymphatic Vessels. Biomolecules 2023; 13:1194. [PMID: 37627259 PMCID: PMC10452152 DOI: 10.3390/biom13081194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/22/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Lymphocyte function-associated antigen-1 (LFA-1) and its endothelial ligand intercellular adhesion molecule-1 (ICAM-1) are important for the migration of lymphocytes from blood vessels into lymph nodes. However, it is largely unknown whether these molecules mediate the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes through lymphatic vessels. In this study, we find that, in naive mice, ICAM-1 is expressed on the sinus endothelia of lymph nodes, but not on the lymphatic vessels of peripheral tissues. In in vivo lymphocyte migration assays, memory CD4+ T cells migrated to lymph nodes from peripheral tissues much more efficiently than from blood vessels, as compared to naive CD4+ T cells. Moreover, ICAM-1 deficiency in host mice significantly inhibited the migration of adoptively transferred wild-type donor lymphocytes from peripheral tissues, but not from blood vessels, into lymph nodes. The migration of LFA-1-deficient donor lymphocytes from peripheral tissues into the lymph nodes of wild-type host mice was also significantly reduced as compared to wild-type donor lymphocytes. Furthermore, the number of memory T cells in lymph nodes was significantly reduced in the absence of ICAM-1 or LFA-1. Thus, our study extends the functions of the LFA-1/ICAM-1 adhesion pathway, indicating its novel role in controlling the homeostatic migration of lymphocytes from peripheral tissues into lymph nodes and maintaining memory T cellularity in lymph nodes.
Collapse
Affiliation(s)
- Jia Guo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan 030012, China
| | - Zeyu Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Rachel C. Gunderson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
| | - Baohui Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sara A. Michie
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; (J.G.); (Z.X.); (R.C.G.)
| |
Collapse
|
10
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
11
|
Zhang Q, Zhang S, Chen J, Xie Z. The Interplay between Integrins and Immune Cells as a Regulator in Cancer Immunology. Int J Mol Sci 2023; 24:6170. [PMID: 37047140 PMCID: PMC10093897 DOI: 10.3390/ijms24076170] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Integrins are a group of heterodimers consisting of α and β subunits that mediate a variety of physiological activities of immune cells, including cell migration, adhesion, proliferation, survival, and immunotolerance. Multiple types of integrins act differently on the same immune cells, while the same integrin may exert various effects on different immune cells. In the development of cancer, integrins are involved in the regulation of cancer cell proliferation, invasion, migration, and angiogenesis; conversely, integrins promote immune cell aggregation to mediate the elimination of tumors. The important roles of integrins in cancer progression have provided valuable clues for the diagnosis and targeted treatment of cancer. Furthermore, many integrin inhibitors have been investigated in clinical trials to explore effective regimens and reduce side effects. Due to the complexity of the mechanism of integrin-mediated cancer progression, challenges remain in the research and development of cancer immunotherapies (CITs). This review enumerates the effects of integrins on four types of immune cells and the potential mechanisms involved in the progression of cancer, which will provide ideas for more optimal CIT in the future.
Collapse
Affiliation(s)
- Qingfang Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Shuo Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Takeda A, Salmi M, Jalkanen S. Lymph node lymphatic endothelial cells as multifaceted gatekeepers in the immune system. Trends Immunol 2023; 44:72-86. [PMID: 36463086 DOI: 10.1016/j.it.2022.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 12/03/2022]
Abstract
Single-cell technologies have recently allowed the identification of multiple lymphatic endothelial cell (LEC) subsets in subcapsular, paracortical, medullary, and other lymph node (LN) sinus systems in mice and humans. New analyses show that LECs serve key immunological functions in the LN stroma during immune responses. We discuss the roles of different LEC types in guiding leukocyte and cancer cell trafficking to and from the LN parenchyma, in capturing microbes, and in transporting, presenting, and storing lymph-borne antigens in distinct types of lymphatic sinuses. We underscore specific adaptations of human LECs and raise unanswered questions concerning LEC functions in human disease. Despite our limited understanding of human lymphatics - hampering clinical translation in inflammation and metastasis - we support the potential of LN LECs as putative targets for boosting/inhibiting immunoreactivity.
Collapse
Affiliation(s)
- Akira Takeda
- MediCity and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Marko Salmi
- MediCity and InFLAMES Flagship, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- MediCity and InFLAMES Flagship, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
13
|
New definitions of human lymphoid and follicular cell entities in lymphatic tissue by machine learning. Sci Rep 2022; 12:18991. [PMID: 36347879 PMCID: PMC9643435 DOI: 10.1038/s41598-022-18097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
Histological sections of the lymphatic system are usually the basis of static (2D) morphological investigations. Here, we performed a dynamic (4D) analysis of human reactive lymphoid tissue using confocal fluorescent laser microscopy in combination with machine learning. Based on tracks for T-cells (CD3), B-cells (CD20), follicular T-helper cells (PD1) and optical flow of follicular dendritic cells (CD35), we put forward the first quantitative analysis of movement-related and morphological parameters within human lymphoid tissue. We identified correlations of follicular dendritic cell movement and the behavior of lymphocytes in the microenvironment. In addition, we investigated the value of movement and/or morphological parameters for a precise definition of cell types (CD clusters). CD-clusters could be determined based on movement and/or morphology. Differentiating between CD3- and CD20 positive cells is most challenging and long term-movement characteristics are indispensable. We propose morphological and movement-related prototypes of cell entities applying machine learning models. Finally, we define beyond CD clusters new subgroups within lymphocyte entities based on long term movement characteristics. In conclusion, we showed that the combination of 4D imaging and machine learning is able to define characteristics of lymphocytes not visible in 2D histology.
Collapse
|
14
|
Abstract
To ensure proper immune function, most leukocytes constantly move within tissues or between them using the blood and lymphatic vessels as transport routes. While afferent lymphatic vessels transfer leukocytes from peripheral tissues to draining lymph nodes (dLNs), efferent lymphatics return lymphocytes from LNs back into the blood vascular circulation. Over the last decades, great progress has been made in our understanding of leukocyte migration into and within the lymphatic compartment, leading to the approval of new drugs targeting this process. In this review, we first introduce the anatomy of the lymphatic vasculature and the main cell types migrating through lymphatics. We primarily focus on dendritic cells (DCs) and T cells, the most prominent lymph-borne cell types, and discuss the functional significance as well as the main molecules and steps involved in their migration. Additionally, we provide an overview of the different techniques used to study lymphatic trafficking.
Collapse
Affiliation(s)
- Aline Bauer
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Hazal Tatliadim
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, CH-8093 Zurich, Switzerland
| |
Collapse
|
15
|
Deseke M, Rampoldi F, Sandrock I, Borst E, Böning H, Ssebyatika GL, Jürgens C, Plückebaum N, Beck M, Hassan A, Tan L, Demera A, Janssen A, Steinberger P, Koenecke C, Viejo-Borbolla A, Messerle M, Krey T, Prinz I. A CMV-induced adaptive human Vδ1+ γδ T cell clone recognizes HLA-DR. J Exp Med 2022; 219:213357. [PMID: 35852466 PMCID: PMC9301659 DOI: 10.1084/jem.20212525] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/02/2022] [Accepted: 06/30/2022] [Indexed: 01/21/2023] Open
Abstract
The innate and adaptive roles of γδ T cells and their clonal γδ T cell receptors (TCRs) in immune responses are still unclear. Recent studies of γδ TCR repertoire dynamics showed massive expansion of individual Vδ1+ γδ T cell clones during viral infection. To judge whether such expansion is random or actually represents TCR-dependent adaptive immune responses, information about their cognate TCR ligands is required. Here, we used CRISPR/Cas9-mediated screening to identify HLA-DRA, RFXAP, RFX5, and CIITA as required for target cell recognition of a CMV-induced Vγ3Vδ1+ TCR, and further characterization revealed a direct interaction of this Vδ1+ TCR with the MHC II complex HLA-DR. Since MHC II is strongly upregulated by interferon-γ, these results suggest an inflammation-induced MHC-dependent immune response of γδ T cells.
Collapse
Affiliation(s)
- Malte Deseke
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover, Germany
| | | | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Eva Borst
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Heike Böning
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - George Liam Ssebyatika
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - Carina Jürgens
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Nina Plückebaum
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Maleen Beck
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Ahmed Hassan
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Likai Tan
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Abdi Demera
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Peter Steinberger
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany,German Center for Infection Research, Partner Site Hamburg-Luebeck-Borstel-Riems, Hamburg, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany,German Center for Infection Research, Partner Site Hamburg-Luebeck-Borstel-Riems, Hamburg, Germany
| | - Thomas Krey
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover, Germany,Institute of Virology, Hannover Medical School, Hannover, Germany,Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, Lübeck, Germany,Center for Structural Systems Biology, Hamburg, Germany,German Center for Infection Research, Partner Site Hamburg-Luebeck-Borstel-Riems, Hamburg, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany,Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover, Germany,Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Correspondence to Immo Prinz:
| |
Collapse
|
16
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
17
|
Friess MC, Kritikos I, Schineis P, Medina-Sanchez JD, Gkountidi AO, Vallone A, Sigmund EC, Schwitter C, Vranova M, Matti C, Arasa J, Saygili Demir C, Bovay E, Proulx ST, Tomura M, Rot A, Legler DF, Petrova TV, Halin C. Mechanosensitive ACKR4 scavenges CCR7 chemokines to facilitate T cell de-adhesion and passive transport by flow in inflamed afferent lymphatics. Cell Rep 2022; 38:110334. [PMID: 35108538 DOI: 10.1016/j.celrep.2022.110334] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 12/02/2021] [Accepted: 01/12/2022] [Indexed: 11/03/2022] Open
Abstract
T cell migration via afferent lymphatics to draining lymph nodes (dLNs) depends on expression of CCR7 in T cells and CCL21 in the lymphatic vasculature. Once T cells have entered lymphatic capillaries, they slowly migrate into contracting collecting vessels. Here, lymph flow picks up, inducing T cell detachment and rapid transport to the dLNs. We find that the atypical chemokine receptor 4 (ACKR4), which binds and internalizes CCL19 and CCL21, is induced by lymph flow in endothelial cells lining lymphatic collectors, enabling them to scavenge these chemokines. In the absence of ACKR4, migration of T cells to dLNs in TPA-induced inflammation is significantly reduced. While entry into capillaries is not impaired, T cells accumulate in the ACKR4-deficient dermal collecting vessel segments. Overall, our findings identify an ACKR4-mediated mechanism by which lymphatic collectors facilitate the detachment of lymph-borne T cells in inflammation and their transition from crawling to free-flow toward the dLNs.
Collapse
Affiliation(s)
- Mona C Friess
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Ioannis Kritikos
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Philipp Schineis
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | | | - Angela Vallone
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Elena C Sigmund
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Corina Schwitter
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Martina Vranova
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Christoph Matti
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Jorge Arasa
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cansaran Saygili Demir
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Lausanne, Epalinges, Switzerland
| | - Esther Bovay
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Lausanne, Epalinges, Switzerland
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland; Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Centre for Inflammation and Therapeutic Innovation, Queen Mary University London, London, UK; Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland; Theodor Kocher Institute, University of Bern, Bern, Switzerland; Faculty of Biology, University of Konstanz, Konstanz, Germany
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Lausanne, Epalinges, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Rezalotfi A, Fritz L, Förster R, Bošnjak B. Challenges of CRISPR-Based Gene Editing in Primary T Cells. Int J Mol Sci 2022; 23:ijms23031689. [PMID: 35163611 PMCID: PMC8835901 DOI: 10.3390/ijms23031689] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/29/2022] [Indexed: 12/30/2022] Open
Abstract
Adaptive T-cell immunotherapy holds great promise for the successful treatment of leukemia, as well as other types of cancers. More recently, it was also shown to be an effective treatment option for chronic virus infections in immunosuppressed patients. Autologous or allogeneic T cells used for immunotherapy are usually genetically modified to express novel T-cell or chimeric antigen receptors. The production of such cells was significantly simplified with the CRISPR/Cas system, allowing for the deletion or insertion of novel genes at specific locations within the genome. In this review, we describe recent methodological breakthroughs that were important for the conduction of these genetic modifications, summarize crucial points to be considered when conducting such experiments, and highlight the potential pitfalls of these approaches.
Collapse
Affiliation(s)
- Alaleh Rezalotfi
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.R.); (L.F.); (R.F.)
| | - Lea Fritz
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.R.); (L.F.); (R.F.)
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.R.); (L.F.); (R.F.)
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover, 30625 Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; (A.R.); (L.F.); (R.F.)
- Correspondence: ; Tel.: +49-511-532-9731
| |
Collapse
|
19
|
Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, Bruno TC, Vignali DAA, Hand TW. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 2021; 54:2812-2824.e4. [PMID: 34861182 PMCID: PMC8865366 DOI: 10.1016/j.immuni.2021.11.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/19/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
The composition of the intestinal microbiota is associated with both the development of tumors and the efficacy of anti-tumor immunity. Here, we examined the impact of microbiota-specific T cells in anti-colorectal cancer (CRC) immunity. Introduction of Helicobacter hepaticus (Hhep) in a mouse model of CRC did not alter the microbial landscape but increased tumor infiltration by cytotoxic lymphocytes and inhibited tumor growth. Anti-tumor immunity was independent of CD8+ T cells but dependent upon CD4+ T cells, B cells, and natural killer (NK) cells. Hhep colonization induced Hhep-specific T follicular helper (Tfh) cells, increased the number of colon Tfh cells, and supported the maturation of Hhep+ tumor-adjacent tertiary lymphoid structures. Tfh cells were necessary for Hhep-mediated tumor control and immune infiltration, and adoptive transfer of Hhep-specific CD4+ T cells to Tfh cell-deficient Bcl6fl/flCd4Cre mice restored anti-tumor immunity. Thus, introduction of immunogenic intestinal bacteria can promote Tfh-associated anti-tumor immunity in the colon, suggesting therapeutic approaches for the treatment of CRC.
Collapse
Affiliation(s)
- Abigail E Overacre-Delgoffe
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA
| | - Hannah J Bumgarner
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program of Microbiology and Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ansen H P Burr
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program of Microbiology and Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Justin T Tometich
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Amrita Bhattacharjee
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Timothy W Hand
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
20
|
Miller EB, Karlen SJ, Ronning KE, Burns ME. Tracking distinct microglia subpopulations with photoconvertible Dendra2 in vivo. J Neuroinflammation 2021; 18:235. [PMID: 34654439 PMCID: PMC8520240 DOI: 10.1186/s12974-021-02285-x] [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: 08/17/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The ability to track individual immune cells within the central nervous system has revolutionized our understanding of the roles that microglia and monocytes play in synaptic maintenance, plasticity, and neurodegenerative diseases. However, distinguishing between similar subpopulations of mobile immune cells over time during episodes of neuronal death and tissue remodeling has proven to be challenging. METHODS We recombineered a photoconvertible fluorescent protein (Dendra2; D2) downstream of the Cx3cr1 promoter commonly used to drive expression of fluorescent markers in microglia and monocytes. Like the popular Cx3cr1-GFP line (Cx3cr1+/GFP), naïve microglia in Cx3cr1-Dendra2 mice (Cx3cr1+/D2) fluoresce green and can be noninvasively imaged in vivo throughout the CNS. In addition, individual D2-expressing cells can be photoconverted, resulting in red fluorescence, and tracked unambiguously within a field of green non-photoconverted cells for several days in vivo. RESULTS Dendra2-expressing retinal microglia were noninvasively photoconverted in both ex vivo and in vivo conditions. Local in vivo D2 photoconversion was sufficiently robust to quantify cell subpopulations by flow cytometry, and the protein was stable enough to survive tissue processing for immunohistochemistry. Simultaneous in vivo fluorescence imaging of Dendra2 and light scattering measurements (Optical Coherence Tomography, OCT) were used to assess responses of individual microglial cells to localized neuronal damage and to identify the infiltration of monocytes from the vasculature in response to large scale neurodegeneration. CONCLUSIONS The ability to noninvasively and unambiguously track D2-expressing microglia and monocytes in vivo through space and time makes the Cx3cr1-Dendra2 mouse model a powerful new tool for disentangling the roles of distinct immune cell subpopulations in neuroinflammation.
Collapse
Affiliation(s)
- Eric B Miller
- Center for Neuroscience, University of California, 1544 Newton Court, Davis, CA, 95618, USA
| | - Sarah J Karlen
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, USA
| | - Kaitryn E Ronning
- Center for Neuroscience, University of California, 1544 Newton Court, Davis, CA, 95618, USA
| | - Marie E Burns
- Center for Neuroscience, University of California, 1544 Newton Court, Davis, CA, 95618, USA.
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, USA.
- Department of Ophthalmology & Vision Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
21
|
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: 82] [Impact Index Per Article: 20.5] [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
|
22
|
Cousin N, Cap S, Dihr M, Tacconi C, Detmar M, Dieterich LC. Lymphatic PD-L1 Expression Restricts Tumor-Specific CD8 + T-cell Responses. Cancer Res 2021; 81:4133-4144. [PMID: 34099493 PMCID: PMC9398148 DOI: 10.1158/0008-5472.can-21-0633] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 01/07/2023]
Abstract
Lymph node (LN)-resident lymphatic endothelial cells (LEC) mediate peripheral tolerance by self-antigen presentation on MHC-I and constitutive expression of T-cell inhibitory molecules, including PD-L1 (CD274). Tumor-associated LECs also upregulate PD-L1, but the specific role of lymphatic PD-L1 in tumor immunity is not well understood. In this study, we generated a mouse model lacking lymphatic PD-L1 expression and challenged these mice with two orthotopic tumor models, B16F10 melanoma and MC38 colorectal carcinoma. Lymphatic PD-L1 deficiency resulted in consistent expansion of tumor-specific CD8+ T cells in tumor-draining LNs in both tumor models, reduced primary tumor growth in the MC38 model, and increased efficacy of adoptive T-cell therapy in the B16F10 model. Strikingly, lymphatic PD-L1 acted primarily by inducing apoptosis in tumor-specific CD8+ central memory T cells. Overall, these findings demonstrate that LECs restrain tumor-specific immunity via PD-L1, which may explain why some patients with cancer without PD-L1 expression in the tumor microenvironment still respond to PD-L1/PD-1-targeted immunotherapy. SIGNIFICANCE: A new lymphatic-specific PD-L1 knockout mouse model reveals that lymphatic endothelial PD-L1 expression reduces tumor immunity, inducing apoptosis in tumor-specific CD8+ central memory cells in tumor-draining lymph nodes.
Collapse
Affiliation(s)
| | | | | | | | | | - Lothar C. Dieterich
- Corresponding Author: Lothar C. Dieterich, ETH Zurich, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 1–5/10, 8093 Zurich, Switzerland. Phone: 41-44-63-37392; Fax: 41-44-63-31344; E-mail:
| |
Collapse
|
23
|
Yu K, Hammerschmidt SI, Permanyer M, Galla M, Rothe M, Zheng X, Werth K, Martens R, Lueder Y, Janssen A, Friedrichsen M, Bernhardt G, Förster R. Targeted delivery of regulatory macrophages to lymph nodes interferes with T cell priming by preventing the formation of stable immune synapses. Cell Rep 2021; 35:109273. [PMID: 34161766 DOI: 10.1016/j.celrep.2021.109273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 02/05/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Immunosuppressive myeloid cells are frequently induced in tumors and attenuate anti-tumor effector functions. In this study, we differentiate immunosuppressive regulatory macrophages (Mregs) from hematopoietic progenitors and test their potential to suppress adaptive immune responses in lymph nodes. Targeted delivery of Mregs to lymph nodes is facilitated by retroviral overexpression of the chemokine receptor CCR7 and intra-lymphatic cell application. Delivery of Mregs completely abolishes the priming of cognate CD8 cells and strongly reduces delayed-type hypersensitivity reactions. Mreg-mediated T cell suppression requires cell-cell contact-regulated nitric oxide production. Two-photon microscopy reveals that nitric oxide produced by Mregs reduces the interaction duration between dendritic cells and T cells. Exposure of activated T cells to nitric oxide strongly reduces their binding to ICAM-1, indicating that nitrosylation of proteins involved in cell adhesion affects synapse formation. Thus, this study identifies a mechanism of myeloid cell-mediated immune suppression and provides an approach for its therapeutic use.
Collapse
Affiliation(s)
- Kai Yu
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Xiang Zheng
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Rieke Martens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Yvonne Lueder
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany.
| |
Collapse
|
24
|
Lymph-Derived Neutrophils Primarily Locate to the Subcapsular and Medullary Sinuses in Resting and Inflamed Lymph Nodes. Cells 2021; 10:cells10061486. [PMID: 34204825 PMCID: PMC8231499 DOI: 10.3390/cells10061486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Neutrophils are the first immune cells to be recruited from the blood to the tissue site of an infection or inflammation. It has been suggested that neutrophils are capable of migrating from the infected tissue via lymphatic vessels to the draining lymph nodes. However, it remains elusive as to which areas within the lymph nodes can be reached by such reversely migrating cells. To address this question, we applied a model for adoptive neutrophil transfer into the afferent lymphatic vessel that drains towards the popliteal lymph node in mice. We showed that resting and in vitro-activated neutrophils did not enter the lymph node parenchyma but localized primarily in the subcapsular and medullary sinuses. Within the medulla, neutrophils show random migration and are able to sense laser-induced sterile tissue injury by massively swarming to the damaged tissue site. Co-injected dendritic cells supported the entry of resting neutrophils into the lymph node parenchyma via the subcapsular sinus. In contrast, in vivo-activated adoptively transferred neutrophils were capable of migrating into the interfollicular areas of the lymph node. Collectively, the data presented here give further insights into the functional behavior of neutrophils within the lymph nodes.
Collapse
|
25
|
Sibler E, He Y, Ducoli L, Keller N, Fujimoto N, Dieterich LC, Detmar M. Single-Cell Transcriptional Heterogeneity of Lymphatic Endothelial Cells in Normal and Inflamed Murine Lymph Nodes. Cells 2021; 10:cells10061371. [PMID: 34199492 PMCID: PMC8229892 DOI: 10.3390/cells10061371] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/22/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022] Open
Abstract
The lymphatic system plays a crucial role in immunity and lymph nodes (LNs) undergo drastic remodeling during inflammation. Here, we used single-cell RNA sequencing to investigate transcriptional changes in lymphatic endothelial cells (LECs) in LNs draining naïve and inflamed skin. We found that subsets of LECs lining the different LN sinuses responded individually to skin inflammation, suggesting that they exert distinct functions under pathological conditions. Among the genes dysregulated during inflammation, we confirmed an up-regulation of CD200 in the LECs lining the subcapsular sinus floor with a possible function in immune regulation. Furthermore, by in silico analysis, we predicted numerous possible interactions of LECs with diverse immune cells in the LNs and found similarities in the transcriptional changes of LN LECs in different skin inflammation settings. In summary, we provide an in-depth analysis of the transcriptional landscape of LN LECs in the naïve state and in skin inflammation.
Collapse
Affiliation(s)
- Eliane Sibler
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
| | - Yuliang He
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
| | - Nadja Keller
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu 520-2192, Japan;
| | - Lothar C. Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland; (E.S.); (Y.H.); (L.D.); (N.K.); (L.C.D.)
- Correspondence:
| |
Collapse
|
26
|
Yeo KP, Lim HY, Angeli V. Leukocyte Trafficking via Lymphatic Vessels in Atherosclerosis. Cells 2021; 10:cells10061344. [PMID: 34072313 PMCID: PMC8229118 DOI: 10.3390/cells10061344] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 02/03/2023] Open
Abstract
In recent years, lymphatic vessels have received increasing attention and our understanding of their development and functional roles in health and diseases has greatly improved. It has become clear that lymphatic vessels are critically involved in acute and chronic inflammation and its resolution by supporting the transport of immune cells, fluid, and macromolecules. As we will discuss in this review, the involvement of lymphatic vessels has been uncovered in atherosclerosis, a chronic inflammatory disease of medium- and large-sized arteries causing deadly cardiovascular complications worldwide. The progression of atherosclerosis is associated with morphological and functional alterations in lymphatic vessels draining the diseased artery. These defects in the lymphatic vasculature impact the inflammatory response in atherosclerosis by affecting immune cell trafficking, lymphoid neogenesis, and clearance of macromolecules in the arterial wall. Based on these new findings, we propose that targeting lymphatic function could be considered in conjunction with existing drugs as a treatment option for atherosclerosis.
Collapse
|
27
|
Structure and Immune Function of Afferent Lymphatics and Their Mechanistic Contribution to Dendritic Cell and T Cell Trafficking. Cells 2021; 10:cells10051269. [PMID: 34065513 PMCID: PMC8161367 DOI: 10.3390/cells10051269] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Afferent lymphatic vessels (LVs) mediate the transport of antigen and leukocytes to draining lymph nodes (dLNs), thereby serving as immunologic communication highways between peripheral tissues and LNs. The main cell types migrating via this route are antigen-presenting dendritic cells (DCs) and antigen-experienced T cells. While DC migration is important for maintenance of tolerance and for induction of protective immunity, T cell migration through afferent LVs contributes to immune surveillance. In recent years, great progress has been made in elucidating the mechanisms of lymphatic migration. Specifically, time-lapse imaging has revealed that, upon entry into capillaries, both DCs and T cells are not simply flushed away with the lymph flow, but actively crawl and patrol and even interact with each other in this compartment. Detachment and passive transport to the dLN only takes place once the cells have reached the downstream, contracting collecting vessel segments. In this review, we describe how the anatomy of the lymphatic network supports leukocyte trafficking and provide updated knowledge regarding the cellular and molecular mechanisms responsible for lymphatic migration of DCs and T cells. In addition, we discuss the relevance of DC and T cell migration through afferent LVs and its presumed implications on immunity.
Collapse
|
28
|
Trac N, Chung EJ. Overcoming physiological barriers by nanoparticles for intravenous drug delivery to the lymph nodes. Exp Biol Med (Maywood) 2021; 246:2358-2371. [PMID: 33957802 DOI: 10.1177/15353702211010762] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The lymph nodes are major sites of cancer metastasis and immune activity, and thus represent important clinical targets. Although not as well-studied compared to subcutaneous administration, intravenous drug delivery is advantageous for lymph node delivery as it is commonly practiced in the clinic and has the potential to deliver therapeutics systemically to all lymph nodes. However, rapid clearance by the mononuclear phagocyte system, tight junctions of the blood vascular endothelium, and the collagenous matrix of the interstitium can limit the efficiency of lymph node drug delivery, which has prompted research into the design of nanoparticle-based drug delivery systems. In this mini review, we describe the physiological and biological barriers to lymph node targeting, how they inform nanoparticle design, and discuss the future outlook of lymph node targeting.
Collapse
Affiliation(s)
- Noah Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, Los Angeles, CA 90033, USA.,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
29
|
In Sickness and in Health: The Immunological Roles of the Lymphatic System. Int J Mol Sci 2021; 22:ijms22094458. [PMID: 33923289 PMCID: PMC8123157 DOI: 10.3390/ijms22094458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 02/06/2023] Open
Abstract
The lymphatic system plays crucial roles in immunity far beyond those of simply providing conduits for leukocytes and antigens in lymph fluid. Endothelial cells within this vasculature are distinct and highly specialized to perform roles based upon their location. Afferent lymphatic capillaries have unique intercellular junctions for efficient uptake of fluid and macromolecules, while expressing chemotactic and adhesion molecules that permit selective trafficking of specific immune cell subsets. Moreover, in response to events within peripheral tissue such as inflammation or infection, soluble factors from lymphatic endothelial cells exert “remote control” to modulate leukocyte migration across high endothelial venules from the blood to lymph nodes draining the tissue. These immune hubs are highly organized and perfectly arrayed to survey antigens from peripheral tissue while optimizing encounters between antigen-presenting cells and cognate lymphocytes. Furthermore, subsets of lymphatic endothelial cells exhibit differences in gene expression relating to specific functions and locality within the lymph node, facilitating both innate and acquired immune responses through antigen presentation, lymph node remodeling and regulation of leukocyte entry and exit. This review details the immune cell subsets in afferent and efferent lymph, and explores the mechanisms by which endothelial cells of the lymphatic system regulate such trafficking, for immune surveillance and tolerance during steady-state conditions, and in response to infection, acute and chronic inflammation, and subsequent resolution.
Collapse
|
30
|
van de Pavert SA. Lymphoid Tissue inducer (LTi) cell ontogeny and functioning in embryo and adult. Biomed J 2021; 44:123-132. [PMID: 33849806 PMCID: PMC8178546 DOI: 10.1016/j.bj.2020.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Innate Lymphoid Cells (ILC) are involved in homeostasis and immunity. Their dynamic differentiation and characterization depend on their tissue of residency and is adapted to their role within these tissues. Lymphoid Tissue inducer (LTi) cells are an ILC member and essential for embryonic lymph node (LN) formation. LNs are formed at pre-defined and strategic positions throughout the body and how LTi cells are initially attracted towards these areas is under debate. Besides their role in LN formation, LTi-like and the closely related ILC type 3 (ILC3) cells have been observed within the embryonic gut. New studies have now shown more information on their origin and differentiation within the embryo. This review will evaluate the embryonic LTi cell origin from a specific embryonic hemogenic wave, which has recently been described in mouse. Moreover, I will discuss their differentiation and similarities with the closely related ILC3 cells in embryo and adult.
Collapse
Affiliation(s)
- Serge A van de Pavert
- Aix-Marseille University, Centre National de la Recherche Scientifique (CNRS), National Institute for Health and Medical Research (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France.
| |
Collapse
|
31
|
Sommer K, Wiendl M, Müller TM, Heidbreder K, Voskens C, Neurath MF, Zundler S. Intestinal Mucosal Wound Healing and Barrier Integrity in IBD-Crosstalk and Trafficking of Cellular Players. Front Med (Lausanne) 2021; 8:643973. [PMID: 33834033 PMCID: PMC8021701 DOI: 10.3389/fmed.2021.643973] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelial barrier is carrying out two major functions: restricting the entry of potentially harmful substances while on the other hand allowing the selective passage of nutrients. Thus, an intact epithelial barrier is vital to preserve the integrity of the host and to prevent development of disease. Vice versa, an impaired intestinal epithelial barrier function is a hallmark in the development and perpetuation of inflammatory bowel disease (IBD). Besides a multitude of genetic, molecular and cellular alterations predisposing for or driving barrier dysintegrity in IBD, the appearance of intestinal mucosal wounds is a characteristic event of intestinal inflammation apparently inducing breakdown of the intestinal epithelial barrier. Upon injury, the intestinal mucosa undergoes a wound healing process counteracting this breakdown, which is controlled by complex mechanisms such as epithelial restitution, proliferation and differentiation, but also immune cells like macrophages, granulocytes and lymphocytes. Consequently, the repair of mucosal wounds is dependent on a series of events including coordinated trafficking of immune cells to dedicated sites and complex interactions among the cellular players and other mediators involved. Therefore, a better understanding of the crosstalk between epithelial and immune cells as well as cell trafficking during intestinal wound repair is necessary for the development of improved future therapies. In this review, we summarize current concepts on intestinal mucosal wound healing introducing the main cellular mediators and their interplay as well as their trafficking characteristics, before finally discussing the clinical relevance and translational approaches to therapeutically target this process in a clinical setting.
Collapse
Affiliation(s)
- Katrin Sommer
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Wiendl
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tanja M Müller
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Karin Heidbreder
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Caroline Voskens
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
32
|
Tomura M, Ikebuchi R, Moriya T, Kusumoto Y. Tracking the fate and migration of cells in live animals with cell-cycle indicators and photoconvertible proteins. J Neurosci Methods 2021; 355:109127. [PMID: 33722643 DOI: 10.1016/j.jneumeth.2021.109127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/13/2022]
Abstract
Cell migration and cell proliferation are the basic principles that make up a living organism, and both biologically and medically. In order to understand living organism and biological phenomena, it is essential to track the migration, proliferation, and fate of cells in living cells and animals and to clarify the properties and molecular expression of cells. Recent developments in novel fluorescent proteins have made it possible to observe cell migration and proliferation as the cell cycle at the single-cell level in living individuals and tissues. Here, we introduce cell cycle visualization of living cells and animals by Fucci (Fluorescent Ubiquitination-based Cell Cycle Indicator) system and in situ cell labeling of cells and tracking cell migration by photoactivatable and photoconvertible proteins. In addition, we will present our established methods as an example of combines above tools with single-cell molecular expression analysis to reveal the fate of migrating cells at single cell level.
Collapse
Affiliation(s)
- Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan.
| | - Ryoyo Ikebuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan; Research Fellow of Japan Society for the Promotion of Science, Japan; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Taiki Moriya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan
| | - Yutaka Kusumoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan
| |
Collapse
|
33
|
Künnapuu J, Bokharaie H, Jeltsch M. Proteolytic Cleavages in the VEGF Family: Generating Diversity among Angiogenic VEGFs, Essential for the Activation of Lymphangiogenic VEGFs. BIOLOGY 2021; 10:167. [PMID: 33672235 PMCID: PMC7926383 DOI: 10.3390/biology10020167] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/24/2022]
Abstract
Specific proteolytic cleavages turn on, modify, or turn off the activity of vascular endothelial growth factors (VEGFs). Proteolysis is most prominent among the lymph-angiogenic VEGF-C and VEGF-D, which are synthesized as precursors that need to undergo enzymatic removal of their C- and N-terminal propeptides before they can activate their receptors. At least five different proteases mediate the activating cleavage of VEGF-C: plasmin, ADAMTS3, prostate-specific antigen, cathepsin D, and thrombin. All of these proteases except for ADAMTS3 can also activate VEGF-D. Processing by different proteases results in distinct forms of the "mature" growth factors, which differ in affinity and receptor activation potential. The "default" VEGF-C-activating enzyme ADAMTS3 does not activate VEGF-D, and therefore, VEGF-C and VEGF-D do function in different contexts. VEGF-C itself is also regulated in different contexts by distinct proteases. During embryonic development, ADAMTS3 activates VEGF-C. The other activating proteases are likely important for non-developmental lymphangiogenesis during, e.g., tissue regeneration, inflammation, immune response, and pathological tumor-associated lymphangiogenesis. The better we understand these events at the molecular level, the greater our chances of developing successful therapies targeting VEGF-C and VEGF-D for diseases involving the lymphatics such as lymphedema or cancer.
Collapse
Affiliation(s)
- Jaana Künnapuu
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Honey Bokharaie
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
| | - Michael Jeltsch
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; (J.K.); (H.B.)
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Wihuri Research Institute, 00290 Helsinki, Finland
| |
Collapse
|
34
|
Cai C, Sun H, Hu L, Fan Z. Visualization of integrin molecules by fluorescence imaging and techniques. ACTA ACUST UNITED AC 2021; 45:229-257. [PMID: 34219865 PMCID: PMC8249084 DOI: 10.32604/biocell.2021.014338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Integrin molecules are transmembrane αβ heterodimers involved in cell adhesion, trafficking, and signaling. Upon activation, integrins undergo dynamic conformational changes that regulate their affinity to ligands. The physiological functions and activation mechanisms of integrins have been heavily discussed in previous studies and reviews, but the fluorescence imaging techniques -which are powerful tools for biological studies- have not. Here we review the fluorescence labeling methods, imaging techniques, as well as Förster resonance energy transfer assays used to study integrin expression, localization, activation, and functions.
Collapse
Affiliation(s)
- Chen Cai
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, 92093, USA
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450051, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| |
Collapse
|
35
|
Jin Z, Li X, Zhang X, Paul D, Xu T, Wu A. Engineering the fate and function of human T-cells via 3D bioprinting. Biofabrication 2020; 13. [PMID: 33348331 DOI: 10.1088/1758-5090/abd56b] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/21/2020] [Indexed: 12/28/2022]
Abstract
T-cell immunotherapy holds promise for the treatment of cancer, infection, and autoimmune diseases. Nevertheless, T-cell therapy is limited by low cell expansion efficiency ex vivo and functional deficits. Here we describe two 3D bioprinting systems made by different biomaterials that mimic the in vivo formation of natural lymph vessels and lymph nodes which modulate T-cell with distinct fates and functions. We observe that coaxial alginate fibers promote T-cell expansion, less exhausted and enable CD4+ T-cell differentiation into central memory-like phenotype (Tcm), CD8+ T-cells differentiation into effector memory subsets (Tem), while alginate-gelatin scaffolds bring T-cells into a relatively resting state. Both of the two bioprinting methods are strikingly different from a standard suspension system. The former bioprinting method yields a new system for T-cell therapy and the latter method can be useful for making an immune-chip to elucidate links between immune response and disease.
Collapse
Affiliation(s)
- Zhizhong Jin
- The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China., Shenyang, Liaoning, 110001, CHINA
| | - Xinda Li
- Department of Mechanical Engineering, Tsinghua University, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China., Beijing, 100084, CHINA
| | - Xinzhi Zhang
- Tsinghua University, East China Institute of Digital Medical Engineering, Shangrao, 334000, China., Medprin Regenerative Medical Technologies Co., Ltd, Shenzhen, 518102, China., Beijing, 334000, CHINA
| | - Desousa Paul
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK., University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK., Edinburgh, EH16 4SB, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Tao Xu
- Institute of Materials Processing Equipment and Automation, Department of Mechanical Engneering,, Tsinghua University, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China., Department of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, 518055, China., Beijing, 100084, CHINA
| | - Anhua Wu
- Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, China., Shenyang, 110001, CHINA
| |
Collapse
|
36
|
Doustar J, Rentsendorj A, Torbati T, Regis GC, Fuchs D, Sheyn J, Mirzaei N, Graham SL, Shah PK, Mastali M, Van Eyk JE, Black KL, Gupta VK, Mirzaei M, Koronyo Y, Koronyo‐Hamaoui M. Parallels between retinal and brain pathology and response to immunotherapy in old, late-stage Alzheimer's disease mouse models. Aging Cell 2020; 19:e13246. [PMID: 33090673 PMCID: PMC7681044 DOI: 10.1111/acel.13246] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/14/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Despite growing evidence for the characteristic signs of Alzheimer's disease (AD) in the neurosensory retina, our understanding of retina-brain relationships, especially at advanced disease stages and in response to therapy, is lacking. In transgenic models of AD (APPSWE/PS1∆E9; ADtg mice), glatiramer acetate (GA) immunomodulation alleviates disease progression in pre- and early-symptomatic disease stages. Here, we explored the link between retinal and cerebral AD-related biomarkers, including response to GA immunization, in cohorts of old, late-stage ADtg mice. This aged model is considered more clinically relevant to the age-dependent disease. Levels of synaptotoxic amyloid β-protein (Aβ)1-42, angiopathic Aβ1-40, non-amyloidogenic Aβ1-38, and Aβ42/Aβ40 ratios tightly correlated between paired retinas derived from oculus sinister (OS) and oculus dexter (OD) eyes, and between left and right posterior brain hemispheres. We identified lateralization of Aβ burden, with one-side dominance within paired retinal and brain tissues. Importantly, OS and OD retinal Aβ levels correlated with their cerebral counterparts, with stronger contralateral correlations and following GA immunization. Moreover, immunomodulation in old ADtg mice brought about reductions in cerebral vascular and parenchymal Aβ deposits, especially of large, dense-core plaques, and alleviation of microgliosis and astrocytosis. Immunization further enhanced cerebral recruitment of peripheral myeloid cells and synaptic preservation. Mass spectrometry analysis identified new parallels in retino-cerebral AD-related pathology and response to GA immunization, including restoration of homeostatic glutamine synthetase expression. Overall, our results illustrate the viability of immunomodulation-guided CNS repair in old AD model mice, while shedding light onto similar retino-cerebral responses to intervention, providing incentives to explore retinal AD biomarkers.
Collapse
Affiliation(s)
- Jonah Doustar
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Altan Rentsendorj
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Tania Torbati
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
- College of Osteopathic Medicine of the PacificWestern University of Health SciencesPomonaCAUSA
| | - Giovanna C. Regis
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Dieu‐Trang Fuchs
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Julia Sheyn
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Nazanin Mirzaei
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Stuart L. Graham
- Department of Clinical MedicineMacquarie UniversitySydneyNSWAustralia
- Save Sight InstituteSydney UniversitySydneyNSWAustralia
| | - Prediman K. Shah
- Oppenheimer Atherosclerosis Research CenterCedars‐Sinai Heart InstituteLos AngelesCAUSA
| | - Mitra Mastali
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCAUSA
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCAUSA
| | - Jennifer E. Van Eyk
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCAUSA
- Barbara Streisand Women’s Heart CenterCedars‐Sinai Medical CenterLos AngelesCAUSA
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Keith L. Black
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Vivek K. Gupta
- Department of Molecular SciencesMacquarie UniversitySydneyNSWAustralia
| | - Mehdi Mirzaei
- Department of Clinical MedicineMacquarie UniversitySydneyNSWAustralia
- Department of Molecular SciencesMacquarie UniversitySydneyNSWAustralia
- Australian Proteome Analysis FacilityMacquarie UniversitySydneyNSWAustralia
| | - Yosef Koronyo
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
| | - Maya Koronyo‐Hamaoui
- Department of NeurosurgeryCedars‐Sinai Medical CenterMaxine Dunitz Neurosurgical Research InstituteLos AngelesCAUSA
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCAUSA
| |
Collapse
|
37
|
Principles of Leukocyte Migration Strategies. Trends Cell Biol 2020; 30:818-832. [DOI: 10.1016/j.tcb.2020.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022]
|
38
|
Oliver G, Kipnis J, Randolph GJ, Harvey NL. The Lymphatic Vasculature in the 21 st Century: Novel Functional Roles in Homeostasis and Disease. Cell 2020; 182:270-296. [PMID: 32707093 PMCID: PMC7392116 DOI: 10.1016/j.cell.2020.06.039] [Citation(s) in RCA: 430] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/17/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Mammals have two specialized vascular circulatory systems: the blood vasculature and the lymphatic vasculature. The lymphatic vasculature is a unidirectional conduit that returns filtered interstitial arterial fluid and tissue metabolites to the blood circulation. It also plays major roles in immune cell trafficking and lipid absorption. As we discuss in this review, the molecular characterization of lymphatic vascular development and our understanding of this vasculature's role in pathophysiological conditions has greatly improved in recent years, changing conventional views about the roles of the lymphatic vasculature in health and disease. Morphological or functional defects in the lymphatic vasculature have now been uncovered in several pathological conditions. We propose that subtle asymptomatic alterations in lymphatic vascular function could underlie the variability seen in the body's response to a wide range of human diseases.
Collapse
Affiliation(s)
- Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
39
|
Differential retention of lymph-borne CD8 memory T cell subsets in the subcapsular sinus of resting and inflamed lymph nodes. Cell Mol Immunol 2020; 18:1317-1319. [PMID: 32398805 DOI: 10.1038/s41423-020-0451-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 11/08/2022] Open
|