Loss of adult skeletal muscle stem cells drives age-related neuromuscular junction degeneration

  1. Wenxuan Liu
  2. Alanna Klose
  3. Sophie Forman
  4. Nicole D Paris
  5. Lan Wei-LaPierre
  6. Mariela Cortés-Lopéz
  7. Aidi Tan
  8. Morgan Flaherty
  9. Pedro Miura
  10. Robert T Dirksen
  11. Joe V Chakkalakal  Is a corresponding author
  1. University of Rochester Medical Center, United States
  2. University of Rochester, United States
  3. University of Nevada, United States
  4. Tsinghua University, China

Abstract

Neuromuscular junction degeneration is a prominent aspect of sarcopenia, the age-associated loss of skeletal muscle integrity. Previously, we showed that muscle stem cells activate and contribute to mouse neuromuscular junction regeneration in response to denervation (1). Here, we examined gene expression profiles and neuromuscular junction integrity in aged mouse muscles, and unexpectedly found limited denervation despite a high level of degenerated neuromuscular junctions. Instead, degenerated neuromuscular junctions were associated with reduced contribution from muscle stem cells. Indeed, muscle stem cell depletion was sufficient to induce neuromuscular junction degeneration at a younger age. Conversely, prevention of muscle stem cell and derived myonuclei loss was associated with attenuation of age-related neuromuscular junction degeneration, muscle atrophy, and the promotion of aged muscle force generation. Our observations demonstrate that deficiencies in muscle stem cell fate and post-synaptic myogenesis provide a cellular basis for age-related neuromuscular junction degeneration and associated skeletal muscle decline.

Data availability

The following data sets were generated

Article and author information

Author details

  1. Wenxuan Liu

    Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  2. Alanna Klose

    Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  3. Sophie Forman

    Department of Biology, University of Rochester, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Nicole D Paris

    Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0654-0983
  5. Lan Wei-LaPierre

    Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Mariela Cortés-Lopéz

    Department of Biology, University of Nevada, Reno, United States
    Competing interests
    The authors declare that no competing interests exist.
  7. Aidi Tan

    Bioinformatics Division and Center for Synthetic and Systems Biology, TNLIST/Department of Automation, Tsinghua University, Beijing, China
    Competing interests
    The authors declare that no competing interests exist.
  8. Morgan Flaherty

    Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Pedro Miura

    Department of Biology, University of Nevada, Reno, United States
    Competing interests
    The authors declare that no competing interests exist.
  10. Robert T Dirksen

    Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
    Competing interests
    The authors declare that no competing interests exist.
  11. Joe V Chakkalakal

    Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, United States
    For correspondence
    joe_chakkalakal@urmc.rochester.edu
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8440-7312

Funding

National Institute on Aging (RO1AG051456)

  • Joe V Chakkalakal

Congressionally Directed Medical Research Programs (W81XWH-14-1-0454)

  • Joe V Chakkalakal

NYSTEM (C026877)

  • Wenxuan Liu

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

Reviewing Editor

  1. Amy J Wagers, Harvard University, United States

Ethics

Animal experimentation: Animal experimentation: This study was performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. Work with mice was conducted in accordance with protocols approved by the University Committee on Animal Resources, University of Rochester Medical Center protocol(#101565/2013-002)

Version history

  1. Received: March 3, 2017
  2. Accepted: May 20, 2017
  3. Accepted Manuscript published: June 6, 2017 (version 1)
  4. Version of Record published: June 7, 2017 (version 2)

Copyright

© 2017, Liu 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

  • 6,165
    views
  • 1,007
    downloads
  • 113
    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. Wenxuan Liu
  2. Alanna Klose
  3. Sophie Forman
  4. Nicole D Paris
  5. Lan Wei-LaPierre
  6. Mariela Cortés-Lopéz
  7. Aidi Tan
  8. Morgan Flaherty
  9. Pedro Miura
  10. Robert T Dirksen
  11. Joe V Chakkalakal
(2017)
Loss of adult skeletal muscle stem cells drives age-related neuromuscular junction degeneration
eLife 6:e26464.
https://doi.org/10.7554/eLife.26464

Share this article

https://doi.org/10.7554/eLife.26464

Further reading

    1. Stem Cells and Regenerative Medicine
    Jing-Ping Wang, Chun-Hao Hung ... C-K James Shen
    Research Article

    A causal relationship exists among the aging process, organ decay and disfunction, and the occurrence of various diseases including cancer. A genetically engineered mouse model, termed Klf1K74R/K74R or Klf1(K74R), carrying mutation on the well-conserved sumoylation site of the hematopoietic transcription factor KLF1/EKLF has been generated that possesses extended lifespan and healthy characteristics, including cancer resistance. We show that the healthy longevity characteristics of the Klf1(K74R) mice, as exemplified by their higher anti-cancer capability, are likely gender-, age-, and genetic background-independent. Significantly, the anti-cancer capability, in particular that against melanoma as well as hepatocellular carcinoma, and lifespan-extending property of Klf1(K74R) mice, could be transferred to wild-type mice via transplantation of their bone marrow mononuclear cells at a young age of the latter. Furthermore, NK(K74R) cells carry higher in vitro cancer cell-killing ability than wild-type NK cells. Targeted/global gene expression profiling analysis has identified changes in the expression of specific proteins, including the immune checkpoint factors PDCD and CD274, and cellular pathways in the leukocytes of the Klf1(K74R) that are in the directions of anti-cancer and/or anti-aging. This study demonstrates the feasibility of developing a transferable hematopoietic/blood system for long-term anti-cancer and, potentially, for anti-aging.

    1. Neuroscience
    2. Stem Cells and Regenerative Medicine
    Pascal Forcella, Niklas Ifflander ... Verdon Taylor
    Research Article

    Neural stem cells (NSCs) are multipotent and correct fate determination is crucial to guarantee brain formation and homeostasis. How NSCs are instructed to generate neuronal or glial progeny is not well understood. Here we addressed how murine adult hippocampal NSC fate is regulated and describe how Scaffold Attachment Factor B (SAFB) blocks oligodendrocyte production to enable neuron generation. We found that SAFB prevents NSC expression of the transcription factor Nuclear Factor I/B (NFIB) by binding to sequences in the Nfib mRNA and enhancing Drosha-dependent cleavage of the transcripts. We show that increasing SAFB expression prevents oligodendrocyte production by multipotent adult NSCs, and conditional deletion of Safb increases NFIB expression and oligodendrocyte formation in the adult hippocampus. Our results provide novel insights into a mechanism that controls Drosha functions for selective regulation of NSC fate by modulating the post-transcriptional destabilization of Nfib mRNA in a lineage-specific manner.