Bio


Dr. McIlwain studies host-response to infectious disease using high dimensional single-cell and spatial proteomics tools. He trained for his Ph.D. at the University of Toronto exploring mouse biology using reverse genetics with renowned immunologist Dr. Tak W. Mak. His doctoral work yielded insights into alternative mRNA splicing and an important discovery about iRhom2 as a new factor controlling the production of inflammatory mediator TNF. As a post-doctoral fellow, Dr. McIlwain investigated host response to viral infection in animal models at the University of Dusseldorf in Germany before moving to Stanford University where along with Dr. Garry Nolan, he leads a team executing research contracted by the FDA’s medical countermeasures initiative to study emerging pathogens. This work includes mass cytometry (CyTOF) and spatial proteomic (CODEX) single-cell analysis of human and animal model influenza, Ebola, zika, and SARS-CoVs infections.

Honors & Awards


  • Canadian Institutes of Health Research Fellow, Canadian Institutes of Health Research (2014-2017)

All Publications


  • Human influenza virus challenge identifies cellular correlates of protection for oral vaccination. Cell host & microbe McIlwain, D. R., Chen, H., Rahil, Z., Bidoki, N. H., Jiang, S., Bjornson, Z., Kolhatkar, N. S., Martinez, C. J., Gaudillière, B., Hedou, J., Mukherjee, N., Schürch, C. M., Trejo, A., Affrime, M., Bock, B., Kim, K., Liebowitz, D., Aghaeepour, N., Tucker, S. N., Nolan, G. P. 2021

    Abstract

    Developing new influenza vaccines with improved performance and easier administration routes hinges on defining correlates of protection. Vaccine-elicited cellular correlates of protection for influenza in humans have not yet been demonstrated. A phase-2 double-blind randomized placebo and active (inactivated influenza vaccine) controlled study provides evidence that a human-adenovirus-5-based oral influenza vaccine tablet (VXA-A1.1) can protect from H1N1 virus challenge in humans. Mass cytometry characterization of vaccine-elicited cellular immune responses identified shared and vaccine-type-specific responses across B and T cells. For VXA-A1.1, the abundance of hemagglutinin-specific plasmablasts and plasmablasts positive for integrin α4β7, phosphorylated STAT5, or lacking expression of CD62L at day 8 were significantly correlated with protection from developing viral shedding following virus challenge at day 90 and contributed to an effective machine learning model of protection. These findings reveal the characteristics of vaccine-elicited cellular correlates of protection for an oral influenza vaccine.

    View details for DOI 10.1016/j.chom.2021.10.009

    View details for PubMedID 34784508

  • Single-Cell Profiling of Ebola Virus Disease InVivo Reveals Viral and Host Dynamics. Cell Kotliar, D., Lin, A. E., Logue, J., Hughes, T. K., Khoury, N. M., Raju, S. S., Wadsworth, M. H., Chen, H., Kurtz, J. R., Dighero-Kemp, B., Bjornson, Z. B., Mukherjee, N., Sellers, B. A., Tran, N., Bauer, M. R., Adams, G. C., Adams, R., Rinn, J. L., Mele, M., Schaffner, S. F., Nolan, G. P., Barnes, K. G., Hensley, L. E., McIlwain, D. R., Shalek, A. K., Sabeti, P. C., Bennett, R. S. 2020

    Abstract

    Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.

    View details for DOI 10.1016/j.cell.2020.10.002

    View details for PubMedID 33159858

  • Landscape of coordinated immune responses to H1N1 challenge in humans. The Journal of clinical investigation Rahil, Z. n., Leylek, R. n., Schürch, C. M., Chen, H. n., Bjornson-Hooper, Z. n., Christensen, S. R., Gherardini, P. F., Bhate, S. S., Spitzer, M. H., Fragiadakis, G. K., Mukherjee, N. n., Kim, N. n., Jiang, S. n., Yo, J. n., Gaudilliere, B. n., Affrime, M. n., Bock, B. n., Hensley, S. E., Idoyaga, J. n., Aghaeepour, N. n., Kim, K. n., Nolan, G. P., McIlwain, D. R. 2020

    Abstract

    Influenza is a significant cause of morbidity and mortality worldwide. Here we show changes in the abundance and activation states of more than 50 immune cell subsets in 35 individuals over 11 time points during human A/California/2009 (H1N1) virus challenge monitored using mass cytometry along with other clinical assessments. Peak change in monocyte, B cell, and T cell subset frequencies coincided with peak virus shedding, followed by marked activation of T and NK cells. Results led to the identification of CD38 as a critical regulator of plasmacytoid dendritic cell function in response to influenza virus. Machine learning using study-derived clinical parameters and single-cell data effectively classified and predicted susceptibility to infection. The coordinated immune cell dynamics defined in this study provide a framework for identifying novel correlates of protection in the evaluation of future influenza therapeutics.

    View details for DOI 10.1172/JCI137265

    View details for PubMedID 33044226

  • iRhom2 Regulation of TACE Controls TNF-Mediated Protection Against Listeria and Responses to LPS SCIENCE McIlwain, D. R., Lang, P. A., Maretzky, T., Hamada, K., Ohishi, K., Maney, S. K., Berger, T., Murthy, A., Duncan, G., Xu, H. C., Lang, K. S., Haeussinger, D., Wakeham, A., Itie-Youten, A., Khokha, R., Ohashi, P. S., Blobel, C. P., Mak, T. W. 2012; 335 (6065): 229-232

    Abstract

    Innate immune responses are vital for pathogen defense but can result in septic shock when excessive. A key mediator of septic shock is tumor necrosis factor-α (TNFα), which is shed from the plasma membrane after cleavage by the TNFα convertase (TACE). We report that the rhomboid family member iRhom2 interacted with TACE and regulated TNFα shedding. iRhom2 was critical for TACE maturation and trafficking to the cell surface in hematopoietic cells. Gene-targeted iRhom2-deficient mice showed reduced serum TNFα in response to lipopolysaccharide (LPS) and could survive a lethal LPS dose. Furthermore, iRhom2-deficient mice failed to control the replication of Listeria monocytogenes. Our study has identified iRhom2 as a regulator of innate immunity that may be an important target for modulating sepsis and pathogen defense.

    View details for DOI 10.1126/science.1214448

    View details for Web of Science ID 000299033100056

    View details for PubMedID 22246778

  • Determinants of SARS-CoV-2 entry and replication in airway mucosal tissue and susceptibility in smokers. Cell reports. Medicine Nakayama, T., Lee, I. T., Jiang, S., Matter, M. S., Yan, C. H., Overdevest, J. B., Wu, C., Goltsev, Y., Shih, L., Liao, C., Zhu, B., Bai, Y., Lidsky, P., Xiao, Y., Zarabanda, D., Yang, A., Easwaran, M., Schurch, C. M., Chu, P., Chen, H., Stalder, A. K., McIlwain, D. R., Borchard, N. A., Gall, P. A., Dholakia, S. S., Le, W., Xu, L., Tai, C., Yeh, T., Erickson-Direnzo, E., Duran, J. M., Mertz, K. D., Hwang, P. H., Haslbauer, J. D., Jackson, P. K., Menter, T., Andino, R., Canoll, P. D., DeConde, A. S., Patel, Z. M., Tzankov, A., Nolan, G. P., Nayak, J. V. 2021: 100421

    Abstract

    Understanding viral tropism is an essential step towards reducing SARS-CoV-2 transmission, decreasing mortality from COVID-19, and limiting opportunities for mutant strains to arise. Currently, little is known about the extent to which distinct tissue sites in the human head & neck region and proximal respiratory tract selectively permit SARS-CoV-2 infection and replication. In this translational study, we discover key variabilities in the expression of ACE2 and TMPRSS2, essential SARS-CoV-2 entry factors, among the mucosal tissues of the human proximal airways. We show that SARS-CoV-2 infection is present in all examined head & neck tissues, with a notable tropism for the nasal cavity and tracheal mucosa. Finally, we uncover an association between smoking and higher SARS-CoV-2 viral infection in the human proximal airway, which may explain the increased susceptibility of smokers to developing severe COVID-19. This is at least partially explained by differences in IFN-beta1 levels between smokers and non-smokers.

    View details for DOI 10.1016/j.xcrm.2021.100421

    View details for PubMedID 34604819

  • SARS-CoV-2 entry factors are expressed in nasal, ocular, and oral tissues: implications for COVID-19 prophylaxes/therapeutics Lee, I., Nakayama, T., Jiang, S., Goltsev, Y., Schurch, C., Zhu, B., McIlwain, D., Chu, P., Chen, H., Tzankov, A., Matter, M., Nayak, J., Nolan, G. MOSBY-ELSEVIER. 2021: AB2
  • Zoonotic risk factors associated with seroprevalence of Ebola virus GP antibodies in the absence of diagnosed Ebola virus disease in the Democratic Republic of Congo. PLoS neglected tropical diseases Bratcher, A., Hoff, N. A., Doshi, R. H., Gadoth, A., Halbrook, M., Mukadi, P., Musene, K., Ilunga-Kebela, B., Spencer, D., Bramble, M. S., McIlwan, D., Kelly, J. D., Mukadi, D., Kingebeni, P. M., Ahuka, S., Okitolonda-Wemakoy, E., Muyembe-Tamfum, J. J., Rimoin, A. W. 2021; 15 (8): e0009566

    Abstract

    Ebola virus (EBOV) is a zoonotic filovirus spread through exposure to infected bodily fluids of a human or animal. Though EBOV is capable of causing severe disease, referred to as Ebola Virus Disease (EVD), individuals who have never been diagnosed with confirmed, probable or suspected EVD can have detectable EBOV antigen-specific antibodies in their blood. This study aims to identify risk factors associated with detectable antibody levels in the absence of an EVD diagnosis.Data was collected from September 2015 to August 2017 from 1,366 consenting individuals across four study sites in the DRC (Boende, Kabondo-Dianda, Kikwit, and Yambuku). Seroreactivity was determined to EBOV GP IgG using Zaire Ebola Virus Glycoprotein (EBOV GP antigen) ELISA kits (Alpha Diagnostic International, Inc.) in Kinshasa, DRC; any result above 4.7 units/mL was considered seroreactive. Among the respondents, 113 (8.3%) were considered seroreactive. Several zoonotic exposures were associated with EBOV seroreactivity after controlling for age, sex, healthcare worker status, location, and history of contact with an EVD case, namely: ever having contact with bats, ever having contact with rodents, and ever eating non-human primate meat. Contact with monkeys or non-human primates was not associated with seroreactivity.This analysis suggests that some zoonotic exposures that have been linked to EVD outbreaks can also be associated with EBOV GP seroreactivity in the absence of diagnosed EVD. Future investigations should seek to clarify the relationships between zoonotic exposures, seroreactivity, asymptomatic infection, and EVD.

    View details for DOI 10.1371/journal.pntd.0009566

    View details for PubMedID 34383755

  • Adjacent Cell Marker Lateral Spillover Compensation and Reinforcement for Multiplexed Images. Frontiers in immunology Bai, Y., Zhu, B., Rovira-Clave, X., Chen, H., Markovic, M., Chan, C. N., Su, T., McIlwain, D. R., Estes, J. D., Keren, L., Nolan, G. P., Jiang, S. 2021; 12: 652631

    Abstract

    Multiplex imaging technologies are now routinely capable of measuring more than 40 antibody-labeled parameters in single cells. However, lateral spillage of signals in densely packed tissues presents an obstacle to the assignment of high-dimensional spatial features to individual cells for accurate cell-type annotation. We devised a method to correct for lateral spillage of cell surface markers between adjacent cells termed REinforcement Dynamic Spillover EliminAtion (REDSEA). The use of REDSEA decreased contaminating signals from neighboring cells. It improved the recovery of marker signals across both isotopic (i.e., Multiplexed Ion Beam Imaging) and immunofluorescent (i.e., Cyclic Immunofluorescence) multiplexed images resulting in a marked improvement in cell-type classification.

    View details for DOI 10.3389/fimmu.2021.652631

    View details for PubMedID 34295327

  • Performance of BioFire array or QuickVue influenza A+B test versus a validation qPCR assay for detection of influenza A during a volunteer A/California/2009/H1N1 challenge study. Virology journal McIlwain, D. R., Chen, H., Apkarian, M., Affrime, M., Bock, B., Kim, K., Mukherjee, N., Nolan, G. P., McNeal, M. M. 2021; 18 (1): 45

    Abstract

    BACKGROUND: Influenza places a significant burden on global health and economics. Individual case management and public health efforts to mitigate the spread of influenza are both strongly impacted by our ability to accurately and efficiently detect influenza viruses in clinical samples. Therefore, it is important to understand the performance characteristics of available assays to detect influenza in a variety of settings. We provide the first report of relative performance between two products marketed to streamline detection of influenza virus in the context of a highly controlled volunteer influenza challenge study.METHODS: Nasopharyngeal swab samples were collected during a controlled A/California/2009/H1N1 influenza challenge study and analyzed for detection of virus shedding using a validated qRT-PCR (qPCR) assay, a sample-to-answer qRT-PCR device (BioMerieux BioFire FilmArray RP), and an immunoassay based rapid test kit (Quidel QuickVue Influenza A+B Test).RESULTS: Relative to qPCR, the sensitivity and specificity of the BioFire assay was 72.1% [63.7-79.5%, 95% confidence interval (CI)] and 93.5% (89.3-96.4%, 95% CI) respectively. For the QuickVue rapid test the sensitivity was 8.5% (4.8-13.7%, 95% CI) and specificity was 99.2% (95.6-100%, 95% CI).CONCLUSION: Relative to qPCR, the BioFire assay had superior performance compared to rapid test in the context of a controlled influenza challenge study.

    View details for DOI 10.1186/s12985-021-01516-0

    View details for PubMedID 33632249

  • Role of iRhoms 1 and 2 in Endochondral Ossification. International journal of molecular sciences Fang, R., Haxaire, C., Otero, M., Lessard, S., Weskamp, G., McIlwain, D. R., Mak, T. W., Lichtenthaler, S. F., Blobel, C. P. 2020; 21 (22)

    Abstract

    Growth of the axial and appendicular skeleton depends on endochondral ossification, which is controlled by tightly regulated cell-cell interactions in the developing growth plates. Previous studies have uncovered an important role of a disintegrin and metalloprotease 17 (ADAM17) in the normal development of the mineralized zone of hypertrophic chondrocytes during endochondral ossification. ADAM17 regulates EGF-receptor signaling by cleaving EGFR-ligands such as TGFalpha from their membrane-anchored precursor. The activity of ADAM17 is controlled by two regulatory binding partners, the inactive Rhomboids 1 and 2 (iRhom1, 2), raising questions about their role in endochondral ossification. To address this question, we generated mice lacking iRhom2 (iR2-/-) with floxed alleles of iRhom1 that were specifically deleted in chondrocytes by Col2a1-Cre (iR1∆Ch). The resulting iR2-/-iR1∆Ch mice had retarded bone growth compared to iR2-/- mice, caused by a significantly expanded zone of hypertrophic mineralizing chondrocytes in the growth plate. Primary iR2-/-iR1∆Ch chondrocytes had strongly reduced shedding of TGFalpha and other ADAM17-dependent EGFR-ligands. The enlarged zone of mineralized hypertrophic chondrocytes in iR2-/-iR1∆Ch mice closely resembled the abnormal growth plate in A17∆Ch mice and was similar to growth plates in Tgfalpha-/- mice or mice with EGFR mutations. These data support a model in which iRhom1 and 2 regulate bone growth by controlling the ADAM17/TGFalpha/EGFR signaling axis during endochondral ossification.

    View details for DOI 10.3390/ijms21228732

    View details for PubMedID 33227998

  • ACE2 localizes to the respiratory cilia and is not increased by ACE inhibitors or ARBs. Nature communications Lee, I. T., Nakayama, T., Wu, C., Goltsev, Y., Jiang, S., Gall, P. A., Liao, C., Shih, L., Schurch, C. M., McIlwain, D. R., Chu, P., Borchard, N. A., Zarabanda, D., Dholakia, S. S., Yang, A., Kim, D., Chen, H., Kanie, T., Lin, C., Tsai, M., Phillips, K. M., Kim, R., Overdevest, J. B., Tyler, M. A., Yan, C. H., Lin, C., Lin, Y., Bau, D., Tsay, G. J., Patel, Z. M., Tsou, Y., Tzankov, A., Matter, M. S., Tai, C., Yeh, T., Hwang, P. H., Nolan, G. P., Nayak, J. V., Jackson, P. K. 2020; 11 (1): 5453

    Abstract

    The coronavirus SARS-CoV-2 is the causative agent of the ongoing severe acute respiratory disease pandemic COVID-19. Tissue and cellular tropism is one key to understanding the pathogenesis of SARS-CoV-2. We investigate the expression and subcellular localization of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), within the upper (nasal) and lower (pulmonary) respiratory tracts of human donors using a diverse panel of banked tissues. Here, we report our discovery that the ACE2 receptor protein robustly localizes within the motile cilia of airway epithelial cells, which likely represents the initial or early subcellular site of SARS-CoV-2 viral entry during host respiratory transmission. We further determine whether ciliary ACE2 expression in the upper airway is influenced by patient demographics, clinical characteristics, comorbidities, or medication use, and show the first mechanistic evidence that the use of angiotensin-converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARBs) does not increase susceptibility to SARS-CoV-2 infection through enhancing the expression of ciliary ACE2 receptor. These findings are crucial to our understanding of the transmission of SARS-CoV-2 for prevention and control of this virulent pathogen.

    View details for DOI 10.1038/s41467-020-19145-6

    View details for PubMedID 33116139

  • Integration of mechanistic immunological knowledge into a machine learning pipeline improves predictions NATURE MACHINE INTELLIGENCE Culos, A., Tsai, A. S., Stanley, N., Becker, M., Ghaemi, M. S., McIlwain, D. R., Fallahzadeh, R., Tanada, A., Nassar, H., Espinosa, C., Xenochristou, M., Ganio, E., Peterson, L., Han, X., Stelzer, I. A., Ando, K., Gaudilliere, D., Phongpreecha, T., Maric, I., Chang, A. L., Shaw, G. M., Stevenson, D. K., Bendall, S., Davis, K. L., Fantl, W., Nolan, G. P., Hastie, T., Tibshirani, R., Angst, M. S., Gaudilliere, B., Aghaeepour, N. 2020
  • Integration of mechanistic immunological knowledge into a machine learning pipeline improves predictions. Nature machine intelligence Culos, A., Tsai, A. S., Stanley, N., Becker, M., Ghaemi, M. S., McIlwain, D. R., Fallahzadeh, R., Tanada, A., Nassar, H., Espinosa, C., Xenochristou, M., Ganio, E., Peterson, L., Han, X., Stelzer, I. A., Ando, K., Gaudilliere, D., Phongpreecha, T., Marić, I., Chang, A. L., Shaw, G. M., Stevenson, D. K., Bendall, S., Davis, K. L., Fantl, W., Nolan, G. P., Hastie, T., Tibshirani, R., Angst, M. S., Gaudilliere, B., Aghaeepour, N. 2020; 2 (10): 619-628

    Abstract

    The dense network of interconnected cellular signalling responses that are quantifiable in peripheral immune cells provides a wealth of actionable immunological insights. Although high-throughput single-cell profiling techniques, including polychromatic flow and mass cytometry, have matured to a point that enables detailed immune profiling of patients in numerous clinical settings, the limited cohort size and high dimensionality of data increase the possibility of false-positive discoveries and model overfitting. We introduce a generalizable machine learning platform, the immunological Elastic-Net (iEN), which incorporates immunological knowledge directly into the predictive models. Importantly, the algorithm maintains the exploratory nature of the high-dimensional dataset, allowing for the inclusion of immune features with strong predictive capabilities even if not consistent with prior knowledge. In three independent studies our method demonstrates improved predictions for clinically relevant outcomes from mass cytometry data generated from whole blood, as well as a large simulated dataset. The iEN is available under an open-source licence.

    View details for DOI 10.1038/s42256-020-00232-8

    View details for PubMedID 33294774

    View details for PubMedCentralID PMC7720904

  • Immunologic timeline of Ebola virus disease and recovery in humans. JCI insight McElroy, A. K., Akondy, R. S., Mcllwain, D. R., Chen, H., Bjornson-Hooper, Z., Mukherjee, N., Mehta, A. K., Nolan, G., Nichol, S. T., Spiropoulou, C. F. 2020; 5 (10)

    Abstract

    A complete understanding of human immune responses to Ebola virus infection is limited by the availability of specimens and the requirement for biosafety level 4 (BSL-4) containment. In an effort to bridge this gap, we evaluated cryopreserved PBMCs from 4 patients who survived Ebola virus disease (EVD) using an established mass cytometry antibody panel to characterize various cell populations during both the acute and convalescent phases. Acute loss of nonclassical monocytes and myeloid DCs, especially CD1c+ DCs, was noted. Classical monocyte proliferation and CD38 upregulation on plasmacytoid DCs coincided with declining viral load. Unsupervised analysis of cell abundance demonstrated acute declines in monocytic, NK, and T cell populations, but some populations, many of myeloid origin, increased in abundance during the acute phase, suggesting emergency hematopoiesis. Despite cell losses during the acute phase, upregulation of Ki-67 correlated with recovery of cell populations over time. These data provide insights into the human immune response during EVD.

    View details for DOI 10.1172/jci.insight.137260

    View details for PubMedID 32434986

  • Functional comparison of PBMCs isolated by Cell Preparation Tubes (CPT) vs. Lymphoprep Tubes. BMC immunology Chen, H., Schurch, C. M., Noble, K., Kim, K., Krutzik, P. O., O'Donnell, E., Vander Tuig, J., Nolan, G. P., McIlwain, D. R. 2020; 21 (1): 15

    Abstract

    BACKGROUND: Cryopreserved human peripheral blood mononuclear cells (PBMCs) are a commonly used sample type for a variety of immunological assays. Many factors can affect the quality of PBMCs, and careful consideration and validation of an appropriate PBMC isolation and cryopreservation method is important for well-designed clinical studies. A major point of divergence in PBMC isolation protocols is the collection of blood, either directly into vacutainers pre-filled with density gradient medium or the use of conical tubes containing a porous barrier to separate the density gradient medium from blood. To address potential differences in sample outcome, we isolated, cryopreserved, and compared PBMCs using parallel protocols differing only in the use of one of two common tube types for isolation.METHODS: Whole blood was processed in parallel using both Cell Preparation Tubes (CPT, BD Biosciences) and Lymphoprep Tubes (Axis-Shield) and assessed for yield and viability prior to cryopreservation. After thawing, samples were further examined by flow cytometry for cell yield, cell viability, frequency of 10 cell subsets, and capacity for stimulation-dependent CD4+ and CD8+ T cell intracellular cytokine production.RESULTS: No significant differences in cell recovery, viability, frequency of immune cell subsets, or T cell functionality between PBMC samples isolated using CPT or Lymphoprep tubes were identified.CONCLUSION: CPT and Lymphoprep tubes are effective and comparable methods for PBMC isolation for immunological studies.

    View details for DOI 10.1186/s12865-020-00345-0

    View details for PubMedID 32228458

  • Substrate-selective protein ectodomain shedding by ADAM17 and iRhom2 depends on their juxtamembrane and transmembrane domains. FASEB journal : official publication of the Federation of American Societies for Experimental Biology Tang, B., Li, X., Maretzky, T., Perez-Aguilar, J. M., McIlwain, D., Xie, Y., Zheng, Y., Mak, T. W., Weinstein, H., Blobel, C. P. 2020

    Abstract

    The metalloprotease ADAM17 (a disintegrin and metalloprotease 17) regulates EGF-receptor and TNFalpha signaling, thereby not only protecting the skin and intestinal barrier, but also contributing to autoimmunity. ADAM17 can be rapidly activated by many stimuli through its transmembrane domain (TMD), with the seven membrane-spanning inactive Rhomboids (iRhom) 1 and 2 implicated as candidate regulatory partners. However, several alternative models of ADAM17 regulation exist that do not involve the iRhoms, such as regulation through disulfide bond exchange or through interaction with charged phospholipids. Here, we report that a non-activatable mutant of ADAM17 with the TMD of betacellulin (BTC) can be rescued by restoring residues from the ADAM17 TMD, but only in Adam17-/- cells, which contain iRhoms, not in iRhom1/2-/- cells. We also provide the first evidence that the extracellular juxtamembrane domains (JMDs) of ADAM17 and iRhom2 regulate the stimulation and substrate selectivity of ADAM17. Interestingly, a point mutation in the ADAM17 JMD identified in a patient with Tetralogy of Fallot, a serious heart valve defect, affects the substrate selectivity of ADAM17 toward Heparin-binding epidermal growth factor like growth factor (HB-EGF), a crucial regulator of heart valve development in mice. These findings provide new insights into the regulation of ADAM17 through an essential interaction with the TMD1 and JMD1 of iRhom2.

    View details for DOI 10.1096/fj.201902649R

    View details for PubMedID 32103528

  • Efficacy, immunogenicity, and safety of an oral influenza vaccine: a placebo-controlled and active-controlled phase 2 human challenge study. The Lancet. Infectious diseases Liebowitz, D., Gottlieb, K., Kolhatkar, N. S., Garg, S. J., Asher, J. M., Nazareno, J., Kim, K., McIlwain, D. R., Tucker, S. N. 2020

    Abstract

    BACKGROUND: Influenza is an important public health problem and existing vaccines are not completely protective. New vaccines that protect by alternative mechanisms are needed to improve efficacy of influenza vaccines. In 2015, we did a phase 1 trial of an oral influenza vaccine, VXA-A1.1. A favourable safety profile and robust immunogenicity results in that trial supported progression of the vaccine to the current phase 2 trial. The aim of this study was to evaluate efficacy of the vaccine in a human influenza challenge model.METHODS: We did a single-site, placebo-controlled and active-controlled, phase 2 study at WCCT Global, Costa Mesa, CA, USA. Eligible individuals had an initial A/California/H1N1 haemagglutination inhibition titre of less than 20 and were aged 18-49 years and in good health. Individuals were randomly assigned (2:2:1) to receive a single immunisation of either 1011 infectious units of VXA-A1.1 (a monovalent tablet vaccine) orally, a full human dose of quadrivalent inactivated influenza vaccine (IIV) via intramuscular injection, or matched placebo. Randomisation was done by computer-generated assignments with block size of five. An unmasked pharmacist provided the appropriate vaccines and placebos to the administrating nurse. Individuals receiving the treatments, investigators, and staff were all masked to group assignments. 90 days after immunisation, individuals without clinically significant symptoms or signs of influenza, an oral temperature of higher than 37·9°C, a positive result for respiratory viral shedding on a Biofire test, and any investigator-assessed contraindications were challenged intranasally with 0·5 mL wild-type A/CA/like(H1N1)pdm09 influenza virus. The primary outcomes were safety, which was assessed in all immunised participants through 365 days, and influenza-positive illness after viral challenge, which was assessed in individuals that received the viral challenge and the required number of assessments post viral challenge. This trial is registered with ClinicalTrials.gov, number NCT02918006.RESULTS: Between Aug 31, 2016, and Jan 23, 2017, 374 individuals were assessed for eligibility, of whom 179 were randomly assigned to receive either VXA-A1.1 (n=71 [one individual did not provide a diary card, thus the solicited events were assessed in 70 individuals]), IIV (n=72), or placebo (n=36). Between Dec 2, 2016, and April 26, 2017, 143 eligible individuals (58 in the VXA-A1.1 group, 54 in the IIV group, and 31 in the placebo group) were challenged with influenza virus. VXA-A1.1 was well tolerated with no serious or medically significant adverse events. The most prevalent solicited adverse events for each of the treatment groups after immunisation were headache in the VXA-A1.1 (in five [7%] of 70 participants) and placebo (in seven [19%] of 36 participants) groups and tenderness at injection site in the IIV group (in 19 [26%] of 72 participants) Influenza-positive illness after challenge was detected in 17 (29%) of 58 individuals in the VXA-A1.1 group, 19 (35%) of 54 in the IIV group, and 15 (48%) of 31 in the placebo group.INTERPRETATION: Orally administered VXA-A1.1 was well tolerated and generated protective immunity against virus shedding, similar to a licensed intramuscular IIV. These results represent a major step forward in developing a safe and effective oral influenza vaccine.FUNDING: Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, and Biomedical Advanced Research and Development Authority.

    View details for DOI 10.1016/S1473-3099(19)30584-5

    View details for PubMedID 31978354

  • ADAM17 stabilizes its interacting partner inactive Rhomboid 2 (iRhom2) but not inactive Rhomboid 1 (iRhom1). The Journal of biological chemistry Weskamp, G. n., Tüshaus, J. n., Li, D. n., Feederle, R. n., Maretzky, T. n., Swendeman, S. n., Falck-Pedersen, E. n., McIlwain, D. R., Mak, T. W., Salmon, J. E., Lichtenthaler, S. F., Blobel, C. P. 2020

    Abstract

    The metalloprotease ADAM17 (a disintegrin and metalloprotease 17) is a key regulator of tumor necrosis factor α (TNFα), interleukin 6 receptor (IL-6R), and epidermal growth factor receptor (EGFR) signaling. ADAM17 maturation and function depend on the seven membrane-spanning inactive rhomboid-like proteins 1 and 2 (iRhom1/2 or Rhbdf1/2). Most studies to date have focused on overexpressed iRhom1 and 2, so only little is known about the properties of the endogenous proteins. Here, we show that endogenous iRhom1 and 2 can be cell-surface biotinylated on mouse embryonic fibroblasts (mEFs), revealing that endogenous iRhom1 and 2 proteins are present on the cell surface, and that iRhom2 also is present on the surface of lipopolysaccharide (LPS)-stimulated primary bone marrow-derived macrophages (BMDM). Interestingly, very little, if any iRhom2 was detectable in mEFs or BMDMs lacking ADAM17, suggesting that iRhom2 is stabilized by ADAM17. By contrast, the levels of iRhom1 were slightly increased in the absence of ADAM17 in mEFs, indicating that its stability does not depend on ADAM17. These findings support a model in which iRhom2 and ADAM17 are obligate binding partners and indicate that iRhom2 stability requires the presence of ADAM17, whereas iRhom1 is stable in the absence of ADAM17.

    View details for DOI 10.1074/jbc.RA119.011136

    View details for PubMedID 32060096

  • Coordinated Cellular Neighborhoods Orchestrate Antitumoral Immunity at the Colorectal Cancer Invasive Front. Cell Schürch, C. M., Bhate, S. S., Barlow, G. L., Phillips, D. J., Noti, L. n., Zlobec, I. n., Chu, P. n., Black, S. n., Demeter, J. n., McIlwain, D. R., Samusik, N. n., Goltsev, Y. n., Nolan, G. P. 2020

    Abstract

    Antitumoral immunity requires organized, spatially nuanced interactions between components of the immune tumor microenvironment (iTME). Understanding this coordinated behavior in effective versus ineffective tumor control will advance immunotherapies. We re-engineered co-detection by indexing (CODEX) for paraffin-embedded tissue microarrays, enabling simultaneous profiling of 140 tissue regions from 35 advanced-stage colorectal cancer (CRC) patients with 56 protein markers. We identified nine conserved, distinct cellular neighborhoods (CNs)-a collection of components characteristic of the CRC iTME. Enrichment of PD-1+CD4+ T cells only within a granulocyte CN positively correlated with survival in a high-risk patient subset. Coupling of tumor and immune CNs, fragmentation of T cell and macrophage CNs, and disruption of inter-CN communication was associated with inferior outcomes. This study provides a framework for interrogating how complex biological processes, such as antitumoral immunity, occur through concerted actions of cells and spatial domains.

    View details for DOI 10.1016/j.cell.2020.07.005

    View details for PubMedID 32763154

  • Robust ACE2 protein expression localizes to the motile cilia of the respiratory tract epithelia and is not increased by ACE inhibitors or angiotensin receptor blockers. medRxiv : the preprint server for health sciences Lee, I. T., Nakayama, T. n., Wu, C. T., Goltsev, Y. n., Jiang, S. n., Gall, P. A., Liao, C. K., Shih, L. C., Schürch, C. M., McIlwain, D. R., Chu, P. n., Borchard, N. A., Zarabanda, D. n., Dholakia, S. S., Yang, A. n., Kim, D. n., Kanie, T. n., Lin, C. D., Tsai, M. H., Phillips, K. M., Kim, R. n., Overdevest, J. B., Tyler, M. A., Yan, C. H., Lin, C. F., Lin, Y. T., Bau, D. T., Tsay, G. J., Patel, Z. M., Tsou, Y. A., Tai, C. J., Yeh, T. H., Hwang, P. H., Nolan, G. P., Nayak, J. V., Jackson, P. K. 2020

    Abstract

    We investigated the expression and subcellular localization of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), within the upper (nasal) and lower (pulmonary) respiratory tracts of healthy human donors. We detected ACE2 protein expression within the cilia organelle of ciliated airway epithelial cells, which likely represents the initial or early subcellular site of SARS-CoV-2 viral entry during respiratory transmission. We further determined whether ACE2 expression in the cilia of upper respiratory cells was influenced by patient demographics, clinical characteristics, co-morbidities, or medication use, and found no evidence that the use of angiotensin-converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARBs) increases ACE2 protein expression.

    View details for DOI 10.1101/2020.05.08.20092866

    View details for PubMedID 32511516

    View details for PubMedCentralID PMC7273284

  • ADAM17 stabilizes its interacting partner inactive Rhomboid 2 (iRhom2) but not inactive Rhomboid 1 (iRhom1). The Journal of biological chemistry Weskamp, G. n., Tüshaus, J. n., Li, D. n., Feederle, R. n., Maretzky, T. n., Swendemann, S. n., Falck-Pedersen, E. n., McIlwain, D. R., Mak, T. W., Salmon, J. E., Lichtenthaler, S. F., Blobel, C. P. 2020; 295 (13): 4350–58

    Abstract

    The metalloprotease ADAM17 (a disintegrin and metalloprotease 17) is a key regulator of tumor necrosis factor α (TNFα), interleukin 6 receptor (IL-6R), and epidermal growth factor receptor (EGFR) signaling. ADAM17 maturation and function depend on the seven-membrane-spanning inactive rhomboid-like proteins 1 and 2 (iRhom1/2 or Rhbdf1/2). Most studies to date have focused on overexpressed iRhom1 and -2, so only little is known about the properties of the endogenous proteins. Here, we show that endogenous iRhom1 and -2 can be cell surface-biotinylated on mouse embryonic fibroblasts (mEFs), revealing that endogenous iRhom1 and -2 proteins are present on the cell surface and that iRhom2 also is present on the surface of lipopolysaccharide-stimulated primary bone marrow-derived macrophages. Interestingly, very little, if any, iRhom2 was detectable in mEFs or bone marrow-derived macrophages lacking ADAM17, suggesting that iRhom2 is stabilized by ADAM17. By contrast, the levels of iRhom1 were slightly increased in the absence of ADAM17 in mEFs, indicating that its stability does not depend on ADAM17. These findings support a model in which iRhom2 and ADAM17 are obligate binding partners and indicate that iRhom2 stability requires the presence of ADAM17, whereas iRhom1 is stable in the absence of ADAM17.

    View details for DOI 10.1074/jbc.RA119.011136

    View details for PubMedID 33506789

  • Multiplexed Imaging for the simultaneous detection of nucleic acids and proteins to dissect the tissue immune landscape and microenvironment of viral diseases Jiang, S., Clave, X., Chan, C., Zhu, B., Bai, Y., Bosse, M., McIlwain, D., Bendall, S., Angelo, M., Estes, J., Nolan, G. BMC. 2019
  • Neurological, Cognitive, and Psychological Findings Among Survivors of Ebola Virus Disease From the 1995 Ebola Outbreak in Kikwit, Democratic Republic of Congo: A Cross-sectional Study CLINICAL INFECTIOUS DISEASES Kelly, J., Hoff, N. A., Spencer, D., Musene, K., Bramble, M. S., McIlwain, D., Okitundu, D., Porco, T. C., Rutherford, G. W., Glymour, M., Bjornson, Z., Mukadi, P., Okitolonda-Wemakoy, E., Nolan, G. P., Muyembe-Tamfum, J., Rimoin, A. W. 2019; 68 (8): 1388–93

    View details for DOI 10.1093/cid/ciy677

    View details for Web of Science ID 000464937800019

  • Multiomics modeling of the immunome, transcriptome, microbiome, proteome and metabolome adaptations during human pregnancy. Bioinformatics (Oxford, England) Ghaemi, M. S., DiGiulio, D. B., Contrepois, K., Callahan, B., Ngo, T. T., Lee-McMullen, B., Lehallier, B., Robaczewska, A., Mcilwain, D., Rosenberg-Hasson, Y., Wong, R. J., Quaintance, C., Culos, A., Stanley, N., Tanada, A., Tsai, A., Gaudilliere, D., Ganio, E., Han, X., Ando, K., McNeil, L., Tingle, M., Wise, P., Maric, I., Sirota, M., Wyss-Coray, T., Winn, V. D., Druzin, M. L., Gibbs, R., Darmstadt, G. L., Lewis, D. B., Partovi Nia, V., Agard, B., Tibshirani, R., Nolan, G., Snyder, M. P., Relman, D. A., Quake, S. R., Shaw, G. M., Stevenson, D. K., Angst, M. S., Gaudilliere, B., Aghaeepour, N. 2019; 35 (1): 95–103

    Abstract

    Motivation: Multiple biological clocks govern a healthy pregnancy. These biological mechanisms produce immunologic, metabolomic, proteomic, genomic and microbiomic adaptations during the course of pregnancy. Modeling the chronology of these adaptations during full-term pregnancy provides the frameworks for future studies examining deviations implicated in pregnancy-related pathologies including preterm birth and preeclampsia.Results: We performed a multiomics analysis of 51 samples from 17 pregnant women, delivering at term. The datasets included measurements from the immunome, transcriptome, microbiome, proteome and metabolome of samples obtained simultaneously from the same patients. Multivariate predictive modeling using the Elastic Net (EN) algorithm was used to measure the ability of each dataset to predict gestational age. Using stacked generalization, these datasets were combined into a single model. This model not only significantly increased predictive power by combining all datasets, but also revealed novel interactions between different biological modalities. Future work includes expansion of the cohort to preterm-enriched populations and in vivo analysis of immune-modulating interventions based on the mechanisms identified.Availability and implementation: Datasets and scripts for reproduction of results are available through: https://nalab.stanford.edu/multiomics-pregnancy/.Supplementary information: Supplementary data are available at Bioinformatics online.

    View details for PubMedID 30561547

  • Innovative Technologies for Advancement of WHO Risk Group 4 Pathogens Research GLOBAL VIROLOGY III: VIROLOGY IN THE 21ST CENTURY Logue, J., Solomon, J., Niemeyer, B. F., Benam, K. H., Lin, A. E., Bjornson, Z., Jiang, S., McIlwain, D. R., Nolan, G. P., Palacios, G., Kuhn, J. H., Shapshak, P., Balaji, S., Kangueane, P., Chiappelli, F., Somboonwit, C., Menezes, L. J., Sinnott, J. T. 2019: 437–69
  • Multiomics modeling of the immunome, transcriptome, microbiome, proteome and metabolome adaptations during human pregnancy BIOINFORMATICS Ghaemi, M., DiGiulio, D. B., Contrepois, K., Callahan, B., Ngo, T. M., Lee-McMullen, B., Lehallier, B., Robaczewska, A., Mcilwain, D., Rosenberg-Hasson, Y., Wong, R. J., Quaintance, C., Culos, A., Stanley, N., Tanada, A., Tsai, A., Gaudilliere, D., Ganio, E., Han, X., Ando, K., McNeil, L., Tingle, M., Wise, P., Maric, I., Sirota, M., Wyss-Coray, T., Winn, V. D., Druzin, M. L., Gibbs, R., Darmstadt, G. L., Lewis, D. B., Nia, V., Agard, B., Tibshirani, R., Nolan, G., Snyder, M. P., Relman, D. A., Quake, S. R., Shaw, G. M., Stevenson, D. K., Angst, M. S., Gaudilliere, B., Aghaeepour, N. 2019; 35 (1): 95–103
  • Blood-Induced Bone Loss In A Mouse Model Of Hemophilic Arthropathy Is Prevented By Blocking The iRhom2/ADAM17/TNF alpha Pathway Haxaire, C., Hakobyan, N., Panellini, T., Carballo, C., Mcilwain, D., Mak, T. W., Acharya, S., Li, D., Szymonifka, J., Rodeo, S., Song, X., Monette, S., Srivastava, A., Salmon, J., Blobel, C. WILEY. 2018: 369
  • Blood-induced bone loss in murine hemophilic arthropathy is prevented by blocking the iRhom2/ADAM17/TNF-alpha pathway BLOOD Haxaire, C., Hakobyan, N., Pannellini, T., Carballo, C., McIlwain, D., Mak, T. W., Rodeo, S., Acharya, S., Li, D., Szymonifka, J., Song, X., Monette, S., Srivastava, A., Salmon, J. E., Blobel, C. P. 2018; 132 (10): 1064–74

    Abstract

    Hemophilic arthropathy (HA) is a debilitating degenerative joint disease that is a major manifestation of the bleeding disorder hemophilia A. HA typically begins with hemophilic synovitis that resembles inflammatory arthritides, such as rheumatoid arthritis, and frequently results in bone loss in patients. A major cause of rheumatoid arthritis is inappropriate release of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) by the TNF-α convertase (TACE; also referred to as ADAM17) and its regulator, iRhom2. Therefore, we hypothesized that iRhom2/ADAM17-dependent shedding of TNF-α also has a pivotal role in mediating HA. Here, we show that addition of blood or its components to macrophages activates iRhom2/ADAM17-dependent TNF-α shedding, providing the premise to study the activation of this pathway by blood in the joint in vivo. For this, we turned to hemophilic FVIII-deficient mice (F8 -/- mice), which develop a hemarthrosis following needle puncture injury with synovial inflammation and significant osteopenia adjacent to the affected joint. We found that needle puncture-induced bleeding leads to increased TNF-α levels in the affected joint of F8 -/- mice. Moreover, inactivation of TNF-α or iRhom2 in F8 -/- mice reduced the osteopenia and synovial inflammation that develops in this mouse model for HA. Taken together, our results suggest that blood entering the joint activates the iRhom2/ADAM17/TNF-α pathway, thereby contributing to osteopenia and synovitis in mice. Therefore, this proinflammatory signaling pathway could emerge as an attractive new target to prevent osteoporosis and joint damage in HA patients.

    View details for PubMedID 29776906

    View details for PubMedCentralID PMC6128089

  • Neurological, cognitive, and psychological findings among survivors of Ebola virus disease from the 1995 Ebola outbreak in Kikwit, Democratic Republic of Congo: a cross-sectional study. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America Kelly, J. D., Hoff, N. A., Spencer, D., Musene, K., Bramble, M. S., McIlwain, D., Okitundu, D., Porco, T. C., Rutherford, G. W., Glymour, M. M., Bjornson, Z., Mukadi, P., Okitolonda-Wemakoy, E., Nolan, G. P., Muyembe-Tamfum, J. J., Rimoin, A. W. 2018

    Abstract

    Background: Clinical sequelae of Ebola virus disease (EVD) have not been described more than three years post-outbreak. We examined survivors and close contacts from the 1995 Ebola outbreak in Kikwit, Democratic Republic of Congo (DRC), and determined prevalence of abnormal neurological, cognitive, and psychological findings and their association with EVD survivorship.Methods: From August to September 2017, we conducted a cross-sectional study in Kikwit, DRC. Over two decades after the EVD outbreak, we recruited EVD survivors and close contacts from the outbreak to undergo physical examination and culturally adapted versions of the Folstein mini-mental status exam (MMSE) and Goldberg Anxiety and Depression Scale (GADS). We estimated the strength of relationships between EVD survivorship and health outcomes using linear regression models by comparing survivors vs. close contacts, adjusting for age, sex, educational level, marital status and healthcare worker status.Results: We enrolled 20 EVD survivors and 187 close contacts. Among the 20 EVD survivors, 4 (20%) reported at least one abnormal neurological symptom and 3 (15%) had an abnormal neurological examination. Among the 187 close contacts, 14 (11%) reported at least one abnormal neurologic symptom and 9 (5%) had an abnormal neurological examination. EVD survivors had lower mean MMSE and higher mean GADS scores as compared to close contacts (MMSE: adjusted coefficient: -1.85; 95% CI: -3.63, -0.07; GADS: adjusted coefficient: 3.91; 95% CI: 1.76, 6.04).Conclusions: EVD survivors can have lower cognitive scores and more symptoms of depression and anxiety than close contacts more than two decades after Ebola virus outbreaks.

    View details for PubMedID 30107392

  • The xenoestrogens biphenol-A and nonylphenol differentially regulate metalloprotease-mediated shedding of EGFR ligands JOURNAL OF CELLULAR PHYSIOLOGY Urriola-Munoz, P., Li, X., Maretzky, T., McIlwain, D. R., Mak, T. W., Reyes, J. G., Blobel, C. P., Moreno, R. D. 2018; 233 (3): 2247–56

    View details for DOI 10.1002/jcp.26097

    View details for Web of Science ID 000433519300042

  • The xenoestrogens biphenol-A and nonylphenol differentially regulate metalloprotease-mediated shedding of EGFR ligands. Journal of cellular physiology Urriola-Muñoz, P., Li, X., Maretzky, T., McIlwain, D. R., Mak, T. W., Reyes, J. G., Blobel, C. P., Moreno, R. D. 2018; 233 (3): 2247-2256

    Abstract

    The xenoestrogens bisphenol-A (BPA) and nonylphenol (NP) are endocrine disruptors used in the plastic polymer industry to manufacture different products for human use. Previous studies have suggested a role of these compounds in the shedding of signaling molecules, such as tumor necrosis factor α (TNF-α). The aim of this work was to evaluate the effect of BPA and NP on the sheddase ADAM17 and its newly discovered regulators iRhom1 and iRhom2 in the release of EGFR-ligands. We report that BPA and NP can stimulate the release of the ADAM17-substrates HB-EGF and TGF-α. In cells lacking ADAM17 (Adam17-/- mEFs) BPA-stimulated release of HB-EGF, but not TGF-α, was strongly reduced, whereas NP-stimulated shedding of HB-EGF and TGF-α was completely abolished. Inactivation of both ADAM17 and the related ADAM10 (Adam10/17-/- mEFs) completely prevented the release of these substrates. In the absence of iRhom1, BPA- or NP-stimulated release of HB-EGF or TGF-α was comparable to wild-type control mEFs, conversely the BPA-induced release of HB-EGF was abolished in iRhom2-/- mEFs. The defect in shedding of HB-EGF in iRhom2-/- mEF cells could be rescued by overexpressing iRhom2. Interestingly, the NP-stimulated release of HB-EGF was not affected by the absence of iRhom2, suggesting that NP could potentially activate both ADAM10 and ADAM17. We tested this hypothesis using betacellulin (BTC), an EGFR-ligand that is a substrate for ADAM10. We found that NP, but not BPA stimulated the release of BTC in Adam17-/- , iRhom2-/- , or iRhom1/2-/- , but not in Adam10/17-/- cells. Taken together, our results suggest that BPA and NP stimulate the release of EGFR-ligands by differentially activating ADAM17 or ADAM10. The identification of specific effects of these endocrine disruptors on ADAM10 and ADAM17 will help to provide a better understanding of their roles in cell signaling and proinflammatory processes, and provide new potential targets for treatment of reproductive or inflammatory diseases such as asthma or breast cancer that are promoted by xenoestrogens.

    View details for DOI 10.1002/jcp.26097

    View details for PubMedID 28703301

    View details for PubMedCentralID PMC5705578

  • NEUROLOGICAL, COGNITIVE, AND PSYCHOLOGICAL FINDINGS AMONG SURVIVORS OF EBOLA VIRUS DISEASE FROM THE 1995 KIKWIT OUTBREAK IN DEMOCRATIC REPUBLIC OF CONGO: A CROSS-SECTIONAL STUDY Kelly, J., Hoff, N., Spencer, D., Bramble, M., McIlwain, D., Musene, K., Okitolonda, E., Porco, T., Rutherford, G., Glymour, M., Mukadi, P., Rimoin, A. AMER SOC TROP MED & HYGIENE. 2018: 214
  • iRhom2 Deficiency Protects Fcgr2b-/- Lupus-Prone Mice from Kidney Damage By Modulating ADAM17-Dependent Shedding of TNF-alpha and EGFR Ligand Qing, X., Chinenov, Y., Redecha, P. M., Madaio, M., Issuree, P., McIlwain, D., Mak, T., Blobel, C., Salmon, J. E. WILEY. 2017
  • An immune clock of human pregnancy. Science immunology Aghaeepour, N. n., Ganio, E. A., Mcilwain, D. n., Tsai, A. S., Tingle, M. n., Van Gassen, S. n., Gaudilliere, D. K., Baca, Q. n., McNeil, L. n., Okada, R. n., Ghaemi, M. S., Furman, D. n., Wong, R. J., Winn, V. D., Druzin, M. L., El-Sayed, Y. Y., Quaintance, C. n., Gibbs, R. n., Darmstadt, G. L., Shaw, G. M., Stevenson, D. K., Tibshirani, R. n., Nolan, G. P., Lewis, D. B., Angst, M. S., Gaudilliere, B. n. 2017; 2 (15)

    Abstract

    The maintenance of pregnancy relies on finely tuned immune adaptations. We demonstrate that these adaptations are precisely timed, reflecting an immune clock of pregnancy in women delivering at term. Using mass cytometry, the abundance and functional responses of all major immune cell subsets were quantified in serial blood samples collected throughout pregnancy. Cell signaling-based Elastic Net, a regularized regression method adapted from the elastic net algorithm, was developed to infer and prospectively validate a predictive model of interrelated immune events that accurately captures the chronology of pregnancy. Model components highlighted existing knowledge and revealed previously unreported biology, including a critical role for the interleukin-2-dependent STAT5ab signaling pathway in modulating T cell function during pregnancy. These findings unravel the precise timing of immunological events occurring during a term pregnancy and provide the analytical framework to identify immunological deviations implicated in pregnancy-related pathologies.

    View details for PubMedID 28864494

  • iRhom2 regulates CSF1R cell surface expression and non-steady state myelopoiesis in mice EUROPEAN JOURNAL OF IMMUNOLOGY Qing, X., Rogers, L. D., Mortha, A., Lavin, Y., Redecha, P., Issuree, P. D., Maretzky, T., Merad, M., McIlwain, D. R., Mak, T. W., Overall, C. M., Blobel, C. P., Salmon, J. E. 2016; 46 (12): 2737-2748

    Abstract

    CSF1R (colony stimulating factor 1 receptor) is the main receptor for CSF1 and has crucial roles in regulating myelopoeisis. CSF1R can be proteolytically released from the cell surface by ADAM17 (A disintegrin and metalloprotease 17). Here, we identified CSF1R as a major substrate of ADAM17 in an unbiased degradomics screen. We explored the impact of CSF1R shedding by ADAM17 and its upstream regulator, inactive rhomboid protein 2 (iRhom2, gene name Rhbdf2), on homeostatic development of mouse myeloid cells. In iRhom2-/- mice, we found constitutive accumulation of membrane-bound CSF1R on myeloid cells at steady state, although cell numbers of these populations were not altered. However, in the context of mixed bone marrow (BM) chimera, under competitive pressure, iRhom2-/- BM progenitor-derived monocytes, tissue macrophages and lung DCs showed a repopulation advantage over those derived from wild-type (WT) BM progenitors, suggesting enhanced CSF1R signaling in the absence of iRhom2. In vitro experiments indicate that iRhom2-/- Lin(-) SCA-1(+) c-Kit(+) (LSKs) cells, but not granulocyte-macrophage progenitors (GMPs), had faster growth rates than WT cells in response to CSF1. Our results shed light on an important role of iRhom2/ADAM17 pathway in regulation of CSF1R shedding and repopulation of monocytes, macrophages and DCs.

    View details for DOI 10.1002/eji.201646482

    View details for Web of Science ID 000392940400013

    View details for PubMedID 27601030

    View details for PubMedCentralID PMC5149455

  • iRhoms 1 and 2 are essential upstream regulators of ADAM17-dependent EGFR signaling PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Li, X., Maretzky, T., Weskamp, G., Monette, S., Qing, X., Issuree, P. D., Crawford, H. C., McIlwain, D. R., Mak, T. W., Salmon, J. E., Blobel, C. P. 2015; 112 (19): 6080-6085

    Abstract

    The metalloproteinase ADAM17 (a disintegrin and metalloprotease 17) controls EGF receptor (EGFR) signaling by liberating EGFR ligands from their membrane anchor. Consequently, a patient lacking ADAM17 has skin and intestinal barrier defects that are likely caused by lack of EGFR signaling, and Adam17(-/-) mice die perinatally with open eyes, like Egfr(-/-) mice. A hallmark feature of ADAM17-dependent EGFR ligand shedding is that it can be rapidly and posttranslationally activated in a manner that requires its transmembrane domain but not its cytoplasmic domain. This suggests that ADAM17 is regulated by other integral membrane proteins, although much remains to be learned about the underlying mechanism. Recently, inactive Rhomboid 2 (iRhom2), which has seven transmembrane domains, emerged as a molecule that controls the maturation and function of ADAM17 in myeloid cells. However, iRhom2(-/-) mice appear normal, raising questions about how ADAM17 is regulated in other tissues. Here we report that iRhom1/2(-/-) double knockout mice resemble Adam17(-/-) and Egfr(-/-) mice in that they die perinatally with open eyes, misshapen heart valves, and growth plate defects. Mechanistically, we show lack of mature ADAM17 and strongly reduced EGFR phosphorylation in iRhom1/2(-/-) tissues. Finally, we demonstrate that iRhom1 is not essential for mouse development but regulates ADAM17 maturation in the brain, except in microglia, where ADAM17 is controlled by iRhom2. These results provide genetic, cell biological, and biochemical evidence that a principal function of iRhoms1/2 during mouse development is to regulate ADAM17-dependent EGFR signaling, suggesting that iRhoms1/2 could emerge as novel targets for treatment of ADAM17/EGFR-dependent pathologies.

    View details for DOI 10.1073/pnas.1505649112

    View details for Web of Science ID 000354390600066

    View details for PubMedID 25918388

    View details for PubMedCentralID PMC4434755

  • Deficiency of the B Cell-Activating Factor Receptor Results in Limited CD169+ Macrophage Function during Viral Infection. Journal of virology Xu, H. C., Huang, J., Khairnar, V., Duhan, V., Pandyra, A. A., Grusdat, M., Shinde, P., McIlwain, D. R., Maney, S. K., Gommerman, J., Löhning, M., Ohashi, P. S., Mak, T. W., Pieper, K., Sic, H., Speletas, M., Eibel, H., Ware, C. F., Tumanov, A. V., Kruglov, A. A., Nedospasov, S. A., Häussinger, D., Recher, M., Lang, K. S., Lang, P. A. 2015; 89 (9): 4748-4759

    Abstract

    The B cell-activating factor (BAFF) is critical for B cell development and humoral immunity in mice and humans. While the role of BAFF in B cells has been widely described, its role in innate immunity remains unknown. Using BAFF receptor (BAFFR)-deficient mice, we characterized BAFFR-related innate and adaptive immune functions following infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). We identified a critical role for BAFFR signaling in the generation and maintenance of the CD169(+) macrophage compartment. Consequently, Baffr(-) (/) (-) mice exhibited limited induction of innate type I interferon production after viral infection. Lack of BAFFR signaling reduced virus amplification and presentation following viral infection, resulting in highly reduced antiviral adaptive immune responses. As a consequence, BAFFR-deficient mice showed exacerbated and fatal disease after viral infection. Mechanistically, transient lack of B cells in Baffr(-) (/) (-) animals resulted in limited lymphotoxin expression, which is critical for maintenance of CD169(+) cells. In conclusion, BAFFR signaling affects both innate and adaptive immune activation during viral infections.Viruses cause acute and chronic infections in humans resulting in millions of deaths every year. Innate immunity is critical for the outcome of a viral infection. Innate type I interferon production can limit viral replication, while adaptive immune priming by innate immune cells induces pathogen-specific immunity with long-term protection. Here, we show that BAFFR deficiency not only perturbed B cells, but also resulted in limited CD169(+) macrophages. These macrophages are critical in amplifying viral particles to trigger type I interferon production and initiate adaptive immune priming. Consequently, BAFFR deficiency resulted in reduced enforced viral replication, limited type I interferon production, and reduced adaptive immunity compared to BAFFR-competent controls. As a result, BAFFR-deficient mice were predisposed to fatal viral infections. Thus, BAFFR expression is critical for innate immune activation and antiviral immunity.

    View details for DOI 10.1128/JVI.02976-14

    View details for PubMedID 25673724

  • Deficiency of the B Cell-Activating Factor Receptor Results in Limited CD169(+) Macrophage Function during Viral Infection JOURNAL OF VIROLOGY Xu, H. C., Huang, J., Khairnar, V., Duhan, V., Pandyra, A. A., Grusdat, M., Shinde, P., McIlwain, D. R., Maney, S. K., Gommerman, J., Loehning, M., Ohashi, P. S., Mak, T. W., Pieper, K., Sic, H., Speletas, M., Eibel, H., Ware, C. F., Tumanov, A. V., Kruglov, A. A., Nedospasov, S. A., Haeussinger, D., Recher, M., Lang, K. S., Lang, P. A. 2015; 89 (9): 4748-4759
  • T-cell STAT3 is required for the maintenance of humoral immunity to LCMV. European journal of immunology McIlwain, D. R., Grusdat, M., Pozdeev, V. I., Xu, H. C., Shinde, P., Reardon, C., Hao, Z., Beyer, M., Bergthaler, A., Häussinger, D., Nolan, G. P., Lang, K. S., Lang, P. A. 2015; 45 (2): 418-427

    Abstract

    STAT3 is a critical transcription factor activated downstream of cytokine signaling and is integral for the function of multiple immune cell types. Human mutations in STAT3 cause primary immunodeficiency resulting in impaired control of a variety of infections, including reactivation of latent viruses. In this study, we investigate how T-cell functions of STAT3 contribute to responses to viral infection by inducing chronic lymphocytic choriomeningitis virus (LCMV) infection in mice lacking STAT3 specifically in T cells. Although mice with conditional disruption of STAT3 in T cells were able to mount early responses to viral infection similar to control animals, including expansion of effector T cells, we found generation of T-follicular helper (Tfh) cells to be impaired. As a result, STAT3 T cell deficient mice produced attenuated germinal center reactions, and did not accumulate bone marrow virus specific IgG-secreting cells, resulting in failure to maintain levels of virus-specific IgG or mount neutralizing responses to LCMV in the serum. These effects were associated with reduced control of viral replication and prolonged infection. Our results demonstrate the importance of STAT3 in T cells for the generation of functional long-term humoral immunity to viral infections.

    View details for DOI 10.1002/eji.201445060

    View details for PubMedID 25393615

  • T-cell STAT3 is required for the maintenance of humoral immunity to LCMV. European journal of immunology McIlwain, D. R., Grusdat, M., Pozdeev, V. I., Xu, H. C., Shinde, P., Reardon, C., Hao, Z., Beyer, M., Bergthaler, A., Häussinger, D., Nolan, G. P., Lang, K. S., Lang, P. A. 2015; 45 (2): 418-427

    Abstract

    STAT3 is a critical transcription factor activated downstream of cytokine signaling and is integral for the function of multiple immune cell types. Human mutations in STAT3 cause primary immunodeficiency resulting in impaired control of a variety of infections, including reactivation of latent viruses. In this study, we investigate how T-cell functions of STAT3 contribute to responses to viral infection by inducing chronic lymphocytic choriomeningitis virus (LCMV) infection in mice lacking STAT3 specifically in T cells. Although mice with conditional disruption of STAT3 in T cells were able to mount early responses to viral infection similar to control animals, including expansion of effector T cells, we found generation of T-follicular helper (Tfh) cells to be impaired. As a result, STAT3 T cell deficient mice produced attenuated germinal center reactions, and did not accumulate bone marrow virus specific IgG-secreting cells, resulting in failure to maintain levels of virus-specific IgG or mount neutralizing responses to LCMV in the serum. These effects were associated with reduced control of viral replication and prolonged infection. Our results demonstrate the importance of STAT3 in T cells for the generation of functional long-term humoral immunity to viral infections.

    View details for DOI 10.1002/eji.201445060

    View details for PubMedID 25393615

  • Deletions in the cytoplasmic domain of iRhom1 and iRhom2 promote shedding of the TNF receptor by the protease ADAM17. Science signaling Maney, S. K., McIlwain, D. R., Polz, R., Pandyra, A. A., Sundaram, B., Wolff, D., Ohishi, K., Maretzky, T., Brooke, M. A., Evers, A., Vasudevan, A. A., Aghaeepour, N., Scheller, J., Münk, C., Häussinger, D., Mak, T. W., Nolan, G. P., Kelsell, D. P., Blobel, C. P., Lang, K. S., Lang, P. A. 2015; 8 (401): ra109-?

    Abstract

    The protease ADAM17 (a disintegrin and metalloproteinase 17) catalyzes the shedding of various transmembrane proteins from the surface of cells, including tumor necrosis factor (TNF) and its receptors. Liberation of TNF receptors (TNFRs) from cell surfaces can dampen the cellular response to TNF, a cytokine that is critical in the innate immune response and promotes programmed cell death but can also promote sepsis. Catalytically inactive members of the rhomboid family of proteases, iRhom1 and iRhom2, mediate the intracellular transport and maturation of ADAM17. Using a genetic screen, we found that the presence of either iRhom1 or iRhom2 lacking part of their extended amino-terminal cytoplasmic domain (herein referred to as ΔN) increases ADAM17 activity, TNFR shedding, and resistance to TNF-induced cell death in fibrosarcoma cells. Inhibitors of ADAM17, but not of other ADAM family members, prevented the effects of iRhom-ΔN expression. iRhom1 and iRhom2 were functionally redundant, suggesting a conserved role for the iRhom amino termini. Cells from patients with a dominantly inherited cancer susceptibility syndrome called tylosis with esophageal cancer (TOC) have amino-terminal mutations in iRhom2. Keratinocytes from TOC patients exhibited increased TNFR1 shedding compared with cells from healthy donors. Our results explain how loss of the amino terminus in iRhom1 and iRhom2 impairs TNF signaling, despite enhancing ADAM17 activity, and may explain how mutations in the amino-terminal region contribute to the cancer predisposition syndrome TOC.

    View details for DOI 10.1126/scisignal.aac5356

    View details for PubMedID 26535007

  • IRF4 and BATF are critical for CD8(+) T-cell function following infection with LCMV CELL DEATH AND DIFFERENTIATION Grusdat, M., McIlwain, D. R., Xu, H. C., Pozdeev, V. I., Knievel, J., Crome, S. Q., Robert-Tissot, C., Dress, R. J., Pandyra, A. A., Speiser, D. E., Lang, E., Maney, S. K., Elford, A. R., Hamilton, S. R., Scheu, S., Pfeffer, K., Bode, J., Mittruecker, H., Lohoff, M., Huber, M., Haeussinger, D., Ohashi, P. S., Mak, T. W., Lang, K. S., Lang, P. A. 2014; 21 (7): 1050-1060

    Abstract

    CD8(+) T-cell functions are critical for preventing chronic viral infections by eliminating infected cells. For healthy immune responses, beneficial destruction of infected cells must be balanced against immunopathology resulting from collateral damage to tissues. These processes are regulated by factors controlling CD8(+) T-cell function, which are still incompletely understood. Here, we show that the interferon regulatory factor 4 (IRF4) and its cooperating binding partner B-cell-activating transcription factor (BATF) are necessary for sustained CD8(+) T-cell effector function. Although Irf4(-/-) CD8(+) T cells were initially capable of proliferation, IRF4 deficiency resulted in limited CD8(+) T-cell responses after infection with the lymphocytic choriomeningitis virus. Consequently, Irf4(-/-) mice established chronic infections, but were protected from fatal immunopathology. Absence of BATF also resulted in reduced CD8(+) T-cell function, limited immunopathology, and promotion of viral persistence. These data identify the transcription factors IRF4 and BATF as major regulators of antiviral cytotoxic T-cell immunity.

    View details for DOI 10.1038/cdd.2014.19

    View details for Web of Science ID 000337234200003

    View details for PubMedID 24531538

  • iRhom2 controls the substrate selectivity of stimulated ADAM17-dependent ectodomain shedding PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Maretzky, T., McIlwain, D. R., Issuree, P. D., Li, X., Malapeira, J., Amin, S., Lang, P. A., Mak, T. W., Blobel, C. P. 2013; 110 (28): 11433-11438

    Abstract

    Protein ectodomain shedding by ADAM17 (a disintegrin and metalloprotease 17), a principal regulator of EGF-receptor signaling and TNFα release, is rapidly and posttranslationally activated by a variety of signaling pathways, and yet little is known about the underlying mechanism. Here, we report that inactive rhomboid protein 2 (iRhom2), recently identified as essential for the maturation of ADAM17 in hematopoietic cells, is crucial for the rapid activation of the shedding of some, but not all substrates of ADAM17. Mature ADAM17 is present in mouse embryonic fibroblasts (mEFs) lacking iRhom2, and yet ADAM17 is unable to support stimulated shedding of several of its substrates, including heparin-binding EGF and Kit ligand 2 in this context. Stimulated shedding of other ADAM17 substrates, such as TGFα, is not affected in iRhom2(-/-) mEFs but can be strongly reduced by treating iRhom2(-/-) mEFs with siRNA against iRhom1. Activation of heparin-binding EGF or Kit ligand 2 shedding by ADAM17 in iRhom2(-/-) mEFs can be rescued by wild-type iRhom2 but not by iRhom2 lacking its N-terminal cytoplasmic domain. The requirement for the cytoplasmic domain of iRhom2 for stimulated shedding by ADAM17 may help explain why the cytoplasmic domain of ADAM17 is not required for stimulated shedding. The functional relevance of iRhom2 in regulating shedding of EGF receptor (EGFR) ligands is established by a lack of lysophasphatidic acid/ADAM17/EGFR-dependent crosstalk with ERK1/2 in iRhom2(-/-) mEFs, and a significant reduction of FGF7/ADAM17/EGFR-stimulated migration of iRhom2(-/-) keratinocytes. Taken together, these findings uncover functions for iRhom2 in the regulation of EGFR signaling and in controlling the activation and substrate selectivity of ADAM17-dependent shedding events.

    View details for DOI 10.1073/pnas.1302553110

    View details for Web of Science ID 000321827000058

    View details for PubMedID 23801765

  • Caspase Functions in Cell Death and Disease COLD SPRING HARBOR PERSPECTIVES IN BIOLOGY McIlwain, D. R., Berger, T., Mak, T. W. 2013; 5 (4)

    Abstract

    Caspases are a family of endoproteases that provide critical links in cell regulatory networks controlling inflammation and cell death. The activation of these enzymes is tightly controlled by their production as inactive zymogens that gain catalytic activity following signaling events promoting their aggregation into dimers or macromolecular complexes. Activation of apoptotic caspases results in inactivation or activation of substrates, and the generation of a cascade of signaling events permitting the controlled demolition of cellular components. Activation of inflammatory caspases results in the production of active proinflammatory cytokines and the promotion of innate immune responses to various internal and external insults. Dysregulation of caspases underlies human diseases including cancer and inflammatory disorders, and major efforts to design better therapies for these diseases seek to understand how these enzymes work and how they can be controlled.

    View details for DOI 10.1101/cshperspect.a008656

    View details for Web of Science ID 000317175200008

    View details for PubMedID 23545416

  • Reactive oxygen species delay control of lymphocytic choriomeningitis virus. Cell death and differentiation Lang, P. A., Xu, H. C., Grusdat, M., McIlwain, D. R., Pandyra, A. A., Harris, I. S., Shaabani, N., HONKE, N., Maney, S. K., Lang, E., Pozdeev, V. I., Recher, M., Odermatt, B., Brenner, D., Häussinger, D., Ohashi, P. S., Hengartner, H., Zinkernagel, R. M., Mak, T. W., Lang, K. S. 2013; 20 (4): 649-658

    Abstract

    Cluster of differentiation (CD)8(+) T cells are like a double edged sword during chronic viral infections because they not only promote virus elimination but also induce virus-mediated immunopathology. Elevated levels of reactive oxygen species (ROS) have been reported during virus infections. However, the role of ROS in T-cell-mediated immunopathology remains unclear. Here we used the murine lymphocytic choriomeningitis virus to explore the role of ROS during the processes of virus elimination and induction of immunopathology. We found that virus infection led to elevated levels of ROS producing granulocytes and macrophages in virus-infected liver and spleen tissues that were triggered by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Lack of the regulatory subunit p47phox of the NADPH oxidase diminished ROS production in these cells. While CD8(+) T cells exhibited ROS production that was independent of NADPH oxidase expression, survival and T-cell function was elevated in p47phox-deficient (Ncf1(-/-)) mice. In the absence of p47phox, enhanced T-cell immunity promoted virus elimination and blunted corresponding immunopathology. In conclusion, we find that NADPH-mediated production of ROS critically impairs the immune response, impacting elimination of virus and outcome of liver cell damage.

    View details for DOI 10.1038/cdd.2012.167

    View details for PubMedID 23328631

  • Involvement of Toso in activation of monocytes, macrophages, and granulocytes PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Lang, K. S., Lang, P. A., Meryk, A., Pandyra, A. A., Boucher, L., Pozdeev, V. I., Tusche, M. W., Goethert, J. R., Haight, J., Wakeham, A., You-Ten, A. J., McIlwain, D. R., Merches, K., Khairnar, V., Recher, M., Nolan, G. P., Hitoshi, Y., Funkner, P., Navarini, A. A., Verschoor, A., Shaabani, N., Honke, N., Penn, L. Z., Ohashi, P. S., Haeussinger, D., Lee, K., Mak, T. W. 2013; 110 (7): 2593-2598

    Abstract

    Rapid activation of immune responses is necessary for antibacterial defense, but excessive immune activation can result in life-threatening septic shock. Understanding how these processes are balanced may provide novel therapeutic potential in treating inflammatory disease. Fc receptors are crucial for innate immune activation. However, the role of the putative Fc receptor for IgM, known as Toso/Faim3, has to this point been unclear. In this study, we generated Toso-deficient mice and used them to uncover a critical regulatory function of Toso in innate immune activation. Development of innate immune cells was intact in the absence of Toso, but Toso-deficient neutrophils exhibited more reactive oxygen species production and reduced phagocytosis of pathogens compared with controls. Cytokine production was also decreased in Toso(-/-) mice compared with WT animals, rendering them resistant to septic shock induced by lipopolysaccharide. However, Toso(-/-) mice also displayed limited cytokine production after infection with the bacterium Listeria monocytogenes that was correlated with elevated presence of Listeria throughout the body. Accordingly, Toso(-/-) mice succumbed to infections of L. monocytogenes, whereas WT mice successfully eliminated the infection. Taken together, our data reveal Toso to be a unique regulator of innate immune responses during bacterial infection and septic shock.

    View details for DOI 10.1073/pnas.1222264110

    View details for Web of Science ID 000315812800047

    View details for PubMedID 23359703

    View details for PubMedCentralID PMC3574925

  • iRHOM2 is a critical pathogenic mediator of inflammatory arthritis JOURNAL OF CLINICAL INVESTIGATION Issuree, P. D., Maretzky, T., McIlwain, D. R., Monette, S., Qing, X., Lang, P. A., Swendeman, S. L., Park-Min, K., Binder, N., Kalliolias, G. D., Yarilina, A., Horiuchi, K., Ivashkiv, L. B., Mak, T. W., Salmon, J. E., Blobel, C. P. 2013; 123 (2): 928-932

    Abstract

    iRHOM2, encoded by the gene Rhbdf2, regulates the maturation of the TNF-α convertase (TACE), which controls shedding of TNF-α and its biological activity in vivo. TACE is a potential target to treat TNF-α-dependent diseases, such as rheumatoid arthritis, but there are concerns about potential side effects, because TACE also protects the skin and intestinal barrier by activating EGFR signaling. Here we report that inactivation of Rhbdf2 allows tissue-specific regulation of TACE by selectively preventing its maturation in immune cells, without affecting its homeostatic functions in other tissues. The related iRHOM1, which is widely expressed, except in hematopoietic cells, supported TACE maturation and shedding of the EGFR ligand TGF-α in Rhbdf2-deficient cells. Remarkably, mice lacking Rhbdf2 were protected from K/BxN inflammatory arthritis to the same extent as mice lacking TACE in myeloid cells or Tnfa-deficient mice. In probing the underlying mechanism, we found that two main drivers of K/BxN arthritis, complement C5a and immune complexes, stimulated iRHOM2/TACE-dependent shedding of TNF-α in mouse and human cells. These data demonstrate that iRHOM2 and myeloid-expressed TACE play a critical role in inflammatory arthritis and indicate that iRHOM2 is a potential therapeutic target for selective inactivation of TACE in myeloid cells.

    View details for DOI 10.1172/JCI66168

    View details for Web of Science ID 000314553600045

    View details for PubMedID 23348744

  • Smg1 is required for embryogenesis and regulates diverse genes via alternative splicing coupled to nonsense-mediated mRNA decay PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA McIlwain, D. R., Pan, Q., Reilly, P. T., Elia, A. J., McCracken, S., Wakeham, A. C., Itie-Youten, A., Blencowe, B. J., Mak, T. W. 2010; 107 (27): 12186-12191

    Abstract

    Smg1 is a PI3K-related kinase (PIKK) associated with multiple cellular functions, including DNA damage responses, telomere maintenance, and nonsense-mediated mRNA decay (NMD). NMD degrades transcripts that harbor premature termination codons (PTCs) as a result of events such as mutation or alternative splicing (AS). Recognition of PTCs during NMD requires the action of the Upstream frameshift protein Upf1, which must first be phosphorylated by Smg1. However, the physiological function of mammalian Smg1 is not known. By using a gene-trap model of Smg1 deficiency, we show that this kinase is essential for mouse embryogenesis such that Smg1 loss is lethal at embryonic day 8.5. High-throughput RNA sequencing (RNA-Seq) of RNA from cells of Smg1-deficient embryos revealed that Smg1 depletion led to pronounced accumulation of PTC-containing splice variant transcripts from approximately 9% of genes predicted to contain AS events capable of eliciting NMD. Among these genes are those involved in splicing itself, as well as genes not previously known to be subject to AS-coupled NMD, including several involved in transcription, intracellular signaling, membrane dynamics, cell death, and metabolism. Our results demonstrate a critical role for Smg1 in early mouse development and link the loss of this NMD factor to major and widespread changes in the mammalian transcriptome.

    View details for DOI 10.1073/pnas.1007336107

    View details for Web of Science ID 000279572100028

    View details for PubMedID 20566848