Network


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

Hotspot


Dive into the research topics where Lisa M. Minter is active.

Publication


Featured researches published by Lisa M. Minter.


Nature Immunology | 2005

The nervous system and innate immunity: the neuropeptide connection

Lisa M. Minter; Danielle M. Turley; Pritam Das; Hyun Mu Shin; Ila Joshi; Rebecca G. Lawlor; Ok Hyun Cho; Tanapat Palaga; Sridevi Gottipati; Janice C. Telfer; Lisa Kostura; Abdul H. Fauq; Katherine Simpson; Kimberly A Such; Lucio Miele; Todd E. Golde; Stephen D. Miller; Barbara A. Osborne

Notch receptors are processed by γ-secretase acting in synergy with T cell receptor signaling to sustain peripheral T cell activation. Activated CD4+ T cells differentiate into T helper type 1 (TH1) or TH2 subsets. Molecular cues directing TH1 differentiation include expression of the TH1-specific transcription factor T-bet, encoded by Tbx21. However, the regulation of Tbx21 remains incompletely defined. Here we report that Notch1 can directly regulate Tbx21 through complexes formed on the Tbx21 promoter. In vitro, γ-secretase inhibitors extinguished expression of Notch, interferon-γ and Tbx21 in TH1-polarized CD4+ cells, whereas ectopic expression of activated Notch1 restored Tbx21 transcription. In vivo, administration of γ-secretase inhibitors substantially impeded TH1-mediated disease progression in the mouse experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, using γ-secretase inhibitors to modulate Notch signaling may prove beneficial in treating TH1-mediated autoimmunity.


The EMBO Journal | 2006

Notch1 augments NF-κB activity by facilitating its nuclear retention

Hyun Mu Shin; Lisa M. Minter; Ok Hyun Cho; Sridevi Gottipati; Abdul H. Fauq; Todd E. Golde; Gail E. Sonenshein; Barbara A. Osborne

Notch1 specifically upregulates expression of the cytokine interferon‐γ in peripheral T cells through activation of NF‐κB. However, how Notch mediates NF‐κB activation remains unclear. Here, we examined the temporal relationship between Notch signaling and NF‐κB induction during T‐cell activation. NF‐κB activation occurs within minutes of T‐cell receptor (TCR) engagement and this activation is sustained for at least 48 h following TCR signaling. We used γ‐secretase inhibitor (GSI) to prevent the cleavage and subsequent activation of Notch family members. We demonstrate that GSI blocked the later, sustained NF‐κB activation, but did not affect the initial activation of NF‐κB. Using biochemical approaches, as well as confocal microscopy, we show that the intracellular domain of Notch1 (N1IC) directly interacts with NF‐κB and competes with IκBα, leading to retention of NF‐κB in the nucleus. Additionally, we show that N1IC can directly regulate IFN‐γ expression through complexes formed on the IFN‐γ promoter. Taken together, these data suggest that there are two ‘waves’ of NF‐κB activation: an initial, Notch‐independent phase, and a later, sustained activation of NF‐κB, which is Notch dependent.


Nature Reviews Immunology | 2007

Notch signalling during peripheral T-cell activation and differentiation

Barbara A. Osborne; Lisa M. Minter

For many years, researchers have focused on the contribution of Notch signalling to lymphoid development. Only recently have investigators begun to ask what role, if any, Notch has during the activation and differentiation of naive CD4+ T cells in the periphery. As interest in this issue grows, it is becoming increasingly clear that the main role of Notch signalling, to regulate cell-fate decisions, might also be influential in peripheral T cells.


Blood | 2009

Notch signaling mediates G1/S cell-cycle progression in T cells via cyclin D3 and its dependent kinases

Ila Joshi; Lisa M. Minter; Janice C. Telfer; Renee M. Demarest; Anthony J. Capobianco; Piotr Sicinski; Abdul H. Fauq; Todd E. Golde; Barbara A. Osborne

Notch signaling plays a role in normal lymphocyte development and function. Activating Notch1-mutations, leading to aberrant downstream signaling, have been identified in human T-cell acute lymphoblastic leukemia (T-ALL). While this highlights the contribution of Notch signaling to T-ALL pathogenesis, the mechanisms by which Notch regulates proliferation and survival in normal and leukemic T cells are not fully understood. Our findings identify a role for Notch signaling in G(1)-S progression of cell cycle in T cells. Here we show that expression of the G(1) proteins, cyclin D3, CDK4, and CDK6, is Notch-dependent both in vitro and in vivo, and we outline a possible mechanism for the regulated expression of cyclin D3 in activated T cells via CSL (CBF-1, mammals; suppressor of hairless, Drosophila melanogaster; Lag-1, Caenorhabditis elegans), as well as a noncanonical Notch signaling pathway. While cyclin D3 expression contributes to cell-cycle progression in Notch-dependent human T-ALL cell lines, ectopic expression of CDK4 or CDK6 together with cyclin D3 shows partial rescue from gamma-secretase inhibitor (GSI)-induced G(1) arrest in these cell lines. Importantly, cyclin D3 and CDK4 are highly overexpressed in Notch-dependent T-cell lymphomas, justifying the combined use of cell-cycle inhibitors and GSI in treating human T-cell malignancies.


Journal of Immunology | 2011

Notch Signaling Regulates Mouse and Human Th17 Differentiation

Shilpa Keerthivasan; Reem Suleiman; Rebecca G. Lawlor; Justine E. Roderick; Tonya C. Bates; Lisa M. Minter; Juan Anguita; Ignacio J. Juncadella; Brian J. Nickoloff; I. Caroline Le Poole; Lucio Miele; Barbara A. Osborne

Th17 cells are known to play a critical role in adaptive immune responses to several important extracellular pathogens. Additionally, Th17 cells are implicated in the pathogenesis of several autoimmune and inflammatory disorders as well as in cancer. Therefore, it is essential to understand the mechanisms that regulate Th17 differentiation. Notch signaling is known to be important at several stages of T cell development and differentiation. In this study, we report that Notch1 is activated in both mouse and human in vitro-polarized Th17 cells and that blockade of Notch signaling significantly downregulates the production of Th17-associated cytokines, suggesting an intrinsic requirement for Notch during Th17 differentiation in both species. We also present evidence, using promoter reporter assays, knockdown studies, as well as chromatin immunoprecipitation, that IL-17 and retinoic acid-related orphan receptor γt are direct transcriptional targets of Notch signaling in Th17 cells. Finally, in vivo inhibition of Notch signaling reduced IL-17 production and Th17-mediated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. Thus, this study highlights the importance of Notch signaling in Th17 differentiation and indicates that selective targeted therapy against Notch may be an important tool to treat autoimmune disorders, including multiple sclerosis.


Journal of Immunology | 2009

Notch Regulates Cytolytic Effector Function in CD8+ T Cells

Ok Hyun Cho; Hyun Mu Shin; Lucio Miele; Todd E. Golde; Abdul H. Fauq; Lisa M. Minter; Barbara A. Osborne

The maturation of naive CD8+ T cells into effector CTLs is a critical feature of a functional adaptive immune system. Development of CTLs depends, in part, upon the expression of the transcriptional regulator eomesodermin (EOMES), which is thought to regulate expression of two key effector molecules, perforin and granzyme B. Although EOMES is important for effector CTL development, the precise mechanisms regulating CD8+ effector cell maturation remains poorly understood. In this study, we show that Notch1 regulates the expression of EOMES, perforin, and granzyme B through direct binding to the promoters of these crucial effector molecules. By abrogating Notch signaling, both biochemically as well as genetically, we conclude that Notch activity mediates CTL activity through direct regulation of EOMES, perforin, and granzyme B.


Clinical and Experimental Immunology | 2003

Hyperbaric oxygen inhibits stimulus-induced proinflammatory cytokine synthesis by human blood-derived monocyte-macrophages

Rm Benson; Lisa M. Minter; Barbara A. Osborne; Eric V. Granowitz

Hyperbaric oxygen (HBO) is 100% oxygen administered at elevated atmospheric pressure to patients with inflammatory diseases. We developed an in vitro model to investigate the effects of HBO on stimulus‐induced proinflammatory cytokine transcription and translation. Human blood‐derived monocyte‐macrophages were stimulated before being transferred to an HBO chamber where they were incubated at 97·9% O2, 2·1% CO2, 2·4 atmospheres absolute, 37°C. Controls were maintained in the same warm room at normoxia at sea level, hyperoxia or increased pressure alone. A 90‐min HBO exposure inhibited IL‐1β synthesized in response to lipopolysaccharide by 23%, lipid A by 45%, phytohaemagglutinin A (PHA) by 68%, and tumour necrosis factor (TNF)‐α by 27%. HBO suppressed lipopolysaccharide‐, lipid A‐ and PHA‐induced TNF‐α by 29%, 31% and 62%, respectively. HBO transiently reduced PHA‐induced steady state IL‐1β mRNA levels. Hyperoxia alone and pressure alone did not affect cytokine production. The immunosuppressive effect of HBO was no longer evident in monocyte‐macrophages exposed to HBO for more than 3 h. Interestingly, cells exposed to HBO for 12 h synthesized more IL‐1β than cells cultured under control conditions. In summary, HBO exposure transiently suppresses stimulus‐induced proinflammatory cytokine production and steady state RNA levels.


Frontiers in Immunology | 2014

Non-Canonical Notch Signaling Drives Activation and Differentiation of Peripheral CD4+ T Cells

Anushka Dongre; Lalitha Surampudi; Rebecca G. Lawlor; Abdul H. Fauq; Lucio Miele; Todd E. Golde; Lisa M. Minter; Barbara A. Osborne

Cleavage of the Notch receptor via a γ-secretase, results in the release of the active intra-cellular domain of Notch that migrates to the nucleus and interacts with RBP-Jκ, resulting in the activation of downstream target genes. This canonical Notch signaling pathway has been documented to influence T cell development and function. However, the mechanistic details underlying this process remain obscure. In addition to RBP-Jκ, the intra-cellular domain of Notch also interacts with other proteins in the cytoplasm and nucleus, giving rise to the possibility of an alternate, RBP-Jκ independent Notch pathway. However, the contribution of such RBP-Jκ independent, “non-canonical” Notch signaling in regulating peripheral T cell responses is unknown. In this report, we specifically demonstrate the requirement of Notch1 for regulating signal strength and signaling events distal to the T cell receptor in peripheral CD4+ T cells. By using mice with a conditional deletion in Notch1 or RBP-Jκ, we show that Notch1 regulates activation and proliferation of CD4+ T cells independently of RBP-Jκ. Furthermore, differentiation to TH1 and iTreg lineages although Notch dependent, is RBP-Jκ independent. Our striking observations demonstrate that many of the cell-intrinsic functions of Notch occur independently of RBP-Jκ. Such non-canonical regulation of these processes likely occurs through NF-κ B. This reveals a previously unknown, novel role of non-canonical Notch signaling in regulating peripheral T cell responses.


Molecular Therapy | 2013

Novel Protein Transduction Domain Mimics as Nonviral Delivery Vectors for siRNA Targeting NOTCH1 in Primary Human T cells

A. Özgül Tezgel; Gabriela Gonzalez-Perez; Janice C. Telfer; Barbara A. Osborne; Lisa M. Minter; Gregory N. Tew

RNA interference technology has recently been highlighted as a powerful research method as well as a potential therapeutic treatment for several diseases. However, the delivery of small interfering RNA (siRNA) into T cell lines and primary blood cells is exceedingly challenging, as they are resistant to transfection by conventional reagents. As a result, there is an unmet need for nonviral, efficient, and easily prepared carriers for siRNA delivery into hard-to-transfect cell types. Here, we report a novel system based on protein transduction domain mimics (PTDMs), generated by ring opening metathesis polymerization, for intracellular delivery of siRNA molecules. PTDM-based siRNA delivery induced efficient NOTCH1 knockdown in Jurkat T cells and human peripheral blood mononuclear cells without any measured toxicity. Furthermore, delivering siRNA to NOTCH1 in human peripheral blood cells modulated cell proliferation and differentiation of T cells into T(H)1 cells.


Journal of Experimental Medicine | 2013

Therapeutic targeting of NOTCH signaling ameliorates immune-mediated bone marrow failure of aplastic anemia

Justine E. Roderick; Gabriela Gonzalez-Perez; Christina Arieta Kuksin; Anushka Dongre; Emily R. Roberts; Janani Srinivasan; Chester Andrzejewski; Abdul H. Fauq; Todd E. Golde; Lucio Miele; Lisa M. Minter

Notch1 signaling sustains the proinflammatory behavior of Th1 cells, implicated in the development of aplastic anemia in humans and mice.

Collaboration


Dive into the Lisa M. Minter's collaboration.

Top Co-Authors

Avatar

Barbara A. Osborne

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Gregory N. Tew

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucio Miele

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriela Gonzalez-Perez

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Brittany M. deRonde

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Christina Arieta Kuksin

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Federica Sgolastra

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Rebecca G. Lawlor

University of Massachusetts Amherst

View shared research outputs
Researchain Logo
Decentralizing Knowledge