Network


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

Hotspot


Dive into the research topics where Lisa Mathews is active.

Publication


Featured researches published by Lisa Mathews.


Journal of Immunology | 2014

IL-33 Is an Unconventional Alarmin That Stimulates IL-2 Secretion by Dendritic Cells To Selectively Expand IL-33R/ST2+ Regulatory T Cells

Benjamin M. Matta; Jeremy M. Lott; Lisa Mathews; Quan Liu; Brian R. Rosborough; Bruce R. Blazar; Hēth R. Turnquist

IL-33 is a recently characterized IL-1 family member that is proposed to function as an alarmin, or endogenous signal of cellular damage, as well as act as a pleiotropic cytokine. The ability of IL-33 to potentiate both Th1 and Th2 immunity supports its role in pathogen clearance and disease immunopathology. Yet, IL-33 restrains experimental colitis and transplant rejection by expanding regulatory T cells (Treg) via an undefined mechanism. We sought to determine the influence of IL-33 on hematopoietic cells that drives Treg expansion and underlies the therapeutic benefit of IL-33 administration. In this study, we identify a feedback loop in which conventional mouse CD11c+ dendritic cells (DC) stimulated by IL-33 secrete IL-2 to selectively expand IL-33R(ST2+)– suppressive CD4+Foxp3+ Treg. Interestingly, this occurs in the absence of classical DC maturation, and DC-derived (innate) IL-2 increases ST2 expression on both DC and interacting Treg. ST2+ Treg represent an activated subset of Foxp3+ cells, demonstrated to be ICOShighCD44high compared with their ST2− counterparts. Furthermore, although studies have shown that IL-33–exposed DC promote Th2 responses, we reveal that ST2+ DC are required for IL-33–mediated in vitro and in vivo Treg expansion. Thus, we have uncovered a relationship between IL-33 and innate IL-2 that promotes the selective expansion of ST2+ Treg over non-Treg. These findings identify a novel regulatory pathway driven by IL-33 in immune cells that may be harnessed for therapeutic benefit or for robust expansion of Treg in vitro and in vivo.


Blood | 2016

Peri-alloHCT IL-33 administration expands recipient T-regulatory cells that protect mice against acute GVHD

Benjamin M. Matta; Dawn K. Reichenbach; Xiaoli Zhang; Lisa Mathews; Brent H. Koehn; Gaelen K. Dwyer; Jeremy M. Lott; Franziska Maria Uhl; Dietmar Pfeifer; Colby J. Feser; Michelle J. Smith; Quan Liu; Robert Zeiser; Bruce R. Blazar; Hēth R. Turnquist

During allogeneic hematopoietic cell transplantation (alloHCT), nonhematopoietic cell interleukin-33 (IL-33) is augmented and released by recipient conditioning to promote type 1 alloimmunity and lethal acute graft-versus-host disease (GVHD). Yet, IL-33 is highly pleiotropic and exhibits potent immunoregulatory properties in the absence of coincident proinflammatory stimuli. We tested whether peri-alloHCT IL-33 delivery can protect against development of GVHD by augmenting IL-33-associated regulatory mechanisms. IL-33 administration augmented the frequency of regulatory T cells (Tregs) expressing the IL-33 receptor, suppression of tumorigenicity-2 (ST2), which persist following total body irradiation. ST2 expression is not exclusive to Tregs and IL-33 expands innate immune cells with regulatory or reparative properties. However, selective depletion of recipient Foxp3(+) cells concurrent with peri-alloHCT IL-33 administration accelerated acute GVHD lethality. IL-33-expanded Tregs protected recipients from GVHD by controlling macrophage activation and preventing accumulation of effector T cells in GVHD-target tissue. IL-33 stimulation of ST2 on Tregs activates p38 MAPK, which drives expansion of the ST2(+) Treg subset. Associated mechanistic studies revealed that proliferating Tregs exhibit IL-33-independent upregulation of ST2 and the adoptive transfer of st2(+) but not st2(-) Tregs mediated GVHD protection. In total, these data demonstrate the protective capacity of peri-alloHCT administration of IL-33 and IL-33-responsive Tregs in mouse models of acute GVHD. These findings provide strong support that the immunoregulatory relationship between IL-33 and Tregs can be harnessed therapeutically to prevent GVHD after alloHCT for treatment of malignancy or as a means for tolerance induction in solid organ transplantation.


Journal of Immunology | 2014

Cutting Edge: Flt3 Ligand Mediates STAT3-Independent Expansion but STAT3-Dependent Activation of Myeloid-Derived Suppressor Cells

Brian R. Rosborough; Lisa Mathews; Benjamin M. Matta; Quan Liu; Dàlia Raïch-Regué; Angus W. Thomson; Hēth R. Turnquist

The Flt3–Flt3 ligand (Flt3L) pathway is critically involved in the differentiation and homeostasis of myeloid cells, including dendritic cells (DC); however, its role in the expansion and function of myeloid-derived suppressor cells (MDSC) has not been determined. In this article, we describe the ability of Flt3L to expand and activate murine MDSC capable of suppressing allograft rejection upon adoptive transfer. Although Flt3L expands and augments the stimulatory capacity of myeloid DC, MDSC expanded by Flt3L have increased suppressive activity. Although STAT3 is considered the central transcription factor for MDSC expansion, inhibition and genetic ablation of STAT3 did not block, but rather augmented, Flt3L-mediated MDSC expansion. MDSC suppressive function, preserved when STAT3 inhibition was removed, was reduced by genetic STAT3 deletion. Both STAT3 inhibition and deletion reduced Flt3L-mediated DC expansion, signifying that STAT3 had reciprocal effects on suppressive MDSC and immunostimulatory DC expansion. Together, these findings enhance our understanding of the immunomodulatory properties of Flt3L.


Biology of Blood and Marrow Transplantation | 2014

IL-12hi Rapamycin-Conditioned Dendritic Cells Mediate IFN-γ–Dependent Apoptosis of Alloreactive CD4+ T Cells In Vitro and Reduce Lethal Graft-Versus-Host Disease

Elizabeth O. Stenger; Brian R. Rosborough; Lisa Mathews; Huihui Ma; Markus Y. Mapara; Angus W. Thomson; Hēth R. Turnquist

Rapamycin (RAPA) inhibits the mechanistic target of rapamycin (mTOR), a crucial immune system regulator. Dendritic cells (DC) generated in RAPA (RAPA-DC) enrich for CD4(+) forkhead box p3 (FoxP3(+)) regulatory T cells and induce T cell apoptosis by an unknown mechanism. RAPA-DC also promote experimental allograft survival, yet paradoxically secrete increased IL-12, crucial for the generation of IFN-γ(+) CD4(+) T cells. However, IFN-γ is pro-apoptotic and IL-12-driven IFN-γ inhibits experimental graft-versus-host disease (GVHD). We hypothesized that IL-12(hi) RAPA-DC would facilitate IFN-γ-mediated apoptosis of alloreactive T cells and, unlike control (CTR)-DC, would reduce lethal GVHD. Following LPS stimulation, RAPA-DC exhibited decreased MHCII and co-stimulatory molecules and contained a significant population of CD86(lo) IL-12(hi) cells. Consistent with our hypothesis, both unstimulated and LPS-stimulated RAPA-DC enhanced alloreactive CD4(+) T cell apoptosis in culture. Augmented T cell apoptosis was ablated by IFN-γ neutralization or using T cells lacking the IFN-γ receptor, and it was associated with increased expression of Fas and cleaved caspase 8. DC production or responses to IFN-γ were not important to increased apoptotic functions of RAPA-DC. LPS-stimulated IL-12p40(-/-) RAPA-DC induced lower levels of T cell apoptosis in culture, which was further decreased with addition of anti-IFN-γ. Finally, whereas CTR-DC accelerated mortality from GVHD, LPS-treated RAPA-DC significantly prolonged host survival. In conclusion, increased apoptosis of allogeneic CD4(+) T cells induced by LPS-stimulated IL-12(hi) RAPA-DC is mediated in vitro through IFN-γ and in part by increased IL-12 expression. Enhanced production of IL-12, the predominant inducer of IFN-γ by immune cells, is a probable mechanism underlying the capacity of LPS-treated RAPA-DC to reduce GVHD.


Transplantation | 2017

IL-33 Augments the Capacity of Human Dendritic Cells to Expand Suppressive, but IFN-Gamma hi Regulatory T Cells

Yifei Zhao; Hong Zhang; Lisa Mathews; Angus W. Thomson; Heth R. Turnquist


Transplantation | 2017

The “Type 2” Cytokine, Interlukin-33, Generates Potent CD4 + ST2 + Tbet + Th1 Responses Independently of IL-12 under Lymphopenic Conditions

Gaelen K. Dwyer; Lisa Mathews; Anna Lucas; Bruce R. Blazar; Heth R. Turnquist


Transplantation | 2014

IL-33 Promotes Selective Expansion of ST2+Foxp3+ Regulatory T Cells Dependent On Dendritic Cell Secretion of IL-2.: Abstract# A91

B. Matta; J. Lott; Lisa Mathews; Brian R. Rosborough; Bruce R. Blazar; Hēth R. Turnquist


Transplantation | 2014

Myeloid-Specific STAT3 Deletion Augments Flt3 Ligand-Driven Myeloid-Derived Suppressor Cell Expansion But Limits Their Suppressor Function.: Abstract# A99

B. Matta; Brian R. Rosborough; Lisa Mathews; Q. Liu; Dàlia Raïch-Regué; Angus W. Thomson; Hēth R. Turnquist


Journal of Immunology | 2014

Conditional STAT3-deficiency augments Flt3 ligand-driven myeloid-derived suppressor cell expansion but limits their suppressor function (IRM7P.487)

Benjamin M. Matta; Brian R. Rosborough; Lisa Mathews; Quan Liu; Dàlia Raïch-Regué; Thomson Angus; Heth R. Turnquist


Journal of Immunology | 2014

Metabolic mechanisms underlying IL-33 mediated Th2 polarization and allergic airway disease (IRM7P.494)

Jeremy M. Lott; Benjamin M. Matta; Lisa Mathews; Anne I. Sperling; Heth R. Turnquist

Collaboration


Dive into the Lisa Mathews's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy M. Lott

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Quan Liu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge