Heparan sulfate-dependent RAGE oligomerization is indispensable for pathophysiological functions of RAGE

  1. Miaomiao Li
  2. Chih Yean Ong
  3. Christophe J Langouët-Astrié
  4. Lisi Tan
  5. Ashwni Verma
  6. Yimu Yang
  7. Xiaoxiao Zhang
  8. Dhaval K Shah
  9. Eric P Schmidt
  10. Ding Xu  Is a corresponding author
  1. University at Buffalo, State University of New York, United States
  2. University of Colorado Anschutz Medical Campus, United States
  3. China Medical University, China

Abstract

RAGE, a druggable inflammatory receptor, is known to function as an oligomer but the exact oligomerization mechanism remains poorly understood. Previously we have shown that heparan sulfate (HS) plays an active role in RAGE oligomerization. To understand the physiological significance of HS-induced RAGE oligomerization in vivo, we generated RAGE knock-in mice (AgerAHA/AHA) by introducing point mutations to specifically disrupt HS-RAGE interaction. The RAGE mutant demonstrated normal ligand-binding but impaired capacity of HS-binding and oligomerization. Remarkably, AgerAHA/AHA mice phenocopied Ager-/- mice in two different pathophysiological processes, namely bone remodeling and neutrophil-mediated liver injury, which demonstrates that HS-induced RAGE oligomerization is essential for RAGE signaling. Our findings suggest that it should be possible to block RAGE signaling by inhibiting HS-RAGE interaction. To test this, we generated a monoclonal antibody that targets the HS-binding site of RAGE. This antibody blocks RAGE signaling in vitro and in vivo, recapitulating the phenotype of AgerAHA/AHA mice. By inhibiting HS-RAGE interaction genetically and pharmacologically, our work validated an alternative strategy to antagonize RAGE. Finally, we have performed RNA-seq analysis of neutrophils and lungs and found that while Ager -/- mice had a broad alteration of transcriptome in both tissues compared to wild-type mice, the changes of transcriptome in AgerAHA/AHA mice were much more restricted. This unexpected finding suggests that by preserving the expression of RAGE protein (in a dominant-negative form), AgerAHA/AHA mouse might represent a cleaner genetic model to study physiological roles of RAGE in vivo compared to Ager -/- mice.

Data availability

PMN and lung RNA-sequencing data have been deposited into the NCBI Gene Expression Omnibus database (accession number GSE174178). All data generated or analysed during this study are included in the manuscript and supporting files. Source data files have been provided for all the main Figures 1-7, Figure S1, S2, S3, S5 and S7.

The following data sets were generated

Article and author information

Author details

  1. Miaomiao Li

    Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, United States
    Competing interests
    Miaomiao Li, is one of the inventors for an international patent (pending, WO 2021/087462) that covers the sequence and use of anti-RAGE mAb B2..
  2. Chih Yean Ong

    Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, United States
    Competing interests
    No competing interests declared.
  3. Christophe J Langouët-Astrié

    Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, United States
    Competing interests
    No competing interests declared.
  4. Lisi Tan

    Department of Periodontics, China Medical University, Shenyang, China
    Competing interests
    No competing interests declared.
  5. Ashwni Verma

    Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3717-0233
  6. Yimu Yang

    Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, United States
    Competing interests
    No competing interests declared.
  7. Xiaoxiao Zhang

    Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-1321-0798
  8. Dhaval K Shah

    Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, United States
    Competing interests
    No competing interests declared.
  9. Eric P Schmidt

    Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, United States
    Competing interests
    No competing interests declared.
  10. Ding Xu

    Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, United States
    For correspondence
    dingxu@buffalo.edu
    Competing interests
    Ding Xu, is one of the inventors for an international patent (pending, WO 2021/087462) that covers the sequence and use of anti-RAGE mAb B2.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-9380-2712

Funding

National Institute of Arthritis and Musculoskeletal and Skin Diseases (R01AR07017)

  • Ding Xu

National Heart, Lung, and Blood Institute (R01HL094463)

  • Ding Xu

Buffalo Accelerator Funds

  • Ding Xu

National Institute of General Medical Sciences (R01GM125095)

  • Eric P Schmidt

National Institute of General Medical Sciences (R01GM114179)

  • Dhaval K Shah

National Institute of Allergy and Infectious Diseases (R21AI138195)

  • Dhaval K Shah

National Cancer Institute (R01CA246785)

  • Dhaval K Shah

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Dolores Shoback, University of California, San Francisco, United States

Ethics

Animal experimentation: All animal works in this study have been approved by the institutional animal care and use committee of the University at Buffalo (protocol number: ORB14126N and ORB18018).

Version history

  1. Preprint posted: June 17, 2021 (view preprint)
  2. Received: June 18, 2021
  3. Accepted: February 1, 2022
  4. Accepted Manuscript published: February 9, 2022 (version 1)
  5. Version of Record published: February 22, 2022 (version 2)

Copyright

© 2022, Li et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 759
    views
  • 131
    downloads
  • 4
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Miaomiao Li
  2. Chih Yean Ong
  3. Christophe J Langouët-Astrié
  4. Lisi Tan
  5. Ashwni Verma
  6. Yimu Yang
  7. Xiaoxiao Zhang
  8. Dhaval K Shah
  9. Eric P Schmidt
  10. Ding Xu
(2022)
Heparan sulfate-dependent RAGE oligomerization is indispensable for pathophysiological functions of RAGE
eLife 11:e71403.
https://doi.org/10.7554/eLife.71403

Share this article

https://doi.org/10.7554/eLife.71403

Further reading

    1. Cell Biology
    2. Neuroscience
    Marcos Moreno-Aguilera, Alba M Neher ... Carme Gallego
    Research Article Updated

    Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.

    1. Cell Biology
    Ang Li, Jianxun Yi ... Jingsong Zhou
    Research Article

    Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disorder characterized by progressive weakness of almost all skeletal muscles, whereas extraocular muscles (EOMs) are comparatively spared. While hindlimb and diaphragm muscles of end-stage SOD1G93A (G93A) mice (a familial ALS mouse model) exhibit severe denervation and depletion of Pax7+satellite cells (SCs), we found that the pool of SCs and the integrity of neuromuscular junctions (NMJs) are maintained in EOMs. In cell sorting profiles, SCs derived from hindlimb and diaphragm muscles of G93A mice exhibit denervation-related activation, whereas SCs from EOMs of G93A mice display spontaneous (non-denervation-related) activation, similar to SCs from wild-type mice. Specifically, cultured EOM SCs contain more abundant transcripts of axon guidance molecules, including Cxcl12, along with more sustainable renewability than the diaphragm and hindlimb counterparts under differentiation pressure. In neuromuscular co-culture assays, AAV-delivery of Cxcl12 to G93A-hindlimb SC-derived myotubes enhances motor neuron axon extension and innervation, recapitulating the innervation capacity of EOM SC-derived myotubes. G93A mice fed with sodium butyrate (NaBu) supplementation exhibited less NMJ loss in hindlimb and diaphragm muscles. Additionally, SCs derived from G93A hindlimb and diaphragm muscles displayed elevated expression of Cxcl12 and improved renewability following NaBu treatment in vitro. Thus, the NaBu-induced transcriptomic changes resembling the patterns of EOM SCs may contribute to the beneficial effects observed in G93A mice. More broadly, the distinct transcriptomic profile of EOM SCs may offer novel therapeutic targets to slow progressive neuromuscular functional decay in ALS and provide possible ‘response biomarkers’ in pre-clinical and clinical studies.