Network


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

Hotspot


Dive into the research topics where Heather Maecker is active.

Publication


Featured researches published by Heather Maecker.


Journal of Biological Chemistry | 2005

Molecular determinants of kinase pathway activation by apo2 ligand/tumor necrosis factor related apoptosis-inducing ligand

Eugene Varfolomeev; Heather Maecker; Darcie Sharp; David S. Lawrence; Mark Renz; Domagoj Vucic; Avi Ashkenazi

Apo2 ligand/tumor necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL) mainly activates programmed cell death through caspases. By contrast, TNF primarily induces gene transcription through the inhibitor of κB kinase (IKK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase pathways. Apo2L/TRAIL also can stimulate these kinases, albeit less strongly; however, the underlying mechanisms of this stimulation and its relation to apoptosis are not well understood. Here we show that Apo2L/TRAIL activates kinase pathways by promoting the association of a secondary signaling complex, subsequent to assembly of a primary, death-inducing signaling complex (DISC). The secondary complex retained the DISC components FADD and caspase-8, but recruited several factors involved in kinase activation by TNF, namely, RIP1, TRAF2, and NEMO/IKKγ. Secondary complex formation required Fas-associated death domain (FADD), as well as caspase-8 activity. Apo2L/TRAIL stimulation of JNK and p38 further depended on RIP1 and TRAF2, whereas IKK activation required NEMO. Apo2L/TRAIL induced secretion of interleukin-8 and monocyte chemoattractant protein-1, augmenting macrophage migration. Thus, Apo2L/TRAIL and TNF organize common molecular determinants in distinct signaling complexes to stimulate similar kinase pathways. One function of kinase stimulation by Apo2L/TRAIL may be to promote phagocytic engulfment of apoptotic cells.


Cell | 2005

TWEAK Attenuates the Transition from Innate to Adaptive Immunity

Heather Maecker; Eugene Varfolomeev; Frank C. Kischkel; David A. Lawrence; Heidi LeBlanc; Wyne P. Lee; Stephen Hurst; Dimitry M. Danilenko; Jun Li; Ellen Filvaroff; Becky Yang; Dylan Daniel; Avi Ashkenazi

Innate immunity is the first line of defense against infection, protecting the host during the development of adaptive immunity and critically affecting the nature of the adaptive response. We show that, in contrast to tumor necrosis factor alpha (TNF-alpha), the related protein TWEAK attenuates the transition from innate to adaptive mechanisms. TWEAK-/- mice had overabundant natural killer (NK) cells and displayed hypersensitivity to bacterial endotoxin, with their innate immune cells producing excess interferon (IFN)-gamma and interleukin (IL)-12. TWEAK inhibited stimulation of the transcriptional activator STAT-1 and induced p65 nuclear factor (NF)-kappaB association with histone deacetylase 1, repressing cytokine production. TWEAK-/- mice developed oversized spleens with expanded memory and T helper 1 (TH1) subtype cells upon aging and mounted stronger innate and adaptive TH1-based responses against tumor challenge. Thus, TWEAK suppresses production of IFN-gamma and IL-12, curtailing the innate response and its transition to adaptive TH1 immunity.


Cell | 2011

USP1 Deubiquitinates ID Proteins to Preserve a Mesenchymal Stem Cell Program in Osteosarcoma

Samuel A. Williams; Heather Maecker; Dorothy French; Jinfeng Liu; Andrew Gregg; Leah B. Silverstein; Tim C. Cao; Richard A. D. Carano; Vishva M. Dixit

Inhibitors of DNA binding (IDs) antagonize basic-helix-loop-helix (bHLH) transcription factors to inhibit differentiation and maintain stem cell fate. ID ubiquitination and proteasomal degradation occur in differentiated tissues, but IDs in many neoplasms appear to escape degradation. We show that the deubiquitinating enzyme USP1 promotes ID protein stability and stem cell-like characteristics in osteosarcoma. USP1 bound, deubiquitinated, and thereby stabilized ID1, ID2, and ID3. A subset of primary human osteosarcomas coordinately overexpressed USP1 and ID proteins. USP1 knockdown in osteosarcoma cells precipitated ID protein destabilization, cell-cycle arrest, and osteogenic differentiation. Conversely, ectopic USP1 expression in mesenchymal stem cells stabilized ID proteins, inhibited osteoblastic differentiation, and enhanced proliferation. Consistent with USP1 functioning in normal mesenchymal stem cells, USP1-deficient mice were osteopenic. Our observations implicate USP1 in preservation of the stem cell state that characterizes osteosarcoma and identify USP1 as a target for differentiation therapy.


Science Signaling | 2012

Cellular Inhibitors of Apoptosis Are Global Regulators of NF-κB and MAPK Activation by Members of the TNF Family of Receptors

Eugene Varfolomeev; Tatiana Goncharov; Heather Maecker; Kerry Zobel; Laszlo Komuves; Kurt Deshayes; Domagoj Vucic

Signaling downstream of tumor necrosis factor family receptors hinges on the presence or absence of c-IAP proteins. Directing TNFR Signaling with c-IAP Binding of ligands to tumor necrosis factor receptors (TNFRs) recruits adaptor proteins and E3 ubiquitin ligases to form signaling complexes that activate NF-κB and MAPK signaling, which are important in development and immunity. Varfolomeev et al. defined the roles of the E3 ubiquitin ligases c-IAP1 and c-IAP2, which are recruited to the TNFR1 complex by the adaptor protein TRAF2. The c-IAPs were critical for NF-κB and MAPK activation and for recruiting distal signaling components to specific TNFRs, and loss of c-IAPs resulted in diminished signaling by these TNFRs. Conversely, TNFR family members that stimulated noncanonical NF-κB signaling, which is inhibited by a complex containing c-IAP proteins, caused the translocation of c-IAP proteins from the cytosol to the plasma membrane, resulting in their degradation. Together, these data suggest that c-IAP proteins regulate canonical and noncanonical NF-κB signaling as well as MAPK activation by TNFR family members. Tumor necrosis factor (TNF) family members are essential for the development and proper functioning of the immune system. TNF receptor (TNFR) signaling is mediated through the assembly of protein signaling complexes that activate the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in a ubiquitin-dependent manner. The cellular inhibitor of apoptosis (c-IAP) proteins c-IAP1 and c-IAP2 are E3 ubiquitin ligases that are recruited to TNFR signaling complexes through their constitutive association with the adaptor protein TNFR-associated factor 2 (TRAF2). We demonstrated that c-IAP1 and c-IAP2 were required for canonical activation of NF-κB and MAPK by members of the TNFR family. c-IAPs were required for the recruitment of inhibitor of κB kinase β (IKKβ), the IKK regulatory subunit NF-κB essential modulator (NEMO), and RBCK1/Hoil1-interacting protein (HOIP) to TNFR signaling complexes and the induction of gene expression by TNF family members. In contrast, TNFRs that stimulated the noncanonical NF-κB pathway triggered translocation of c-IAPs, TRAF2, and TRAF3 from the cytosol to membrane fractions, which led to their proteasomal and lysosomal degradation. Finally, we established that signaling by B cell–activating factor receptor 3 induced the cytosolic depletion of TRAF3, which enabled noncanonical NF-κB activation. These results define c-IAP proteins as critical regulators of the activation of NF-κB and MAPK signaling pathways by members of the TNFR superfamily.


Immunity | 2016

MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade

Peter J.R. Ebert; Jeanne Cheung; Yagai Yang; Erin McNamara; Rebecca Hong; Marina Moskalenko; Stephen E. Gould; Heather Maecker; Bryan Irving; Jeong M. Kim; Marcia Belvin; Ira Mellman

Targeted inhibition of mitogen-activated protein kinase (MAPK) kinase (MEK) can induce regression of tumors bearing activating mutations in the Ras pathway but rarely leads to tumor eradication. Although combining MEK inhibition with T-cell-directed immunotherapy might lead to more durable efficacy, T cell responses are themselves at least partially dependent on MEK activity. We show here that MEK inhibition did profoundly block naive CD8(+) T cell priming in tumor-bearing mice, but actually increased the number of effector-phenotype antigen-specific CD8(+) T cells within the tumor. MEK inhibition protected tumor-infiltrating CD8(+) T cells from death driven by chronic TCR stimulation while sparing cytotoxic activity. Combining MEK inhibition with anti-programmed death-ligand 1 (PD-L1) resulted in synergistic and durable tumor regression even where either agent alone was only modestly effective. Thus, despite the central importance of the MAP kinase pathway in some aspects of T cell function, MEK-targeted agents can be compatible with T-cell-dependent immunotherapy.


Journal for ImmunoTherapy of Cancer | 2013

MAP kinase inhibitors stimulate T cell and anti-tumor activity in combination with blockade of the PD-L1/PD-1 interaction

Bryan Irving; Jeanne Cheung; Yagai Yang; Marina Moskalenka; Marcin Kowanetz; Heather Maecker; Ira Mellman

Pharmacological inhibition of the MAPK pathway with MEK or BRAF antagonists has proved successful in inducing regression of melanoma tumors bearing the targeted activating mutations. Moreover, antibodies targeting T-cell immune checkpoint inhibitors CTLA-4 or PD-L1/PD-1 have demonstrated the capacity to generate durable responses in patients with multiple cancer types. Thus, combining MAPK pathway-targeted agents with antibodies that enhance anti-tumor immunity represents an increasingly attractive treatment paradigm for cancer. However, little is known about the impact of tumor-targeted agents on immune function as similar signaling pathways drive both T-cell activation and cancer cell proliferation. Accordingly, agents targeting MAPK-dependent tumor growth would be predicted to also inhibit T-cell immunity. Here we show that, unexpectedly, potent suppression of T-cell receptor (TCR) function by MEK inhibition can be largely overcome in the presence of co-stimulation by anti-CD28 in vitro or blockade of the inhibitory PD-L1/PD-1 pathway in T cells in vivo. The ability of anti-CD28 to override suppression of T-cell activation by MEK inhibitors was dependent on the PI3K/mTOR pathway. Enhanced anti-tumor activity was also observed combining MEK inhibition with PD-L1 blockade, which was likely potentiated by upregulation of tumor MHC Class I expression through inhibition of MEK. Interestingly, inhibitors targeting BRAF V600E mutations actually augmented TCR-driven proliferation in vitro and T-cell function in vivo when combined with a vaccine or blockade of PD-L1 exclusively in the context of a wildtype BRAF background. These data demonstrate that targeting the MAPK pathway can be compatible with or even enhance T-cell function and provide rationale for combining these inhibitors with immunotherapy in clinical trials.


Cancer Research | 2013

Abstract 2849: Pharmacodynamic biomarkers of anti-PD-L1 activity in pre-clinical mouse tumor models.

Marigold Boe; Mahrukh Huseni; Brittany Jiang; Hartmut Koeppen; Heather Maecker; Bryan Irving; Priti Hegde; Marcin Kowanetz

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Programmed cell death 1 ligand 1 (PD-L1, CD274, B7-H1) is an immune checkpoint molecule expressed in various tissues, including multiple peripheral blood mononuclear cells and placenta. PD-L1 binds its receptor PD-1 on activated T cells to negatively regulate their function in both physiological and pathological conditions. PD-L1 also binds B7.1 (CD80), further down modulating immune responses. Importantly, numerous cancers resist T-cell mediated cytotoxic activity by over-expressing PD-L1. The goal of this study was to identify pharmacodynamic biomarkers of response to agents blocking PD-L1/PD-1 pathway in pre-clinical models. CT-26 and MC-38 murine mouse tumor models were selected for this analysis, based on their PD-L1 expression and in-vivo sensitivity to anti-PD-L1 treatment. Tumors have been collected at various time points following treatment with an antibody blocking PD-L1 binding to PD-1 and B7.1. Subsequently, FACS, IHC, qPCR and a multiplex qPCR-based expression assay measuring a panel of ∼90 immune-related biomarkers have been used to evaluate changes in major immune cell subtypes and their activation status. We show that the intra-tumoral immune environment undergoes a time-sensitive dynamic modulation in response to PD-L1 pathway blockade, including an early and potent Th1 driven CTL response, as measured by relevant markers. Additionally, our findings show that targeting PD-L1 in pre-clinical tumor models can reinvigorate the host immune responses against the tumor and lead to T-cell mediated tumor killing. Our data also support identification of potential pharmacodynamic biomarkers that can be used to monitor activity of agents targeting the PD-L1 pathway in the clinic. Citation Format: Marigold Boe, Mahrukh Huseni, Brittany Jiang, Hartmut Koeppen, Heather Maecker, Bryan Irving, Priti Hegde, Marcin Kowanetz. Pharmacodynamic biomarkers of anti-PD-L1 activity in pre-clinical mouse tumor models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2849. doi:10.1158/1538-7445.AM2013-2849


Cancer Research | 2012

Abstract 2151: Activation of NF-kB and MAP kinases by TNF Family Receptors is regulated by cellular IAPs

Evgeny Varfolomeev; Tatiana Goncharov; Heather Maecker; Kerry Zobel; Laszlo Komuves; Kurt Deshayes; Domagoj Vucic

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Activation of NF-kB, JNK and p38 MAPK signaling pathways is innermost activity of TNF receptor protein signaling complexes that is important for proper function of the immune system. However, excessive activation of these signaling pathways may result in tumor development and progression. Therefore, elucidation of the detailed molecular mechanisms of TNF receptor signaling should gain advantage in our understanding of the tumor biology and invention of the novel anticancer therapies. The formation of TNF receptor complexes, as well structural and functional associations between different signaling molecules is regulated by ubiquitination in TRAF-dependent fashion. TNF receptors employ several proteins possessing ubiquitin E3 ligase activities including cellular Inhibitor of Apoptosis, c-IAP1 and c-IAP2 proteins. We demonstrate that c-IAPs are required for canonical NF-kB and MAPK activation by TNF receptors. By regulating the recruitment of Nemo, IKK2 and HOIP to TNFR signaling complexes c-IAPs promote induction of gene expression by TNF ligands. We also found that TNF receptors that stimulate the noncanonical NF-kB pathway trigger c-IAPs, TRAF2 and TRAF3 intracellular translocation followed by their proteasomal and lysosomal degradation. Finally, we establish that BR3-induced cytosolic depletion of TRAF3 results in noncanonical NF-κB activation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2151. doi:1538-7445.AM2012-2151


Archive | 2009

Anti-PD-L1 antibodies, compositions and articles of manufacture

Bryan Irving; Henry Chiu; Heather Maecker; Sanjeev Mariathasan; Sophie M. Lehar; Yan Wu; Jeanne Cheung


Archive | 2012

METHODS OF TREATING CANCER USING PD-1 AXIS BINDING ANTAGONISTS AND MEK INHIBITORS

Heather Maecker; Bryan Irving

Collaboration


Dive into the Heather Maecker'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
Researchain Logo
Decentralizing Knowledge