MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis

  1. Alireza Ghasemizadeh
  2. Emilie Christin
  3. Alexandre Guiraud
  4. Nathalie Couturier
  5. Marie Abitbol
  6. Valerie Risson
  7. Emmanuelle Girard
  8. Krzysztof Jagla
  9. Cedric Soler
  10. lilia Laddada
  11. Colline Sanchez
  12. Francisco-Ignacio Jaque-Fernandez
  13. Vincent Jacquemond
  14. Jean-Luc Thomas
  15. Marine Lanfranchi
  16. Julien Courchet
  17. Julien Gondin
  18. Laurent Schaeffer
  19. Vincent Gache  Is a corresponding author
  1. INMG, France
  2. GReD Laboratory/Clermont-Auvergne University, France
  3. Institut Génétique Reproduction et Développement, France
  4. Centre de Génétique et de Physiologie Moléculaire et Cellulaire, France

Abstract

Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.

Data availability

All data generated or analysed during this study are included in the manuscript and supporting files. Source data files have been provided for all Figures

Article and author information

Author details

  1. Alireza Ghasemizadeh

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8202-8546
  2. Emilie Christin

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  3. Alexandre Guiraud

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  4. Nathalie Couturier

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  5. Marie Abitbol

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  6. Valerie Risson

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, LYON, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-6812-6297
  7. Emmanuelle Girard

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, LYON, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-6730-9596
  8. Krzysztof Jagla

    GReD Laboratory/Clermont-Auvergne University, Clermont-Ferrandf, France
    Competing interests
    The authors declare that no competing interests exist.
  9. Cedric Soler

    Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-2312-5370
  10. lilia Laddada

    GReD Laboratory/Clermont-Auvergne University, Clermont-Ferrandf, France
    Competing interests
    The authors declare that no competing interests exist.
  11. Colline Sanchez

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  12. Francisco-Ignacio Jaque-Fernandez

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, LYON, France
    Competing interests
    The authors declare that no competing interests exist.
  13. Vincent Jacquemond

    Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  14. Jean-Luc Thomas

    MNCA team, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  15. Marine Lanfranchi

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, LYON, France
    Competing interests
    The authors declare that no competing interests exist.
  16. Julien Courchet

    INSERM U1217, CNRS UMR5310, Université Claude Bernard Lyon I, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-1199-9329
  17. Julien Gondin

    INSERM U1217, CNRS UMR5310, Université Claude Bernard Lyon I, INMG, Lyon, France
    Competing interests
    The authors declare that no competing interests exist.
  18. Laurent Schaeffer

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, LYON, France
    Competing interests
    The authors declare that no competing interests exist.
  19. Vincent Gache

    INSERM U1217,CNRS UMR5310, Université Claude Bernard Lyon I, INMG, Lyon, France
    For correspondence
    vincent.gache@inserm.fr
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-2928-791X

Funding

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

Reviewing Editor

  1. Mohammed Akaaboune, University of Michigan, United States

Ethics

Animal experimentation: All of the experiments and procedures were conducted in accordance with the guidelines of the local animal ethics committee of the University Claude Bernard - Lyon 1 and in accordance with French and European legislation on animal experimentation and approved by the ethics committee CECCAPP (ref APAFIS#17455-2018091216033835v5) and the French ministry of research.

Version history

  1. Preprint posted: May 15, 2019 (view preprint)
  2. Received: May 18, 2021
  3. Accepted: August 10, 2021
  4. Accepted Manuscript published: August 27, 2021 (version 1)
  5. Version of Record published: October 8, 2021 (version 2)

Copyright

© 2021, Ghasemizadeh 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

  • 2,378
    views
  • 448
    downloads
  • 6
    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. Alireza Ghasemizadeh
  2. Emilie Christin
  3. Alexandre Guiraud
  4. Nathalie Couturier
  5. Marie Abitbol
  6. Valerie Risson
  7. Emmanuelle Girard
  8. Krzysztof Jagla
  9. Cedric Soler
  10. lilia Laddada
  11. Colline Sanchez
  12. Francisco-Ignacio Jaque-Fernandez
  13. Vincent Jacquemond
  14. Jean-Luc Thomas
  15. Marine Lanfranchi
  16. Julien Courchet
  17. Julien Gondin
  18. Laurent Schaeffer
  19. Vincent Gache
(2021)
MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis
eLife 10:e70490.
https://doi.org/10.7554/eLife.70490

Share this article

https://doi.org/10.7554/eLife.70490

Further reading

    1. Cell Biology
    Yoko Nakai-Futatsugi, Jianshi Jin ... Masayo Takahashi
    Research Article

    Retinal pigment epithelium (RPE) cells show heterogeneous levels of pigmentation when cultured in vitro. To know whether their color in appearance is correlated with the function of the RPE, we analyzed the color intensities of human-induced pluripotent stem cell-derived RPE cells (iPSC-RPE) together with the gene expression profile at the single-cell level. For this purpose, we utilized our recent invention, Automated Live imaging and cell Picking System (ALPS), which enabled photographing each cell before RNA-sequencing analysis to profile the gene expression of each cell. While our iPSC-RPE were categorized into four clusters by gene expression, the color intensity of iPSC-RPE did not project any specific gene expression profiles. We reasoned this by less correlation between the actual color and the gene expressions that directly define the level of pigmentation, from which we hypothesized the color of RPE cells may be a temporal condition not strongly indicating the functional characteristics of the RPE.

    1. Cancer Biology
    2. Cell Biology
    Savvas Nikolaou, Amelie Juin ... Laura M Machesky
    Research Article

    Pancreatic ductal adenocarcinoma carries a dismal prognosis, with high rates of metastasis and few treatment options. Hyperactivation of KRAS in almost all tumours drives RAC1 activation, conferring enhanced migratory and proliferative capacity as well as macropinocytosis. Macropinocytosis is well understood as a nutrient scavenging mechanism, but little is known about its functions in trafficking of signaling receptors. We find that CYRI-B is highly expressed in pancreatic tumours in a mouse model of KRAS and p53-driven pancreatic cancer. Deletion of Cyrib (the gene encoding CYRI-B protein) accelerates tumourigenesis, leading to enhanced ERK and JNK-induced proliferation in precancerous lesions, indicating a potential role as a buffer of RAC1 hyperactivation in early stages. However, as disease progresses, loss of CYRI-B inhibits metastasis. CYRI-B depleted tumour cells show reduced chemotactic responses to lysophosphatidic acid, a major driver of tumour spread, due to impaired macropinocytic uptake of the lysophosphatidic acid receptor-1. Overall, we implicate CYRI-B as a mediator of growth and signaling in pancreatic cancer, providing new insights into pathways controlling metastasis.