Network


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

Hotspot


Dive into the research topics where Margaret Alexander is active.

Publication


Featured researches published by Margaret Alexander.


Nature Communications | 2015

Exosome-delivered microRNAs modulate the inflammatory response to endotoxin.

Margaret Alexander; Ruozhen Hu; Marah C. Runtsch; Dominique A. Kagele; Timothy L. Mosbruger; Tanya Tolmachova; Miguel C. Seabra; June L. Round; Diane M. Ward; Ryan M. O'Connell

MicroRNAs regulate gene expression posttranscriptionally and function within the cells in which they are transcribed. However, recent evidence suggests that microRNAs can be transferred between cells and mediate target gene repression. We find that endogenous miR-155 and miR-146a, two critical microRNAs that regulate inflammation, are released from dendritic cells within exosomes and are subsequently taken up by recipient dendritic cells. Following uptake, exogenous microRNAs mediate target gene repression and can reprogramme the cellular response to endotoxin, where exosome-delivered miR-155 enhances while miR-146a reduces inflammatory gene expression. We also find that miR-155 and miR-146a are present in exosomes and pass between immune cells in vivo, as well as demonstrate that exosomal miR-146a inhibits while miR-155 promotes endotoxin-induced inflammation in mice. Together, our findings provide strong evidence that endogenous microRNAs undergo a functional transfer between immune cells and constitute a mechanism of regulating the inflammatory response.


Immunity | 2014

miR-155 Promotes T Follicular Helper Cell Accumulation during Chronic, Low-Grade Inflammation

Ruozhen Hu; Dominique A. Kagele; Thomas B. Huffaker; Marah C. Runtsch; Margaret Alexander; Jin Liu; Erin Bake; Wei Su; Matthew A. Williams; Dinesh S. Rao; Thomas Möller; Gwenn A. Garden; June L. Round; Ryan M. O’Connell

Chronic inflammation is a contributing factor to most life-shortening human diseases. However, the molecular and cellular mechanisms that sustain chronic inflammatory responses remain poorly understood, making it difficult to treat this deleterious condition. Using a mouse model of age-dependent inflammation that results from a deficiency in miR-146a, we demonstrate that miR-155 contributed to the progressive inflammatory disease that emerged as Mir146a(-/-) mice grew older. Upon analyzing lymphocytes from inflamed versus healthy middle-aged mice, we found elevated numbers of T follicular helper (Tfh) cells, germinal center (GC) B cells, and autoantibodies, all occurring in a miR-155-dependent manner. Further, Cd4-cre Mir155(fl/fl) mice were generated and demonstrated that miR-155 functions in T cells, in addition to its established role in B cells, to promote humoral immunity in a variety of contexts. Taken together, our study discovers that miR-146a and miR-155 counterregulate Tfh cell development that drives aberrant GC reactions during chronic inflammation.


Blood | 2017

miR-155 promotes FLT3-ITD–induced myeloproliferative disease through inhibition of the interferon response

Jared Wallace; Dominique A. Kagele; Anna M. Eiring; Carissa N. Kim; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Soh Hyun Lee; Ami B. Patel; Timothy L. Mosbruger; Dinesh S. Rao; Rodney R. Miles; June L. Round; Michael W. Deininger; Ryan M. O'Connell

FLT3-ITD+ acute myeloid leukemia (AML) accounts for ∼25% of all AML cases and is a subtype that carries a poor prognosis. microRNA-155 (miR-155) is specifically overexpressed in FLT3-ITD+ AML compared with FLT3 wild-type (FLT3-WT) AML and is critical for the growth of FLT3-ITD+ AML cells in vitro. However, miR-155s role in regulating FLT3-ITD-mediated disease in vivo remains unclear. In this study, we used a genetic mouse model to determine whether miR-155 influences the development of FLT3-ITD-induced myeloproliferative disease. Results indicate that miR-155 promotes FLT3-ITD-induced myeloid expansion in the bone marrow, spleen, and peripheral blood. Mechanistically, miR-155 increases proliferation of the hematopoietic stem and progenitor cell compartments by reducing the growth-inhibitory effects of the interferon (IFN) response, and this involves targeting of Cebpb. Consistent with our observations in mice, primary FLT3-ITD+ AML clinical samples have significantly higher miR-155 levels and a lower IFN response compared with FLT3-WT AML samples. Further, inhibition of miR-155 in FLT3-ITD+ AML cell lines using CRISPR/Cas9, or primary FLT3-ITD+ AML samples using locked nucleic acid antisense inhibitors, results in an elevated IFN response and reduces colony formation. Altogether, our data reveal that miR-155 collaborates with FLT3-ITD to promote myeloid cell expansion in vivo and that this involves a multitarget mechanism that includes repression of IFN signaling.


BioEssays | 2015

Noncoding RNAs and chronic inflammation: Micro-managing the fire within

Margaret Alexander; Ryan M. O'Connell

Inflammatory responses are essential for the clearance of pathogens and the repair of injured tissues; however, if these responses are not properly controlled chronic inflammation can occur. Chronic inflammation is now recognized as a contributing factor to many age‐associated diseases including metabolic disorders, arthritis, neurodegeneration, and cardiovascular disease. Due to the connection between chronic inflammation and these diseases, it is essential to understand underlying mechanisms behind this process. In this review, factors that contribute to chronic inflammation are discussed. Further, we emphasize the emerging roles of microRNAs (miRNAs) and other noncoding RNAs (ncRNA) in regulating chronic inflammatory states, making them important future diagnostic markers and therapeutic targets. Copyright Line:


Journal of Immunology | 2017

Rab27-Dependent exosome production inhibits chronic inflammation and enables acute responses to inflammatory stimuli

Margaret Alexander; Andrew G. Ramstead; Kaylyn M. Bauer; Soh-Hyun Lee; Marah C. Runtsch; Jared Wallace; Thomas B. Huffaker; Dane K. Larsen; Tanya Tolmachova; Miguel C. Seabra; June L. Round; Diane M. Ward; Ryan M. O’Connell

Extracellular vesicles, including exosomes, have recently been implicated as novel mediators of immune cell communication in mammals. However, roles for endogenously produced exosomes in regulating immune cell functions in vivo are just beginning to be identified. In this article, we demonstrate that Rab27a and Rab27b double-knockout (Rab27DKO) mice that are deficient in exosome secretion have a chronic, low-grade inflammatory phenotype characterized by elevated inflammatory cytokines and myeloproliferation. Upon further investigation, we found that some of these phenotypes could be complemented by wild-type (WT) hematopoietic cells or administration of exosomes produced by GM-CSF–expanded bone marrow cells. In addition, chronically inflamed Rab27DKO mice had a blunted response to bacterial LPS, resembling endotoxin tolerance. This defect was rescued by bone marrow exosomes from WT, but not miR-155−/−, cells, suggesting that uptake of miR-155–containing exosomes is important for a proper LPS response. Further, we found that SHIP1 and IRAK-M, direct targets of miR-155 that are known negative regulators of the LPS response, were elevated in Rab27DKO mice and decreased after treatment with WT, but not miR-155−/−, exosomes. Together, our study finds that Rab27-dependent exosome production contributes to homeostasis within the hematopoietic system and appropriate responsiveness to inflammatory stimuli.


Journal of Biological Chemistry | 2017

Antitumor immunity is defective in T cell–specific microRNA-155–deficient mice and is rescued by immune checkpoint blockade

Thomas B. Huffaker; Soh-Hyun Lee; William W. Tang; Jared Wallace; Margaret Alexander; Marah C. Runtsch; Dane K. Larsen; Jacob Thompson; Andrew G. Ramstead; Ruozhen Hu; June L. Round; Matthew A. Williams; Ryan M. O'Connell

MicroRNA-155 (miR-155) regulates antitumor immune responses. However, its specific functions within distinct immune cell types have not been delineated in conditional KO mouse models. In this study, we investigated the role of miR-155 specifically within T cells during the immune response to syngeneic tumors. We found that miR-155 expression within T cells is required to limit syngeneic tumor growth and promote IFNγ production by T cells within the tumor microenvironment. Consequently, we found that miR-155 expression by T cells is necessary for proper tumor-associated macrophage expression of IFNγ-inducible genes. We also found that immune checkpoint–blocking (ICB) antibodies against programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte–associated protein 4 (CTLA-4) restored antitumor immunity in miR-155 T cell–conditional KO mice. We noted that these ICB antibodies rescued the levels of IFNγ-expressing T cells, expression of multiple activation and effector genes expressed by tumor-infiltrating CD8+ and CD4+ T cells, and tumor-associated macrophage activation. Moreover, the ICB approach partially restored expression of several derepressed miR-155 targets in tumor-infiltrating, miR-155–deficient CD8+ T cells, suggesting that miR-155 and ICB regulate overlapping pathways to promote antitumor immunity. Taken together, our findings highlight the multifaceted role of miR-155 in T cells, in which it promotes antitumor immunity. These results suggest that the augmentation of miR-155 expression could be used to improve anticancer immunotherapies.


Oncotarget | 2015

MicroRNA-146a constrains multiple parameters of intestinal immunity and increases susceptibility to DSS colitis

Marah C. Runtsch; Ruozhen Hu; Margaret Alexander; Jared Wallace; Dominique A. Kagele; Charisse Petersen; John F. Valentine; Noah C. Welker; Mary P. Bronner; Xinjian Chen; Daniel P. Smith; Nadim J. Ajami; Joseph F. Petrosino; June L. Round; Ryan M. O'Connell


Archive | 2016

Exosome delivery of micrornas

Ryan M. O'Connell; Margaret Alexander


Blood | 2016

MiR-155 Promotes FLT3-ITD-Induced Myeloproliferative Disease through Inhibition of Interferon Signaling

Jared Wallace; Dominique A. Kagele; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Timothy L. Mosbruger; Dinesh S. Rao; Rodney R. Miles; June L. Round; Ryan M. O'Connell


Blood | 2015

Genome-Wide Crispr-Cas9 Screen Identifies Functionally Relevant Micro-RNAs in FLT3-ITD+ AML

Jared Wallace; Dominique A. Kagele; Ruozhen Hu; Marah C. Runtsch; Margaret Alexander; Thomas B. Huffaker; Timothy L. Mosbruger; Timothy J. Dahlem; W. Zac Stephens; Dinesh S. Rao; Rodney R. Miles; June L. Round; Ryan M. O'Connell

Collaboration


Dive into the Margaret Alexander'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

Dinesh S. Rao

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge