Network


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

Hotspot


Dive into the research topics where Matthew T. Stier is active.

Publication


Featured researches published by Matthew T. Stier.


The Journal of Allergy and Clinical Immunology | 2016

Respiratory syncytial virus infection activates IL-13–producing group 2 innate lymphoid cells through thymic stromal lymphopoietin

Matthew T. Stier; Melissa H. Bloodworth; Shinji Toki; Dawn C. Newcomb; Kasia Goleniewska; Kelli L. Boyd; Marc Quitalig; Anne L. Hotard; Martin L. Moore; Tina V. Hartert; Baohua Zhou; Andrew N. J. McKenzie; R. Stokes Peebles

Background Respiratory syncytial virus (RSV) is a major health care burden with a particularly high worldwide morbidity and mortality rate among infants. Data suggest that severe RSV-associated illness is in part caused by immunopathology associated with a robust type 2 response. Objective We sought to determine the capacity of RSV infection to stimulate group 2 innate lymphoid cells (ILC2s) and the associated mechanism in a murine model. Methods Wild-type (WT) BALB/c, thymic stromal lymphopoietin receptor (TSLPR) knockout (KO), or WT mice receiving an anti-TSLP neutralizing antibody were infected with the RSV strain 01/2-20. During the first 4 to 6 days of infection, lungs were collected for evaluation of viral load, protein concentration, airway mucus, airway reactivity, or ILC2 numbers. Results were confirmed with 2 additional RSV clinical isolates, 12/11-19 and 12/12-6, with known human pathogenic potential. Results RSV induced a 3-fold increase in the number of IL-13–producing ILC2s at day 4 after infection, with a concurrent increase in total lung IL-13 levels. Both thymic stromal lymphopoietin (TSLP) and IL-33 levels were increased 12 hours after infection. TSLPR KO mice did not mount an IL-13–producing ILC2 response to RSV infection. Additionally, neutralization of TSLP significantly attenuated the RSV-induced IL-13–producing ILC2 response. TSLPR KO mice displayed reduced lung IL-13 protein levels, decreased airway mucus and reactivity, attenuated weight loss, and similar viral loads as WT mice. Both 12/11-19 and 12/12-6 similarly induced IL-13–producing ILC2s through a TSLP-dependent mechanism. Conclusion These data demonstrate that multiple pathogenic strains of RSV induce IL-13–producing ILC2 proliferation and activation through a TSLP-dependent mechanism in a murine model and suggest the potential therapeutic targeting of TSLP during severe RSV infection.


American Journal of Respiratory and Critical Care Medicine | 2015

Prostaglandin I2 Signaling and Inhibition of Group 2 Innate Lymphoid Cell Responses

Weisong Zhou; Shinji Toki; Jian Zhang; Goleniewksa K; Dawn C. Newcomb; Jacqueline Yvonne Cephus; Daniel E. Dulek; Melissa H. Bloodworth; Matthew T. Stier; Polosuhkin; Rama Gangula; S. Mallal; Broide Dh; R.S. Peebles

RATIONALE Group 2 innate lymphoid cells (ILC2s) robustly produce IL-5 and IL-13, cytokines central to the asthma phenotype; however, the effect of prostaglandin (PG) I2 on ILC2 function is unknown. OBJECTIVES To determine the effect of PGI2 on mouse and human ILC2 cytokine expression in vitro and the effect of endogenous PGI2 and the PGI2 analog cicaprost on lung ILC2s in vivo. METHODS Flow-sorted bone marrow ILC2s of wild-type (WT) and PGI2 receptor-deficient (IP(-/-)) mice were cultured with IL-33 and treated with the PGI2 analog cicaprost. WT and IP(-/-) mice were challenged intranasally with Alternaria alternata extract for 4 consecutive days to induce ILC2 responses, and these were quantified. Prior to A. alternata extract, challenged WT mice were treated with cicaprost. Human flow-sorted peripheral blood ILC2s were cultured with IL-33 and IL-2 and treated with the PGI2 analog cicaprost. MEASUREMENT AND MAIN RESULTS We demonstrate that PGI2 inhibits IL-5 and IL-13 protein expression by IL-33-stimulated ILC2s purified from mouse bone marrow in a manner that was dependent on signaling through the PGI2 receptor IP. In a mouse model of 4 consecutive days of airway challenge with an extract of A. alternata, a fungal aeroallergen associated with severe asthma exacerbations, endogenous PGI2 signaling significantly inhibited lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s, as well as the mean fluorescence intensity of IL-5 and IL-13 staining. In addition, exogenous administration of a PGI2 analog inhibited Alternaria extract-induced lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s and the mean fluorescence intensity of IL-5 and IL-13 staining. Finally, a PGI2 analog inhibited IL-5 and IL-13 expression by human ILC2s that were stimulated with IL-2 and IL-33. CONCLUSIONS These results suggest that PGI2 may be a potential therapy to reduce the ILC2 response to protease-containing aeroallergens, such as Alternaria.


Thorax | 2016

The histone deacetylase inhibitor trichostatin A suppresses murine innate allergic inflammation by blocking group 2 innate lymphoid cell (ILC2) activation

Shinji Toki; Kasia Goleniewska; Sara Reiss; Weisong Zhou; Dawn C. Newcomb; Melissa H. Bloodworth; Matthew T. Stier; Kelli L. Boyd; Vasiliy V. Polosukhin; Sriram Subramaniam; R. Stokes Peebles

Background Group 2 innate lymphoid cells (ILC2) are an important source of the type 2 cytokines interleukin (IL)-5 and IL-13 that are critical to the allergic airway phenotype. Previous studies reported that histone deacetylase (HDAC) inhibition by trichostatin A (TSA) downregulated adaptive allergic immune responses; however, the effect of HDAC inhibition on the early innate allergic immune response is unknown. Therefore, we investigated the effect of TSA on innate airway inflammation mediated by ILC2 activation. Methods BALB/c mice were challenged intranasally with Alternaria extract, exogenous recombinant mouse IL-33 (rmIL-33) or the respective vehicles for four consecutive days following TSA or vehicle treatment. Bronchoalveolar lavage (BAL) fluids and lungs were harvested 24 h after the last challenge. Results We found that TSA treatment significantly decreased the number of ILC2 expressing IL-5 and IL-13 in the lungs challenged with Alternaria extract or rmIL-33 compared with vehicle treatment (p<0.05). TSA treatment significantly decreased protein expression of IL-5, IL-13, CCL11 and CCL24 in the lung homogenates from Alternaria extract-challenged mice or rmIL-33-challenged mice compared with vehicle treatment (p<0.05). Further, TSA treatment significantly decreased the number of perivascular eosinophils and mucus production in the large airways that are critical components of the asthma phenotype (p<0.05). TSA did not change early IL-33 release in the BAL fluids; however, TSA decreased lung IL-33 expression from epithelial cells 24 h after last Alternaria extract challenge compared with vehicle treatment (p<0.05). Conclusions These results reveal that TSA reduces allergen-induced ILC2 activation and the early innate immune responses to an inhaled protease-containing aeroallergen.


Journal of Experimental Medicine | 2018

IL-33 promotes the egress of group 2 innate lymphoid cells from the bone marrow

Matthew T. Stier; Jian Zhang; Kasia Goleniewska; Jacqueline Yvonne Cephus; Mark Rusznak; Lan Wu; Luc Van Kaer; Baohua Zhou; Dawn C. Newcomb; R. Stokes Peebles

Group 2 innate lymphoid cells (ILC2s) are effector cells within the mucosa and key participants in type 2 immune responses in the context of allergic inflammation and infection. ILC2s develop in the bone marrow from common lymphoid progenitor cells, but little is known about how ILC2s egress from the bone marrow for hematogenous trafficking. In this study, we identified a critical role for IL-33, a hallmark peripheral ILC2-activating cytokine, in promoting the egress of ILC2 lineage cells from the bone marrow. Mice lacking IL-33 signaling had normal development of ILC2s but retained significantly more ILC2 progenitors in the bone marrow via augmented expression of CXCR4. Intravenous injection of IL-33 or pulmonary fungal allergen challenge mobilized ILC2 progenitors to exit the bone marrow. Finally, IL-33 enhanced ILC2 trafficking to the lungs in a parabiosis mouse model of tissue disruption and repopulation. Collectively, these data demonstrate that IL-33 plays a critical role in promoting ILC2 egress from the bone marrow.


Journal of Immunology | 2017

STAT1 Represses Cytokine-Producing Group 2 and Group 3 Innate Lymphoid Cells during Viral Infection

Matthew T. Stier; Kasia Goleniewska; Jacqueline Yvonne Cephus; Dawn C. Newcomb; Taylor P. Sherrill; Kelli L. Boyd; Melissa H. Bloodworth; Martin L. Moore; Kong Chen; Jay K. Kolls; R. Stokes Peebles

The appropriate orchestration of different arms of the immune response is critical during viral infection to promote efficient viral clearance while limiting immunopathology. However, the signals and mechanisms that guide this coordination are not fully understood. IFNs are produced at high levels during viral infection and have convergent signaling through STAT1. We hypothesized that STAT1 signaling during viral infection regulates the balance of innate lymphoid cells (ILC), a diverse class of lymphocytes that are poised to respond to environmental insults including viral infections with the potential for both antiviral or immunopathologic functions. During infection with respiratory syncytial virus (RSV), STAT1-deficient mice had reduced numbers of antiviral IFN-γ+ ILC1 and increased numbers of immunopathologic IL-5+ and IL-13+ ILC2 and IL-17A+ ILC3 compared with RSV-infected wild-type mice. Using bone marrow chimeric mice, we found that both ILC-intrinsic and ILC-extrinsic factors were responsible for this ILC dysregulation during viral infection in STAT1-deficient mice. Regarding ILC-extrinsic mechanisms, we found that STAT1-deficient mice had significantly increased expression of IL-33 and IL-23, cytokines that promote ILC2 and ILC3, respectively, compared with wild-type mice during RSV infection. Moreover, disruption of IL-33 or IL-23 signaling attenuated cytokine-producing ILC2 and ILC3 responses in STAT1-deficient mice during RSV infection. Collectively, these data demonstrate that STAT1 is a key orchestrator of cytokine-producing ILC responses during viral infection via ILC-extrinsic regulation of IL-33 and IL-23.


Mammalian Genome | 2012

Polymorphisms in Ly6 genes in Msq1 encoding susceptibility to mouse adenovirus type 1

Matthew T. Stier; Katherine R. Spindler

Strain-specific differences in susceptibility to mouse adenovirus type 1 (MAV-1) are linked to the quantitative trait locus Msq1 on mouse chromosome 15. This region contains 14 Ly6 or Ly6-related genes, many of which are known to be expressed on the surface of immune cells, suggesting a possible role in host defense. We analyzed these genes for polymorphisms between MAV-1-susceptible and MAV-1-resistant inbred mouse strains. Sequencing of cDNAs identified 12 coding-region polymorphisms in 2010109I03Rik, Ly6e, Ly6a, Ly6c1, and Ly6c2, six of which were nonsynonymous and five of which were previously unlisted in dbSNP Build 132. We also clarified sequence discrepancies in GenBank for the coding regions of I830127L07Rik and Ly6g. Additionally, Southern blotting revealed size polymorphisms within the DNA regions of Ly6e, Ly6a, and Ly6g. Collectively, these genetic variations have implications for the structure, function, and/or expression of Ly6 and Ly6-related genes that may contribute to the observed strain-specific differences in susceptibility to MAV-1.


Annals of Allergy Asthma & Immunology | 2017

Innate lymphoid cells and allergic disease

Matthew T. Stier; R. Stokes Peebles

Allergic diseases—including asthma, atopic dermatitis, and allergic rhinitis—are substantial causes of morbidity and mortality in both developing and developed countries. An expansive set of cellular mediators and mechanisms have been identified that contribute to the initiation or exacerbation of allergic disease. Over the last decade, the discovery of a new subset of leukocytes known as innate lymphoid cells (ILC) has great expanded our understanding of the pathogenesis of allergic disease. ILC are effector lymphocytes that are embedded at mucosal sites and are involved in tissue homeostasis and response to diverse environmental insults. Derived from common lymphoid progenitors, ILC develop into three major cell subsets—group 1, 2, and 3 ILC.1 Group 1 ILC (ILC1) are driven by the transcription factor T-bet and produce IFN-γ. Group 2 ILC (ILC2) are governed by the transcription factor GATA3 and express IL-4, IL-5, IL-9, IL-13, and amphiregulin. Group 3 ILC (ILC3) are controlled by the transcription factor RORγt and produce IL-17 and/or IL-22. These transcription factor signatures and effector functions largely mirror CD4+ T helper (Th) 1, Th2, and Th17 cells, respectively. However, ILC exhibit several key differences that make them distinct from their CD4+ Th cell counterparts. In contrast to CD4+ T cells, ILC are primarily at mucosal sites even at baseline and are only found in low numbers in secondary lymphoid organs such as lymph nodes and the spleen.2 Moreover, even in ILC-rich tissues, ILC are found in significantly fewer numbers than CD4+ T cells. ILC do not depend on antigen presentation and can be activated immediately from stimuli in the tissue milieu such as cytokines, positioning them as innate counterparts to CD4+ T cells.3–5 Herein, we discuss the development and homeostasis of ILC, mechanistic insights into the role of ILC in the pathogenesis of allergy, and the contribution of ILC to human allergic disease, with a particular focus on ILC2 and their contributions to allergic disease pathogenesis.


The Journal of Allergy and Clinical Immunology | 2018

Glucagon-like peptide 1 receptor signaling attenuates respiratory syncytial virus–induced type 2 responses and immunopathology

Melissa H. Bloodworth; Mark Rusznak; Connor C. Pfister; Jian Zhang; Sandra Alvarez Calvillo; James D. Chappell; Kelli L. Boyd; Shinji Toki; Dawn C. Newcomb; Matthew T. Stier; Weisong Zhou; Kasia Goleniewska; Martin L. Moore; Tina V. Hartert; Kevin D. Niswender; R. Stokes Peebles

GLP-1R signaling, an emerging anti-inflammatory therapeutic target, attenuated type 2-associated immunopathology in mice infected with a strain of RSV that was isolated from a hospitalized infant with severe lower respiratory tract infection and bronchiolitis.


Cell Reports | 2017

Testosterone Attenuates Group 2 Innate Lymphoid Cell-Mediated Airway Inflammation

Jacqueline Yvonne Cephus; Matthew T. Stier; Hubaida Fuseini; Jeffrey A. Yung; Shinji Toki; Melissa H. Bloodworth; Weisong Zhou; Kasia Goleniewska; Jian Zhang; Sarah L. Garon; Robert G. Hamilton; Vasiliy V. Poloshukin; Kelli L. Boyd; R. Stokes Peebles; Dawn C. Newcomb


Cell Host & Microbe | 2017

Dietary Manganese Promotes Staphylococcal Infection of the Heart

Lillian J. Juttukonda; Evelien T.M. Berends; Joseph P Zackular; Jessica L. Moore; Matthew T. Stier; Yaofang Zhang; Jonathan E. Schmitz; William N. Beavers; Christiaan D. Wijers; Benjamin A. Gilston; Thomas E. Kehl-Fie; James B. Atkinson; Mary Kay Washington; R. Stokes Peebles; Walter J. Chazin; Victor J. Torres; Richard M. Caprioli; Eric P. Skaar

Collaboration


Dive into the Matthew T. Stier'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
Top Co-Authors

Avatar

Kelli L. Boyd

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Yvonne Cephus

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge