1
|
Huang HW, Wu S, Chowdhury EA, Shah DK. Expansion of platform physiologically-based pharmacokinetic model for monoclonal antibodies towards different preclinical species: cats, sheep, and dogs. J Pharmacokinet Pharmacodyn 2024; 51:621-638. [PMID: 37947924 DOI: 10.1007/s10928-023-09893-5] [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: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Monoclonal antibodies (mAbs) are becoming an important therapeutic option in veterinary medicine, and understanding the pharmacokinetic (PK) of mAbs in higher-order animal species is also important for human drug development. To better understand the PK of mAbs in these animals, here we have expanded a platform physiological-based pharmacokinetic (PBPK) model to characterize the disposition of mAbs in three different preclinical species: cats, sheep, and dogs. We obtained PK data for mAbs and physiological parameters for the three different species from the literature. We were able to describe the PK of mAbs following intravenous (IV) or subcutaneous administration in cats, IV administration in sheep, and IV administration dogs reasonably well by fixing the physiological parameters and just estimating the parameters related to the binding of mAbs to the neonatal Fc receptor. The platform PBPK model presented here provides a quantitative tool to predict the plasma PK of mAbs in dogs, cats, and sheep. The model can also predict mAb PK in different tissues where the site of action might be located. As such, the mAb PBPK model presented here can facilitate the discovery, development, and preclinical-to-clinical translation of mAbs for veterinary and human medicine. The model can also be modified in the future to account for more detailed compartments for certain organs, different pathophysiology in the animals, and target-mediated drug disposition.
Collapse
Affiliation(s)
- Hsien-Wei Huang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Shengjia Wu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Ekram A Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
2
|
Nikmaneshi MR, Baish JW, Zhou H, Padera TP, Munn LL. Transport Barriers Influence the Activation of Anti-Tumor Immunity: A Systems Biology Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304076. [PMID: 37949675 PMCID: PMC10754116 DOI: 10.1002/advs.202304076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/07/2023] [Indexed: 11/12/2023]
Abstract
Effective anti-cancer immune responses require activation of one or more naïve T cells. If the correct naïve T cell encounters its cognate antigen presented by an antigen presenting cell, then the T cell can activate and proliferate. Here, mathematical modeling is used to explore the possibility that immune activation in lymph nodes is a rate-limiting step in anti-cancer immunity and can affect response rates to immune checkpoint therapy. The model provides a mechanistic framework for optimizing cancer immunotherapy and developing testable solutions to unleash anti-tumor immune responses for more patients with cancer. The results show that antigen production rate and trafficking of naïve T cells into the lymph nodes are key parameters and that treatments designed to enhance tumor antigen production can improve immune checkpoint therapies. The model underscores the potential of radiation therapy in augmenting tumor immunogenicity and neoantigen production for improved ICB therapy, while emphasizing the need for careful consideration in cases where antigen levels are already sufficient to avoid compromising the immune response.
Collapse
Affiliation(s)
- Mohammad R. Nikmaneshi
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - James W. Baish
- Biomedical EngineeringBucknell UniversityLewisburgPA17837USA
| | - Hengbo Zhou
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Timothy P. Padera
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Lance L. Munn
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| |
Collapse
|
3
|
Jiang S, Sun B, Zhang Y, Han J, Zhou Y, Pan C, Wang H, Si N, Bian B, Wang L, Wang L, Wei X, Zhao H. The immediate adverse drug reactions induced by ShenMai Injection are mediated by thymus-derived T cells and associated with RhoA/ROCK signaling pathway. Front Immunol 2023; 14:1135701. [PMID: 37026017 PMCID: PMC10070857 DOI: 10.3389/fimmu.2023.1135701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction The mechanism of the immediate adverse drug reactions (ADRs) induced by ShenMai injection (SMI) has not been completely elucidated. Within 30 minutes, the ears and lungs of mice injected with SMI for the first time showed edema and exudation reactions. These reactions were different from the IV hypersensitivity. The theory of pharmacological interaction with immune receptor (p-i) offered a new insight into the mechanisms of immediate ADRs induced by SMI. Methods In this study, we determined that the ADRs were mediated by thymus-derived T cells through the different reactions of BALB/c mice (thymus-derived T cell normal) and BALB/c nude mice (thymus-derived T cell deficient) after injecting SMI. The flow cytometric analysis, cytokine bead array (CBA) assay and untargeted metabolomics were used to explain the mechanisms of the immediate ADRs. Moreover, the activation of the RhoA/ROCK signaling pathway was detected by western blot analysis. Results In BALB/c mice, the vascular leakage and histopathology results showed the occurrence of the immediate ADRs induced by SMI. The flow cytometric analysis revealed that CD4+ T cell subsets (Th1/Th2, Th17/Treg) were imbalanced. And the levels of cytokines such as IL-2, IL-4, IL12P70 and INF-γ increased significantly. However, in BALB/c nude mice, all the indicators mentioned above have not changed significantly. The metabolic profile of both BALB/c mice and BALB/c nude mice was significantly changed after injecting SMI, and the notable increase in lysolecithin level might have a greater association with the immediate ADRs induced by SMI. The Spearman correlation analysis revealed that LysoPC (18:3(6Z,9Z,12Z)/0:0) showed a significant positive correlation with cytokines. After injecting SMI, the levels of RhoA/ROCK signaling pathway-related protein increased significantly in BALB/c mice. Protein-protein interaction (PPI) showed that the increased lysolecithin levels might be related to the activation of the RhoA/ROCK signaling pathway. Discussion Together, the results of our study revealed that the immediate ADRs induced by SMI were mediated by thymus-derived T cells, and elucidated the mechanisms of such ADRs. This study provided new insights into the underlying mechanism of immediate ADRs induced by SMI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiaolu Wei
- *Correspondence: Xiaolu Wei, ; Haiyu Zhao,
| | - Haiyu Zhao
- *Correspondence: Xiaolu Wei, ; Haiyu Zhao,
| |
Collapse
|
4
|
Milutinovic S, Abe J, Jones E, Kelch I, Smart K, Lauder SN, Somerville M, Ware C, Godkin A, Stein JV, Bogle G, Gallimore A. Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells. CANCER RESEARCH COMMUNICATIONS 2022; 2:1641-1656. [PMID: 36704666 PMCID: PMC7614106 DOI: 10.1158/2767-9764.crc-21-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
High endothelial venules (HEV) are specialized post capillary venules that recruit naïve T cells and B cells into secondary lymphoid organs (SLO) such as lymph nodes (LN). Expansion of HEV networks in SLOs occurs following immune activation to support development of an effective immune response. In this study, we used a carcinogen-induced model of fibrosarcoma to examine HEV remodeling after depletion of regulatory T cells (Treg). We used light sheet fluorescence microscopy imaging to visualize entire HEV networks, subsequently applying computational tools to enable topological mapping and extraction of numerical descriptors of the networks. While these analyses revealed profound cancer- and immune-driven alterations to HEV networks within LNs, these changes did not identify successful responses to treatment. The presence of HEV networks within tumors did however clearly distinguish responders from nonresponders. Finally, we show that a successful treatment response is dependent on coupling tumor-associated HEV (TA-HEV) development to T-cell activation implying that T-cell activation acts as the trigger for development of TA-HEVs which subsequently serve to amplify the immune response by facilitating extravasation of T cells into the tumor mass.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Emma Jones
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Inken Kelch
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn Smart
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sarah N. Lauder
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michelle Somerville
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Carl Ware
- Laboratory of Molecular Immunology, Sanford Burnham Prebys, La Jolla, California
| | - Andrew Godkin
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gib Bogle
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Awen Gallimore
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
5
|
Johnson SC, Frattolin J, Edgar LT, Jafarnejad M, Moore Jr JE. Lymph node swelling combined with temporary effector T cell retention aids T cell response in a model of adaptive immunity. J R Soc Interface 2021; 18:20210464. [PMID: 34847790 PMCID: PMC8633806 DOI: 10.1098/rsif.2021.0464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Swelling of lymph nodes (LNs) is commonly observed during the adaptive immune response, yet the impact on T cell (TC) trafficking and subsequent immune response is not well known. To better understand the effect of macro-scale alterations, we developed an agent-based model of the LN paracortex, describing the TC proliferative response to antigen-presenting dendritic cells alongside inflammation-driven and swelling-induced changes in TC recruitment and egress, while also incorporating regulation of the expression of egress-modulating TC receptor sphingosine-1-phosphate receptor-1. Analysis of the effector TC response under varying swelling conditions showed that swelling consistently aided TC activation. However, subsequent effector CD8+ TC production was reduced in scenarios where swelling occurred too early in the TC proliferative phase or when TC cognate frequency was low due to increased opportunity for TC exit. Temporarily extending retention of newly differentiated effector TCs, mediated by sphingosine-1-phosphate receptor-1 expression, mitigated any negative effects of swelling by allowing facilitation of activation to outweigh increased access to exit areas. These results suggest that targeting temporary effector TC retention and egress associated with swelling offers new ways to modulate effector TC responses in, for example, immuno-suppressed patients and to optimize of vaccine design.
Collapse
Affiliation(s)
- Sarah C. Johnson
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Lowell T. Edgar
- Department of Bioengineering, Imperial College London, London, UK
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
6
|
Vella G, Guelfi S, Bergers G. High Endothelial Venules: A Vascular Perspective on Tertiary Lymphoid Structures in Cancer. Front Immunol 2021; 12:736670. [PMID: 34484246 PMCID: PMC8416033 DOI: 10.3389/fimmu.2021.736670] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023] Open
Abstract
High endothelial venules (HEVs) are specialized postcapillary venules composed of cuboidal blood endothelial cells that express high levels of sulfated sialomucins to bind L-Selectin/CD62L on lymphocytes, thereby facilitating their transmigration from the blood into the lymph nodes (LN) and other secondary lymphoid organs (SLO). HEVs have also been identified in human and murine tumors in predominantly CD3+T cell-enriched areas with fewer CD20+B-cell aggregates that are reminiscent of tertiary lymphoid-like structures (TLS). While HEV/TLS areas in human tumors are predominantly associated with increased survival, tumoral HEVs (TU-HEV) in mice have shown to foster lymphocyte-enriched immune centers and boost an immune response combined with different immunotherapies. Here, we discuss the current insight into TU-HEV formation, function, and regulation in tumors and elaborate on the functional implication, opportunities, and challenges of TU-HEV formation for cancer immunotherapy.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sophie Guelfi
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Neurological Surgery, UCSF Comprehensive Cancer Center, University of California San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
7
|
Milutinovic S, Abe J, Godkin A, Stein JV, Gallimore A. The Dual Role of High Endothelial Venules in Cancer Progression versus Immunity. Trends Cancer 2021; 7:214-225. [PMID: 33132107 PMCID: PMC9213382 DOI: 10.1016/j.trecan.2020.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/17/2022]
Abstract
Secondary lymphoid organs (SLOs) are important initiators and regulators of immunity. To carry out this function, the blood vasculature must deliver oxygen and nutrients and recruit circulating lymphocytes into the SLO parenchyma, where they encounter cognate antigen. High endothelial venules (HEVs) are specialised postcapillary venules that specifically serve this function and are found in all SLOs except spleen. It is becoming clear that alterations to HEV network density and/or morphology can result in immune activation or, as recently implicated, in providing an exit route for tumour cell dissemination and metastases. In this review, the structural plasticity of HEVs, the regulatory pathways underpinning this plasticity, and the relevance of these pathways to cancer progression will be discussed.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Andrew Godkin
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Awen Gallimore
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
8
|
Scholz EMB, Kashuba ADM. The Lymph Node Reservoir: Physiology, HIV Infection, and Antiretroviral Therapy. Clin Pharmacol Ther 2021; 109:918-927. [PMID: 33529355 DOI: 10.1002/cpt.2186] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022]
Abstract
Despite advances in treatment, finding a cure for HIV remains a top priority. Chronic HIV infection is associated with increased risk of comorbidities, such as diabetes and cardiovascular disease. Additionally, people living with HIV must remain adherent to daily antiretroviral therapy, because lapses in medication adherence can lead to viral rebound and disease progression. Viral recrudescence occurs from cellular reservoirs in lymphoid tissues. In particular, lymph nodes are central to the pathology of HIV due to their unique architecture and compartmentalization of immune cells. Understanding how antiretrovirals (ARVs) penetrate lymph nodes may explain why these tissues are maintained as HIV reservoirs, and how they contribute to viral rebound upon treatment interruption. In this report, we review (i) the physiology of the lymph nodes and their function as part of the immune and lymphatic systems, (ii) the pathogenesis and outcomes of HIV infection in lymph nodes, and (iii) ARV concentrations and distribution in lymph nodes, and the relationship between ARVs and HIV in this important reservoir.
Collapse
Affiliation(s)
- Erin M B Scholz
- Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA
| | - Angela D M Kashuba
- Eshelman School of Pharmacy, The University of North Carolina, Chapel Hill, North Carolina, USA.,School of Medicine, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
9
|
Sawada J, Perrot CY, Chen L, Fournier-Goss AE, Oyer J, Copik A, Komatsu M. High Endothelial Venules Accelerate Naive T Cell Recruitment by Tumor Necrosis Factor-Mediated R-Ras Upregulation. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:396-414. [PMID: 33159887 DOI: 10.1016/j.ajpath.2020.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023]
Abstract
Recruitment of naive T cells to lymph nodes is essential for the development of adaptive immunity. Upon pathogen infection, lymph nodes promptly increase the influx of naive T cells from the circulation in order to screen and prime the T cells. The precise contribution of the lymph node vasculature to the regulation of this process remains unclear. Here we show a role for the Ras GTPase, R-Ras, in the functional adaptation of high endothelial venules to increase naive T cell trafficking to the lymph nodes. R-Ras is transiently up-regulated in the endothelium of high endothelial venules by the inflammatory cytokine tumor necrosis factor (TNF) within 24 hours of pathogen inoculation. TNF induces R-Ras upregulation in endothelial cells via JNK and p38 mitogen-activated protein kinase but not NF-κB. Studies of T cell trafficking found that the loss of function of endothelial R-Ras impairs the rapid acceleration of naive T cell recruitment to the lymph nodes upon inflammation. This defect diminished the ability of naive OT-1 T cells to develop antitumor activity against ovalbumin-expressing melanoma. Proteomic analyses suggest that endothelial R-Ras facilitates TNF-dependent transendothelial migration (diapedesis) of naive T cells by modulating molecular assembly the at T cell-endothelial cell interface. These findings give new mechanistic insights into the functional adaptation of high endothelial venules to accelerate naive T cell recruitment to the lymph nodes.
Collapse
Affiliation(s)
- Junko Sawada
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Carole Y Perrot
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Linyuan Chen
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Ashley E Fournier-Goss
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Jeremiah Oyer
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida; Burnett School of Biomedical Sciences, University of Central Florida, Orlando Florida
| | - Alicja Copik
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando Florida
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla California.
| |
Collapse
|
10
|
Plasma DNA as a "liquid biopsy" incompletely complements tumor biopsy for identification of mutations in a case series of four patients with oligometastatic breast cancer. Breast Cancer Res Treat 2020; 182:665-677. [PMID: 32562118 DOI: 10.1007/s10549-020-05714-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Circulating tumor DNA in plasma may present a minimally invasive opportunity to identify tumor-derived mutations to inform selection of targeted therapies for individual patients, particularly in cases of oligometastatic disease where biopsy of multiple tumors is impractical. To assess the utility of plasma DNA as a "liquid biopsy" for precision oncology, we tested whether sequencing of plasma DNA is a reliable surrogate for sequencing of tumor DNA to identify targetable genetic alterations. METHODS Blood and biopsies of 1-3 tumors were obtained from 4 evaluable patients with advanced breast cancer. One patient provided samples from an additional 7 tumors post-mortem. DNA extracted from plasma, tumor tissues, and buffy coat of blood were used for probe-directed capture of all exons in 149 cancer-related genes and massively parallel sequencing. Somatic mutations in DNA from plasma and tumors were identified by comparison to buffy coat DNA. RESULTS Sequencing of plasma DNA identified 27.94 ± 11.81% (mean ± SD) of mutations detected in a tumor(s) from the same patient; such mutations tended to be present at high allelic frequency. The majority of mutations found in plasma DNA were not found in tumor samples. Mutations were also found in plasma that matched clinically undetectable tumors found post-mortem. CONCLUSIONS The incomplete overlap of genetic alteration profiles of plasma and tumors warrants caution in the sole reliance of plasma DNA to identify therapeutically targetable alterations in patients and indicates that analysis of plasma DNA complements, but does not replace, tumor DNA profiling. TRIAL REGISTRATION Subjects were prospectively enrolled in trial NCT01836640 (registered April 22, 2013).
Collapse
|
11
|
Absence of hepatic activity in lymphoscintigraphy performed with Tc-99m-Nanoscan. Nucl Med Commun 2020; 41:505-509. [DOI: 10.1097/mnm.0000000000001181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Computer-assembled cross-species/cross-modalities two-pore physiologically based pharmacokinetic model for biologics in mice and rats. J Pharmacokinet Pharmacodyn 2019; 46:339-359. [PMID: 31079322 DOI: 10.1007/s10928-019-09640-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/05/2019] [Indexed: 12/11/2022]
Abstract
Two-pore physiologically-based pharmacokinetic (PBPK) models can be expected to describe the tissue distribution and elimination kinetics of soluble proteins, endogenous or dosed, as function of their size. In this work, we amalgamated our previous two-pore PBPK model for an inert domain antibody (dAb) in mice with the cross-species platform PBPK model for monoclonal antibodies described in literature into a unified two-pore platform that describes protein modalities of different sizes and includes neonatal Fc receptor (FcRn) mediated recycling. This unified PBPK model was parametrized for organ-specific lymph flow rates and the endosomal recycling rate constant using an extended tissue distribution time-course dataset that included an inert dAb, albumin and IgG in rats and mice. The model was evaluated by comparing the ab initio predictions for the tissue distribution and elimination properties of albumin-binding dAbs (AlbudAbsTM) in mice and rats with the experimental observations. Due to the large number of molecular species and reactions involved in large-scale PBPK models, we have also developed and deployed a MatlabTM script for automating the assembly of SimBiologyTM-based two-pore biologics PBPK models which drastically cuts the time and effort required for model building.
Collapse
|
13
|
Application of Pharmacokinetics Modelling to Predict Human Exposure of a Cationic Liposomal Subunit Antigen Vaccine System. Pharmaceutics 2017; 9:pharmaceutics9040057. [PMID: 29215597 PMCID: PMC5750663 DOI: 10.3390/pharmaceutics9040057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/18/2017] [Accepted: 12/04/2017] [Indexed: 12/02/2022] Open
Abstract
The pharmacokinetics of a liposomal subunit antigen vaccine system composed of the cationic lipid dimethyldioctadecylammonium bromide (DDA) and the immunostimulatory agent trehalose 6,6-dibehenate (TDB) (8:1 molar ratio) combined with the Ag85B-ESAT-6 (H1) antigen were modelled using mouse in-vivo data. Compartment modelling and physiologically based pharmacokinetics (PBPK) were used to predict the administration site (muscle) and target site (lymph) temporal concentration profiles and factors governing these. Initial estimates using compartmental modelling established that quadriceps pharmacokinetics for the liposome demonstrated a long half-life (22.6 days) compared to the associated antigen (2.62 days). A mouse minimal-PBPK model was developed and successfully predicted quadriceps liposome and antigen pharmacokinetics. Predictions for the popliteal lymph node (PLN) aligned well at earlier time-points. A local sensitivity analysis highlighted that the predicted AUCmuscle was sensitive to the antigen degradation constant kdeg (resulting in a 3-log change) more so than the fraction escaping the quadriceps (fe) (resulting in a 10-fold change), and the predicted AUCPLN was highly sensitive to fe. A global sensitivity analysis of the antigen in the muscle demonstrated that model predictions were within the 50th percentile for predictions and showed acceptable fits. To further translate in-vitro data previously generated by our group, the mouse minimal-PBPK model was extrapolated to humans and predictions made for antigen pharmacokinetics in muscle and PLN. Global analysis demonstrated that both kdeg and fe had a minimal impact on the resulting simulations in the muscle but a greater impact in the PLN. In summary, this study has predicted the in-vivo fate of DDA:TDB:H1 in humans and demonstrated the roles that formulation degradation and fraction escaping the depot site can play upon the overall depot effect within the site of administration.
Collapse
|
14
|
Ager A. High Endothelial Venules and Other Blood Vessels: Critical Regulators of Lymphoid Organ Development and Function. Front Immunol 2017; 8:45. [PMID: 28217126 PMCID: PMC5289948 DOI: 10.3389/fimmu.2017.00045] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/11/2017] [Indexed: 12/30/2022] Open
Abstract
The blood vasculature regulates both the development and function of secondary lymphoid organs by providing a portal for entry of hemopoietic cells. During the development of lymphoid organs in the embryo, blood vessels deliver lymphoid tissue inducer cells that initiate and sustain the development of lymphoid tissues. In adults, the blood vessels are structurally distinct from those in other organs due to the requirement for high levels of lymphocyte recruitment under non-inflammatory conditions. In lymph nodes (LNs) and Peyer's patches, high endothelial venules (HEVs) especially adapted for lymphocyte trafficking form a spatially organized network of blood vessels, which controls both the type of lymphocyte and the site of entry into lymphoid tissues. Uniquely, HEVs express vascular addressins that regulate lymphocyte entry into lymphoid organs and are, therefore, critical to the function of lymphoid organs. Recent studies have demonstrated important roles for CD11c+ dendritic cells in the induction, as well as the maintenance, of vascular addressin expression and, therefore, the function of HEVs. Tertiary lymphoid organs (TLOs) are HEV containing LN-like structures that develop inside organized tissues undergoing chronic immune-mediated inflammation. In autoimmune lesions, the development of TLOs is thought to exacerbate disease. In cancerous tissues, the development of HEVs and TLOs is associated with improved patient outcomes in several cancers. Therefore, it is important to understand what drives the development of HEVs and TLOs and how these structures contribute to pathology. In several human diseases and experimental animal models of chronic inflammation, there are some similarities between the development and function of HEVs within LN and TLOs. This review will summarize current knowledge of how hemopoietic cells with lymphoid tissue-inducing, HEV-inducing, and HEV-maintaining properties are recruited from the bloodstream to induce the development and control the function of lymphoid organs.
Collapse
Affiliation(s)
- Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
15
|
Yen HH, Washington E, Kimpton W, Hallein E, Allen J, Lin SY, Barber S. Development of an ovine efferent mammary lymphatic cannulation model with minimal tissue damage. BMC Vet Res 2016; 12:285. [PMID: 27955650 PMCID: PMC5153879 DOI: 10.1186/s12917-016-0908-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/01/2016] [Indexed: 02/04/2023] Open
Abstract
Background Two mammary lymphatic cannulation models in sheep have been described with minimal use in the past 50 years. The purpose of this study was to investigate a new surgical technique to allow long term monitoring of mammary lymph flow and composition from the mammary glands, with rapid ewe recovery and minimal complications post-surgery. Results We developed a modified methodology for cannulating the efferent mammary lymphatic from the mammary lymph node with minimum tissue damage. Compared to the previous models, our method required only a small incision on the aponeurosis of the external abdominal oblique muscles and thus reduced the difficulties in suturing the aponeurosis. It allowed for lymph collection and assessment for at least one week post-surgery with concurrent milk collection. Conclusion This method allows for good ewe recovery post-surgery and in vivo sampling of efferent mammary lymph from the mammary lymph nodes in real-time and comparison with milk parameters.
Collapse
Affiliation(s)
- Hung-Hsun Yen
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Elizabeth Washington
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wayne Kimpton
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Evan Hallein
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joanne Allen
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Silk Yu Lin
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stuart Barber
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
16
|
Manzo A, Benaglio F, Vitolo B, Bortolotto C, Zibera F, Todoerti M, Alpini C, Bugatti S, Caporali R, Calliada F, Montecucco C. Power Doppler ultrasonographic assessment of the joint-draining lymph node complex in rheumatoid arthritis: a prospective, proof-of-concept study on treatment with tumor necrosis factor inhibitors. Arthritis Res Ther 2016; 18:242. [PMID: 27770827 PMCID: PMC5075165 DOI: 10.1186/s13075-016-1142-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 09/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Emerging research on the mechanisms of disease chronicity in experimental arthritis has included a new focus on the draining lymph node (LN). Here, we combined clinical-serological analyses and power Doppler ultrasound (PDUS) imaging to delineate noninvasively the reciprocal relationship in vivo between the joint and the draining LN in patients with rheumatoid arthritis (RA). METHODS Forty consecutive patients refractory to conventional synthetic disease-modifying anti-rheumatic drugs were examined through parallel PDUS of the hand-wrist joints and axillary LNs and compared with 20 healthy subjects. A semiquantitative score for LN gray-scale (GS) parameters (nodal hypertrophy and cortical structure) and LN PD signal was developed. A 6-month follow-up study with serial sonographic assessments was then performed on initiation of tumor necrosis factor (TNF) inhibitors. RESULTS PDUS analysis of RA axillary LNs revealed the existence of marked inter-individual heterogeneity and of quantitative differences compared with healthy individuals in both GS and PD characteristics. RA LN changes were plastic, responsive to anti-TNF treatment, and displayed a degree of concordance with synovitis activity in peripheral joints. However, low LN PD signal at baseline despite active arthritis was strongly associated with a poor clinical response to TNF blockade. CONCLUSIONS PDUS analysis of the draining LN in RA allows capture of measurable inter-individual differences and dynamic changes linked to the underlying pathologic process. LN and joint sonographic assessments are nonredundant approaches that may provide independent perspectives on peripheral disease and its evolution over time.
Collapse
Affiliation(s)
- Antonio Manzo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy.
| | - Francesca Benaglio
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| | - Barbara Vitolo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| | - Chandra Bortolotto
- Division of Radiology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Pavia, Italy
| | - Francesca Zibera
- Division of Radiology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Pavia, Italy
| | - Monica Todoerti
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| | - Claudia Alpini
- Laboratory of Biochemical-Clinical Analyses, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Serena Bugatti
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| | - Roberto Caporali
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| | - Fabrizio Calliada
- Division of Radiology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Pavia, Italy
| | - Carlomaurizio Montecucco
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Biologic Therapy Unit, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, P.le Golgi 19, 27100, Pavia, Italy
| |
Collapse
|
17
|
Dasoveanu DC, Shipman WD, Chia JJ, Chyou S, Lu TT. Regulation of Lymph Node Vascular-Stromal Compartment by Dendritic Cells. Trends Immunol 2016; 37:764-777. [PMID: 27638128 DOI: 10.1016/j.it.2016.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022]
Abstract
During normal and pathologic immune responses, lymph nodes can swell considerably. The lymph node vascular-stromal compartment supports and regulates the developing immune responses and undergoes dynamic expansion and remodeling. Recent studies have shown that dendritic cells (DCs), best known for their antigen presentation roles, can directly regulate the vascular-stromal compartment, pointing to a new perspective on DCs as facilitators of lymphoid tissue function. Here, we review the phases of lymph node vascular-stromal growth and remodeling during immune responses, discuss the roles of DCs, and discuss how this understanding can potentially be used for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Dragos C Dasoveanu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Physiology, Biophysics and Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - William D Shipman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Jennifer J Chia
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Susan Chyou
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
18
|
Short Intracellular HIV-1 Transcripts as Biomarkers of Residual Immune Activation in Patients on Antiretroviral Therapy. J Virol 2016; 90:5665-5676. [PMID: 27030274 DOI: 10.1128/jvi.03158-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 03/25/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED HIV-1 patients continue to remain at an abnormal immune status despite prolonged combination antiretroviral therapy (cART), which results in an increased risk of non-AIDS-related diseases. Given the growing recognition of the importance of understanding and controlling the residual virus in patients, additional virological markers to monitor infected cells are required. However, viral replication in circulating cells is much poorer than that in lymph nodes, which results in the absence of markers to distinguish these cells from uninfected cells in the blood. In this study, we identified prematurely terminated short HIV-1 transcripts (STs) in peripheral blood mononuclear cells (PBMCs) as an efficient intracellular biomarker to monitor viral activation and immune status in patients with cART-mediated full viral suppression in plasma. STs were detected in PBMCs obtained from both treated and untreated patients. ST levels in untreated patients generally increased with disease progression and decreased after treatment initiation. However, some patients exhibited sustained high levels of ST and low CD4(+) cell counts despite full viral suppression by treatment. The levels of STs strongly reflected chronic immune activation defined by coexpression of HLA-DR and CD38 on CD8(+) T cells, rather than circulating proviral load. These observations represent evidence for a relationship between viral persistence and host immune activation, which in turn results in the suboptimal increase in CD4(+) cells despite suppressive antiretroviral therapy. This cell-based measurement of viral persistence contributes to an improved understanding of the dynamics of viral persistence in cART patients and will guide therapeutic approaches targeting viral reservoirs. IMPORTANCE Combination antiretroviral therapy (cART) suppresses HIV-1 load to below the detectable limit in plasma. However, the virus persists, and patients remain at an abnormal immune status, which results in an increased risk of non-AIDS-related complications. To achieve a functional cure for HIV-1 infection, activities of viral reservoirs must be quantified and monitored. However, latently infected cells are difficult to be monitored. Here, we identified prematurely terminated short HIV-1 transcripts (STs) as an efficient biomarker for monitoring viral activation and immune status in patients with cART-mediated full viral suppression in plasma. This cell-based measurement of viral persistence will contribute to our understanding of the impact of residual virus on chronic immune activation in HIV-1 patients during cART.
Collapse
|
19
|
Tegenge MA, Mitkus RJ. A first-generation physiologically based pharmacokinetic (PBPK) model of alpha-tocopherol in human influenza vaccine adjuvant. Regul Toxicol Pharmacol 2015; 71:353-64. [DOI: 10.1016/j.yrtph.2015.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 12/30/2022]
|
20
|
The draining lymph node in rheumatoid arthritis: current concepts and research perspectives. BIOMED RESEARCH INTERNATIONAL 2015; 2015:420251. [PMID: 25793195 PMCID: PMC4352497 DOI: 10.1155/2015/420251] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/29/2014] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease of unknown aetiology, leading to progressive damage of bone and cartilage with functional impairment and disability. Whilst the synovial membrane represents the epicentre of the immune-inflammatory process, there is growing evidence indicating the potential involvement of additional anatomical compartments, such as the lung, bone marrow, and secondary lymphoid tissues. Draining lymph nodes represent the elective site for tissue immune-surveillance, for the generation of adaptive immune responses and a candidate compartment for the maintenance of peripheral tolerance. Despite the precise role of the juxta- and extra-articular lymph node stations in the pathogenesis of RA remaining poorly defined, several lines of research exploiting new technological approaches are now focusing on their assessment as a potential new source of pathobiologic information, biomarkers, and complementary therapeutic targets. In this review we present an updated overview of the main concepts driving lymph node research in RA, highlighting the most relevant findings, current hypothesis, and translational perspectives.
Collapse
|
21
|
Sung HC, Lemos S, Ribeiro-Santos P, Kozyrytska K, Vasseur F, Legrand A, Charbit A, Rocha B, Evaristo C. Cognate antigen stimulation generates potent CD8(+) inflammatory effector T cells. Front Immunol 2013; 4:452. [PMID: 24379814 PMCID: PMC3863990 DOI: 10.3389/fimmu.2013.00452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/28/2013] [Indexed: 11/13/2022] Open
Abstract
Inflammatory reactions are believed to be triggered by innate signals and have a major protective role by recruiting innate immunity cells, favoring lymphocyte activation and differentiation, and thus contributing to the sequestration and elimination of the injurious stimuli. Although certain lymphocyte types such as TH17 cells co-participate in inflammatory reactions, their generation from the naïve pool requires the pre-existence of an inflammatory milieu. In this context, inflammation is always regarded as beginning with an innate response that may be eventually perpetuated and amplified by certain lymphocyte types. In contrast, we here show that even in sterile immunizations or in MyD88-deficient mice, CD8 T cells produce a burst of pro-inflammatory cytokines and chemokines. These functions follow opposite rules to the classic CD8 effector functions since they are generated prior to cell expansion and decline before antigen elimination. As few as 56 CD8(+) inflammatory effector cells in a lymph node can mobilize 10(7) cells in 24 h, including lymphocytes, natural killer cells, and several accessory cell types involved in inflammatory reactions. Thus, although inflammation modulates cognate responses, CD8 cognate responses also initiate local inflammatory reactions.
Collapse
Affiliation(s)
- Hsueh-Cheng Sung
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | - Sara Lemos
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | | | - Kateryna Kozyrytska
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | - Florence Vasseur
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | - Agnès Legrand
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | - Alain Charbit
- Faculté de Médecine, U1002, Université Paris-Descartes, INSERM , Paris , France
| | - Benedita Rocha
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| | - César Evaristo
- Faculté de Médecine, U1020, Université Paris-Descartes, INSERM , Paris , France
| |
Collapse
|
22
|
Masopust D, Schenkel JM. The integration of T cell migration, differentiation and function. Nat Rev Immunol 2013; 13:309-20. [PMID: 23598650 DOI: 10.1038/nri3442] [Citation(s) in RCA: 441] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
T cells function locally. Accordingly, T cells' recognition of antigen, their subsequent activation and differentiation, and their role in the processes of infection control, tumour eradication, autoimmunity, allergy and alloreactivity are intrinsically coupled with migration. Recent discoveries revise our understanding of the regulation and patterns of T cell trafficking and reveal limitations in current paradigms. Here, we review classic and emerging concepts, highlight the challenge of integrating new observations with existing T cell classification schemes and summarize the heuristic framework provided by viewing T cell differentiation and function first through the prism of migration.
Collapse
Affiliation(s)
- David Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
23
|
Jalkanen S, Salmi M. Lymphocyte adhesion and trafficking. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
24
|
Aarntzen EHJG, Srinivas M, Radu CG, Punt CJA, Boerman OC, Figdor CG, Oyen WJG, de Vries IJM. In vivo imaging of therapy-induced anti-cancer immune responses in humans. Cell Mol Life Sci 2012; 70:2237-57. [PMID: 23052208 PMCID: PMC3676735 DOI: 10.1007/s00018-012-1159-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/27/2012] [Accepted: 09/03/2012] [Indexed: 12/16/2022]
Abstract
Immunotherapy aims to re-engage and revitalize the immune system in the fight against cancer. Research over the past decades has shown that the relationship between the immune system and human cancer is complex, highly dynamic, and variable between individuals. Considering the complexity, enormous effort and costs involved in optimizing immunotherapeutic approaches, clinically applicable tools to monitor therapy-induced immune responses in vivo are most warranted. However, the development of such tools is complicated by the fact that a developing immune response encompasses several body compartments, e.g., peripheral tissues, lymph nodes, lymphatic and vascular systems, as well as the tumor site itself. Moreover, the cells that comprise the immune system are not static but constantly circulate through the vascular and lymphatic system. Molecular imaging is considered the favorite candidate to fulfill this task. The progress in imaging technologies and modalities has provided a versatile toolbox to address these issues. This review focuses on the detection of therapy-induced anticancer immune responses in vivo and provides a comprehensive overview of clinically available imaging techniques as well as perspectives on future developments. In the discussion, we will focus on issues that specifically relate to imaging of the immune system and we will discuss the strengths and limitations of the current clinical imaging techniques. The last section provides future directions that we envision to be crucial for further development.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Bogle G, Dunbar PR. On-lattice simulation of T cell motility, chemotaxis, and trafficking in the lymph node paracortex. PLoS One 2012; 7:e45258. [PMID: 23028887 PMCID: PMC3447002 DOI: 10.1371/journal.pone.0045258] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/15/2012] [Indexed: 01/02/2023] Open
Abstract
Agent-based simulation is a powerful method for investigating the complex interplay of the processes occurring in a lymph node during an adaptive immune response. We have previously established an agent-based modeling framework for the interactions between T cells and dendritic cells within the paracortex of lymph nodes. This model simulates in three dimensions the “random-walk” T cell motility observed in vivo, so that cells interact in space and time as they process signals and commit to action such as proliferation. On-lattice treatment of cell motility allows large numbers of densely packed cells to be simulated, so that the low frequency of T cells capable of responding to a single antigen can be dealt with realistically. In this paper we build on this model by incorporating new numerical methods to address the crucial processes of T cell ingress and egress, and chemotaxis, within the lymph node. These methods enable simulation of the dramatic expansion and contraction of the T cell population in the lymph node paracortex during an immune response. They also provide a novel probabilistic method to simulate chemotaxis that will be generally useful in simulating other biological processes in which chemotaxis is an important feature.
Collapse
Affiliation(s)
- Gib Bogle
- Maurice Wilkins Centre, University of Auckland, Aukland, New Zealand.
| | | |
Collapse
|
26
|
Liersch R, Shin JW, Bayer M, Schwöppe C, Schliemann C, Berdel WE, Mesters R, Detmar M. Analysis of a novel highly metastatic melanoma cell line identifies osteopontin as a new lymphangiogenic factor. Int J Oncol 2012; 41:1455-63. [PMID: 22797548 PMCID: PMC3583651 DOI: 10.3892/ijo.2012.1548] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 04/30/2012] [Indexed: 12/15/2022] Open
Abstract
Tumor cell invasion and metastasis are hallmarks of malignancy. Despite recent advances in the understanding of lymphatic spread, the mechanisms by which tumors metastasize to sentinel/distant lymph nodes and beyond are poorly understood. To gain new insights into this complex process, we established highly metastatic melanoma cell lines by in vivo passaging the B16 parental cell line through the lymphatic system. In this study we characterized morphology, rate of cell proliferation, colony formation, migration, tumorigenicity, lymph flow, and capacities to induce tumor- and sentinel lymph node-lymphangiogenesis. Furthermore, microarray-based comparative analysis between parental and passaged cell lines was performed to identify specific gene expression profiles. The most differentially expressed gene was SPP (osteopontin), a secreted glycophosphoprotein which is known to be involved in cancer metastasis. Overexpression of osteopontin in B16 F1-variant was confirmed by western blot analysis and quantitative RT-PCR. Treatment of cultured lymphatic endothelial cells (LECs) with osteopontin promoted cell migration mediated by the integrin α9 pathway. Our results identify osteopontin as a novel lymphangiogenic factor.
Collapse
Affiliation(s)
- Ruediger Liersch
- Department of Medicine, Hematology and Oncology, University Hospital Muenster, D-48129 Muenster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Tan KW, Yeo KP, Wong FHS, Lim HY, Khoo KL, Abastado JP, Angeli V. Expansion of cortical and medullary sinuses restrains lymph node hypertrophy during prolonged inflammation. THE JOURNAL OF IMMUNOLOGY 2012; 188:4065-80. [PMID: 22430738 DOI: 10.4049/jimmunol.1101854] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During inflammation, accumulation of immune cells in activated lymph nodes (LNs), coupled with a transient shutdown in lymphocyte exit, results in dramatic cellular expansion. Counter-regulatory measures to restrain LN expansion must exist and may include re-establishment of lymphocyte egress to steady-state levels. Indeed, we show in a murine model that egress of lymphocytes from LNs was returned to steady-state levels during prolonged inflammation following initial retention. This restoration in lymphocyte egress was supported by a preferential expansion of cortical and medullary sinuses during late inflammation. Cortical and medullary sinus remodeling during late inflammation was dependent on temporal and spatial changes in vascular endothelial growth factor-A distribution. Specifically, its expression was restricted to the subcapsular space of the LN during early inflammation, whereas its expression was concentrated in the paracortical and medullary regions of the LN at later stages. We next showed that this process was mostly driven by the synergistic cross-talk between fibroblastic reticular cells and interstitial flow. Our data shed new light on the biological significance of LN lymphangiogenesis during prolonged inflammation and further underscore the collaborative roles of stromal cells, immune cells, and interstitial flow in modulating LN plasticity and function.
Collapse
Affiliation(s)
- Kar Wai Tan
- Department of Microbiology, Immunology Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
28
|
Systems biology approaches for understanding cellular mechanisms of immunity in lymph nodes during infection. J Theor Biol 2011; 287:160-70. [PMID: 21798267 DOI: 10.1016/j.jtbi.2011.06.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 06/30/2011] [Accepted: 06/30/2011] [Indexed: 12/20/2022]
Abstract
Adaptive immunity is initiated in secondary lymphoid tissues when naive T cells recognize foreign antigen presented as MHC-bound peptide on the surface of dendritic cells. Only a small fraction of T cells in the naive repertoire will express T cell receptors specific for a given epitope, but antigen recognition triggers T cell activation and proliferation, thus greatly expanding antigen-specific clones. Expanded T cells can serve a helper function for B cell responses or traffic to sites of infection to secrete cytokines or kill infected cells. Over the past decade, two-photon microscopy of lymphoid tissues has shed important light on T cell development, antigen recognition, cell trafficking and effector functions. These data have enabled the development of sophisticated quantitative and computational models that, in turn, have been used to test hypotheses in silico that would otherwise be impossible or difficult to explore experimentally. Here, we review these models and their principal findings and highlight remaining questions where modeling approaches are poised to advance our understanding of complex immunological systems.
Collapse
|
29
|
Manzo A, Caporali R, Vitolo B, Alessi S, Benaglio F, Todoerti M, Bugatti S, Calliada F, Montecucco C. Subclinical remodelling of draining lymph node structure in early and established rheumatoid arthritis assessed by power Doppler ultrasonography. Rheumatology (Oxford) 2011; 50:1395-400. [PMID: 21378108 DOI: 10.1093/rheumatology/ker076] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE To investigate the suitability of power Doppler ultrasonography (PD-US) for the assessment of lymph node (LN) status in RA, evaluating the existence of structural and dynamic modifications in well-characterized stages of the disease. METHODS Ten patients with active disease and five patients in clinical remission underwent complete clinical and PD-US examination of hands, wrists, axillary and cervical LNs on the same day. Synovitis and PD were graded 0-3. LN assessment included maximum short axis, cortical hypertrophy (CH) and PD signal distribution. All patients with active disease were re-evaluated prospectively 3 months after initiation of therapy. RESULTS PD-US signs of axillary LN remodelling were observed in 7 out of 10 patients with active disease despite the absence of clinical lymphoadenopathy. Subclinical alterations were detected in both early untreated RA and in established disease. Characteristic structural changes consisted of hypertrophy of the LN cortex and PD signal amplification in cortical and hilar regions. Cervical LNs in active disease and axillary LNs in clinical remission were unaffected. LN PD amplification returned to normal ranges in patients with baseline alterations re-evaluated 3 months after therapy with TNF-α blocking agents and/or MTX. CONCLUSION Draining LNs in RA are subjected to subclinical intra-parenchymal changes and vascular flow modulation detectable by PD-US. Sonographic signs of LN involvement associate with disease activity and are reversible upon treatment. These data point at LN reactivity as a dynamic component of RA inflammatory cascade and an attractive platform to be explored in prognostic and response to therapy evaluations.
Collapse
Affiliation(s)
- Antonio Manzo
- Division of Rheumatology, Laboratory of Rheumatology, University of Pavia School of Medicine and IRCCS Policlinico San Matteo Foundation, P.le Golgi 2, 27100 Pavia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bogle G, Dunbar PR. T cell responses in lymph nodes. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 2:107-116. [PMID: 20836014 DOI: 10.1002/wsbm.47] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Activation of T cells by antigen-presenting cells (APCs) in lymph nodes (LNs) is a key initiating event in many immune responses. Our understanding of this process has been both improved and complicated in recent years by evidence from techniques such as intravital microscopy that has revealed new levels of dynamism in the interaction of T cells and APCs. In particular, the complex motility of T cells within LNs, and their serial interactions with many APCs, imply that earlier static models of T cell activation need to be updated. Here we review the first attempts to model T cell interactions with APCs in LNs that incorporate simulations of T cell motility, based on experimental observations. We show that lattice-based modeling approaches are the dominant trend in these models, and then chart a possible course for development of these models toward spatially-resolved models of immune responses within LNs.
Collapse
Affiliation(s)
- Gib Bogle
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Liersch R, Biermann C, Mesters RM, Berdel WE. Lymphangiogenesis in cancer: current perspectives. Recent Results Cancer Res 2010; 180:115-35. [PMID: 20033381 DOI: 10.1007/978-3-540-78281-0_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although the lymphatic system has been initially described in the sixteenth century, basic research has been limited. Despite its importance for the maintenance of tissue fluid homeostasis and for the afferent immune response, research of the molecular mechanisms of lymphatic vessel formation and function has for a long time been hampered. One reason could be because of the difficulties of visibility due to the lack of lymphatic markers. But since the discovery of several molecules specifically expressed in lymphatic endothelial cells, a rediscovery of the lymphatic vasculature has taken place. New scientific insights has facilitated detailed analysis of the nature and organization of the lymphatic system in physiological and pathophysiological conditions, such as in chronic inflammation and metastatic cancer spread. Knowledge about the molecules that control lymphangiogenesis and tumor-associated lymphangiogenesis is now expanding, allowing better opportunities for the development of drugs interfering with the relevant signaling pathways. Advances in our understanding of the mechanisms have translated into a number of novel therapeutic studies.
Collapse
Affiliation(s)
- Rüediger Liersch
- Department of Medicine, Hematology/Oncology, University Hospital Münster, Albert-Schweitzer-str. 33, 48129, Münster, Germany.
| | | | | | | |
Collapse
|
32
|
Rossi-Schneider TR, Verli FD, Marinho SA, Yurgel LS, De Souza MAL. Study of intussusceptive angiogenesis in inflammatory regional lymph nodes by scanning electron microscopy. Microsc Res Tech 2010; 73:14-9. [PMID: 19526518 DOI: 10.1002/jemt.20747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of the present study was to verify the occurrence of intussusceptive angiogenesis in blood vessels from submandibular lymph nodes responsible for lymphatic drainage of the tongue. A surgical wound inflicted on the ventral tongue of male Wistar rats and submandibular regional lymph nodes were evaluated at different postoperative periods. Scanning electron microscopy (SEM) was used to observe 123 lymph nodes at times 2, 3, 7, 10, 14, and 21 postoperative days. During the analysis of the vascular models with SEM, intussusceptive angiogenesis was observed in all groups evaluated. This was more extensive on the second and third postoperative days (83.33% and 80%, respectively), representing in these groups the expansion of the vascular chain of lymph nodes. At 21 postoperative days, intussusceptive angiogenesis (42.85%) was suggestive of vascular remodeling.
Collapse
Affiliation(s)
- Tíssiana Rachel Rossi-Schneider
- School of Dentistry, Morphological Science Department, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.
| | | | | | | | | |
Collapse
|
33
|
Agent-based simulation of T-cell activation and proliferation within a lymph node. Immunol Cell Biol 2009; 88:172-9. [PMID: 19884904 DOI: 10.1038/icb.2009.78] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recent intravital microscopy experiments have revealed the complex behavior of T cells within lymph nodes. Modeling T-cell responses in lymph nodes now requires integration of cell trafficking and motility with the molecular processes involved in T-cell activation. We describe an agent-based model that allows such integration, in which T cells undertake a random walk through a three-dimensional representation of the lymph node paracortex, integrating signals from dendritic cells (DCs), and proliferating in response. The model accommodates simulation of a large number of T cells packed at realistic densities, and includes dynamic cell trafficking that allows the lymph nodes to swell and shrink as the immune response progresses. The results from the model, including the kinetics of cognate T-cell proliferation and release, and the changes in their avidity profile, are similar to those observed in vivo. We therefore propose that this modeling framework is capable of successfully simulating T-cell activation while also accounting for new spatiotemporal knowledge of how T cells and DCs interact. Although some of the parameters used to drive the model are not yet experimentally validated, the model is capable of testing the effects of alternative values for any parameter on the T-cell response. We intend to refine each aspect of the model in collaboration with both theoreticians and experimentalists.
Collapse
|
34
|
Turhan A, Lin M, Lee GS, Miele LF, Tsuda A, Konerding MA, Mentzer SJ. Vascular microarchitecture of murine colitis-associated lymphoid angiogenesis. Anat Rec (Hoboken) 2009; 292:621-32. [PMID: 19382226 DOI: 10.1002/ar.20902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In permissive tissues, such as the gut and synovium, chronic inflammation can result in the ectopic development of anatomic structures that resemble lymph nodes. These inflammation-induced structures, termed lymphoid neogenesis or tertiary lymphoid organs, may reflect differential stromal responsiveness to the process of lymphoid neogenesis. To investigate the structural reorganization of the microcirculation involved in colonic lymphoid neogenesis, we studied a murine model of dextran sodium sulfate (DSS)-induced colitis. Standard 2-dimensional histology demonstrated both submucosal and intramucosal lymphoid structures in DSS-induced colitis. A spatial frequency analysis of serial histologic sections suggested that most intramucosal lymphoid aggregates developed de novo. Intravital microscopy of intravascular tracers confirmed that the developing intramucosal aggregates were supplied by capillaries arising from the quasi-polygonal mucosal plexus. Confocal optical sections and whole mount morphometry demonstrated capillary networks (185 +/- 46 microm diameter) involving six to ten capillaries with a luminal diameter of 6.8 +/- 1.1 microm. Microdissection and angiogenesis PCR array analysis demonstrated enhanced expression of multiple angiogenic genes including CCL2, CXCL2, CXCL5, Il-1b, MMP9, and TNF within the mucosal plexus. Intravital microscopy of tracer particle flow velocities demonstrated a marked decrease in flow velocity from 808 +/- 901 microm/sec within the feeding mucosal plexus to 491 +/- 155 microm/sec within the capillary structures. We conclude that the development of ectopic lymphoid tissue requires significant structural remodeling of the stromal microcirculation. A feature of permissive tissues may be the capacity for lymphoid angiogenesis.
Collapse
Affiliation(s)
- Aslihan Turhan
- Laboratory of Adaptive and Regenerative Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
McConnell I, Hopkins J, Lachmann P. Lymphocyte traffic through lymph nodes during cell shutdown. CIBA FOUNDATION SYMPOSIUM 2008; 71:167-95. [PMID: 6154566 DOI: 10.1002/9780470720547.ch10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Antigenic challenge of lymph nodes in sheep has marked effects on lymphocyte traffic through lymph nodes. The non-specific effects include a marked reduction in lymphocyte output in efferent lymph without a corresponding decrease in lymph flow--a phenomenon known as cell shutdown. With certain antigens there is a total disappearance of B lymphocytes during cell shutdown. The phenomenon can be reproduced in unprimed lymph nodes whenever localized complement activation occurs within the node. This also induces the release of prostaglandins, particularly PGE2. These results suggest that cell shutdown might be a two-step process involving both complement and prostaglandins. Repeated stimulation of nodes with antigen also has considerable effects on the traffic of antigen-specific lymphocytes. Antigen localized within the node can promote the selective entry into the node of T lymphocytes specific for the challenge antigen. Consequently there is a net loss from the whole animal of T cells reactive to the challenge antigen. These results are discussed in relation to lymphocyte recirculation through antigen-stimulated lymph nodes.
Collapse
|
36
|
Andrews P, Ford WL, Stoddart RW. Metabolic studies of high-walled endothelium of postcapillary venules in rat lymph nodes. CIBA FOUNDATION SYMPOSIUM 2008; 71:211-30. [PMID: 6899990 DOI: 10.1002/9780470720547.ch12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
37
|
Abe H, Schmidt RA, Sennett CA, Shimauchi A, Newstead GM. US-guided Core Needle Biopsy of Axillary Lymph Nodes in Patients with Breast Cancer: Why and How to Do It. Radiographics 2007; 27 Suppl 1:S91-9. [DOI: 10.1148/rg.27si075502] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Liao S, Ruddle NH. Synchrony of High Endothelial Venules and Lymphatic Vessels Revealed by Immunization. THE JOURNAL OF IMMUNOLOGY 2006; 177:3369-79. [PMID: 16920978 DOI: 10.4049/jimmunol.177.5.3369] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The mature phenotype of peripheral lymph node (LN) high endothelial venules (HEVs), defined as MAdCAM-1(low) PNAd(high) LTbetaR(high) HEC-6ST(high), is dependent on signaling through the lymphotoxin-beta receptor (LTbetaR). Plasticity of PLN HEVs during immunization with oxazolone was apparent as a reversion to an immature phenotype (MAdCAM-1(high) PNAd(low) LTbetaR(low) HEC-6ST(low)) followed by recovery to the mature phenotype. The recovery was dependent on B cells and was inhibited by LTbetaR-Ig treatment. Concurrent with HEV reversion, at day 4 following oxazolone or OVA immunization, reduced accumulation of Evans blue dye and newly activated DCs in the draining LNs revealed a temporary afferent lymphatic vessel (LV) functional insufficiency. T cell priming to a second Ag was temporarily inhibited. At day 7, lymphangiogenesis peaked in both the skin and draining LN, and afferent LV function was restored at the same time as HEV phenotype recovery. This process was delayed in the absence of B cells. LV and HEV both express the LTbetaR. During lymphangiogenesis in the draining LN, HEV, and LV were directly apposed; some vessels appeared to express both PNAd and LYVE-1. Pretreatment with LTbetaR-Ig drastically reduced the number of PNAd+ LYVE-1+ vessels, suggesting a reduction in LV and HEV cross-talk. The concordance in time and function and the close physical contact between LVs and HEVs in the remodeling process after immunization indicate that the two vascular systems are in synchrony and engage in cross-talk through B cells and LTbetaR.
Collapse
Affiliation(s)
- Shan Liao
- Department of Epidemiology and Public Health and Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
39
|
Webster B, Ekland EH, Agle LM, Chyou S, Ruggieri R, Lu TT. Regulation of lymph node vascular growth by dendritic cells. ACTA ACUST UNITED AC 2006; 203:1903-13. [PMID: 16831898 PMCID: PMC2118366 DOI: 10.1084/jem.20052272] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lymph nodes grow rapidly and robustly at the initiation of an immune response, and this growth is accompanied by growth of the blood vessels. Although the vessels are critical for supplying nutrients and for controlling cell trafficking, the regulation of lymph node vascular growth is not well understood. We show that lymph node endothelial cells begin to proliferate within 2 d of immunization and undergo a corresponding expansion in cell numbers. Endothelial cell proliferation is dependent on CD11c+ dendritic cells (DCs), and the subcutaneous injection of DCs is sufficient to trigger endothelial cell proliferation and growth. Lymph node endothelial cell proliferation is dependent on vascular endothelial growth factor (VEGF), and DCs are associated with increased lymph node VEGF levels. DC-induced endothelial cell proliferation and increased VEGF levels are mediated by DC-induced recruitment of blood-borne cells. Vascular growth in the draining lymph node includes the growth of high endothelial venule endothelial cells and is functionally associated with increased cell entry into the lymph node. Collectively, our results suggest a scenario whereby endothelial cell expansion in the draining lymph node is induced by DCs as part of a program that optimizes the microenvironment for the ensuing immune response.
Collapse
Affiliation(s)
- Brian Webster
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
40
|
Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol 2006; 7:344-53. [PMID: 16550197 DOI: 10.1038/ni1330] [Citation(s) in RCA: 534] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of lymphoid organs can be viewed as a continuum. At one end are the 'canonical' secondary lymphoid organs, including lymph nodes and spleen; at the other end are 'ectopic' or tertiary lymphoid organs, which are cellular accumulations arising during chronic inflammation by the process of lymphoid neogenesis. Secondary lymphoid organs are genetically 'preprogrammed' and 'prepatterned' during ontogeny, whereas tertiary lymphoid organs arise under environmental influences and are not restricted to specific developmental 'windows' or anatomic locations. Between these two boundaries are other types of lymphoid tissues that are less developmentally but more environmentally regulated, such as Peyer's patches, nasal-associated lymphoid tissue, bronchial-associated lymphoid tissue and inducible bronchial-associated lymphoid tissue. Their regulation, functions and potential effects are discussed here.
Collapse
Affiliation(s)
- Danielle L Drayton
- Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520-8034, USA
| | | | | | | |
Collapse
|
41
|
Soderberg KA, Payne GW, Sato A, Medzhitov R, Segal SS, Iwasaki A. Innate control of adaptive immunity via remodeling of lymph node feed arteriole. Proc Natl Acad Sci U S A 2005; 102:16315-20. [PMID: 16260739 PMCID: PMC1283434 DOI: 10.1073/pnas.0506190102] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The adaptive immune system relies on rare cognate lymphocytes to detect pathogen-derived antigens. Naïve lymphocytes recirculate through secondary lymphoid organs in search of cognate antigen. Here, we show that the naïve-lymphocyte recirculation pattern is controlled at the level of innate immune recognition, independent of antigen-specific stimulation. We demonstrate that inflammation-induced lymphocyte recruitment to the lymph node is mediated by the remodeling of the primary feed arteriole, and that its physiological role is to increase the efficiency of screening for rare antigen-specific lymphocytes. Our data reveal a mechanism of innate control of adaptive immunity: by increasing the pool of naïve lymphocytes for detection of foreign antigens via regulation of vascular input to the local lymph node.
Collapse
Affiliation(s)
- Kelly A Soderberg
- Section of Immunobiology, John B. Pierce Laboratory and Department of Cellular and Molecular Physiology, and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
42
|
Mena A, Nichani AK, Popowych Y, Ioannou XP, Godson DL, Mutwiri GK, Hecker R, Babiuk LA, Griebel P. Bovine and ovine blood mononuclear leukocytes differ markedly in innate immune responses induced by Class A and Class B CpG-oligodeoxynucleotide. Oligonucleotides 2005; 13:245-59. [PMID: 15000839 DOI: 10.1089/154545703322460621] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cytosine-phosphate-guanosine (CpG)-DNA can induce an impressive array of innate immune responses that may directly or indirectly contribute to the clearance of infectious agents. Assays, such as lymphocyte proliferative responses, have been used to demonstrate that the immunostimulatory activity of CpG-DNA is conserved among a broad range of vertebrate species, but no studies have been completed to determine if qualitative differences exist among species for CpG-oligodeoxynucleotide (ODN)-induced innate immune responses. In this study, we assessed the capacity of a Class A (ODN 2216) and a Class B (ODN 2007) CpG-ODN to induce innate immune responses in two closely related species, ovine (n = 28) and bovine (n = 29). The secretion of interferon (IFN)-alpha and IFN-gamma and non-major histocompatability complex (MHC)-restricted cytotoxic activity were assayed with CpG-ODN-stimulated peripheral blood mononuclear cells (PBMC). These investigations revealed significant interspecies and intraspecies variation in the responses. As expected, ODN 2216 was a potent inducer of IFN-alpha secretion by both bovine and ovine PBMC, but ODN 2007 also induced dose-dependent, CpG-specific IFN-alpha secretion by ovine PBMC. In contrast, a significant dose-dependent, CpG-specific IFN-gamma secretion response was only observed following ODN 2216 stimulation of bovine PBMC. Furthermore, both ODN 2216 and ODN 2007 induced CpG-specific non-MHC-restricted cytotoxicity with ovine but not bovine PBMC. Finally, there was not a single assay in which PBMC from all sheep or cattle responded at a detectable level. A striking aspect of these results is that such marked differences in CpG-ODN induced innate responses existed both between and within two closely related species.
Collapse
Affiliation(s)
- Angelo Mena
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK. Canada S7N 5E3
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Butler JE. Collection, Handling, and Analysis of Specimens for Studies of Mucosal Immunity in Large Animals. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
44
|
Tanaka T, Ebisuno Y, Kanemitsu N, Umemoto E, Yang BG, Jang MH, Miyasaka M. Molecular Determinants Controlling Homeostatic Recirculation and Tissue-Specific Trafficking of Lymphocytes. Int Arch Allergy Immunol 2004; 134:120-34. [PMID: 15153792 DOI: 10.1159/000078497] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The homeostasis of the immune system is maintained by the recirculation of naïve lymphocytes through the secondary lymphoid tissues, such as the lymph nodes, Peyer's patches, and spleen. Upon insult by pathogens or antigens, lymphocytes become activated, and the regulated trafficking of these cells results in the integration of systemic and regional immune responses. The exquisite specificity of such lymphocyte trafficking is determined by tissue-specific guidance signals expressed by the endothelial cells of postcapillary venules, combined with counterreceptors expressed by the circulating lymphocytes. The high endothelial venules can selectively guide naïve lymphocytes into the lymph nodes and Peyer's patches by expressing a unique combination of vascular addressins, lymphocyte-specific chemokines, and chemokine-binding molecules. The inflamed postcapillary venules in extralymphoid tissues, such as the skin and intestinal lamina propria, also use a distinct array of endothelial adhesion molecules and tissue- selective chemokines, and support the recruitment of effector and memory lymphocytes that express the appropriate receptors for tissue-specific trafficking. In this review, we summarize the present understanding of the homeostatic recirculation of naïve lymphocytes through the secondary lymphoid tissues and the specific targeting of antigen-experienced lymphocytes into the effector sites. We also revisit some previous studies that reported apparently conflicting observations.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Molecular and Cellular Recognition, Osaka University Graduate School of Medicine (C8), 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Miyasaka M, Tanaka T. Lymphocyte trafficking across high endothelial venules: dogmas and enigmas. Nat Rev Immunol 2004; 4:360-70. [PMID: 15122201 DOI: 10.1038/nri1354] [Citation(s) in RCA: 340] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Masayuki Miyasaka
- Laboratory of Molecular and Cellular Recognition, Osaka University Graduate School of Medicine, 2-2, Yamada-oka, Suita 565-0871, Japan.
| | | |
Collapse
|
46
|
Au B, McCulloch CAG, Hay JB. Quantitative studies on the movement of fluid and lymphocytes through periodontal tissue and into the draining lymph. Microsc Res Tech 2002; 56:66-71. [PMID: 11810708 DOI: 10.1002/jemt.10004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Chronic lymph drainage techniques in sheep have been used to map the pathways and to quantify the fluid and cell traffic through periodontal tissues. The continuous collection of cervical and prescapular lymph has demonstrated that 65% of labelled protein tracer injected into the periodontal tissues could be found in lymph over a period of 7.5 hours. Nearly 90% of the total radioactivity could be accounted for between the lymph and the tissue site. When silk was impregnated with radiolabelled albumin and a tooth ligated, the kinetics of the subsequent appearance of the tracer in lymph emphasized the ease with which macromolecules surrounding the teeth gain access to the lymphatics, regional lymph nodes, and immune apparatus. Animals were primed with BCG and then tuberculin (delayed hypersensitivity) lesions were simultaneously induced in the skin, bowel, and periodontium. When T cells were labelled with radioisotopes and their migration from blood to lymph measured, the periodontal tissue traffic pattern was distinct from the traffic pattern through DTH in the skin and also distinct from the pattern through the small intestine. This indicates that the lymphocyte traffic through the inflamed periodontium has unique features. This tissue specificity was not apparent when lesions were induced with TNFalpha. The static assessment of lymphocyte subsets within the tissues was also assessed with immunohistochemistry.
Collapse
Affiliation(s)
- Binh Au
- Faculty of Dentistry, University of Toronto, Toronto, Canada M5G 1G6
| | | | | |
Collapse
|
47
|
Uwiera RR, Gerdts V, Pontarollo RA, Babiuk LA, Middleton DM, Griebel PJ. Plasmid DNA induces increased lymphocyte trafficking: a specific role for CpG motifs. Cell Immunol 2001; 214:155-64. [PMID: 12088414 DOI: 10.1006/cimm.2001.1899] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bacterial DNA, primarily through immunostimulatory cytosine-guanine (CpG) motifs, induces the secretion of cytokines and activates a variety of effector cells. We investigated the possibility that CpG motifs might also modulate immunosurveillance by altering cell trafficking through a regional lymph node. Intradermal injection of plasmid DNA induced rapid and prolonged increases in the number of lymphocytes collected in efferent lymph. This effect on cell trafficking was not dependent on the expression of an encoded reporter gene but varied with plasmid construct and required a circular form. Injection of synthetic oligodeoxyribonucleotides containing CpG motifs did not alter lymphocyte trafficking but CpG-enhanced plasmid induced a dose-dependent increase in cell trafficking. Phenotypic analyses revealed that the increase in cell trafficking involved all lymphocyte subpopulations and represented a mass movement of cells. These observations reveal that bacterial DNA, through immunostimulatory CpG motifs, alters immunosurveillance by increasing cell recruitment to a regional lymph node.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Animals
- Chemotaxis, Leukocyte/drug effects
- CpG Islands
- DNA, Bacterial/administration & dosage
- DNA, Bacterial/immunology
- DNA, Bacterial/pharmacology
- DNA, Circular/administration & dosage
- DNA, Circular/immunology
- DNA, Circular/pharmacology
- Female
- Immunologic Surveillance/immunology
- Immunophenotyping
- Injections, Intradermal
- Lymphocyte Subsets/cytology
- Lymphocyte Subsets/immunology
- Oligodeoxyribonucleotides/chemistry
- Oligodeoxyribonucleotides/immunology
- Plasmids/genetics
- Plasmids/immunology
- Sheep
Collapse
Affiliation(s)
- R R Uwiera
- Department of Veterinary Pathology, Veterinary Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Byrne SN, Halliday GM, Johnston LJ, King NJ. Interleukin-1beta but not tumor necrosis factor is involved in West Nile virus-induced Langerhans cell migration from the skin in C57BL/6 mice. J Invest Dermatol 2001; 117:702-9. [PMID: 11564180 DOI: 10.1046/j.0022-202x.2001.01454.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Langerhans cells are bone marrow-derived epidermal dendritic cells. They migrate out of the epidermis into the lymphatics and travel to the draining lymph nodes where they are responsible for the activation of T cells in the primary immune response. Tumor necrosis factor and interleukin-1beta, have previously been shown to be responsible for Langerhans cell migration in response to contact sensitizers in BALB/C mice; however, which cytokines are responsible for mediating Langerhans cell migration in response to a replicating cutaneously acquired virus such as the West Nile Virus, are not known. We have devised a method for identifying Langerhans cells in the draining lymph nodes using E-cadherin labeling and flow cytometry. We infected tumor necrosis factor-deficient gene knockout mice (tumor necrosis factor-/-) intradermally with West Nile Virus and found that levels of Langerhans cell emigration and accumulation in the draining lymph nodes were similar to wild-type C57BL/6 mice. This was borne out by the finding that high levels of systemic neutralizing anti-tumor necrosis factor antibody failed to inhibit the migration of Langerhans cells from the epidermis and their accumulation in the draining lymph nodes in wild-type C57BL/6 mice. In West Nile Virus-infected, tumor necrosis factor-/- mice treated with systemic neutralizing anti-interleukin-1beta antibodies, however, migration of Langerhans cells from the epidermis and their accumulation in the draining lymph nodes were significantly inhibited compared with control antibody-treated, infected animals. The results indicate that Langerhans cell migration, accumulation in the draining lymph nodes and the initiation of lymph node shut-down in response to a cutaneous West Nile Virus infection is dependent on interleukin-1beta and can occur in the absence of tumor necrosis factor.
Collapse
Affiliation(s)
- S N Byrne
- Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
49
|
Abstract
The immune system consists of a complex collection of leukocytes and dendritic cells that surveys most tissues in the body for the appearance of foreign antigens. For an efficient immune response, the interaction and co-localization of antigen-presenting cells, costimulatory helper cells and effector cells are crucial parameters. Therefore, the migration routes of antigen-presenting cells and potential antigen-specific lymphocytes merge in secondary lymphoid organs in order to increase the likelihood and speed of a lymphocyte finding its cognate antigen. Additionally, antigen-primed effector cells are directed to the tissue where they are most likely to encounter their cognate antigen. This highly organized and efficient antigen encounter is based on a continuous recirculation of antigen-specific lymphocytes between blood, peripheral tissue, and secondary lymphoid organs. Moreover, the efficacy of the immune system is further increased by the ability of different lymphocyte subsets to recirculate only through distinct tissues. The scope of this review is to outline the concept and mechanisms of lymphocyte homing and recirculation and to discuss the significance for the immune defense. Current models in leukocyte homing and recirculation and the underlying molecular functions of implicated cell adhesion molecules, chemokines, and chemokine receptors are discussed.
Collapse
Affiliation(s)
- G Wiedle
- Department of Pathology, Centre Medical Universitaire, Geneva, Switzerland
| | | | | |
Collapse
|
50
|
West CA, Young AJ, Mentzer SJ. Lymphocyte traffic into antigen-stimulated tissues. Transplant Rev (Orlando) 2000. [DOI: 10.1053/trre.2000.16512] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|