Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where James L. Reading is active.

Publication


Featured researches published by James L. Reading.


Transplantation | 2013

Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Fourth Meeting: Lessons Learned from First Clinical Trials

Marcella Franquesa; Martin J. Hoogduijn; Marlies E.J. Reinders; Elke Eggenhofer; Anja U. Engela; F. Mensah; Joan Torras; Antonello Pileggi; Cees van Kooten; Bernard P. Mahon; Olivier Detry; Felix C. Popp; Volker Benseler; Federica Casiraghi; Christian L. Johnson; Janis Ancans; Barbara Fillenberg; Olga DelaRosa; Josep M. Aran; Marieke Roemeling-Vanrhijn; Jef Pinxteren; Norberto Perico; Eliana Gotti; B Christ; James L. Reading; Martino Introna; Robert Deans; Murat Shagidulin; Ramon Farre; Alessandro Rambaldi

The Fourth Expert Meeting of the Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Consortium took place in Barcelona on October 19 and 20, 2012. This meeting focused on the translation of preclinical data into early clinical settings. This position paper highlights the main topics explored on the safety and efficacy of mesenchymal stem cells as a therapeutic agent in solid organ transplantation and emphasizes the issues (proper timing, concomitant immunossupression, source and immunogenicity of mesenchymal stem cells, and oncogenicity) that have been addressed and will be followed up by the MiSOT Consortium in future studies.


Journal of Immunology | 2013

Clinical-Grade Multipotent Adult Progenitor Cells Durably Control Pathogenic T Cell Responses in Human Models of Transplantation and Autoimmunity

James L. Reading; Jennie H. M. Yang; Shereen Sabbah; Ania Skowera; Robin R. Knight; Jef Pinxteren; Bart Vaes; Timothy E. Allsopp; Anthony E. Ting; Sarah A. Busch; Amy Raber; Robert Deans; Timothy Tree

A major goal of immunotherapy remains the control of pathogenic T cell responses that drive autoimmunity and allograft rejection. Adherent progenitor cells, including mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs), represent attractive immunomodulatory cell therapy candidates currently active in clinical trials. MAPCs can be distinguished from MSCs on the basis of cellular phenotype, size, transcriptional profile, and expansion capacity. However, despite their ongoing evaluation in autoimmune and allogeneic solid organ transplantation settings, data supporting the immune regulatory potential of clinical-grade MAPCs are limited. In this study, we used allogeneic islet transplantation as a model indication to assess the ability of clinical-grade MAPCs to control T cell responses that drive immunopathology in human autoimmune disease and allograft rejection. MAPCs suppressed T cell proliferation and Th1 and Th17 cytokine production while increasing secretion of IL-10 and were able to suppress effector functions of bona fide autoreactive T cells from individuals with type 1 diabetes mellitus, including killing of human islets. Furthermore, MAPCs favored the proliferation of regulatory T cells during homeostatic expansion driven by γ-chain cytokines and exerted a durable, yet reversible, control of T cell function. MAPC suppression required licensing and proceeded via IDO-mediated tryptophan catabolism. Therefore, the common immune modulatory characteristics of clinical-grade MAPCs shown in this study suggest that they can be regarded as an alternative source of adult progenitor cells with similar clinical usefulness to MSCs. Taken collectively, these findings may guide the successful deployment of both MSCs and MAPCs for the amelioration of human autoimmunity and allograft rejection.


Journal of Virology | 2008

WFDC1/ps20 is a novel innate immunomodulatory signature protein of human immunodeficiency virus (HIV)-Permissive CD4(+) CD45RO(+) memory T cells that promotes infection by upregulating CD54 integrin expression and is elevated in HIV type 1 infection

Raymond Alvarez; James L. Reading; D King; M Hayes; Philippa Easterbrook; Farzin Farzaneh; Steven J. Ressler; Feng Yang; David R. Rowley; Annapurna Vyakarnam

ABSTRACT Understanding why human immunodeficiency virus (HIV) preferentially infects some CD4+ CD45RO+ memory T cells has implications for antiviral immunity and pathogenesis. We report that differential expression of a novel secreted factor, ps20, previously implicated in tissue remodeling, may underlie why some CD4 T cells are preferentially targeted. We show that (i) there is a significant positive correlation between endogenous ps20 mRNA in diverse CD4 T-cell populations and in vitro infection, (ii) a ps20+ permissive cell can be made less permissive by antibody blockade- or small-interference RNA-mediated knockdown of endogenous ps20, and (iii) conversely, a ps20low cell can be more permissive by adding ps20 exogenously or engineering stable ps20 expression by retroviral transduction. ps20 expression is normally detectable in CD4 T cells after in vitro activation and interleukin-2 expansion, and such oligoclonal populations comprise ps20positive and ps20low/negative isogenic clones at an early differentiation stage (CD45RO+/CD25+/CD28+/CD57−). This pattern is altered in chronic HIV infection, where ex vivo CD4+ CD45RO+ T cells express elevated ps20. ps20 promoted HIV entry via fusion and augmented CD54 integrin expression; both of these effects were reversed by anti-ps20 antibody. We therefore propose ps20 to be a novel signature of HIV-permissive CD4 T cells that promotes infection in an autocrine and paracrine manner and that HIV has coopted a fundamental role of ps20 in promoting cell adhesion for its benefit. Disrupting the ps20 pathway may therefore provide a novel anti-HIV strategy.


Molecular Therapy | 2015

Suppression of IL-7-dependent Effector T-cell Expansion by Multipotent Adult Progenitor Cells and PGE2.

James L. Reading; Bart Vaes; Caroline M. Hull; Shereen Sabbah; Thomas Hayday; Nancy Wang; Anthony DiPiero; Nicholas A. Lehman; Jen M Taggart; Fiona Carty; Karen English; Jef Pinxteren; Robert Deans; Anthony E. Ting; Timothy Tree

T-cell depletion therapy is used to prevent acute allograft rejection, treat autoimmunity and create space for bone marrow or hematopoietic cell transplantation. The evolved response to T-cell loss is a transient increase in IL-7 that drives compensatory homeostatic proliferation (HP) of mature T cells. Paradoxically, the exaggerated form of this process that occurs following lymphodepletion expands effector T-cells, often causing loss of immunological tolerance that results in rapid graft rejection, autoimmunity, and exacerbated graft-versus-host disease (GVHD). While standard immune suppression is unable to treat these pathologies, growing evidence suggests that manipulating the incipient process of HP increases allograft survival, prevents autoimmunity, and markedly reduces GVHD. Multipotent adult progenitor cells (MAPC) are a clinical grade immunomodulatory cell therapy known to alter γ-chain cytokine responses in T-cells. Herein, we demonstrate that MAPC regulate HP of human T-cells, prevent the expansion of Th1, Th17, and Th22 effectors, and block the development of pathogenic allograft responses. This occurs via IL-1β-primed secretion of PGE2 and activates T-cell intrinsic regulatory mechanisms (SOCS2, GADD45A). These data provide proof-of-principle that HP of human T-cells can be targeted by cellular and molecular therapies and lays a basis for the development of novel strategies to prevent immunopathology in lymphodepleted patients.


Diabetes | 2015

Natural Variation in Interleukin-2 Sensitivity Influences Regulatory T-Cell Frequency and Function in Individuals With Long-standing Type 1 Diabetes

Jennie H. M. Yang; Antony J. Cutler; Ricardo C. Ferreira; James L. Reading; Nicholas J. Cooper; Chris Wallace; Pamela Clarke; Deborah J. Smyth; Christopher S Boyce; Guo-Jian Gao; John A. Todd; Linda S. Wicker; Timothy Tree

Defective immune homeostasis in the balance between FOXP3+ regulatory T cells (Tregs) and effector T cells is a likely contributing factor in the loss of self-tolerance observed in type 1 diabetes (T1D). Given the importance of interleukin-2 (IL-2) signaling in the generation and function of Tregs, observations that polymorphisms in genes in the IL-2 pathway associate with T1D and that some individuals with T1D exhibit reduced IL-2 signaling indicate that impairment of this pathway may play a role in Treg dysfunction and the pathogenesis of T1D. Here, we have examined IL-2 sensitivity in CD4+ T-cell subsets in 70 individuals with long-standing T1D, allowing us to investigate the effect of low IL-2 sensitivity on Treg frequency and function. IL-2 responsiveness, measured by STAT5a phosphorylation, was a very stable phenotype within individuals but exhibited considerable interindividual variation and was influenced by T1D-associated PTPN2 gene polymorphisms. Tregs from individuals with lower IL-2 signaling were reduced in frequency, were less able to maintain expression of FOXP3 under limiting concentrations of IL-2, and displayed reduced suppressor function. These results suggest that reduced IL-2 signaling may be used to identify patients with the highest Treg dysfunction and who may benefit most from IL-2 immunotherapy.


Current Opinion in Organ Transplantation | 2013

Mesenchymal stromal cells as a means of controlling pathological T-cell responses in allogeneic islet transplantation

James L. Reading; Shereen Sabbah; Sarah A. Busch; Timothy Tree

Purpose of reviewTo evaluate the potential for mesenchymal stromal cells (MSCs) to regulate T-cell responses responsible for graft destruction following allogeneic islet transplantation (AIT). Recent findingsDespite a high level of primary graft function being observed in most individuals following AIT, the majority of recipients require exogenous insulin within 5 years, presumably due to graft attrition. Although this process is not fully understood, recent evidence suggests that a combination of chronic allograft rejection and/or the recrudescence of anti-islet autoimmunity govern islet loss. Emerging reports highlight that the pathology of AIT may involve the proliferation, effector function and homeostatic expansion of alloreactive and autoreactive T-cell pools. MSCs exhibit several desirable characteristics, which may advocate their use in AIT. This includes the capacity to suppress alloreactive and autoimmune T-cell responses, and promote a cytokine environment that is likely to be graft protective. However, further work is needed to clarify if MSCs can function in the setting of immune suppression and discern how they may effect T-cell effector functions and influence homeostatic expansion. SummaryMSCs exhibit the potential to regulate the T-cell-driven processes that underlie disease pathology in AIT, but further study may be required to maximize their therapeutic efficacy.


Retrovirology | 2011

WFDC1 expression identifies memory CD4 T-lymphocytes rendered vulnerable to cell-cell HIV-1 transfer by promoting intercellular adhesive junctions.

Raymond Alvarez; Georgina Thorborn; James L. Reading; Shalini Kamu Reddy; Annapurna Vyakarnam

BackgroundElucidating mechanisms that promote HIV-1 transfer between CD4+ T-lymphocytes and their subsequent loss is of importance to HIV-1 pathogenesis. We recently reported that whey acidic protein, ps20, promotes cell-free HIV-1 spread through ICAM-1 modulation. Since ICAM-1 is pivotal in cell conjugation and intercellular HIV-1 transfer, this study examines ps20 effects on HIV-1 spread between T lymphocytes.ResultsWe demonstrate intrinsic ps20 variability in primary CD4+ T-lymphocyte clonal populations and a significant positive correlation between endogenous ps20 levels and virus transfer involving fusion resulting in a spreading infection that could be reversed by the addition of reverse transcriptase inhibitors. Blocking anti-ps20 antibody or siRNA mediated ps20 knockdown, significantly reduced virus transfer. Conversely, virus transfer was promoted by ectopic ps20 expression or by exogenous addition of recombinant ps20. A higher frequency of virological synapse formation was evident in cocultures of HIV-1 infected donor T-cells with ps20high v ps20low/intermediate targets. Blocking ps20 inhibited T-lymphocyte conjugate formation and ICAM-1 expression, and was as potent as ICAM-1 in inhibiting HIV-1 transfer.ConclusionsTherefore ps20 is a novel marker of CD4+ T-cells rendered vulnerable to HIV-1 infection by regulating the fundamental biologic process of intercellular conjugate formation and consequently of potential importance in HIV-1 pathogenesis.


Current Opinion in Hiv and Aids | 2012

Whey acidic proteins (WAPs): novel modulators of innate immunity to HIV infection.

James L. Reading; Adrienne F.A. Meyers; Annapurna Vyakarnam

PURPOSE OF REVIEW To discuss how whey acidic proteins (WAPs), a new class of immunomodulatory soluble mediators, impact innate immunity to HIV infection. RECENT FINDINGS Innate immunity to HIV infection is increasingly being recognized as critical in determining initial virus transmission and dissemination and may, therefore, be exploited in vaccine and microbicide intervention strategies to combat HIV infection. Several important innate immune mediators have recently been shown to regulate HIV infection in vitro and are, thus, implicated in in vivo immunity to the virus. These include soluble mediators, such as type I interferon, the defensins and more recently WAPs. Recent evidence is discussed, which show that WAPs are pleiotropic soluble mediators that may impact the course of HIV infection in two ways: as regulators of HIV replication and as regulators of innate and adaptive immunity. SUMMARY A better understanding of host factors that regulate HIV transmission is essential in the development of novel therapeutic strategies. This review focuses on recent findings that highlight the HIV regulatory and anti-inflammatory function of WAPs and assesses their potential to be exploited as novel therapeutics.


Frontiers in Immunology | 2018

Multipotent Adult Progenitor Cells Suppress T Cell Activation in In Vivo Models of Homeostatic Proliferation in a Prostaglandin E2-Dependent Manner

Fiona Carty; Jennifer M. Corbett; Joäo P M. C. M. Cunha; James L. Reading; Timothy Tree; Anthony E. Ting; Samantha Stubblefield; Karen English

Lymphodepletion strategies are used in the setting of transplantation (including bone marrow, hematopoietic cell, and solid organ) to create space or to prevent allograft rejection and graft versus host disease. Following lymphodepletion, there is an excess of IL-7 available, and T cells that escape depletion respond to this cytokine undergoing accelerated proliferation. Moreover, this environment promotes the skew of T cells to a Th1 pro-inflammatory phenotype. Existing immunosuppressive regimens fail to control this homeostatic proliferative (HP) response, and thus the development of strategies to successfully control HP while sparing T cell reconstitution (providing a functioning immune system) represents a significant unmet need in patients requiring lymphodepletion. Multipotent adult progenitor cells (MAPC®) have the capacity to control T cell proliferation and Th1 cytokine production. Herein, this study shows that MAPC cells suppressed anti-thymocyte globulin-induced cytokine production but spared T cell reconstitution in a pre-clinical model of lymphodepletion. Importantly, MAPC cells administered intraperitoneally were efficacious in suppressing interferon-γ production and in promoting the expansion of regulatory T cells in the lymph nodes. MAPC cells administered intraperitoneally accumulated in the omentum but were not present in the spleen suggesting a role for soluble factors. MAPC cells suppressed lymphopenia-induced cytokine production in a prostaglandin E2-dependent manner. This study suggests that MAPC cell therapy may be useful as a novel strategy to target lymphopenia-induced pathogenic T cell responses in lymphodepleted patients.


Cytotherapy | 2018

315 - Multipotent Adult Progenitor Cells suppress homeostatic driven activation of T cells in vivo: ISCT 2018 Annual Meeting

Fiona Carty; J. Corbett; J. Cunha; James L. Reading; Anthony E. Ting; S. Stubblefield; Karen English

In the setting of transplantation, T cells are deliberately depleted to prevent allograft rejection. However, this IL-7 activates the accelerated proliferation of effector memory T cells, which are the predominant mediators of graft rejection. Multipotent adult progenitor cells (MAPC) have been previously shown to modulate the response of T cells to IL-7, in vitro but not in vivo. Here, for the first time, we demonstrate that MAPC suppress IL-7 and anti-thymocyte (ATG) driven pro-inflammatory cytokine production by both CD4+ and CD8+ T cells in the spleen. While, MAPC significantly suppressed T cell proliferation in the IL-7 driven model, MAPC did not suppress T cell proliferation in the ATG model when administered on day 4. The effects of routes of administration on the bio-distribution of MAPC in our model of homeostatic proliferation was further clarified using novel 3D whole animal Cryo-imaging technology. MAPC administered i.v. were observed in the spleen 48 hours post injection. However, following i.p. administration, MAPC do not gain access to the spleen but reside in the omentum tissue suggesting that MAPC mediate their effects in the spleen through trophic signalling. Importantly, we demonstrate that MAPC mediate their suppression of pro-inflammatory cytokine production in a PGE-2 dependent manner. Given the importance of successful reconstitution of the immune cell compartment to provide a functional immune system, the fact that MAPC do not suppress this reconstitution but dampen pathogenic cytokine production is a very positive finding. Together this data supports the idea that MAPC may be useful in controlling immune dysregulation via suppression of T cell cytokine production while sparing T cell reconstitution in lymphodepleted patients. (Less)

Collaboration


Dive into the James L. Reading's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Deans

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jef Pinxteren

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martino Introna

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge