32 results on '"Emily Spaulding"'
Search Results
2. Blockade of LAG-3 in PD-L1-Deficient Mice Enhances Clearance of Blood Stage Malaria Independent of Humoral Responses
- Author
-
Raquel Furtado, Laurent Chorro, Natalie Zimmerman, Erik Guillen, Emily Spaulding, Shu Shien Chin, Johanna P. Daily, and Grégoire Lauvau
- Subjects
checkpoint therapeutic blockade ,PD-1/PD-L1/LAG-3 ,inhibitory receptors ,malaria ,humoral immunity ,LAG-3+ CD4+ and CD8+ T cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T cells expressing high levels of inhibitory receptors such as PD-1 and LAG-3 are a hallmark of chronic infections and cancer. Checkpoint blockade therapies targeting these receptors have been largely validated as promising strategies to restore exhausted T cell functions and clearance of chronic infections and tumors. The inability to develop long-term natural immunity in malaria-infected patients has been proposed to be at least partially accounted for by sustained expression of high levels of inhibitory receptors on T and B lymphocytes. While blockade or lack of PD-1/PD-L1 and/or LAG-3 was reported to promote better clearance of Plasmodium parasites in various mouse models, how exactly blockade of these pathways contributes to enhanced protection is not known. Herein, using the mouse model of non-lethal P. yoelii (Py) infection, we reveal that the kinetics of blood parasitemia as well as CD4+ T follicular helper (TFH) and germinal center (GC) B cell responses are indistinguishable between PD-1-/-, PD-L1-/- and WT mice. Yet, we also report that monoclonal antibody (mAb) blockade of LAG-3 in PD-L1-/- mice promotes accelerated control of blood parasite growth and clearance, consistent with prior therapeutic blockade experiments. However, neither CD4+ TFH and GC B cell responses, nor parasite-specific Ab serum titers and capacity to transfer protection differed. We also found that i) the majority of LAG-3+ cells are T cells, ii) selective depletion of CD4+ but not CD8+ T cells prevents anti-LAG-3-mediated protection, and iii) production of effector cytokines by CD4+ T cells is increased in anti-LAG-3-treated versus control mice. Thus, taken together, these results are consistent with a model in which blockade and/or deficiency of PD-L1 and LAG-3 on parasite-specific CD4+ T cells unleashes their ability to effectively clear blood parasites, independently from humoral responses.
- Published
- 2021
- Full Text
- View/download PDF
3. STING-Licensed Macrophages Prime Type I IFN Production by Plasmacytoid Dendritic Cells in the Bone Marrow during Severe Plasmodium yoelii Malaria.
- Author
-
Emily Spaulding, David Fooksman, Jamie M Moore, Alex Saidi, Catherine M Feintuch, Boris Reizis, Laurent Chorro, Johanna Daily, and Grégoire Lauvau
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Malaria remains a global health burden causing significant morbidity, yet the mechanisms underlying disease outcomes and protection are poorly understood. Herein, we analyzed the peripheral blood of a unique cohort of Malawian children with severe malaria, and performed a comprehensive overview of blood leukocytes and inflammatory mediators present in these patients. We reveal robust immune cell activation, notably of CD14+ inflammatory monocytes, NK cells and plasmacytoid dendritic cells (pDCs) that is associated with very high inflammation. Using the Plasmodium yoelii 17X YM surrogate mouse model of lethal malaria, we report a comparable pattern of immune cell activation and inflammation and found that type I IFN represents a key checkpoint for disease outcomes. Compared to wild type mice, mice lacking the type I interferon (IFN) receptor exhibited a significant decrease in immune cell activation and inflammatory response, ultimately surviving the infection. We demonstrate that pDCs were the major producers of systemic type I IFN in the bone marrow and the blood of infected mice, via TLR7/MyD88-mediated recognition of Plasmodium parasites. This robust type I IFN production required priming of pDCs by CD169+ macrophages undergoing activation upon STING-mediated sensing of parasites in the bone marrow. pDCs and macrophages displayed prolonged interactions in this compartment in infected mice as visualized by intravital microscopy. Altogether our findings describe a novel mechanism of pDC activation in vivo and precise stepwise cell/cell interactions taking place during severe malaria that contribute to immune cell activation and inflammation, and subsequent disease outcomes.
- Published
- 2016
- Full Text
- View/download PDF
4. Supplemental Figure 3 from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Figure 3. Representative gating strategy for the analysis of blood and spleen samples from HLA-A2 transgenic mice.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Figure 5 from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Figure 5. Antitumor activity of mCUE-101 in TC-1 tumors.
- Published
- 2023
- Full Text
- View/download PDF
6. Data from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Purpose:To assess the potential for CUE-101, a novel therapeutic fusion protein, to selectively activate and expand HPV16 E711-20-specific CD8+ T cells as an off-the shelf therapy for the treatment of HPV16-driven tumors, including head and neck squamous cell carcinoma (HNSCC), cervical, and anal cancers.Experimental Design:CUE-101 is an Fc fusion protein composed of a human leukocyte antigen (HLA) complex, an HPV16 E7 peptide epitope, reduced affinity human IL2 molecules, and an effector attenuated human IgG1 Fc domain. Human E7-specific T cells and human peripheral blood mononuclear cells (PBMC) were tested to demonstrate cellular activity and specificity of CUE-101, whereas in vivo activity of CUE-101 was assessed in HLA-A2 transgenic mice. Antitumor efficacy with a murine surrogate (mCUE-101) was tested in the TC-1 syngeneic tumor model.Results:CUE-101 demonstrates selective binding, activation, and expansion of HPV16 E711-20-specific CD8+ T cells from PBMCs relative to nontarget cells. Intravenous administration of CUE-101 induced selective expansion of HPV16 E711-20-specific CD8+ T cells in HLA-A2 (AAD) transgenic mice, and anticancer efficacy and immunologic memory was demonstrated in TC-1 tumor-bearing mice treated with mCUE-101. Combination therapy with anti-PD-1 checkpoint blockade further enhanced the observed efficacy.Conclusions:Consistent with its design, CUE-101 demonstrates selective expansion of an HPV16 E711-20-specific population of cytotoxic CD8+ T cells, a favorable safety profile, and in vitro and in vivo evidence supporting its potential for clinical efficacy in an ongoing phase I trial (NCT03978689).
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Materials and Methods from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 4 from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Figure 4. Antitumor activity of mCUE-101 in established TC-1 tumors.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Figure 6 from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Figure 6. Characterization of E7-specific CD8+ T cells in mice bearing TC-1 tumors.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Table 1
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Figure 1 from CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
Mary Ellen Simcox, Kenneth J. Pienta, Saso Cemerski, Anish Suri, Ronald Seidel, Rodolfo Chaparro, Steven Almo, Peter A. Kiener, John F. Ross, Emily Spaulding, Mark Haydock, Jonathan Soriano, Jessica Ryabin, Luke Witt, Dominic R. Beal, Lauren D. Kraemer, Alyssa Nelson, Sandrine Hulot, Paige Ruthardt, Miguel Moreta, Fan Zhao, Alex Histed, Melissa M. Kemp, Zohra Merazga, Dharma R. Thapa, Natasha Girgis, and Steven N. Quayle
- Abstract
Supplementary Figure 1. Characterization of CUE-101 binding properties.
- Published
- 2023
- Full Text
- View/download PDF
12. A longitudinal and cross‐sectional study of plasma neurofilament light chain concentration in <scp>Charcot‐Marie‐Tooth</scp> disease
- Author
-
Matilde Laura, Alexander M. Rossor, Robert W. Burgess, James N. Sleigh, Henny Wellington, Alexa Bacha, Reilly Mm, Mahima Kapoor, Shy Me, Amanda Heslegrave, Henrik Zetterberg, Xingyao Wu, and Emily Spaulding
- Subjects
Adult ,Change over time ,Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Treatment response ,Cross-sectional study ,Neurofilament light ,Intermediate Filaments ,Disease ,Cohort Studies ,Mice ,03 medical and health sciences ,Tooth disease ,0302 clinical medicine ,Charcot-Marie-Tooth Disease ,Neurofilament Proteins ,Internal medicine ,Animals ,Humans ,Medicine ,030304 developmental biology ,0303 health sciences ,business.industry ,General Neuroscience ,3. Good health ,Clinical trial ,Cross-Sectional Studies ,Cohort ,Neurology (clinical) ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Aims Advances in genetic technology and small molecule drug development have paved the way for clinical trials in Charcot-Marie-Tooth disease (CMT), however, the current FDA-approved clinical trial outcome measures are insensitive to detect a meaningful clinical response. There is therefore a need to identify sensitive outcome measures or clinically relevant biomarkers. The aim of this study was to further evaluate plasma neurofilament light chain (NFL) as a disease biomarker in CMT. Methods Plasma NFL was measured using SIMOA technology in both a cross sectional study of a US cohort of CMT patients and longitudinally over six years in a UK CMT cohort. In addition, plasma NFL was measured longitudinally in two mouse models of CMT2D. Results Plasma concentrations of NFL were increased in a US cohort of patients with CMT1B, CMT1X and CMT2A but not CMT2E compared with controls. In a separate UK cohort, over a six-year interval, there was no significant change in plasma NFL concentration in CMT1A or HSN1, but a small but significant reduction in patients with CMT1X. Plasma NFL was increased in wild type compared to GARSC201R mice. There was no significant difference in plasma NFL in GARSP278KY compared to wild type mice. Conclusion In patients with CMT1A, the small difference in cross sectional NFL concentration versus healthy controls and the lack of change over time suggests that plasma NFL may lack sufficient sensitivity to detect a clinically meaningful treatment response in adulthood. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
13. Peptide-HLA-based immunotherapeutics platforms for direct modulation of antigen-specific T cells
- Author
-
Rodolfo J. Chaparro, Paige Ruthardt, Wynona Bautista, R.D. Seidel, John F. Ross, Anish Suri, Zohra Merazga, Dharma Thapa, Emily Spaulding, Peter A. Kiener, Jonathan Soriano, Steven N. Quayle, Ahmet S. Vakkasoglu, Saso Cemerski, Simon Low, and Steven C. Almo
- Subjects
T cell ,Recombinant Fusion Proteins ,Science ,Immunology ,Primary Cell Culture ,Stimulation ,Peptide ,Mice, Transgenic ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Protein Engineering ,Article ,Mice ,Antigen specific ,Neoplasms ,medicine ,Animals ,Humans ,Cells, Cultured ,chemistry.chemical_classification ,Multidisciplinary ,HLA-A Antigens ,Chemistry ,Cancer ,medicine.disease ,In vitro ,medicine.anatomical_structure ,Mutation ,Cancer research ,B7-1 Antigen ,Medicine ,Immunotherapy ,Peptides ,CD80 ,Biotechnology - Abstract
Targeted pharmacologic activation of antigen-specific (AgS) T cells may bypass limitations inherent in current T cell-based cancer therapies. We describe two immunotherapeutics platforms for selective delivery of costimulatory ligands and peptide-HLA (pHLA) to AgS T cells. We engineered and deployed on these platforms an affinity-attenuated variant of interleukin-2, which selectively expands oligoclonal and polyfunctional AgS T cells in vitro and synergizes with CD80 signals for superior proliferation versus peptide stimulation.
- Published
- 2021
14. tRNA overexpression rescues peripheral neuropathy caused by mutations in tRNA synthetase
- Author
-
Divita Kulshrestha, Nick H.M. van Bakel, Erik Storkebaum, Céline Sijlmans, Marica Catinozzi, Abigail L. D. Tadenev, Marije Been, Zoya Ignatova, Sarada Das, Robert W. Burgess, Amila Zuko, Moushami Mallik, Emily Spaulding, Robin Thompson, Anne R. Wienand, Leonardo A. Santos, and Julia Bussmann
- Subjects
Multidisciplinary ,Transgene ,Mutant ,RNA ,Biology ,medicine.disease ,biology.organism_classification ,Molecular biology ,Peripheral neuropathy ,Transfer RNA ,medicine ,Protein biosynthesis ,Drosophila melanogaster ,Gene ,Molecular Neurobiology - Abstract
Defeating peripheral neuropathy The mechanisms underlying peripheral neuropathies are not well understood. Spaulding et al . studied mouse models of the inherited Charcot-Marie-Tooth (CMT) disease, which is caused by mutations in transfer RNA (tRNA) synthetases. Changes in gene expression and the rate of protein synthesis in neurons in the spinal cord triggered the cell stress response activated by the protein sensor GCN2. When GCN2 was genetically deleted or inhibited with drugs, the stress response was blocked, and the neuropathy was much milder. Zuko et al . found that mutant glycyl-tRNA synthetases bind tRNA Gly but fail to release it, thus depleting the cellular tRNA Gly pool. This process caused stalling of translating ribosomes on glycine codons and activated the integrated stress response. Transgenic tRNA Gly overexpression prevented peripheral neuropathy and protein synthesis defects in mouse and fruit fly models. Thus, elevating tRNA Gly levels or targeting GCN2 may have therapeutic potential for this currently untreatable disease (see the Perspective by Mellado and Willis). —SMH
- Published
- 2021
15. CUE-101, a Novel E7-pHLA-IL2-Fc Fusion Protein, Enhances Tumor Antigen-Specific T-Cell Activation for the Treatment of HPV16-Driven Malignancies
- Author
-
John F. Ross, Dominic R. Beal, Jonathan Soriano, R.D. Seidel, Rodolfo J. Chaparro, Natasha Girgis, Kenneth J. Pienta, Zohra Merazga, Paige Ruthardt, Fan Zhao, Peter A. Kiener, Miguel Moreta, Sandrine Hulot, Anish Suri, Steven N. Quayle, Luke Witt, Saso Cemerski, Dharma Thapa, Mark Haydock, Alex Histed, Melissa M. Kemp, Steven C. Almo, Lauren D. Kraemer, Jessica Ryabin, Mary Simcox, Emily Spaulding, and Alyssa Nelson
- Subjects
0301 basic medicine ,Cancer Research ,Papillomavirus E7 Proteins ,T cell ,Mice, Transgenic ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Epitope ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Neoplasms ,HLA-A2 Antigen ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Cells, Cultured ,Chemistry ,Fusion protein ,Healthy Volunteers ,Tumor antigen ,Immunoglobulin Fc Fragments ,Tumor Necrosis Factor Receptor Superfamily, Member 7 ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Leukocytes, Mononuclear ,Cancer research ,Interleukin-2 ,Female ,CD8 - Abstract
Purpose: To assess the potential for CUE-101, a novel therapeutic fusion protein, to selectively activate and expand HPV16 E711-20-specific CD8+ T cells as an off-the shelf therapy for the treatment of HPV16-driven tumors, including head and neck squamous cell carcinoma (HNSCC), cervical, and anal cancers. Experimental Design: CUE-101 is an Fc fusion protein composed of a human leukocyte antigen (HLA) complex, an HPV16 E7 peptide epitope, reduced affinity human IL2 molecules, and an effector attenuated human IgG1 Fc domain. Human E7-specific T cells and human peripheral blood mononuclear cells (PBMC) were tested to demonstrate cellular activity and specificity of CUE-101, whereas in vivo activity of CUE-101 was assessed in HLA-A2 transgenic mice. Antitumor efficacy with a murine surrogate (mCUE-101) was tested in the TC-1 syngeneic tumor model. Results: CUE-101 demonstrates selective binding, activation, and expansion of HPV16 E711-20-specific CD8+ T cells from PBMCs relative to nontarget cells. Intravenous administration of CUE-101 induced selective expansion of HPV16 E711-20-specific CD8+ T cells in HLA-A2 (AAD) transgenic mice, and anticancer efficacy and immunologic memory was demonstrated in TC-1 tumor-bearing mice treated with mCUE-101. Combination therapy with anti-PD-1 checkpoint blockade further enhanced the observed efficacy. Conclusions: Consistent with its design, CUE-101 demonstrates selective expansion of an HPV16 E711-20-specific population of cytotoxic CD8+ T cells, a favorable safety profile, and in vitro and in vivo evidence supporting its potential for clinical efficacy in an ongoing phase I trial (NCT03978689).
- Published
- 2020
- Full Text
- View/download PDF
16. Allele-specific RNA interference prevents neuropathy in Charcot-Marie-Tooth disease type 2D mouse models
- Author
-
Kathryn H. Morelli, Laurie B. Griffin, Allison M. Fowler, Timothy J. Hines, James R. Lupski, Lindsay M. Wallace, Samuel G. Kocen, Scott Q. Harper, Jacob O. Kitzman, Ryuichi Takase, Stephanie N. Oprescu, Alexey I. Nesvizhskii, Rebecca Meyer-Schuman, Nettie K. Pyne, Pedro Mancias, Robert W. Burgess, Dattatreya Mellacheruvu, Ya-Ming Hou, Emily Spaulding, Anthony Antonellis, Na Wei, Xiang-Lei Yang, and Ian J. Butler
- Subjects
Glycine-tRNA Ligase ,0301 basic medicine ,Genetic enhancement ,Mutant ,Charcot-Marie-Tooth Disease Type 2D ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Charcot-Marie-Tooth Disease ,RNA interference ,Mutant protein ,Animals ,Humans ,Medicine ,Allele ,Alleles ,Gene knockdown ,business.industry ,Genetic Therapy ,General Medicine ,medicine.disease ,Disease Models, Animal ,HEK293 Cells ,030104 developmental biology ,Peripheral neuropathy ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,RNA Interference ,business ,Research Article - Abstract
Gene therapy approaches are being deployed to treat recessive genetic disorders by restoring the expression of mutated genes. However, the feasibility of these approaches for dominantly inherited diseases — where treatment may require reduction in the expression of a toxic mutant protein resulting from a gain-of-function allele — is unclear. Here we show the efficacy of allele-specific RNAi as a potential therapy for Charcot-Marie-Tooth disease type 2D (CMT2D), caused by dominant mutations in glycyl-tRNA synthetase (GARS). A de novo mutation in GARS was identified in a patient with a severe peripheral neuropathy, and a mouse model precisely recreating the mutation was produced. These mice developed a neuropathy by 3–4 weeks of age, validating the pathogenicity of the mutation. RNAi sequences targeting mutant GARS mRNA, but not wild-type, were optimized and then packaged into AAV9 for in vivo delivery. This almost completely prevented the neuropathy in mice treated at birth. Delaying treatment until after disease onset showed modest benefit, though this effect decreased the longer treatment was delayed. These outcomes were reproduced in a second mouse model of CMT2D using a vector specifically targeting that allele. The effects were dose dependent, and persisted for at least 1 year. Our findings demonstrate the feasibility of AAV9-mediated allele-specific knockdown and provide proof of concept for gene therapy approaches for dominant neuromuscular diseases.
- Published
- 2019
- Full Text
- View/download PDF
17. Blockade of LAG-3 in PD-L1-deficient mice enhances clearance of blood stage malaria independent of humoral responses
- Author
-
Raquel Furtado, Laurent Chorro, Natalie Zimmerman, Erik Guillen, Emily Spaulding, Shu Shien Chin, Johanna P. Daily, and Grégoire Lauvau
- Subjects
CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,Helper T lymphocyte ,Programmed Cell Death 1 Receptor ,Parasitemia ,B7-H1 Antigen ,Mice ,0302 clinical medicine ,humoral immunity ,Immunology and Allergy ,Cytotoxic T cell ,Malaria, Falciparum ,Receptor ,Immune Checkpoint Inhibitors ,Cells, Cultured ,Original Research ,Mice, Knockout ,checkpoint therapeutic blockade ,biology ,Chemistry ,Antibodies, Monoclonal ,Lymphocyte Activation Gene 3 Protein ,medicine.anatomical_structure ,PD-1/PD-L1/LAG-3 ,Antibody ,LAG-3+ CD4+ and CD8+ T cells ,lcsh:Immunologic diseases. Allergy ,PD-L1 and PD-1 knockout mice ,T cell ,Plasmodium falciparum ,Immunology ,malaria ,03 medical and health sciences ,Antigens, CD ,medicine ,Animals ,Humans ,B cell ,Life Cycle Stages ,Innate immune system ,Germinal center ,Plasmodium yoelii ,medicine.disease ,Immunity, Humoral ,Blockade ,Mice, Inbred C57BL ,Disease Models, Animal ,inhibitory receptors ,030104 developmental biology ,Humoral immunity ,biology.protein ,lcsh:RC581-607 ,CD8 ,030215 immunology - Abstract
T cells expressing high levels of inhibitory receptors such as PD-1 and LAG-3 are a hallmark of chronic infections and cancer. Checkpoint blockade therapies targeting these receptors have been largely validated as promising strategies to restore exhausted T cell functions and clearance of chronic infections and tumors. The inability to develop long-term natural immunity in malaria-infected patients has been proposed to be at least partially accounted for by sustained expression of high levels of inhibitory receptors on T and B lymphocytes. While blockade or lack of PD-1/PD-L1 and/or LAG-3 was reported to promote better clearance of Plasmodium parasites in mice, how exactly these pathways contributes to protection is not known. Herein, using a mouse model of non-lethal P. yoelii (Py) infection, we reveal that the kinetics of blood parasitemia is indistinguishable between PD-1-/-, PD-L1-/- and WT mice. Yet, monoclonal antibody (mAb) blockade of LAG-3 in PD-L1-/- mice promoted accelerated control of blood parasite growth and clearance. We also report that i) the majority of LAG-3+ cells are T cells, ii) selective depletion of CD8+ T cells did not prevent anti-LAG-3-mediated protection, and iii) production of effector cytokines by CD4+ T cells is increased in anti-LAG-3-treated versus control mice. In addition, parasite-specific Ab serum titers and their ability to transfer protection from both groups of mice was comparable and depletion of CD4+ T cells prevented protection. Thus, taken together, these results are consistent with a model in which disruption of PD-L1 and LAG-3 on parasite-specific CD4+ T cells unleashes their ability to effectively clear blood parasites, independently from humoral responses.Author SummaryMalaria, caused by Plasmodium parasites, is a global burden for which an efficacious vaccine is urgently needed. The development of long-term immunity against malaria is unclear, but we know that both T and B (that produce antibodies, Ab) lymphocytes, that are subsets of white blood cells, are required. Studies in mouse models of malaria have suggested that sets of inhibitory receptors, namely LAG-3 and PD-1, expressed on cytotoxic and helper T lymphocytes hamper the development of effective immunity against malaria. Therapeutic blockade of these receptors was reported to enhance blood parasite clearance through the development of more protective parasite-specific helper T lymphocytes and Abs. Herein, we reveal that, while mice genetically deficient for the PD-1 pathway fail to clear blood parasites better than WT counterparts, anti-LAG-3 treatment does. Importantly, we found comparable parasite-specific Ab responses between all mouse groups, and Ab transfers conferred similar protection to newly infected mice. We also show that LAG-3 is mostly expressed on T lymphocytes, and that cytotoxic T lymphocytes are not involved in anti-LAG-3 accelerated clearance of parasites. Our results suggest that LAG-3 blockade acts on helper T lymphocytes to unleash their effector responses and enhance the control of blood-stage malaria, independently from parasite-specific Abs.
- Published
- 2020
- Full Text
- View/download PDF
18. Severity of Demyelinating and Axonal Neuropathy Mouse Models Is Modified by Genes Affecting Structure and Function of Peripheral Nodes
- Author
-
Loiuse A. Dionne, Robert W. Burgess, Kathryn H. Morelli, David G. Schroeder, Emily Spaulding, Gregory A. Cox, and Kevin L. Seburn
- Subjects
0301 basic medicine ,length constant ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,Neuromuscular Junction ,degeneration ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Myelin ,0302 clinical medicine ,Atrophy ,Charcot-Marie-Tooth Disease ,SH3TC2 ,medicine ,Animals ,Peripheral Nerves ,motor neuropathy ,Axon ,hereditary sensory ,lcsh:QH301-705.5 ,Mutation ,Genetic heterogeneity ,Intracellular Signaling Peptides and Proteins ,Heterozygote advantage ,Anatomy ,genetic modifiers ,medicine.disease ,Axons ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Peripheral neuropathy ,lcsh:Biology (General) ,NAV1.6 Voltage-Gated Sodium Channel ,Carrier Proteins ,Cell Adhesion Molecules ,Neuroscience ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous group of inherited polyneuropathies. Mutations in 80 genetic loci can cause forms of CMT, resulting in demyelination and axonal dysfunction. The clinical presentation, including sensory deficits, distal muscle weakness, and atrophy, can vary greatly in severity and progression. Here, we used mouse models of CMT to demonstrate genetic interactions that result in a more severe neuropathy phenotype. The cell adhesion molecule Nrcam and the Na+ channel Scn8a (NaV1.6) are important components of nodes. Homozygous Nrcam and heterozygous Scn8a mutations synergized with both an Sh3tc2 mutation, modeling recessive demyelinating Charcot-Marie-Tooth type 4C, and mutations in Gars, modeling dominant axonal Charcot-Marie-Tooth type 2D. We conclude that genetic variants perturbing the structure and function of nodes interact with mutations affecting the cable properties of axons by thinning myelin or reducing axon diameter. Therefore, genes integral to peripheral nodes are candidate modifiers of peripheral neuropathy.
- Published
- 2017
- Full Text
- View/download PDF
19. Inflammatory monocyte effector mechanisms
- Author
-
Grégoire Lauvau, Emily Spaulding, Laurent Chorro, and Saidi M.Homa Soudja
- Subjects
Inflammation ,CCR2 ,Innate immune system ,Receptors, CCR2 ,Effector ,CD14 ,Immunology ,Antigen presentation ,Antigen-Presenting Cells ,Inflammatory monocyte ,Adaptive Immunity ,Biology ,Immunity, Innate ,Monocytes ,Article ,Microbiology ,Mice ,Immune system ,In vivo ,Animals ,Antigens, Ly ,Humans - Abstract
Monocytes are blood-derived mononuclear phagocytic cells that traffic throughout the body and can provide rapid innate immune effector responses in response to microbial pathogen infections. Among blood monocytes, the most abundant subset in mice is represented by inflammatory Ly6C(+) CCR2(+) monocytes and is the functional equivalent of the CD14(+) monocytes in humans. Herein we focus on published evidence describing the exquisite functional plasticity of these cells, and we extend this overview to their multiples roles in vivo during host immune defenses against microbial pathogen infections, as antigen-presenting cells, inflammatory cells or Trojan horse cells.
- Published
- 2014
- Full Text
- View/download PDF
20. STING-Licensed Macrophages Prime Type I IFN Production by Plasmacytoid Dendritic Cells in the Bone Marrow during Severe Plasmodium yoelii Malaria
- Author
-
Alex Saidi, Boris Reizis, Emily Spaulding, Johanna P. Daily, Grégoire Lauvau, Laurent Chorro, Catherine M. Feintuch, Jamie Moore, and David R. Fooksman
- Subjects
0301 basic medicine ,Physiology ,Cell ,Pathology and Laboratory Medicine ,Monocytes ,Mice ,White Blood Cells ,0302 clinical medicine ,Interferon ,Animal Cells ,Medicine and Health Sciences ,Immune Response ,lcsh:QH301-705.5 ,biology ,Hematology ,Animal Models ,Flow Cytometry ,3. Good health ,Body Fluids ,medicine.anatomical_structure ,Blood ,Interferon Type I ,medicine.symptom ,Anatomy ,Cellular Types ,Plasmodium yoelii ,medicine.drug ,Research Article ,lcsh:Immunologic diseases. Allergy ,CD14 ,Immune Cells ,Immunology ,Inflammation ,Bone Marrow Cells ,Mouse Models ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Model Organisms ,Signs and Symptoms ,Diagnostic Medicine ,Virology ,Genetics ,medicine ,Parasitic Diseases ,Animals ,Humans ,Molecular Biology ,Blood Cells ,Macrophages ,Membrane Proteins ,Biology and Life Sciences ,TLR7 ,Dendritic Cells ,Cell Biology ,Macrophage Activation ,biology.organism_classification ,medicine.disease ,Tropical Diseases ,Malaria ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,lcsh:Biology (General) ,Parasitology ,Bone marrow ,lcsh:RC581-607 ,030215 immunology - Abstract
Malaria remains a global health burden causing significant morbidity, yet the mechanisms underlying disease outcomes and protection are poorly understood. Herein, we analyzed the peripheral blood of a unique cohort of Malawian children with severe malaria, and performed a comprehensive overview of blood leukocytes and inflammatory mediators present in these patients. We reveal robust immune cell activation, notably of CD14+ inflammatory monocytes, NK cells and plasmacytoid dendritic cells (pDCs) that is associated with very high inflammation. Using the Plasmodium yoelii 17X YM surrogate mouse model of lethal malaria, we report a comparable pattern of immune cell activation and inflammation and found that type I IFN represents a key checkpoint for disease outcomes. Compared to wild type mice, mice lacking the type I interferon (IFN) receptor exhibited a significant decrease in immune cell activation and inflammatory response, ultimately surviving the infection. We demonstrate that pDCs were the major producers of systemic type I IFN in the bone marrow and the blood of infected mice, via TLR7/MyD88-mediated recognition of Plasmodium parasites. This robust type I IFN production required priming of pDCs by CD169+ macrophages undergoing activation upon STING-mediated sensing of parasites in the bone marrow. pDCs and macrophages displayed prolonged interactions in this compartment in infected mice as visualized by intravital microscopy. Altogether our findings describe a novel mechanism of pDC activation in vivo and precise stepwise cell/cell interactions taking place during severe malaria that contribute to immune cell activation and inflammation, and subsequent disease outcomes., Author Summary The Plasmodium parasite is the number one killer among human parasitic diseases worldwide. Protection is associated with length of exposure for people living in endemic areas, with severe disease primarily affecting young children. Inflammation is a key component in the pathophysiology in malaria, and disease severity has been linked to the degree of activation of the immune system. However, the underlying mechanisms of protection and disease outcomes remain poorly understood. We provide a comprehensive analysis of peripheral blood immune cells obtained from a cohort of children with severe malaria. Our results show heightened inflammation and immune cell activation, in particular for monocytes, natural killer cells, and plasmacytoid dendritic cells (pDCs). We have also utilized a mouse model of lethal malaria that recapitulates many features identified in this cohort of severe malaria patients to examine drivers of immune cell activation and inflammation. Our studies provide evidence that type I interferon (IFN) acts as an early switch in inducing a potent inflammatory response in the infected host. Type I IFN production is massively produced in the bone marrow and the blood of infected mice by plasmacytoid dendritic cells (pDCs), a subset of DCs. We also demonstrate that resident macrophages in the bone marrow, control type I IFN production by the pDCs. We define how both myeloid cells “sense” the parasite to initiate the host immune response and report a previously uncharacterized physical interaction between pDCs and macrophages in the bone marrow as visualized by intravital microscopy in vivo. Our results define cellular processes underlying the marked inflammation of severe malaria and could open novel therapeutic opportunities to improve outcomes in this important human infectious disease.
- Published
- 2016
21. Sensory neuron fate is developmentally perturbed by Gars mutations causing human neuropathy
- Author
-
Steven J. West, James N. Sleigh, Robert W. Burgess, Giampietro Schiavo, M Z Cader, John M. Dawes, Kevin Talbot, Emily Spaulding, A Gomez-Martin, and David L.H. Bennett
- Subjects
0303 health sciences ,Mechanism (biology) ,Sensory system ,Disease ,Anatomy ,Biology ,Sensory neuron ,Peripheral ,Housekeeping gene ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Allele ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Charcot-Marie-Tooth disease type 2D (CMT2D) is a peripheral nerve disorder caused by dominant, toxic, gain-of-function mutations in the widely expressed, housekeeping gene,GARS. The mechanisms underlying selective nerve pathology in CMT2D remain unresolved, as does the cause of the mild-to-moderate sensory involvement that distinguishes CMT2D from the allelic disorder distal spinal muscular atrophy type V. To elucidate the mechanism responsible for the underlying afferent nerve pathology, we examined the sensory nervous system in CMT2D mice. We show that the equilibrium between functional subtypes of sensory neuron in dorsal root ganglia is distorted byGarsmutations, leading to sensory defects in peripheral tissues and correlating with overall disease severity. CMT2D mice display changes in sensory behaviour concordant with the afferent imbalance, which is present at birth and non-progressive, indicating that sensory neuron identity is prenatally perturbed and that a critical developmental insult is key to the afferent pathology. This suggests that both neurodevelopmental and neurodegenerative mechanisms contribute to CMT2D pathogenesis, and thus has profound implications for the timing of future therapeutic treatments.Significance StatementCharcot-Marie-Tooth disease (CMT) is a collection of genetically diverse inherited nerve disorders with the unifying feature of peripheral neuron degeneration. The mechanisms triggering this motor and sensory nerve dysfunction remain unresolved, as does the reason for the lack of sensory pathology observed in distal hereditary motor neuropathies, which can be associated with CMT genes. To unravel the mechanisms leading to afferent deterioration, we have studied the sensory nervous system of CMT Type 2D mice. Our work indicates that the specific cellular identity of sensory nerves is perturbed in mutant mice pre-natally. CMT therefore manifests through the complex interplay between malfunctioning developmental, maturation, and survival programs, which has important ramifications for therapeutic timing.
- Published
- 2016
- Full Text
- View/download PDF
22. Paclitaxel-induced epithelial damage and ectopic MMP-13 expression promotes neurotoxicity in zebrafish
- Author
-
Leah J. Middleton, Erin V. Carter, Thomas S. Lisse, Emily Spaulding, Olivia Lopes, Elizabeth A. Brochu, Paige B. Martin, Sandra Rieger, Ashley L. Waldron, and Adriana D. Pellegrini
- Subjects
Keratinocytes ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Embryo, Nonmammalian ,Paclitaxel ,Gene Expression ,Antineoplastic Agents ,Matrix Metalloproteinase Inhibitors ,Epithelium ,Epithelial Damage ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Matrix Metalloproteinase 13 ,Toxicity Tests ,medicine ,Animals ,Humans ,Peripheral Nerves ,Zebrafish ,Skin ,Multidisciplinary ,biology ,Epidermis (botany) ,Regeneration (biology) ,Neurotoxicity ,biology.organism_classification ,medicine.disease ,Axons ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,Touch Perception ,chemistry ,Animal Fins ,Cancer research - Abstract
Paclitaxel is a microtubule-stabilizing chemotherapeutic agent that is widely used in cancer treatment and in a number of curative and palliative regimens. Despite its beneficial effects on cancer, paclitaxel also damages healthy tissues, most prominently the peripheral sensory nervous system. The mechanisms leading to paclitaxel-induced peripheral neuropathy remain elusive, and therapies that prevent or alleviate this condition are not available. We established a zebrafish in vivo model to study the underlying mechanisms and to identify pharmacological agents that may be developed into therapeutics. Both adult and larval zebrafish displayed signs of paclitaxel neurotoxicity, including sensory axon degeneration and the loss of touch response in the distal caudal fin. Intriguingly, studies in zebrafish larvae showed that paclitaxel rapidly promotes epithelial damage and decreased mechanical stress resistance of the skin before induction of axon degeneration. Moreover, injured paclitaxel-treated zebrafish skin and scratch-wounded human keratinocytes (HEK001) display reduced healing capacity. Epithelial damage correlated with rapid accumulation of fluorescein-conjugated paclitaxel in epidermal basal keratinocytes, but not axons, and up-regulation of matrix-metalloproteinase 13 (MMP-13, collagenase 3) in the skin. Pharmacological inhibition of MMP-13, in contrast, largely rescued paclitaxel-induced epithelial damage and neurotoxicity, whereas MMP-13 overexpression in zebrafish embryos rendered the skin vulnerable to injury under mechanical stress conditions. Thus, our studies provide evidence that the epidermis plays a critical role in this condition, and we provide a previously unidentified candidate for therapeutic interventions.
- Published
- 2016
- Full Text
- View/download PDF
23. Synaptic Deficits at Neuromuscular Junctions in Two Mouse Models of Charcot-Marie-Tooth Type 2d
- Author
-
Kevin L. Seburn, Martin J. Pinter, Kathryn H. Morelli, Emily Spaulding, Robert W. Burgess, and James N. Sleigh
- Subjects
0301 basic medicine ,Glycine-tRNA Ligase ,Weakness ,Patch-Clamp Techniques ,Neuromuscular transmission ,Neuromuscular Junction ,Aminopyridines ,Stimulation ,Mice, Transgenic ,Nerve Tissue Proteins ,Neurotransmission ,Biology ,Synaptic vesicle ,Motor Endplate ,Synaptic Transmission ,Neuromuscular junction ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Imaging, Three-Dimensional ,Charcot-Marie-Tooth Disease ,medicine ,Animals ,Receptors, Cholinergic ,Muscle Strength ,Muscle, Skeletal ,Denervation ,General Neuroscience ,Age Factors ,Articles ,Synaptic Potentials ,Electric Stimulation ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Mutation ,Synaptic Vesicles ,medicine.symptom ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Patients with Charcot–Marie–Tooth Type 2D (CMT2D), caused by dominant mutations in Glycl tRNA synthetase (GARS), present with progressive weakness, consistently in the hands, but often in the feet also. Electromyography shows denervation, and patients often report that early symptoms include cramps brought on by cold or exertion. Based on reported clinical observations, and studies of mouse models of CMT2D, we sought to determine whether weakened synaptic transmission at the neuromuscular junction (NMJ) is an aspect of CMT2D. Quantal analysis of NMJs in two different mouse models of CMT2D (GarsP278KY,GarsC201R), found synaptic deficits that correlated with disease severity and progressed with age. Results of voltage-clamp studies revealed presynaptic defects characterized by: (1) decreased frequency of spontaneous release without any change in quantal amplitude (miniature endplate current), (2) reduced amplitude of evoked release (endplate current) and quantal content, (3) age-dependent changes in the extent of depression in response to repetitive stimulation, and (4) release failures at some NMJs with high-frequency, long-duration stimulation. Drugs that modify synaptic efficacy were tested to see whether neuromuscular performance improved. The presynaptic action of 3,4 diaminopyridine was not beneficial, whereas postsynaptic-acting physostigmine did improve performance. Smaller mutant NMJs with correspondingly fewer vesicles and partial denervation that eliminates some release sites also contribute to the reduction of release at a proportion of mutant NMJs. Together, these voltage-clamp data suggest that a number of release processes, while essentially intact, likely operate suboptimally at most NMJs of CMT2D mice.SIGNIFICANCE STATEMENTWe have uncovered a previously unrecognized aspect of axonal Charcot–Marie–Tooth disease in mouse models of CMT2D. Synaptic dysfunction contributes to impaired neuromuscular performance and disease progression. This suggests that drugs which improve synaptic efficacy at the NMJ could be considered in treating the pathophysiology of CMT2D patients.
- Published
- 2016
24. Differential responses of FLIPLong and FLIPShort-overexpressing human myeloid leukemia cells to TNF-α and TRAIL-initiated apoptotic signals
- Author
-
H. Joachim Deeg, Hans-Peter Kiem, Nissa Abbassi, David M. Hockenbery, Sudeshna Seal, and Emily Spaulding
- Subjects
Cancer Research ,CASP8 and FADD-Like Apoptosis Regulating Protein ,bcl-X Protein ,Apoptosis ,Caspase 3 ,Biology ,Inhibitor of apoptosis ,Article ,TNF-Related Apoptosis-Inducing Ligand ,Cell Line, Tumor ,Genetics ,Humans ,Molecular Biology ,Gene Expression Regulation, Leukemic ,Tumor Necrosis Factor-alpha ,Lentivirus ,NF-kappa B ,Cell Biology ,Hematology ,Cell biology ,XIAP ,Enzyme Activation ,Leukemia, Myeloid, Acute ,Cell culture ,Flip ,Tumor necrosis factor alpha ,Signal transduction ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
Objective Clonal marrow cells from patients with early myelodysplastic syndrome (MDS) undergo apoptosis in response to tumor necrosis factor (TNF)−related apoptosis-inducing ligand (TRAIL). Cells from advanced MDS are resistant to TRAIL. Two isoforms of the Flice inhibitory protein (FLIP) short (FLIP S ) and FLIP long (FLIP L ), which modulate TRAIL signals, showed disease-stage−dependent differential regulation. Therefore, we aimed at characterizing potential differential effects of FLIP L and FLIP S , on TRAIL and TNF-α−induced apoptosis in model leukemic cell lines. Materials and Methods Using lentiviral constructs, FLIP L and FLIP S , as well as a green fluorescent protein control were overexpressed in ML-1 cells, which constitutively express very low levels of FLIP and are highly sensitive to apoptosis induction. Cells were then exposed to TRAIL or TNF-α, and effects on the extrinsic and intrinsic pathways of apoptosis induction were assessed. Results Overexpression of FLIP reduced TRAIL and TNF-α−induced apoptosis in ML-1 cells. However, while FLIP L completely abrogated apoptosis, FLIP S allowed for BID cleavage and caspase-3 activation. Concurrently, there was a decline of Bcl-xL and X-linked inhibitor of apoptosis protein (XIAP) in FLIP S cells followed by apoptosis. Further, inhibition of nuclear factor-κB (NF-κB) activation in TNF-α−treated cells resulted in profound apoptosis in FLIP S , but not in FLIP L -overexpressing cells, consistent with the observations in patients with early stage MDS. Inhibition of NF-κB had only minimal effects on TRAIL signaling. Conclusion Thus, FLIP L and FLIP S exerted differential effects in myeloid leukemic cell lines in response to TRAIL and TNF-α. It might be possible to therapeutically exploit those differences with effector molecules specific for the FLIP isoforms.
- Published
- 2008
- Full Text
- View/download PDF
25. Trk receptor signalling and sensory neuron fate are perturbed in human neuropathy caused by Gars mutations
- Author
-
Qinghai Zhang, John M. Dawes, James N. Sleigh, Giampietro Schiavo, Yang X-L., A Gomez-Martin, Kevin Talbot, David L.H. Bennett, Na Wei, Emily Spaulding, M Z Cader, Steven J. West, and Robert W. Burgess
- Subjects
Glycine-tRNA Ligase ,Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Sensory Receptor Cells ,Sensory system ,Biology ,medicine.disease_cause ,Pathogenesis ,Glycine—tRNA ligase ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Charcot-Marie-Tooth Disease ,Internal medicine ,medicine ,Animals ,Humans ,Receptor, trkA ,Cells, Cultured ,Genetics (clinical) ,Mice, Knockout ,Mutation ,Multidisciplinary ,medicine.disease ,Sensory neuron ,nervous system diseases ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Signalling ,PNAS Plus ,Neurology ,Trk receptor ,Pediatrics, Perinatology and Child Health ,Female ,Neurology (clinical) ,Hereditary motor and sensory neuropathy ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Charcot-Marie-Tooth disease type 2D (CMT2D) is a peripheral nerve disorder caused by dominant, toxic, gain-of-function mutations in the widely expressed, housekeeping gene, GARS The mechanisms underlying selective nerve pathology in CMT2D remain unresolved, as does the cause of the mild-to-moderate sensory involvement that distinguishes CMT2D from the allelic disorder distal spinal muscular atrophy type V. To elucidate the mechanism responsible for the underlying afferent nerve pathology, we examined the sensory nervous system of CMT2D mice. We show that the equilibrium between functional subtypes of sensory neuron in dorsal root ganglia is distorted by Gars mutations, leading to sensory defects in peripheral tissues and correlating with overall disease severity. CMT2D mice display changes in sensory behavior concordant with the afferent imbalance, which is present at birth and nonprogressive, indicating that sensory neuron identity is prenatally perturbed and that a critical developmental insult is key to the afferent pathology. Through in vitro experiments, mutant, but not wild-type, GlyRS was shown to aberrantly interact with the Trk receptors and cause misactivation of Trk signaling, which is essential for sensory neuron differentiation and development. Together, this work suggests that both neurodevelopmental and neurodegenerative mechanisms contribute to CMT2D pathogenesis, and thus has profound implications for the timing of future therapeutic treatments.
- Published
- 2017
- Full Text
- View/download PDF
26. The T-Cell Inhibitory Molecule Butyrophilin-Like 2 Is Up-regulated in Mild Plasmodium falciparum Infection and Is Protective During Experimental Cerebral Malaria
- Author
-
Kathryn Anastos, Grégoire Lauvau, Catherine M. Feintuch, Ryung S. Kim, Johanna P. Daily, Emily Spaulding, Chen Dong, Emil Ivan, Xikui Liu, Krishanthi Subramaniam, Xingxing Zhang, and Eugene Mutimura
- Subjects
Adult ,Male ,Endemic Diseases ,Plasmodium berghei ,Plasmodium falciparum ,Malaria, Cerebral ,Biology ,Lymphocyte Activation ,Nitric Oxide ,B7-H1 Antigen ,Interferon-gamma ,Mice ,Young Adult ,Major Articles and Brief Reports ,Immune system ,Butyrophilin ,parasitic diseases ,medicine ,Immunology and Allergy ,Animals ,Humans ,Interferon gamma ,Malaria, Falciparum ,Complement Activation ,Mice, Knockout ,Membrane Glycoproteins ,Butyrophilins ,Rwanda ,medicine.disease ,biology.organism_classification ,Malaria ,Up-Regulation ,Mice, Inbred C57BL ,Infectious Diseases ,Cerebral Malaria ,Immunology ,Interferon Type I ,Female ,Interferon type I ,medicine.drug - Abstract
Plasmodium falciparum infection can result in severe disease that is associated with elevated inflammation and vital organ dysfunction; however, malaria-endemic residents gain protection from lethal outcomes and manifest only mild symptoms during infection. To characterize host responses associated with this more effective antimalarial response, we characterized whole-blood transcriptional profiles in Rwandan adults during a mild malaria episode and compared them with findings from a convalescence sample. We observed transcriptional up-regulation in many pathways, including type I interferon, interferon γ, complement activation, and nitric oxide during malaria infection, which provide benchmarks of mild disease physiology. Transcripts encoding negative regulators of T-cell activation, such as programmed death ligand 1 (PD-L1), programmed death 1 ligand 2 (PD-L2), and the butyrophilin family member butyrophilin-like 2 (BTNL2) were also increased. To support an important functional role for BTNL2 during malaria infection, we studied chimeric mice reconstituted with BTNL2(-/-) or wild-type hematopoietic cells that were inoculated with Plasmodium berghei ANKA, a murine model of cerebral malaria. We found that BTNL2(-/-) chimeric mice had a significant decrease in survival compared with wild-type counterparts. Collectively these data characterize the immune responses associated with mild malaria and uncover a novel role for BTNL2 in the host response to malaria.
- Published
- 2014
27. Macrophages and Plasmacytoid Dendritic Cells Initiate Early Immune Response To Severe Malaria In The Bone Marrow
- Author
-
Gregoire Lauvau, Emily Spaulding, David R Fooksman, Boris Reizis, and Johanna P Daily
- Subjects
Immunology ,Immunology and Allergy - Abstract
Malaria still remains a significant global health problem worldwide with approximately 200 million people infected, and half a million deaths every year. Most of these fatalities (80%) occur in children under the age of five, and the latest RTS, S, preventative vaccine trial only exhibited low protective efficacy (below 30%). The blood stage of Plasmodium infection accounts for the clinical symptoms of malaria, yet the immune mechanisms underlying disease outcomes are poorly understood. Here we have analyzed a cohort of severely infected Malawian children and revealed dramatic levels of inflammatory cytokines and extremely robust blood leukocyte activation, notably for CD14+CD16lo CCR2+ inflammatory monocytes, plasmacytoid DCs (pDCs), NK and T cells. We could recapitulate these observations using the Plasmodium yoelii (Py) 17X YM surrogate mouse model of lethal blood stage malaria. Using this experimental system, we demonstrate that type I interferon (IFN) signals as a key cytokine controlling lethal outcomes and immune cell activation. Plasmacytoid dendritic cells represented the main cellular source of type I IFN in the bone marrow and the blood of infected mice. Most interestingly, the activation of pDCs required CD169+ macrophages, and both cell types exhibited prolonged interactions in the bone marrow of infected mice by intravital microscopy. We will present additional data uncovering the molecular sensing pathways controlling these processes. Altogether our study establishes essential molecular pathways and cellular interactions that occur during this significant human parasitic infection, and suggest novel potential therapeutic targets involved in severe malaria.
- Published
- 2016
- Full Text
- View/download PDF
28. Iron overload, hematopoietic cell transplantation, and graft-versus-host disease
- Author
-
Emily Spaulding, Howard M. Shulman, and H. Joachim Deeg
- Subjects
Cancer Research ,Iron Overload ,Transplantation Conditioning ,Anemia ,medicine.medical_treatment ,Graft vs Host Disease ,Apoptosis ,Mice, Inbred Strains ,Hematopoietic stem cell transplantation ,Mice, SCID ,Infections ,Intestinal absorption ,Article ,Diagnosis, Differential ,Mice ,Postoperative Complications ,Phlebotomy ,Hepcidin ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,biology ,Myelodysplastic syndromes ,Hematopoietic Stem Cell Transplantation ,Transferrin ,Hematology ,medicine.disease ,Chelation Therapy ,Transplantation ,Graft-versus-host disease ,surgical procedures, operative ,Oncology ,chemistry ,Intestinal Absorption ,Liver ,Myelodysplastic Syndromes ,Immunology ,biology.protein ,Erythrocyte Transfusion ,Iron, Dietary - Abstract
Many patients who undergo hematopoietic cell transplantation (HCT) present with anemia and have received red blood cell transfusions before HCT. As a result, iron overload is frequent and appears to be particularly prominent in patients with myelodysplastic syndromes (MDS). There is evidence that peritransplant events contribute to further iron accumulation, although the mechanism that disrupts normal iron homeostasis remains to be determined. Recent studies suggest that iron overload, as determined by ferritin levels, a surrogate marker for iron, is a risk factor for increased non-relapse mortality after HCT. Iron overload is associated with an increased rate of infections, in particular with fungal organisms. Furthermore. anecdotal data suggest that increased hepatic iron may mimic the clinical picture of (chronic) graft-versus-host-disease (GVHD). Whether excess iron contributes to GVHD and whether iron depletion, be it by phlebotomy or chelation, reduces the post-transplantation complication rate and improves transplant outcome has yet to be determined.
- Published
- 2009
29. Transplantation of allogeneic T cells alters iron homeostasis in NOD/SCID mice
- Author
-
Kris V. Kowdley, H. Joachim Deeg, Jaakko Parkkinen, Steven M. Bair, François Canonne-Hergaux, Vladimir Lesnikov, Howard M. Shulman, Mary Beauchamp, Emily Spaulding, Center for High Pressure Rheology, Georgia Institute of Technology [Atlanta], Departments of Pediatrics, Ophthalmology, and Pharmacology, Research Center of Hôpital Sainte-Justine, Institut de Chimie des Substances Naturelles (ICSN), Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC), Institut d'Astrophysique de Paris (IAP), and Centre National de la Recherche Scientifique (CNRS)-Institut national des sciences de l'Univers (INSU - CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)
- Subjects
Adoptive cell transfer ,Iron Overload ,Iron ,T-Lymphocytes ,Immunology ,Nod ,Mice, SCID ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Hepcidin ,Mice, Inbred NOD ,medicine ,Animals ,Homeostasis ,Transplantation, Homologous ,chemistry.chemical_classification ,Transplantation ,Mice, Inbred BALB C ,medicine.diagnostic_test ,Transferrin ,Cell Biology ,Hematology ,T lymphocyte ,Adoptive Transfer ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Hepatocyte ,Serum iron ,biology.protein ,Apoproteins ,030215 immunology - Abstract
Iron overload is common in patients undergoing allogeneic hematopoietic cell transplantation (HCT), but the mechanisms leading to overload are unknown. Here, we determined iron levels and the expression of iron regulatory proteins in the liver and gut of nonobese diabetic–severe combined immunodeficient (NOD/SCID) mice that underwent transplantation with syngeneic (histocompatible) or allogeneic (histoincompatible) T lymphocytes. Infusion of histoincompatible T cells resulted in a significant rise in serum iron levels and liver iron content. Iron deposition was accompanied by hepatocyte injury and intestinal villous damage. Feeding of low- or high-iron diet was associated with appropriate ferroportin 1 and hepcidin responses in mice given histocompatible T cells, whereas mice given histoincompatible T cells showed inappropriate up-regulation of duodenal ferroportin 1 and a loss of expression of hepatic hepcidin. These findings suggest that alloreactive T cell–dependent signals induced dysregulation of intestinal iron absorption, which contributed to liver iron overload after HCT.
- Published
- 2009
- Full Text
- View/download PDF
30. Transferrin fails to provide protection against Fas-induced hepatic injury in mice with deletion of functional transferrin-receptor type 2
- Author
-
Howard M. Shulman, Robert E. Fleming, Vladimir Lesnikov, Emily Spaulding, Nelson Fausto, Jean S. Campbell, Nicholas T. Gorden, and H. Joachim Deeg
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Fas Ligand Protein ,Iron ,Clinical Biochemistry ,bcl-X Protein ,Pharmaceutical Science ,Male mice ,Transferrin receptor ,Apoptosis ,Article ,Mice ,Internal medicine ,Receptors, Transferrin ,medicine ,Animals ,Aspartate Aminotransferases ,Pharmacology ,chemistry.chemical_classification ,Mice, Knockout ,Mice, Inbred BALB C ,Sex Characteristics ,biology ,medicine.diagnostic_test ,Liver Diseases ,Biochemistry (medical) ,Apo transferrin ,Transferrin ,Cell Biology ,Molecular biology ,Iron Metabolism Disorders ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,Cytoprotection ,biology.protein ,Serum iron ,Hepatocytes ,Female ,Plasma iron ,Antibody ,Chemical and Drug Induced Liver Injury ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
We reported previously that Fas-induced hepatic failure in normal mice was attenuated or prevented by exogenous transferrin (Tf), particularly apoTf. Here we show in C57BL6J/129 mice with genetic inactivation of transferrin receptor 2 (TfR2(Y245X)), that Fas-induced hepatotoxicity (apoptosis; rise in plasma aspartate aminotransferase (AST) levels) was comparable to that in wild-type mice, but was not modified by pretreatment with Tf. Rises in plasma AST were preceded by a decline in serum iron levels. AST elevations and iron declines were more profound in female than in male mice. Female mice also showed higher baseline levels of Bcl-xL in hepatocytes, which declined significantly upon treatment with agonistic anti-Fas antibody. These data confirm the cytoprotective function of Tf, and show a novel property of TfR2. Both apoptotic Fas responses and cytoprotective effects of Tf were associated with significant shifts in plasma iron levels, which quantitatively differed between male and female mice.
- Published
- 2008
31. Deleterious role for type I interferon during acute blood stage malaria infection (MPF1P.767)
- Author
-
Emily Spaulding and Gregoire Lauvau
- Subjects
Immunology ,Immunology and Allergy - Abstract
Malaria is a severe and life-threatening disease that affects over 219 million people worldwide every year. In patients, a robust induction of the type I IFN pathway is observed, yet studies in mouse models have failed to determine whether type I IFN contributes to host protection or pathogenesis. We thus hypothesized that type I IFN is essential during blood stage malaria infection. Using Plasmodium yoelii (Py) YM which causes an acute lethal blood infection in C57BL/6 WT mice, we found that while WT mice rapidly succumbed, mice lacking the type I IFN receptor (IFNαβR-/-) mostly survived (>80%). Innate immune cells, particularly monocytes and NK cells, underwent early and robust activation and overwhelming production of inflammatory mediators in WT mice, whereas this was prevented in IFNαβR-/- mice. We are currently investigating precisely the cellular source of type I IFN and the mechanisms that regulate the activation of type I IFN-producing cells during this infection. Our results favor a model in which type I IFN promotes over-exuberant activation of innate monocytes and NK cells, leading to overwhelming inflammation and death of Py YM-infected mice. Overall, our work highlights a novel and unsuspected role for type I IFN in this significant human parasitic disease. We believe these results contribute to our understanding of the immunological cues leading to severe disease in malaria patients.
- Published
- 2014
- Full Text
- View/download PDF
32. MDS Marrow Stroma Is Characterized by Distinct Epigenetic Alterations
- Author
-
John M. Greally, Amit Verma, Reid F. Thompson, Emily Spaulding, A. Mario Q. Marcondes, Davendra Sohal, Tushar D. Bhagat, Yongkai Mo, H. Joachim Deeg, and Li Zhou
- Subjects
Stromal cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,HELP assay ,Stroma ,DNA methylation ,medicine ,Bone marrow ,Epigenetics ,Epigenomics - Abstract
The bone marrow microenvironment plays an important role in the pathogenesis and perpetuation of stem cell defects in Myelodysplastic Syndrome (MDS). However, while distinct cytogenetic alterations have been described in the stem cell compartment in MDS, the bone marrow stroma has never been shown to be part of the clone. Thus, aberrant epigenetic alterations may be responsible for altered function of bone marrow stroma in MDS. DNA methyl transferase (DNMT) inhibitors, which are therapeutically effective in MDS, affect both hematopoietic cells and the stroma, providing further rationale for studying DNA methylation profiles of bone marrow stroma in this disease. To accomplish this aim, bone marrow mononuclear cells from MDS patients and controls were grown to form adherent cell layers and then depleted for hematopoietic elements by immunomagnetic CD45 negative selection. CD45 negative adherent cells were subsequently expanded and then used for whole genome methylation studies using a recently described novel method, the HELP assay (HpaII tiny fragment Enrichment by Ligation-mediated PCR; Khulan et al, Genome Res. 2006 Aug;16(8)) which uses differential methylation-specific digestion by HpaII and MspI followed by amplification, two color labeling and hybridization to quantify individual promoter CpG island methylation. A custom whole genome human promoter array (Roche-Nimblegen) was used to determine the level of methylation of 25626 gene promoters by calculating HpaII/MspI cut fragment intensity ratio. Global epigenetic profiling revealed that MDS stroma (n=6) was epigenetically distinct from normal bone marrow stroma (n=4) (ANOVA, P In subsequent studies, we profiled stroma from another set of MDS patients who had been treated with the DNMT inhibitor, 5-Azacytidine (n=4). In contrast to untreated MDS patients, there were no significant epigenetic differences between these 5-Azacytidine treated MDS patients and healthy controls (p = NS). These 5-Azacytidine exposed stroma cells did not demonstrate global hypomethylation (as hypothesized after DNMT inhibitor treatment) and were characterized by both hyper- and hypo-methylated loci similar to healthy controls. Thus our results reveal that MDS is characterized by widespread aberrant epigenetic changes in the bone marrow microenvironment. Our results also demonstrate that DNMT inhibitors can alter the epigenomic profiles of stromal cells, and we hypothesize that those stroma effects contribute in part to their clinical efficacy. Overall, these studies underscore the importance of studying the entire bone marrow, including the microenvironment, if we are to improve our understanding of the pathophysiology of MDS and further improve therapy.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.