Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Elife ; 122023 Dec 21.
Article in English | MEDLINE | ID: mdl-38127067

ABSTRACT

Intestinal intraepithelial lymphocytes (IELs) are characterized by an unusual phenotype and developmental pathway, yet their specific ligands and functions remain largely unknown. Here by analysis of QFL T cells, a population of CD8+ T cells critical for monitoring the MHC I antigen processing pathway, we established that unconventional Qa-1b-restricted CD8+ T cells are abundant in intestinal epithelium. We found that QFL T cells showed a Qa-1b-dependent unconventional phenotype in the spleen and small intestine of naïve wild-type mice. The splenic QFL T cells showed innate-like functionality exemplified by rapid response to cytokines or antigens, while the gut population was refractory to stimuli. Microbiota was required for the maintenance, but not the initial gut homing of QFL T cells. Moreover, monocolonization with Pediococcus pentosaceus, which expresses a peptide that cross-activated QFL T cells, was sufficient to maintain QFL T cells in the intestine. Thus, microbiota is critical for shaping the Qa-1b-restricted IEL landscape.


Subject(s)
Bacteria , CD8-Positive T-Lymphocytes , Animals , Mice , Epithelium , Cytokines , Intestinal Mucosa
2.
bioRxiv ; 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37645777

ABSTRACT

Regulatory T cells (Treg) are crucial immune modulators, yet the exact mechanism of thymic Treg development remains controversial. Here, we present the first direct evidence for H2-O, an MHC class II peptide editing molecular chaperon, on selection of thymic Tregs. We provide evidence that lack of H2-O in the thymic medulla promotes thymic Treg development and leads to an increased peripheral Treg frequency. Single-cell RNA-sequencing (scRNA-seq) analysis of splenic CD4 T cells revealed not only of an enrichment of effector-like Tregs but also of activated CD4 T cells in the absence of H2-O. Our data support two concepts; a) lack of H2-O expression in the thymic medulla creates an environment permissive to Treg development and, b) that loss of H2-O drives increased basal auto-stimulation of CD4 T cells. These findings can help in better understanding of predispositions to autoimmunity and design of therapeutics for treatment of autoimmune diseases.

3.
bioRxiv ; 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-36909616

ABSTRACT

Intestinal intraepithelial lymphocytes (IELs) are characterized by an unusual phenotype and developmental pathway, yet their specific ligands and functions remain largely unknown. Here by analysis of QFL T cells, a population of CD8 + T cells critical for monitoring the MHC I antigen processing pathway, we established that unconventional Qa-1 b -restricted CD8 + T cells are abundant in intestinal epithelium. We found that QFL T cells showed a Qa-1 b -dependent unconventional phenotype in the spleen and small intestine of naïve wild-type mice. The splenic QFL T cells showed innate-like functionality exemplified by rapid response to cytokines or antigen, while the gut population was refractory to stimuli. Microbiota was required for the maintenance, but not the initial gut homing of QFL T cells. Moreover, monocolonization with Pediococcus pentosaceus, which expresses a peptide that cross-activated QFL T cells, was sufficient to maintain QFL T cells in the intestine. Thus, microbiota is critical for shaping the Qa-1 b -restricted IEL landscape.

4.
Front Immunol ; 14: 1304798, 2023.
Article in English | MEDLINE | ID: mdl-38250071

ABSTRACT

Regulatory T cells (Treg) are crucial immune modulators, yet the exact mechanism of thymic Treg development remains controversial. Here, we present the first direct evidence for H2-O, an MHC class II peptide editing molecular chaperon, on selection of thymic Tregs. We identified that lack of H2-O in the thymic medulla promotes thymic Treg development and leads to an increased peripheral Treg frequency. Single-cell RNA-sequencing (scRNA-seq) analysis of splenic CD4 T cells revealed not only an enrichment of effector-like Tregs, but also activated CD4 T cells in the absence of H2-O. Our data support two concepts; a) lack of H2-O expression in the thymic medulla creates an environment permissive to Treg development and, b) that loss of H2-O drives increased basal auto-stimulation of CD4 T cells. These findings can help in better understanding of predispositions to autoimmunity and design of therapeutics for treatment of autoimmune diseases.


Subject(s)
Autoimmune Diseases , CD4-Positive T-Lymphocytes , Humans , Lymphocyte Activation/genetics , T-Lymphocytes, Regulatory , Histocompatibility Antigens Class II , Cell Differentiation
6.
Cell Rep ; 36(3): 109422, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289373

ABSTRACT

Tumor-associated tertiary lymphoid structures (TA-TLS) are associated with enhanced patient survival and responsiveness to cancer therapies, but the mechanisms underlying their development are unknown. We show here that TA-TLS development in murine melanoma is orchestrated by cancer-associated fibroblasts (CAF) with characteristics of lymphoid tissue organizer cells that are induced by tumor necrosis factor receptor signaling. CAF organization into reticular networks is mediated by CD8 T cells, while CAF accumulation and TA-TLS expansion depend on CXCL13-mediated recruitment of B cells expressing lymphotoxin-α1ß2. Some of these elements are also overrepresented in human TA-TLS. Additionally, we demonstrate that immunotherapy induces more and larger TA-TLS that are more often organized with discrete T and B cell zones, and that TA-TLS presence, number, and size are correlated with reduced tumor size and overall response to checkpoint immunotherapy. This work provides a platform for manipulating TA-TLS development as a cancer immunotherapy strategy.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Neoplasms/immunology , Neoplasms/pathology , Tertiary Lymphoid Structures/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Humans , Immunotherapy , Lymphocyte Activation/immunology , Lymphotoxin beta Receptor/metabolism , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Neoplasms/therapy , Peritoneum/pathology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction
7.
Front Immunol ; 10: 816, 2019.
Article in English | MEDLINE | ID: mdl-31057546

ABSTRACT

Lymphatic and blood vessels are formed by specialized lymphatic endothelial cells (LEC) and blood endothelial cells (BEC), respectively. These endothelial populations not only form peripheral tissue vessels, but also critical supporting structures in secondary lymphoid organs, particularly the lymph node (LN). Lymph node LEC (LN-LEC) also have been shown to have important immunological functions that are not observed in LEC from tissue lymphatics. LN-LEC can maintain peripheral tolerance through direct presentation of self-antigen via MHC-I, leading to CD8 T cell deletion; and through transfer of self-antigen to dendritic cells for presentation via MHC-II, resulting in CD4 T cell anergy. LN-LEC also can capture and archive foreign antigens, transferring them to dendritic cells for maintenance of memory CD8 T cells. The molecular basis for these functional elaborations in LN-LEC remain largely unexplored, and it is also unclear whether blood endothelial cells in LN (LN-BEC) might express similar enhanced immunologic functionality. Here, we used RNA-Seq to compare the transcriptomic profiles of freshly isolated murine LEC and BEC from LN with one another and with freshly isolated LEC from the periphery (diaphragm). We show that LN-LEC, LN-BEC, and diaphragm LEC (D-LEC) are transcriptionally distinct from one another, demonstrating both lineage and tissue-specific functional specializations. Surprisingly, tissue microenvironment differences in gene expression profiles were more numerous than those determined by endothelial cell lineage specification. In this regard, both LN-localized endothelial cell populations show a variety of functional elaborations that suggest how they may function as antigen presenting cells, and also point to as yet unexplored roles in both positive and negative regulation of innate and adaptive immune responses. The present work has defined in depth gene expression differences that point to functional specializations of endothelial cell populations in different anatomical locations, but especially the LN. Beyond the analyses provided here, these data are a resource for future work to uncover mechanisms of endothelial cell functionality.


Subject(s)
Blood Vessels/cytology , Endothelial Cells/physiology , Lymph Nodes/cytology , Lymphatic Vessels/cytology , Transcriptome , Animals , Antigen Presentation , Cell Adhesion Molecules/metabolism , Cellular Microenvironment , Chemokines/metabolism , Diaphragm/cytology , Endothelial Cells/immunology , Extracellular Matrix/metabolism , Mice , Mice, Inbred C57BL , RNA-Seq , Signal Transduction
8.
Virchows Arch ; 474(4): 463-474, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30631935

ABSTRACT

For many years, the gold standard cancer grading and staging had focused on the characteristics of the cancer cells and often disregarded the non-tumoral cell compartments. The expansion of research on the tumor immune microenvironment, the successes and dissemination of immunotherapies to treat cancer, and the open access to large -omic databases have allowed the development of novel powerful immune-based prognostic and theranostic biomarkers. Although they often correlate with histopathologic characteristics and TNM staging, in many instances, they are independently associated with, and potentially superior predictors of, the patient's prognosis and response to immunotherapies. As pathologists in the era of precision medicine, we are uniquely positioned to participate in the integration of these histologic and molecular features of the tumor microenvironment to provide the best prognostic information to clinicians and patients. In this review, we summarize some of the most important immune-related prognostic biomarkers in solid cancer, how they integrate with traditional histopathologic (i.e., staging and grading) and novel molecular stratification systems, and their potential role as predictors to response to agents blocking the PD-1/PD-L1 axis.


Subject(s)
Biomarkers, Tumor/analysis , Medical Oncology/trends , Neoplasms/classification , Precision Medicine/trends , Humans , Medical Oncology/methods , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology
9.
Br J Cancer ; 120(1): 45-53, 2019 01.
Article in English | MEDLINE | ID: mdl-30413828

ABSTRACT

The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.


Subject(s)
Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cell Proliferation/genetics , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/pathology
10.
Methods Mol Biol ; 1845: 241-257, 2018.
Article in English | MEDLINE | ID: mdl-30141017

ABSTRACT

Tertiary lymphoid structures (TLS) are transient ectopic lymphoid aggregates that often share structural similarities to conventional secondary lymphoid organs. In a variety of solid cancers, the presence of these structures commonly correlates with high densities of tumor-infiltrating T lymphocytes and prolonged patient survival. These observations suggest that TLS act as sites for the development of beneficial antitumor immune responses. However, few murine tumor models have been described that could enable a more comprehensive understanding of the functionality of TLS in solid cancers. We previously reported that murine B16-F1 melanoma or Lewis lung carcinoma cells transfected to express the model antigen ovalbumin form intratumoral TLS after implantation into the peritoneal cavity of C57BL/6 mice. In this chapter, we describe immunofluorescent microscopy and flow cytometry approaches for identifying and characterizing intratumoral TLS. Additionally, we describe an adoptive transfer method for demonstrating the infiltration of naïve T cells into B16-OVA melanoma tumors via the lymph node-like vasculature, which is an essential functional feature of tumor-associated TLS.


Subject(s)
Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology , Adoptive Transfer , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Flow Cytometry , Fluorescent Antibody Technique , Melanoma, Experimental/metabolism , Mice , Microscopy, Fluorescence , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tertiary Lymphoid Structures/metabolism , Tumor Microenvironment/immunology
11.
J Immunol ; 200(2): 432-442, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29311385

ABSTRACT

Limited representation of intratumoral immune cells is a major barrier to tumor control. However, simply enhancing immune responses in tumor-draining lymph nodes or through adoptive transfer may not overcome the limited ability of tumor vasculature to support effector infiltration. An alternative is to promote a sustained immune response intratumorally. This idea has gained traction with the observation that many tumors are associated with tertiary lymphoid structures (TLS), which organizationally resemble lymph nodes. These peri- and intratumoral structures are usually, but not always, associated with positive prognoses in patients. Preclinical and clinical data support a role for TLS in modulating immunity in the tumor microenvironment. However, there appear to be varied functions of TLS, potentially based on their structure or location in relation to the tumor or the origin or location of the tumor itself. Understanding more about TLS development, composition, and function may offer new therapeutic opportunities to modulate antitumor immunity.


Subject(s)
Immunity , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Tertiary Lymphoid Structures/immunology , Tumor Microenvironment/immunology , Animals , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Prognosis , Tertiary Lymphoid Structures/metabolism
12.
Cancer Immunol Res ; 5(12): 1062-1073, 2017 12.
Article in English | MEDLINE | ID: mdl-29097419

ABSTRACT

Although CD8+ T cells are critical for controlling tumors, how they are recruited and home to primary and metastatic lesions is incompletely understood. We characterized the homing receptor (HR) ligands on tumor vasculature to determine what drives their expression and their role in T-cell entry. The anatomic location of B16-OVA tumors affected the expression of E-selectin, MadCAM-1, and VCAM-1, whereas the HR ligands CXCL9 and ICAM-1 were expressed on the vasculature regardless of location. VCAM-1 and CXCL9 expression was induced by IFNγ-secreting adaptive immune cells. VCAM-1 and CXCL9/10 enabled CD8+ T-cell effectors expressing α4ß1 integrin and CXCR3 to enter both subcutaneous and peritoneal tumors, whereas E-selectin enabled E-selectin ligand+ effectors to enter subcutaneous tumors. However, MadCAM-1 did not mediate α4ß7+ effector entry into peritoneal tumors due to an unexpected lack of luminal expression. These data establish the relative importance of certain HRs expressed on activated effectors and certain HR ligands expressed on tumor vasculature in the effective immune control of tumors. Cancer Immunol Res; 5(12); 1062-73. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/etiology , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Adaptive Immunity , Animals , Biomarkers , Cell Line, Tumor , Chemokine CXCL9/genetics , E-Selectin/genetics , E-Selectin/metabolism , Integrin alpha4beta1/genetics , Ligands , Melanoma, Experimental , Mice , Models, Biological , Receptors, Lymphocyte Homing/metabolism , Vascular Cell Adhesion Molecule-1/genetics
14.
Adv Cancer Res ; 128: 263-307, 2015.
Article in English | MEDLINE | ID: mdl-26216636

ABSTRACT

CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/blood supply , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Tumor Microenvironment/immunology , Animals , Humans
15.
Nature ; 523(7560): 337-41, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26030524

ABSTRACT

One of the characteristics of the central nervous system is the lack of a classical lymphatic drainage system. Although it is now accepted that the central nervous system undergoes constant immune surveillance that takes place within the meningeal compartment, the mechanisms governing the entrance and exit of immune cells from the central nervous system remain poorly understood. In searching for T-cell gateways into and out of the meninges, we discovered functional lymphatic vessels lining the dural sinuses. These structures express all of the molecular hallmarks of lymphatic endothelial cells, are able to carry both fluid and immune cells from the cerebrospinal fluid, and are connected to the deep cervical lymph nodes. The unique location of these vessels may have impeded their discovery to date, thereby contributing to the long-held concept of the absence of lymphatic vasculature in the central nervous system. The discovery of the central nervous system lymphatic system may call for a reassessment of basic assumptions in neuroimmunology and sheds new light on the aetiology of neuroinflammatory and neurodegenerative diseases associated with immune system dysfunction.


Subject(s)
Central Nervous System/anatomy & histology , Central Nervous System/immunology , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/immunology , Animals , Central Nervous System/cytology , Cranial Sinuses/anatomy & histology , Female , Humans , Immune Tolerance/immunology , Immunologic Surveillance/immunology , Lymphatic Vessels/cytology , Male , Meninges/anatomy & histology , Meninges/cytology , Meninges/immunology , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/immunology
16.
Nat Commun ; 6: 7114, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25968334

ABSTRACT

The presence of lymph node (LN)-like vasculature in tumours, characterized by expression of peripheral node addressin and chemokine CCL21, is correlated with T-cell infiltration and positive prognosis in breast cancer and melanoma patients. However, mechanisms controlling the development of LN-like vasculature and how it might contribute to a beneficial outcome for cancer patients are unknown. Here we demonstrate that LN-like vasculature is present in murine models of melanoma and lung carcinoma. It enables infiltration by naive T cells that significantly delay tumour outgrowth after intratumoral activation. Development of this vasculature is controlled by a mechanism involving effector CD8 T cells and NK cells that secrete LTα3 and IFNγ. LN-like vasculature is also associated with organized aggregates of B lymphocytes and gp38(+) fibroblasts, which resemble tertiary lymphoid organs that develop in models of chronic inflammation. These results establish LN-like vasculature as both a consequence of and key contributor to anti-tumour immunity.


Subject(s)
Neoplasms, Experimental/pathology , T-Lymphocytes/physiology , Animals , Antigens, Neoplasm , Chemokine CCL21/genetics , Chemokine CCL21/metabolism , Female , Gene Expression Regulation , Immunoglobulins , Lymphotoxin beta Receptor/immunology , Mice , Mice, Knockout , Tumor Microenvironment
17.
Nat Commun ; 6: 6771, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25857745

ABSTRACT

Lymphatic endothelial cells (LECs) directly express peripheral tissue antigens and induce CD8 T-cell deletional tolerance. LECs express MHC-II molecules, suggesting they might also tolerize CD4 T cells. We demonstrate that when ß-galactosidase (ß-gal) is expressed in LECs, ß-gal-specific CD8 T cells undergo deletion via the PD-1/PD-L1 and LAG-3/MHC-II pathways. In contrast, LECs do not present endogenous ß-gal in the context of MHC-II molecules to ß-gal-specific CD4 T cells. Lack of presentation is independent of antigen localization, as membrane-bound haemagglutinin and I-Eα are also not presented by MHC-II molecules. LECs express invariant chain and cathepsin L, but not H2-M, suggesting that they cannot load endogenous antigenic peptides onto MHC-II molecules. Importantly, LECs transfer ß-gal to dendritic cells, which subsequently present it to induce CD4 T-cell anergy. Therefore, LECs serve as an antigen reservoir for CD4 T-cell tolerance, and MHC-II molecules on LECs are used to induce CD8 T-cell tolerance via LAG-3.


Subject(s)
Antigen Presentation/genetics , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Peripheral Tolerance/genetics , Animals , Antigens/genetics , Antigens/immunology , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cathepsin L/genetics , Cathepsin L/immunology , Clonal Anergy/genetics , Dendritic Cells/cytology , Endothelial Cells/cytology , Gene Expression , Hemagglutinins/genetics , Hemagglutinins/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , beta-Galactosidase/genetics , beta-Galactosidase/immunology , Lymphocyte Activation Gene 3 Protein
18.
Front Immunol ; 4: 241, 2013.
Article in English | MEDLINE | ID: mdl-23966998

ABSTRACT

T cell activation induces homing receptors that bind ligands on peripheral tissue vasculature, programing movement to sites of infection and injury. There are three major types of CD8 effector T cells based on homing receptor expression, which arise in distinct lymphoid organs. Recent publications indicate that naïve, effector, and memory T cell migration is more complex than once thought; while many effectors enter peripheral tissues, some re-enter lymph nodes (LN), and contain central memory precursors. LN re-entry can depend on CD62L or peripheral tissue homing receptors. Memory T cells in LN tend to express the same homing receptors as their forebears, but often are CD62Lneg. Homing receptors also control CD8 T cell tumor entry. Tumor vasculature has low levels of many peripheral tissue homing receptor ligands, but portions of it resemble high endothelial venules (HEV), enabling naïve T cell entry, activation, and subsequent effector activity. This vasculature is associated with positive prognoses in humans, suggesting it may sustain ongoing anti-tumor responses. These findings reveal new roles for homing receptors expressed by naïve, effector, and memory CD8 T cells in controlling entry into lymphoid and non-lymphoid tissues.

SELECTION OF CITATIONS
SEARCH DETAIL
...