Article10 March 2020Open Access Enhancing protective microglial activities with a dual function TREM2 antibody to the stalk region Kai Schlepckow German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Kathryn M Monroe Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Gernot Kleinberger orcid.org/0000-0002-5811-8226 Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Ludovico Cantuti-Castelvetri German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Samira Parhizkar Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Dan Xia Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Michael Willem Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Georg Werner Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Nadine Pettkus Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Bettina Brunner German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Alice Sülzen German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Brigitte Nuscher Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Heike Hampel Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Xianyuan Xiang Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Graduate School of Systemic Neuroscience, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Regina Feederle orcid.org/0000-0002-3981-367X German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Helmholtz Center Munich, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Core Facility Monoclonal Antibody Development, Munich, Germany Search for more papers by this author Sabina Tahirovic orcid.org/0000-0003-4403-9559 German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Joshua I Park Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Rachel Prorok Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Cathal Mahon Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Chun-Chi Liang Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Ju Shi Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Do Jin Kim Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Hanna Sabelström Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Fen Huang Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Gilbert Di Paolo Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Mikael Simons German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Institute of Neuronal Cell Biology (TUM-NZB), Munich, Germany Search for more papers by this author Joseph W Lewcock Corresponding Author [email protected] orcid.org/0000-0003-3012-7881 Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Christian Haass Corresponding Author [email protected] orcid.org/0000-0002-4869-1627 German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Search for more papers by this author Kai Schlepckow German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Kathryn M Monroe Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Gernot Kleinberger orcid.org/0000-0002-5811-8226 Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Ludovico Cantuti-Castelvetri German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Samira Parhizkar Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Dan Xia Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Michael Willem Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Georg Werner Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Nadine Pettkus Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Bettina Brunner German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Alice Sülzen German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Brigitte Nuscher Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Heike Hampel Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Xianyuan Xiang Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Graduate School of Systemic Neuroscience, Ludwig-Maximilians-Universität München, Munich, Germany Search for more papers by this author Regina Feederle orcid.org/0000-0002-3981-367X German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Helmholtz Center Munich, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Core Facility Monoclonal Antibody Development, Munich, Germany Search for more papers by this author Sabina Tahirovic orcid.org/0000-0003-4403-9559 German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Search for more papers by this author Joshua I Park Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Rachel Prorok Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Cathal Mahon Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Chun-Chi Liang Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Ju Shi Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Do Jin Kim Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Hanna Sabelström Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Fen Huang Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Gilbert Di Paolo Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Mikael Simons German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Institute of Neuronal Cell Biology (TUM-NZB), Munich, Germany Search for more papers by this author Joseph W Lewcock Corresponding Author [email protected] orcid.org/0000-0003-3012-7881 Denali Therapeutics Inc., South San Francisco, CA, USA Search for more papers by this author Christian Haass Corresponding Author [email protected] orcid.org/0000-0002-4869-1627 German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany Search for more papers by this author Author Information Kai Schlepckow1,‡, Kathryn M Monroe2,‡, Gernot Kleinberger3,†,‡, Ludovico Cantuti-Castelvetri1, Samira Parhizkar3, Dan Xia2, Michael Willem3, Georg Werner3, Nadine Pettkus3, Bettina Brunner1, Alice Sülzen1, Brigitte Nuscher3, Heike Hampel3, Xianyuan Xiang3,4, Regina Feederle1,5,6, Sabina Tahirovic1, Joshua I Park2, Rachel Prorok2, Cathal Mahon2, Chun-Chi Liang2, Ju Shi2,†, Do Jin Kim2, Hanna Sabelström2, Fen Huang2, Gilbert Di Paolo2, Mikael Simons1,5,7, Joseph W Lewcock *,2 and Christian Haass *,1,3,5 1German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany 2Denali Therapeutics Inc., South San Francisco, CA, USA 3Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany 4Graduate School of Systemic Neuroscience, Ludwig-Maximilians-Universität München, Munich, Germany 5Munich Cluster for Systems Neurology (SyNergy), Munich, Germany 6Helmholtz Center Munich, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Core Facility Monoclonal Antibody Development, Munich, Germany 7Institute of Neuronal Cell Biology (TUM-NZB), Munich, Germany †Present address: ISAR Bioscience GmbH, Planegg, Germany †Present address: Jazz Pharmaceuticals, Palo Alto, CA, USA ‡These authors contributed equally to this work *Correspondence: Tel: +1 (650) 745-5247; E-mail: [email protected] *Correspondence: Tel: +49 (0) 89-4400-46549; E-mail: [email protected] EMBO Mol Med (2020)12:e11227https://doi.org/10.15252/emmm.201911227 PDFDownload PDF of article text and main figures. Peer ReviewDownload a summary of the editorial decision process including editorial decision letters, reviewer comments and author responses to feedback. ToolsAdd to favoritesDownload CitationsTrack CitationsPermissions ShareFacebookTwitterLinked InMendeleyWechatReddit Figures & Info Abstract Triggering receptor expressed on myeloid cells 2 (TREM2) is essential for the transition of homeostatic microglia to a disease-associated microglial state. To enhance TREM2 activity, we sought to selectively increase the full-length protein on the cell surface via reducing its proteolytic shedding by A Disintegrin And Metalloproteinase (i.e., α-secretase) 10/17. We screened a panel of monoclonal antibodies against TREM2, with the aim to selectively compete for α-secretase-mediated shedding. Monoclonal antibody 4D9, which has a stalk region epitope close to the cleavage site, demonstrated dual mechanisms of action by stabilizing TREM2 on the cell surface and reducing its shedding, and concomitantly activating phospho-SYK signaling. 4D9 stimulated survival of macrophages and increased microglial uptake of myelin debris and amyloid β-peptide in vitro. In vivo target engagement was demonstrated in cerebrospinal fluid, where nearly all soluble TREM2 was 4D9-bound. Moreover, in a mouse model for Alzheimer's disease-related pathology, 4D9 reduced amyloidogenesis, enhanced microglial TREM2 expression, and reduced a homeostatic marker, suggesting a protective function by driving microglia toward a disease-associated state. Synopsis This study describes the discovery and characterization of a novel TREM2 antibody, which induces protective microglial functions and provides a basis for the development of human antibodies with a similar mechanistic profile for treatment of Alzheimer's disease. An antibody directed to the stalk region of TREM2 prevents its shedding and increases cell autonomous signaling. Addition of this TREM2 antibody to myeloid cells in vitro stimulates phagocytosis, and improves cell survival. TREM2 antibody treatment increases TREM2 expression on brain microglia, decreases homeostatic markers and reduces amyloid plaque pathology in a mouse model of Alzheimer's disease. Antibody mediated stimulation of TREM2 signaling may be efficacious in Alzheimer's disease as well as other neurodegenerative disorders and obesity-associated metabolic syndromes. Introduction Alzheimer's disease (AD) and related disorders are devastating diseases threatening our aging society, and still, no cure is available. Since a number of recent clinical trials using amyloid β-peptide (Aβ)-based therapeutic approaches failed to improve cognition or even worsened the clinical outcome of patients (Egan et al, 2019), novel targets are desperately needed. Importantly, it appears that anti-Aβ therapeutics may have to be administered decades before symptom onset, since at the time patients are enrolled in clinical studies they are amyloid positron emission tomography (PET)-positive (Sevigny et al, 2016) even if they have not yet developed overt dementia. We therefore need to develop strategies to interfere with the amyloid cascade immediately after it has been initially triggered by Aβ and may become independent of its further de novo production. In addition to the selective deposition of amyloidogenic proteins, neuroinflammation associated with microgliosis is a common feature of many neurodegenerative disorders (Ransohoff, 2016). Recent genome-wide association studies strongly substantiated a central role of innate immunity for neurodegeneration by identifying a number of risk variants in genes that are exclusively expressed within microglia in the brain. Among them, coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk for late-onset AD as much as the apolipoprotein ε4 allele (Guerreiro et al, 2013; Jonsson & Stefansson, 2013; Jonsson et al, 2013; Rayaprolu et al, 2013; Borroni et al, 2014; Cuyvers et al, 2014). The disease-associated variants appear to cause a loss of function of TREM2 through a variety of mechanisms, including inhibition of cellular maturation of TREM2, destroying essential lipid binding sites within the immunoglobulin region of the ectodomain, and enhancing its shedding (Kleinberger et al, 2014, 2017; Mazaheri et al, 2017; Schlepckow et al, 2017; Ulland et al, 2017; Song et al, 2018). Full-length TREM2, in association with DAP12, forms a heteromeric complex required to activate phospho-SYK signaling (Colonna, 2003). Signaling appears to be terminated by α-secretase-mediated shedding of the TREM2 ectodomain (Fig 1A) (Wunderlich et al, 2013; Kleinberger et al, 2014), although very recent data suggest that soluble TREM2 (sTREM2) may also have signaling functions (Zhong et al, 2017, 2019), and requires further investigation. Loss of TREM2-mediated signaling locks microglia in a homeostatic state and inhibits their transition to disease-associated microglia (DAM) (Krasemann et al, 2017; Mazaheri et al, 2017), which are phenotypically characterized by enhanced migration, chemotaxis, and phagocytosis (Keren-Shaul et al, 2017; Mazaheri et al, 2017). Furthermore, lack of functional TREM2 appears to be associated with reduced cellular proliferation and death of phagocytes as well as reduced energy metabolism and cerebral blood flow in mice (Ulland et al, 2015, 2017; Kleinberger et al, 2017). TREM2 has been shown to be involved in lipid sensing, a function which is diminished by the AD-associated TREM2 R47H variant (Wang et al, 2015). Furthermore, loss of TREM2 results in downregulation of genes involved in lipid metabolism (Keren-Shaul et al, 2017; Griciuc et al, 2019). Consistent with these observations, TREM2 appears to be required for elimination of damaged myelin by microglia (Cantoni et al, 2015). Moreover, TREM2-deficient microglia were shown to be overloaded with cholesterol esters (CEs) as well as other lipid species as a result of chronic phagocytic challenge of myelin debris induced by a cuprizone diet (Nugent et al, 2019), indicating that TREM2 is important for lipid processing in addition to phagocytosis. Figure 1. Screening and molecular characterization of anti-mouse TREM2 antibodies Schematic representation of TREM2 processing by ADAM10/17. Cleavage occurs C-terminal of residue His 157. The entire ectodomain (residues 19–171) was used for immunization of rats to generate TREM2 antibodies. CTF, C-terminal fragment; sTREM2, soluble TREM2. Immunoblot analysis of membrane fractions of HEK293 Flp-In cells stably overexpressing both mouse TREM2 and mouse DAP12 upon treatment with 4D9 antibody reveals increased levels of membrane-bound TREM2 similar to what can be achieved by ADAM protease inhibition using the GM6001 inhibitor. An isotype antibody was used as a negative control. Calnexin served as a loading control. Levels of membrane-bound TREM2 were quantified by MSD ELISA. Data represent the mean ± SEM (n = 3). One-way ANOVA, Tukey's post hoc test; P (DMSO vs GM) = 0.0011; P (DMSO vs isotype) = 0.992; P (isotype vs 4D9) = 0.0005; n.s., not significant. Immunoblot analysis of conditioned media from HEK293 Flp-In cells stably overexpressing both mouse TREM2 and mouse DAP12 upon treatment with 4D9 antibody reveals decreased levels of sTREM2 similar to what can be achieved by ADAM protease inhibition using the GM6001 inhibitor. An isotype antibody was used as a negative control. sAPPα served as a loading control. Note that heavy and light chains of the antibodies used for treatment are also detected and annotated. Levels of sTREM2 were quantified by MSD ELISA. Data represent the mean ± SEM (n = 3). One-way ANOVA, Tukey's post hoc test; P (DMSO vs GM) < 0.0001; P (DMSO vs isotype) = 0.6372; P (isotype vs 4D9) < 0.0001; n.s., not significant. 4D9 antibody selectively detects TREM2 on the cell surface of HEK293 Flp-In cells stably overexpressing mouse TREM2 and mouse DAP12. An anti-HA antibody was used as a positive control, while empty vector-transfected HEK293 Flp-In cells were used as a negative control. Scale bar = 10 μm. Peptide ELISAs detect anti-mouse TREM2 antibody binding to tiled stalk region peptides, full-length stalk peptide, or a truncated ADAM cleavage site peptide. The binding epitope of 4D9 antibody is located 12-amino acids N-terminal of the ADAM cleavage site at His 157. Sequence comparison of mouse TREM2 and human TREM2 shows substantial sequence conservation around the 4D9 epitope (upper panel). Immunoblot analysis demonstrates that antibody 4D9 is highly specific for mouse TREM2 and does not detect human TREM2 or mouse TREM1 (lower panel). 4D9 binding to the mouse TREM2 ECD is competed off by a stalk region peptide. A competition ELISA demonstrates that a dose titration of stalk peptide reduces binding of 4D9 to TREM2 ECD with an EC50 of 1.3 μM. Data represent the mean ± SEM (n = 3). ECD, extracellular domain. Surface plasmon resonance binding kinetics of increasing concentrations of 4D9 antibody to mouse TREM2 ECD evaluated by Biacore, kon = 5.9 × 105 M−1 s−1, koff = 4.0 × 10−5 s−1, KD = 68 pM. 4D9 binding to human TREM2 or mouse TREM1 was undetectable. Data information: Statistical evaluations are displayed as follows: **P < 0.01; ***P < 0.001; ****P < 0.0001. Source data are available online for this figure. Source Data for Figure 1 [emmm201911227-sup-0002-SDataFig1.pdf] Download figure Download PowerPoint Quantitative analyses of the cleaved sTREM2 ectodomain in the cerebrospinal fluid of patients with sporadic and autosomal dominant AD (ADAD) revealed enhanced levels at the mild cognitive impairment stage (Suarez-Calvet et al, 2016b, 2019) and up to 5 years before the estimated onset of ADAD (Suarez-Calvet et al, 2016a), respectively. Moreover, sTREM2 positively correlates with total tau and phospho-tau but not with Aβ (Suarez-Calvet et al, 2016b). Thus, CSF sTREM2 seems to be associated with tau pathology. Moreover, microglial activation and associated cellular functions are protective for cognition in a mouse model of AD (Focke et al, 2019), and ameliorate retinal degeneration (O'Koren et al, 2019). Furthermore, high levels of CSF sTREM2 predict a better outcome of cognition and reduced hippocampal shrinkage in the Alzheimer's disease neuroimaging initiative cohort (Ewers et al, 2019). Taken in combination with the observation that TREM2 loss of function increases AD risk, this suggests that enhancing TREM2 signaling represents a therapeutic strategy that is downstream of the initial Aβ-mediated trigger of the amyloid cascade. Since α-secretase-mediated shedding of TREM2 (Kleinberger et al, 2014; Schlepckow et al, 2017; Thornton et al, 2017) is sufficient to abrogate cell-autonomous signaling, we hypothesized that elevating TREM2 levels on the cell surface would enhance potentially disease-modulating microglial functions. Indeed, inhibition of α-secretase activity with a protease inhibitor enhanced the phagocytic capacity of microglial-like BV2 cells (Kleinberger et al, 2014). However, α-secretase competes with the amyloidogenic pathway and limits Aβ production (Haass & Selkoe, 1993); therefore, its inhibition in AD would be not be efficacious. Moreover, a disintegrin and metalloproteinase (ADAM, i.e., α-secretase) 10 and ADAM17 cleave numerous substrates within the brain, many of which stimulate essential physiological signaling pathways (Kuhn et al, 2016). We therefore sought to selectively inhibit TREM2 cleavage without affecting shedding of other ADAM10/17 substrates. We and others have previously identified the cleavage site of TREM2 after His 157 within the stalk region of TREM2 (Feuerbach et al, 2017; Schlepckow et al, 2017; Thornton et al, 2017) (Fig 1A). In this study, we screened for monoclonal antibodies binding to the stalk region encompassing the cleavage site with the purpose of stabilizing membrane-bound TREM2 and selectively enhancing TREM2-dependent protective functions in microglia. Results Screening and molecular characterization of anti-TREM2 antibodies To identify antibodies capable of increasing TREM2 on the cell surface, we raised monoclonal antibodies to the recombinantly expressed ectodomain of mouse TREM2 (amino acids 19–171; Fig 1A). Among the antibodies tested, 4D9, a rat IgG2a antibody, was selected for additional studies based on its ability to significantly enhance cell membrane TREM2 levels and to reduce sTREM2 levels similar to an ADAM protease inhibitor (Fig 1B and C). Immunohistochemistry showed that the 4D9 antibody bound to the cell surface of mouse TREM2-expressing HEK293 cells, while empty vector-expressing control cells did not show staining (Fig 1D). Antigen mapping using tiling peptides along the stalk region of TREM2 revealed specific binding of 4D9 to N-DAGDLWVPE, a 9-amino acid peptide N-terminal to the ADAM cleavage site (Fig 1E). Although this epitope shows substantial sequence similarities to human TREM2 (Fig 1F), 4D9 did not detect recombinant human TREM2 by Western blotting (Fig 1F). Furthermore, consistent with a lack of sequence conservation of the 9-amino acid region between mouse TREM1 and TREM2, 4D9 binding to mouse TREM1 was not detected (Fig 1F), demonstrating that 4D9 is indeed selective for mouse TREM2. To further confirm specific binding of 4D9 to the stalk region epitope, peptide competition experiments were performed. This demonstrated efficient competition of a TREM2 stalk peptide for 4D9 binding to the extracellular protein (Fig 1G). Antibody–antigen affinity was determined by surface plasmon resonance (SPR) demonstrating a monovalent KD of 68pM for mouse TREM2 (Fig 1H). Consistent with the Western blot data (see Fig 1F), human TREM2 and mouse TREM1 did not result in any SPR response (Fig 1H). High-affinity cell-surface TREM2 binding was established for 4D9 using HEK293 cells stably expressing TREM2. A dose response demonstrated a cell binding EC50 of 0.29 nM, while an isotype control antibody did not show measurable binding (Fig 2A). Next, we generated Fab fragments of 4D9, which specifically bound cell-surface TREM2 on HEK293 cells with an EC50 of 0.17 nM (Fig 2A). In vitro peptide cleavage assays using recombinant ADAM17 revealed that the full-length 4D9 antibody, as well as 4D9 Fab, significantly blocked cleavage of a TREM2 stalk peptide substrate (Fig 2B). Thus, 4D9 appears to sterically hinder access of ADAM17 to its substrate. However, in a cell-based assay, only full-length IgG 4D9 antibody, but not 4D9 Fab, potently reduced shedding of TREM2 in a dose-dependent manner with an EC50 of 2.3 nM (Fig 2C). Given that 4D9 reduced shedding and enhanced cell-surface levels of full-length TREM2, we next evaluated the effects on downstream signaling. We therefore investigated p-SYK activity in the presence or absence of 4D9 and an isotype control in HEK293 cells expressing mouse TREM2 and its signaling adapter DAP12. This revealed a dose-dependent increase in p-SYK upon addition of 4D9 but not 4D9 Fab to the culture media of the cells (Fig 2D). Furthermore, anionic liposome ligand (Shirotani et al, 2019) stimulated TREM2 signaling 2.3-fold by full-length 4D9 but not by 4D9 Fab or the isotype control (Fig 2E). These data therefore suggest that the monoclonal antibody 4D9 enhances TREM2-dependent signaling via bivalent binding which cross-links TREM2 on the plasma membrane. Bivalent TREM2 binding is required for both signaling and shedding blocking mechanisms of TREM2 antibody function, potentially as a result of the inability of proteases to accept dimeric substrates (Fig 2F). Figure 2. 4D9 antibody stimulates TREM2-dependent SYK signaling and blocks ADAM17-mediated TREM2 shedding Flow cytometry dose–response curve for cell binding of 4D9 mAb (EC50 = 0.29 nM), 4D9 Fab (EC50 = 0.17 nM), and isotype to HEK cells stably overexpressing mouse TREM2. Data represent the mean ± SEM (n = 2). In vitro ADAM17 sheddase activity is blocked by 4D9-effectorless mAb and 4D9 Fab fragment but not an isotype control. Fluorescence polarization of FAM-conjugated TREM2 stalk peptide was detected in the presence or absence of ADAM17 and 4D9 mAb, 4D9 Fab, and isotype control. Data represent the mean ± SEM (n = 6). One-way ANOVA, Tukey's post hoc test; P (4D9 Fab vs 4D9 mAb) = 0.8855; P (4D9 Fab vs uncleaved) < 0.0001; P (4D9 mAb vs uncleaved) < 0.0001; n.s., not significant. ELISA-mediated quantification of sTREM2 in conditioned media from HEK293 cells stably overexpressing mouse TREM2 treated with a dose titration of 4D9 mAb (EC50 = 2.3 nM), 4D9 Fab, or isotype for 18 h. Data represent the mean ± SEM (n = 3). AlphaLISA-mediated quantification of p-SYK levels in HEK293 Flp-In cells stably overexpressing mouse TREM2 and mouse DAP12 treated with a dose titration of 4D9 mAb, 4D9 Fab, or isotype for 5 min. Data represent the mean ± SEM (n = 3). AlphaLISA-mediated quantification of p-SYK levels in HEK293 Flp-In cells stably overexpressing mouse TREM2 and mouse DAP12 or empty vector treated with 1 mg/ml POPC/POPS liposomes and 20 μg/ml 4D9 mAb, 4D9 Fab, or isotype for 5 min. p-SYK levels were also determined for cells treated with liposomes only. Data represent the mean ± SEM (n = 3). Two-way ANOVA, Tukey's post hoc test (cell line effect: F1,16 = 365.7, P ≤ 0.0001; treatment effect: F3,16 = 39.35, P < 0.0001; cell line × treatment effect: F3,16 = 38.75, P < 0.0001); P (no Ab vs isotype) = 0.6218; P (no Ab vs 4D9 mAb) < 0.0001; P (no Ab vs 4D9 Fab) = 0.7301; P (isotype vs 4D9 mAb) < 0.0001; P (isotype vs 4D9 Fab) > 0.9999; P (4D9 mAb vs 4D9 Fab) < 0.0001; n.s., not significant. Schematic representation of the proposed mechanism of action of antibody 4D9. Binding of 4D9 to TREM2 leads to receptor clustering on the cell surface, thereby driving downstream p-SYK signaling. At the same time, cell-surface levels are enhanced by inhibition of ectodomain shedding, potentially because of the inability of proteases to cleave dimeric substrates. Data information: Statistical evaluations are displayed as follows: ***P < 0.001; ****P < 0.0001. Download figure Download PowerPoint 4D9 antibody modulates TREM2 function in primary macrophages and microglia We next investigated whether TREM2-dependent functions of myeloid cells can be modulated by 4D9. For these studies, 4D9 was sequenced and reformatted onto an effectorless human IgG1 backbone carrying mutations that abolish effector function to avoid confounds of off-target impact of FcR interactions on primary myeloid cells (for details, see Materials and Methods). To demonstrate that 4D9 binds endogenous cell-surface TREM2 in myeloid cells, bone marrow-derived macrophages (BMDMs) were incubated with biotinylated 4D9 or isotype control antibody. Cell-surface