Malatl fine-tunes bone homeostasis by orchestrating cellular crosstalk and the β-catenin-OPG/Jagged1 pathway

  1. Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA.
  2. Department of Medicine, Weill Cornell Medical College, New York, New York, USA.
  3. Department of Cell and Developmental Biology, Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
  4. Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Strasse 42-44, 60596 Frankfurt, Germany
  5. RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
  6. Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, USA
  7. Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA.

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Dolores Shoback
    University of California, San Francisco, San Francisco, United States of America
  • Senior Editor
    Dolores Shoback
    University of California, San Francisco, San Francisco, United States of America

Reviewer #1 (Public Review):

Summary

The authors were trying to discover a novel bone remodeling network system. They found that an IncRNA Malat1 plays a central role in the remodeling by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In addition, Malat1 significantly promotes bone regeneration in fracture healing in vivo. Their findings suggest a new concept of Malat1 function in the skeletal system. One significantly different finding between this manuscript and the competing paper pertains to the role of Malat1 in osteoclast lineage, specifically, whether Malat1 functions intrinsically in osteoclast lineage or not.

Strengths:

This study provides strong genetic evidence demonstrating that Malat1 acts intrinsically in osteoblasts while suppressing osteoclastogenesis in a non-autonomous manner, whereas the other group did not utilize relevant conditional knockout mice. As shown in the results, Malat1 knockout mouse exhibited abnormal bone remodeling and turnover. Furthermore, they elucidated molecular function of Malat1, which is sufficient to understand the phenotype in vivo.

Weaknesses:

Discussing differences between previous paper and their status would be highly informative and beneficial for the field, as it would elucidate the solid underlying mechanisms.

Reviewer #2 (Public Review):

Summary:

The authors investigated the roles of IncRNA Malat1 in bone homeostasis which was initially believed to be non-functional for physiology. They found that both Malat1 KO and conditional KO in osteoblast lineage exhibit significant osteoporosis due to decreased osteoblast bone formation and increased osteoclast resorption. More interestingly they found that deletion of Malat1 in osteoclast lineage cells does not affect osteoclast differentiation and function. Mechanistically, they found that Malat1 acts as a co-activator of b-Catenin directly regulating osteoblast activity and indirectly regulating osteoclast activity via mediating OPG, but not RANKL expression in osteoblast and chondrocyte. Their discoveries establish a previously unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine-tune tissue homeostasis, and remodeling.

Strengths:

The authors generated global and conditional KO mice in osteoblast and osteoclast lineage cells and carefully analyzed the role of Matat1 with both in vivo and in vitro systems. The conclusion of this paper is mostly well supported by data.

Weaknesses:

More objective biological and biochemical analyses are required.

Reviewer #3 (Public Review):

Summary:

In this manuscript, Qin and colleagues study the role of Malat1 in bone biology. This topic is interesting given the role of lncRNAs in multiple physiologic processes. A previous study (PMID 38493144) suggested a role for Malat1 in osteoclast maturation. However, the role of this lncRNA in osteoblast biology was previously not explored. Here, the authors note osteopenia with increased bone resorption in mice lacking Malat1 globally and in osteoblast lineage cells. At the mechanistic level, the authors suggest that Malat1 controls beta-catenin activity. These results advance the field regarding the role of this lncRNA in bone biology.

Strengths:

The manuscript is well-written and data are presented in a clear and easily understandable manner. The bone phenotype of osteoblast-specific Malat1 knockout mice is of high interest. The role of Malat1 in controlling beta-catenin activity and OPG expression is interesting and novel.

Weaknesses:

The lack of a bone phenotype when Malat1 is deleted with LysM-Cre is of interest given the previous report suggesting a role for this lncRNA in osteoclasts. However, to interpret the findings here, the authors should investigate the deletion efficiency of Malat1 in osteoclast lineage cells in their model. The data in the fracture model in Figure 8 seems incomplete in the absence of a more complete characterization of callus histology and a thorough time course. The role of Malat1 and OPG in chondrocytes is unclear since the osteocalcin-Cre mice (which should retain normal Malat1 levels in chondrocytes) have similar bone loss as the global mutants.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation