4.6 Article

Disrupted NOS2 metabolism drives myoblast response to wasting-associated cytokines

期刊

EXPERIMENTAL CELL RESEARCH
卷 407, 期 1, 页码 -

出版社

ELSEVIER INC
DOI: 10.1016/j.yexcr.2021.112779

关键词

NOS2; Nitric oxide; Myoblast; Cancer cachexia; TNF alpha; IFN gamma

资金

  1. National Institutes of Health [R00AR66696, R35GM128594]
  2. Mayo Clinic start-up funds
  3. Mayo Clinic SPORE in Pancreatic Cancer (NIH/National Cancer Institute (NCI)) [CA102701]
  4. American Association for Cancer Research/Pancreatic Cancer Action Network
  5. Glenn Foundation for Medical Research
  6. Mayo Clinic Regenerative Sciences Training Program (RSTP)
  7. Mayo Clinic Metabolomics Resource Core through NIH/National Institute of Diabetes and Digestive and Kidney Disease (NIDDK) from the NIH Director's Common Fund [U24DK10049]

向作者/读者索取更多资源

Skeletal muscle wasting is a key driver of negative clinical outcomes and is associated with a range of pathologies including cancer. Cancer cachexia is a multi-factorial syndrome that includes skeletal muscle wasting, but remains understudied. Research suggests that amino acid metabolic disruption is linked to myoblast defects induced by cancer cell conditioned media.
Skeletal muscle wasting drives negative clinical outcomes and is associated with a spectrum of pathologies including cancer. Cancer cachexia is a multi-factorial syndrome that encompasses skeletal muscle wasting and remains understudied, despite being a frequent and serious co-morbidity. Deviation from the homeostatic balance between breakdown and regeneration leads to muscle wasting disorders, such as cancer cachexia. Muscle stem cells (MuSCs) are the cellular compartment responsible for muscle regeneration, which makes MuSCs an intriguing target in the context of wasting muscle. Molecular studies investigating MuSCs and skeletal muscle wasting largely focus on transcriptional changes, but our group and others propose that metabolic changes are another layer of cellular regulation underlying MuSC dysfunction in cancer cachexia. In the present study, we combined gene expression and non-targeted metabolomic profiling of myoblasts exposed to wasting conditions (cancer cell conditioned media, CC-CM) to derive a more complete picture of the myoblast response to wasting factors. After mapping these features to annotated pathways, we found that more than half of the mapped pathways were amino acid-related, linking global amino acid metabolic disruption to conditioned media-induced myoblast defects. Notably, arginine metabolism was a highly enriched pathway in combined metabolomic and transcriptomic data. Arginine catabolism generates nitric oxide (NO), an important signaling molecule known to have negative effects on mature muscle. We hypothesize that tumor-derived disruptions in Nitric Oxide Synthase (NOS)2-regulated arginine catabolism impair differentiation of MuSCs. The work presented here further investigates the effect of NOS2 overactivity on myoblast proliferation and differentiation. We show that NOS2 inhibition is sufficient to rescue wasting phenotypes associated with inflammatory cytokines. Ultimately, this work provides new insights into MuSC biology and opens up potential therapeutic avenues for addressing disrupted MuSC dynamics in cancer cachexia.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据