Precise let-7 expression levels balance organ regeneration against tumor suppression

  1. Linwei Wu
  2. Liem H Nguyen
  3. Kejin Zhou
  4. T Yvanka de Soysa
  5. Lin Li
  6. Jason B Miller
  7. Jianmin Tian
  8. Joseph Locker
  9. Shuyuan Zhang
  10. Gen Shinoda
  11. Marc T Seligson
  12. Lauren R Zeitels
  13. Asha Acharya
  14. Sam C Wang
  15. Joshua T Mendell
  16. Xiaoshun He
  17. Jinsuke Nishino
  18. Sean J Morrison
  19. Daniel J Siegwart
  20. George Q Daley
  21. Ng Shyh-Chang
  22. Hao Zhu  Is a corresponding author
  1. University of Texas Southwestern Medical Center, United States
  2. Boston Children's Hospital and Dana Farber Cancer Institute, United States
  3. University of Pittsburg, United States
  4. The First Affiliated Hospital of Sun Yat-Sen University, China
  5. Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, United States
  6. Harvard Medical School, United States
  7. Children's Hospital and Dana Farber Cancer Institute, United States

Abstract

The in vivo roles for even the most intensely studied microRNAs remain poorly defined. Here, analysis of mouse models revealed that let-7, a large and ancient microRNA family, performs tumor suppressive roles at the expense of regeneration. Too little or too much let-7 resulted in compromised protection against cancer or tissue damage, respectively. Modest let-7 overexpression abrogated MYC-driven liver cancer by antagonizing multiple let-7 sensitive oncogenes. However, the same level of overexpression blocked liver regeneration, while let-7 deletion enhanced it, demonstrating that distinct let-7 levels can mediate desirable phenotypes. let-7 dependent regeneration phenotypes resulted from influences on the insulin-PI3K-mTOR pathway. We found that chronic high-dose let-7 overexpression caused liver damage and degeneration, paradoxically leading to tumorigenesis. These dose-dependent roles for let-7 in tissue repair and tumorigenesis rationalize the tight regulation of this microRNA in development, and have important implications for let-7 based therapeutics.

Article and author information

Author details

  1. Linwei Wu

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  2. Liem H Nguyen

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  3. Kejin Zhou

    Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  4. T Yvanka de Soysa

    Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States
    Competing interests
    No competing interests declared.
  5. Lin Li

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  6. Jason B Miller

    Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  7. Jianmin Tian

    Department of Pathology, University of Pittsburg, Pittsburg, United States
    Competing interests
    No competing interests declared.
  8. Joseph Locker

    Department of Pathology, University of Pittsburg, Pittsburg, United States
    Competing interests
    No competing interests declared.
  9. Shuyuan Zhang

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  10. Gen Shinoda

    Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States
    Competing interests
    No competing interests declared.
  11. Marc T Seligson

    Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, United States
    Competing interests
    No competing interests declared.
  12. Lauren R Zeitels

    Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  13. Asha Acharya

    Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  14. Sam C Wang

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  15. Joshua T Mendell

    Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  16. Xiaoshun He

    Organ Transplant Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
    Competing interests
    No competing interests declared.
  17. Jinsuke Nishino

    Children's Medical Center Research Institute at UT Southwestern, Department of Pediatrics, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  18. Sean J Morrison

    Children's Medical Center Research Institute at UT Southwestern, Department of Pediatrics, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    Sean J Morrison, Senior editor, eLife.
  19. Daniel J Siegwart

    Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    No competing interests declared.
  20. George Q Daley

    Children's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, United States
    Competing interests
    No competing interests declared.
  21. Ng Shyh-Chang

    Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children's Hospital and Dana Farber Cancer Institute, Boston, United States
    Competing interests
    No competing interests declared.
  22. Hao Zhu

    Children's Research Institute, Departments of Pediatrics and Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
    For correspondence
    Hao.Zhu@utsouthwestern.edu
    Competing interests
    No competing interests declared.

Reviewing Editor

  1. Amy J Wagers, Harvard University, United States

Ethics

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. All of the animals were handled according to approved institutional animal care and use committee (IACUC) protocols (#2012-0143) of the University of Texas Southwestern Medical Center. All surgery was performed under isoflurane anesthesia with appropriate analgesia, and every effort was made to minimize suffering.

Version history

  1. Received: June 15, 2015
  2. Accepted: October 5, 2015
  3. Accepted Manuscript published: October 7, 2015 (version 1)
  4. Version of Record published: December 10, 2015 (version 2)

Copyright

© 2015, Wu 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

  • 3,128
    views
  • 908
    downloads
  • 47
    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. Linwei Wu
  2. Liem H Nguyen
  3. Kejin Zhou
  4. T Yvanka de Soysa
  5. Lin Li
  6. Jason B Miller
  7. Jianmin Tian
  8. Joseph Locker
  9. Shuyuan Zhang
  10. Gen Shinoda
  11. Marc T Seligson
  12. Lauren R Zeitels
  13. Asha Acharya
  14. Sam C Wang
  15. Joshua T Mendell
  16. Xiaoshun He
  17. Jinsuke Nishino
  18. Sean J Morrison
  19. Daniel J Siegwart
  20. George Q Daley
  21. Ng Shyh-Chang
  22. Hao Zhu
(2015)
Precise let-7 expression levels balance organ regeneration against tumor suppression
eLife 4:e09431.
https://doi.org/10.7554/eLife.09431

Share this article

https://doi.org/10.7554/eLife.09431

Further reading

    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.

    1. Cell Biology
    Simona Bolamperti, Hiroaki Saito ... Hanna Taipaleenmäki
    Research Article

    Osteoblast adherence to bone surfaces is important for remodeling bone tissue. This study demonstrates that deficiency of TG-interacting factor 1 (Tgif1) in osteoblasts results in altered cell morphology, reduced adherence to collagen type I-coated surfaces, and impaired migration capacity. Tgif1 is essential for osteoblasts to adapt a regular cell morphology and to efficiently adhere and migrate on collagen type I-rich matrices in vitro. Furthermore, Tgif1 acts as a transcriptional repressor of p21-activated kinase 3 (Pak3), an important regulator of focal adhesion formation and osteoblast spreading. Absence of Tgif1 leads to increased Pak3 expression, which impairs osteoblast spreading. Additionally, Tgif1 is implicated in osteoblast recruitment and activation of bone surfaces in the context of bone regeneration and in response to parathyroid hormone 1–34 (PTH 1–34) treatment in vivo in mice. These findings provide important novel insights in the regulation of the cytoskeletal architecture of osteoblasts.