Network


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

Hotspot


Dive into the research topics where Amber M. Smith is active.

Publication


Featured researches published by Amber M. Smith.


PLOS Pathogens | 2009

Arginase-1–Expressing Macrophages Suppress Th2 Cytokine–Driven Inflammation and Fibrosis

John T. Pesce; Thirumalai R. Ramalingam; Margaret M. Mentink-Kane; Mark S. Wilson; Karim C. El Kasmi; Amber M. Smith; Robert W. Thompson; Allen W. Cheever; Peter J. Murray; Thomas A. Wynn

Macrophage-specific expression of Arginase-1 is commonly believed to promote inflammation, fibrosis, and wound healing by enhancing L-proline, polyamine, and Th2 cytokine production. Here, however, we show that macrophage-specific Arg1 functions as an inhibitor of inflammation and fibrosis following infection with the Th2-inducing pathogen Schistosoma mansoni. Although susceptibility to infection was not affected by the conditional deletion of Arg1 in macrophages, Arg1 −/flox ;LysMcre mice died at an accelerated rate. The mortality was not due to acute Th1/NOS2-mediated hepatotoxicity or endotoxemia. Instead, granulomatous inflammation, liver fibrosis, and portal hypertension increased in infected Arg1 −/flox ;LysMcre mice. Similar findings were obtained with Arg1 flox/flox ;Tie2cre mice, which delete Arg1 in all macrophage populations. Production of Th2 cytokines increased in the infected Arg1 −/flox ;LysMcre mice, and unlike alternatively activated wild-type macrophages, Arg1 −/flox ;LysMcre macrophages failed to inhibit T cell proliferation in vitro, providing an underlying mechanism for the exacerbated Th2 pathology. The suppressive activity of Arg1-expressing macrophages was independent of IL-10 and TGF-β1. However, when exogenous L-arginine was provided, T cell proliferation was restored, suggesting that Arg1-expressing macrophages deplete arginine, which is required to sustain CD4+ T cell responses. These data identify Arg1 as the essential suppressive mediator of alternatively activated macrophages (AAM) and demonstrate that Arg1-expressing macrophages function as suppressors rather than inducers of Th2-dependent inflammation and fibrosis.


Cell Host & Microbe | 2016

A Mouse Model of Zika Virus Pathogenesis

Helen M. Lazear; Jennifer Govero; Amber M. Smith; Derek J. Platt; Estefania Fernandez; Jonathan J. Miner; Michael S. Diamond

The ongoing Zika virus (ZIKV) epidemic and unexpected clinical outcomes, including Guillain-Barré syndrome and birth defects, has brought an urgent need for animal models. We evaluated infection and pathogenesis with contemporary and historical ZIKV strains in immunocompetent mice and mice lacking components of the antiviral response. Four- to six-week-old Irf3(-/-)Irf5(-/-)Irf7(-/-) triple knockout mice, which produce little interferon α/β, and mice lacking the interferon receptor (Ifnar1(-/-)) developed neurological disease and succumbed to ZIKV infection, whereas single Irf3(-/-), Irf5(-/-), and Mavs(-/-) knockout mice exhibited no overt illness. Ifnar1(-/-) mice sustained high viral loads in the brain and spinal cord, consistent with evidence that ZIKV causes neurodevelopmental defects in human fetuses. The testes of Ifnar1(-/-) mice had the highest viral loads, which is relevant to sexual transmission of ZIKV. This model of ZIKV pathogenesis will be valuable for evaluating vaccines and therapeutics as well as understanding disease pathogenesis.


Nature Immunology | 2014

Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages

Stanley Ching-Cheng Huang; Bart Everts; Yulia Ivanova; David O'Sullivan; Marcia Nascimento; Amber M. Smith; Wandy L. Beatty; Latisha Love-Gregory; Wing Y. Lam; Christina M. O'Neill; Cong Yan; Hong Du; Nada A. Abumrad; Joseph F. Urban; Maxim N. Artyomov; Erika L. Pearce; Edward J. Pearce

Alternative (M2) activation of macrophages driven via the α-chain of the receptor for interleukin 4 (IL-4Rα) is important for immunity to parasites, wound healing, the prevention of atherosclerosis and metabolic homeostasis. M2 polarization is dependent on fatty acid oxidation (FAO), but the source of the fatty acids that support this metabolic program has not been clear. We found that the uptake of triacylglycerol substrates via the scavenger receptor CD36 and their subsequent lipolysis by lysosomal acid lipase (LAL) was important for the engagement of elevated oxidative phosphorylation, enhanced spare respiratory capacity (SRC), prolonged survival and expression of genes that together define M2 activation. Inhibition of lipolysis suppressed M2 activation during infection with a parasitic helminth and blocked protective responses to this pathogen. Our findings delineate a critical role for cell-intrinsic lysosomal lipolysis in M2 activation.


Nature Immunology | 2014

TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation

Bart Everts; Eyal Amiel; Stanley Ching-Cheng Huang; Amber M. Smith; Chih-Hao Chang; Wing Y. Lam; Veronika Redmann; Tori C. Freitas; Julianna Blagih; Gerritje J.W. van der Windt; Maxim N. Artyomov; Russell G. Jones; Erika L. Pearce; Edward J. Pearce

The ligation of Toll-like receptors (TLRs) leads to rapid activation of dendritic cells (DCs). However, the metabolic requirements that support this process remain poorly defined. We found that DC glycolytic flux increased within minutes of exposure to TLR agonists and that this served an essential role in supporting the de novo synthesis of fatty acids for the expansion of the endoplasmic reticulum and Golgi required for the production and secretion of proteins that are integral to DC activation. Signaling via the kinases TBK1, IKKɛ and Akt was essential for the TLR-induced increase in glycolysis by promoting the association of the glycolytic enzyme HK-II with mitochondria. In summary, we identified the rapid induction of glycolysis as an integral component of TLR signaling that is essential for the anabolic demands of the activation and function of DCs.


Immunity | 2014

Memory CD8+ T Cells Use Cell-Intrinsic Lipolysis to Support the Metabolic Programming Necessary for Development

David O’Sullivan; Gerritje J.W. van der Windt; Stanley Ching-Cheng Huang; Jonathan D. Curtis; Chih-Hao Chang; Michael D. Buck; Jing Qiu; Amber M. Smith; Wing Y. Lam; Lisa M. DiPlato; Fong-Fu Hsu; Morris J. Birnbaum; Edward J. Pearce; Erika L. Pearce

Generation of CD8(+) memory T cells requires metabolic reprogramming that is characterized by enhanced mitochondrial fatty-acid oxidation (FAO). However, where the fatty acids (FA) that fuel this process come from remains unclear. While CD8(+) memory T cells engage FAO to a greater extent, we found that they acquired substantially fewer long-chain FA from their external environment than CD8(+) effector T (Teff) cells. Rather than using extracellular FA directly, memory T cells used extracellular glucose to support FAO and oxidative phosphorylation (OXPHOS), suggesting that lipids must be synthesized to generate the substrates needed for FAO. We have demonstrated that memory T cells rely on cell intrinsic expression of the lysosomal hydrolase LAL (lysosomal acid lipase) to mobilize FA for FAO and memory T cell development. Our observations link LAL to metabolic reprogramming in lymphocytes and show that cell intrinsic lipolysis is deterministic for memory T cell fate.


Proceedings of the National Academy of Sciences of the United States of America | 2013

CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability

Gerritje J.W. van der Windt; David O’Sullivan; Bart Everts; Stanley Ching-Cheng Huang; Michael D. Buck; Jonathan D. Curtis; Chih-Hao Chang; Amber M. Smith; Teresa Ai; Brandon Faubert; Russell G. Jones; Edward J. Pearce; Erika L. Pearce

A characteristic of memory T (TM) cells is their ability to mount faster and stronger responses to reinfection than naïve T (TN) cells do in response to an initial infection. However, the mechanisms that allow this rapid recall are not completely understood. We found that CD8 TM cells have more mitochondrial mass than CD8 TN cells and, that upon activation, the resulting secondary effector T (TE) cells proliferate more quickly, produce more cytokines, and maintain greater ATP levels than primary effector T cells. We also found that after activation, TM cells increase oxidative phosphorylation and aerobic glycolysis and sustain this increase to a greater extent than TN cells, suggesting that greater mitochondrial mass in TM cells not only promotes oxidative capacity, but also glycolytic capacity. We show that mitochondrial ATP is essential for the rapid induction of glycolysis in response to activation and the initiation of proliferation of both TN and TM cells. We also found that fatty acid oxidation is needed for TM cells to rapidly respond upon restimulation. Finally, we show that dissociation of the glycolysis enzyme hexokinase from mitochondria impairs proliferation and blocks the rapid induction of glycolysis upon T-cell receptor stimulation in TM cells. Our results demonstrate that greater mitochondrial mass endows TM cells with a bioenergetic advantage that underlies their ability to rapidly recall in response to reinfection.


Journal of Immunology | 2006

General Nature of the STAT3-Activated Anti-Inflammatory Response

Karim C. El Kasmi; Jeff Holst; Maryaline Coffre; Lisa A. Mielke; Antoine de Pauw; Nouara Lhocine; Amber M. Smith; Robert Rutschman; Deepak Kaushal; Yuhong Shen; Takashi Suda; Raymond P. Donnelly; Martin G. Myers; Warren S. Alexander; Dario A. A. Vignali; Stephanie S. Watowich; Matthias Ernst; Douglas J. Hilton; Peter J. Murray

Although many cytokine receptors generate their signals via the STAT3 pathway, the IL-10R appears unique in promoting a potent anti-inflammatory response (AIR) via STAT3 to antagonize proinflammatory signals that activate the innate immune response. We found that heterologous cytokine receptor systems that activate STAT3 but are naturally refractory (the IL-22R), or engineered to be refractory (the IL-6, leptin, and erythropoietin receptors), to suppressor of cytokine signaling-3-mediated inhibition activate an AIR indistinguishable from IL-10. We conclude that the AIR is a generic cytokine signaling pathway dependent on STAT3 but not unique to the IL-10R.


Cell Reports | 2016

Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus in Tears.

Jonathan J. Miner; Abdoulaye Sene; Justin M. Richner; Amber M. Smith; Andrea Santeford; Norimitsu Ban; James Weger-Lucarelli; Francesca Manzella; Claudia Rückert; Jennifer Govero; Kevin K. Noguchi; Gregory D. Ebel; Michael S. Diamond; Rajendra S. Apte

Zika virus (ZIKV) is an emerging flavivirus that causes congenital abnormalities and Guillain-Barré syndrome. ZIKV infection also results in severe eye disease characterized by optic neuritis, chorioretinal atrophy, and blindness in newborns and conjunctivitis and uveitis in adults. We evaluated ZIKV infection of the eye by using recently developed mouse models of pathogenesis. ZIKV-inoculated mice developed conjunctivitis, panuveitis, and infection of the cornea, iris, optic nerve, and ganglion and bipolar cells in the retina. This phenotype was independent of the entry receptors Axl or Mertk, given that Axl(-/-), Mertk(-/-), and Axl(-/-)Mertk(-/-) double knockout mice sustained levels of infection similar to those of control animals. We also detected abundant viral RNA in tears, suggesting that virus might be secreted from lacrimal glands or shed from the cornea. This model provides a foundation for studying ZIKV-induced ocular disease, defining mechanisms of viral persistence, and developing therapeutic approaches for viral infections of the eye.


Journal of Experimental Medicine | 2012

Agammaglobulinemia and absent B lineage cells in a patient lacking the p85α subunit of PI3K

Mary Ellen Conley; A. Kerry Dobbs; Anita M. Quintana; Amma Bosompem; Yong Dong Wang; Elaine Coustan-Smith; Amber M. Smith; Elena E. Perez; Peter J. Murray

A patient with a homozygous premature stop codon in PIK3R1 showed an early developmental block in B cell development but minimal effects in other organ systems.


Science Signaling | 2010

Arginine usage in mycobacteria-infected macrophages depends on autocrine-paracrine cytokine signaling.

Joseph E. Qualls; Geoffrey Neale; Amber M. Smith; Mi Sun Koo; Ashley A. DeFreitas; Huiyuan Zhang; Gilla Kaplan; Stephanie S. Watowich; Peter J. Murray

Mycobacteria trigger the production of cytokines that suppress immune responses in infected and uninfected macrophages. Spreading Suppression The amino acid arginine, a critical component in the earliest responses of the immune system to infection, is the substrate for inducible nitric oxide synthase (iNOS), which generates the antimicrobial compound NO in macrophages. In infected macrophages, mycobacteria stimulate a pathway that depends on the Toll-like receptor adaptor protein MyD88 to increase the production of Arg1, an enzyme that breaks down arginine, removing the source of NO and thereby dampening the immune response (see the Perspective by Morris). Qualls et al. showed that MyD88 was required for the production of cytokines that acted in an autocrine manner to drive the expression of Arg1 in the infected cells. However, these cytokines also acted on uninfected macrophages, thus inhibiting their ability to produce NO in the absence of infection. The generation of such a suppressed situation in vivo would enable spread of the infection. Nitric oxide (NO) produced by macrophages is toxic to host tissues and invading pathogens, and its regulation is essential to suppress host cytotoxicity. Macrophage arginase 1 (Arg1) competes with NO synthases for arginine, a substrate common to both types of enzymes, to inhibit NO production. Two signal transduction pathways control the production of Arg1 in macrophages: One pathway dependent on the Toll-like receptor adaptor protein myeloid differentiation marker 88 (MyD88) induces the expression of Arg1 during intracellular infections, whereas another pathway, which depends on signal transducer and activator of transcription 6 (STAT6), is required for Arg1 expression in alternatively activated macrophages. We found that mycobacteria-infected macrophages produced soluble factors, including interleukin-6 (IL-6), IL-10, and granulocyte colony-stimulating factor (G-CSF), that induced expression of Arg1 in an autocrine-paracrine manner. Arg1 expression was controlled by the MyD88-dependent production of these cytokines rather than by cell-intrinsic MyD88 signaling to Arg1. Our study revealed that the MyD88-dependent pathway that induced the expression of Arg1 after infection by mycobacteria required STAT3 activation and that this pathway may cause the development of an immunosuppressive niche in granulomas because of the induced production of Arg1 in surrounding uninfected macrophages.

Collaboration


Dive into the Amber M. Smith's collaboration.

Top Co-Authors

Avatar

Peter J. Murray

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Joseph E. Qualls

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Karim C. El Kasmi

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bart Everts

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stanley Ching-Cheng Huang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Geoffrey Neale

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jonathan J. Miner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Thomas A. Wynn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge