American Association for Cancer Research
Browse
crc-24-0045_fig1.png (1.04 MB)

FIGURE 1 from Mutant RAS-driven Secretome Causes Skeletal Muscle Defects in Breast Cancer

Download (1.04 MB)
figure
posted on 2024-05-15, 07:40 authored by Ruizhong Wang, Aditi S. Khatpe, Brijesh Kumar, Henry Elmer Mang, Katie Batic, Adedeji K. Adebayo, Harikrishna Nakshatri

Tumor subtypes differentially affect skeletal muscle function in mice with PDXs. A, Growth rate of PDXs in NSG mice. B, Histology of PDXs confirming their molecular subtype. C, Impaired rotarod performance of mice with PDXs across all tumor subtypes. D, Differential impact of tumors by subtype on grip strength of mice with PDXs. Mice with TNBC tumors had severe loss of grip strength. E, Loss of grip strength was improved by Curaxin (CBL0137) treatment in nude mice with tumors from MDA-MB-436 cells. F, Impaired rotarod performance was ameliorated by Curaxin (CBL0137) in nude mice with tumors from MDA-MB-436 cells.

Funding

U.S. Department of Veterans Affairs (VA)

Indiana University Precision Health Initiative

History

ARTICLE ABSTRACT

Cancer-induced skeletal muscle defects differ in severity between individuals with the same cancer type. Cancer subtype-specific genomic aberrations are suggested to mediate these differences, but experimental validation studies are very limited. We utilized three different breast cancer patient-derived xenograft (PDX) models to correlate cancer subtype with skeletal muscle defects. PDXs were derived from brain metastasis of triple-negative breast cancer (TNBC), estrogen receptor–positive/progesterone receptor–positive (ER+/PR+) primary breast cancer from a BRCA2-mutation carrier, and pleural effusion from an ER+/PR− breast cancer. While impaired skeletal muscle function as measured through rotarod performance and reduced levels of circulating and/or skeletal muscle miR-486 were common across all three PDXs, only TNBC-derived PDX activated phospho-p38 in skeletal muscle. To further extend these results, we generated transformed variants of human primary breast epithelial cells from healthy donors using HRASG12V or PIK3CAH1047R mutant oncogenes. Mutations in RAS oncogene or its modulators are found in approximately 37% of metastatic breast cancers, which is often associated with skeletal muscle defects. Although cells transformed with both oncogenes generated adenocarcinomas in NSG mice, only HRASG12V-derived tumors caused skeletal muscle defects affecting rotarod performance, skeletal muscle contraction force, and miR-486, Pax7, pAKT, and p53 levels in skeletal muscle. Circulating levels of the chemokine CXCL1 were elevated only in animals with tumors containing HRASG12V mutation. Because RAS pathway aberrations are found in 19% of cancers, evaluating skeletal muscle defects in the context of genomic aberrations in cancers, particularly RAS pathway mutations, may accelerate development of therapeutic modalities to overcome cancer-induced systemic effects. Mutant RAS- and PIK3CA-driven breast cancers distinctly affect the function of skeletal muscle. Therefore, research and therapeutic targeting of cancer-induced systemic effects need to take aberrant cancer genome into consideration.

Usage metrics

    Cancer Research Communications

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC