ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Please note that you are viewing an archived section from 2020 and some content may be unavailable. To unlock all content for 2020, please visit the archives.

Abstract: PO2239

Efficacy of Low-Intensity Pulsed Ultrasound on CKD-Associated Cachexia and Muscle Wasting Prevention in a Mouse Model

Session Information

Category: Pathology and Lab Medicine

  • 1601 Pathology and Lab Medicine: Basic

Authors

  • Liu, Shing-Hwa, Institute of Toxicology, National Taiwan University, Taipei, Taiwan
  • Hung, Kuan-Yu, Superintendent Office, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
  • Chiang, Chih-Kang, Institute of Toxicology, National Taiwan University, Taipei, Taiwan
Background

Low-intensity pulsed ultrasound (LIPUS), a therapeutic ultrasound, is recognized to elevate the bone fracture repair process and help in some soft tissue healing. Here, we tested the prevention of chronic kidney disease (CKD)-associated cachexia and sarcopenia by LIPUS in a renal ischemia/reperfusion injury (IRI) mouse model.

Methods

Adult C57BL/6J male mice were used. A model of unilateral IRI with nephrectomy of the contralateral kidney with or without LIPUS treatment (3 MHz, 0.1 W/cm2, 20 minutes/day) 5 days before and 14 days after surgery was performed. The CKD-related cachexia/muscle wasting in mice was evaluated. Mice were euthanized 14 days after IRI.

Results

LIPUS treatment significantly alleviated the decrease in the serum albumin/globulin (A/G) ratio and the increases in the serum levels of blood urea nitrogen (BUN), creatinine, cystatin C, and fibroblast growth factor (FGF)-23, and the renal pathological changes and fibrosis in CKD mice (p<0.05, n=8; for A/G ratio and FGF-23). The development of epithelial-mesenchymal transition and the induction of senescence-related molecular signals and the decreased protein expressions of α-Klotho and endogenous antioxidant enzymes in the kidneys of CKD mice were significantly alleviated by LIPUS treatment (p<0.05, n=4). LIPUS treatment could also significantly reverse the decreased muscle mass, grip strength, and cross-section areas (CSA) of muscle fibers (p<0.05, n=8; for soleus muscle weight, hindlimb grip strength), and the increased muscular protein expressions of atrogenes, Atrogin1 and MuRF1, and phosphorylated AMP-activated protein kinase (AMPK), and the decreased muscular protein expressions of phosphorylated Akt, peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC-1α), and mitochondrially encoded cytochrome c oxidase I (MT-CO1) in CKD mice (p<0.05, n=5 or 6).

Conclusion

LIPUS treatment showed the benefits for renal and muscular protection in a CKD mouse model via inhibition of renal fibrosis, restoration of antioxidant enzymes, and attenuation renal senescence/aging, and muscle mass loss via prevention of muscular mitochondrial dysfunction, AMPK activation, and Akt downregulation. LIPUS treatment may be potentially applied to an alternative non-invasive therapeutic intervention on CKD-associated cachexia/muscle wasting therapy.