Muscle fibrosis and maladaptation occur progressively in CKD and are rescued by dialysis

Camille R Brightwell, Ameya S Kulkarni, William Paredes, Kehao Zhang, Jaclyn B Perkins, Knubian J Gatlin, Matthew Custodio, Hina Farooq, Bushra Zaidi, Rima Pai, Rupinder S Buttar, Yan Tang, Michal L Melamed, Thomas H Hostetter, Jeffrey E Pessin, Meredith Hawkins, Christopher S Fry, Matthew K Abramowitz, Camille R Brightwell, Ameya S Kulkarni, William Paredes, Kehao Zhang, Jaclyn B Perkins, Knubian J Gatlin, Matthew Custodio, Hina Farooq, Bushra Zaidi, Rima Pai, Rupinder S Buttar, Yan Tang, Michal L Melamed, Thomas H Hostetter, Jeffrey E Pessin, Meredith Hawkins, Christopher S Fry, Matthew K Abramowitz

Abstract

BACKGROUNDSkeletal muscle maladaptation accompanies chronic kidney disease (CKD) and negatively affects physical function. Emphasis in CKD has historically been placed on muscle fiber-intrinsic deficits, such as altered protein metabolism and atrophy. However, targeted treatment of fiber-intrinsic dysfunction has produced limited improvement, whereas alterations within the fiber-extrinsic environment have scarcely been examined.METHODSWe investigated alterations to the skeletal muscle interstitial environment with deep cellular phenotyping of biopsies from patients with CKD and age-matched controls and performed transcriptome profiling to define the molecular underpinnings of CKD-associated muscle impairments. We examined changes in muscle maladaptation following initiation of dialysis therapy for kidney failure.RESULTSPatients with CKD exhibited a progressive fibrotic muscle phenotype, which was associated with impaired regenerative capacity and lower vascular density. The severity of these deficits was strongly associated with the degree of kidney dysfunction. Consistent with these profound deficits, CKD was associated with broad alterations to the muscle transcriptome, including altered ECM organization, downregulated angiogenesis, and altered expression of pathways related to stem cell self-renewal. Remarkably, despite the seemingly advanced nature of this fibrotic transformation, dialysis treatment rescued these deficits, restoring a healthier muscle phenotype. Furthermore, after accounting for muscle atrophy, strength and endurance improved after dialysis initiation.CONCLUSIONThese data identify a dialysis-responsive muscle fibrotic phenotype in CKD and suggest the early dialysis window presents a unique opportunity of improved muscle regenerative capacity during which targeted interventions may achieve maximal impact.TRIAL REGISTRATIONNCT01452412FUNDINGNIH, NIH Clinical and Translational Science Awards (CTSA), and Einstein-Mount Sinai Diabetes Research Center.

Keywords: Chronic kidney disease; Extracellular matrix; Muscle Biology; Nephrology; Skeletal muscle.

Figures

Figure 1. Flow diagram of study participation.
Figure 1. Flow diagram of study participation.
ESRD indicates dialysis patients (transplant patients were not recruited).
Figure 2. Collagen content and density are…
Figure 2. Collagen content and density are progressively elevated in skeletal muscle of patients with CKD.
Proportion of ECM collagen content is elevated in CKD patients (A) and negatively associated with eGFR (B) (n = 33). Densely packed collagen content is elevated in CKD patients (C) and negatively associated with eGFR (D) (n = 33). Loosely packed collagen content was numerically higher but not significantly different in CKD patients (E) and not significantly correlated with eGFR (F) (n = 33). Representative images of picrosirius red staining in control and CKD muscle under both bright-field and polarized light (G). Total skeletal muscle collagen content assayed biochemically is elevated in CKD patients (H) and negatively associated with GFR (I) (n = 28). Comparisons made using 2-tailed t tests or Wilcoxon rank-sum tests. Spearman coefficients calculated to test correlations. Scale bar: 100 μm. *P < 0.05, control compared with CKD.
Figure 3. Satellite cell abundance, proximity of…
Figure 3. Satellite cell abundance, proximity of satellite cells to capillaries, and capillary density are lower in skeletal muscle of patients with CKD.
Satellite cell abundance is lower in CKD patients (A) and positively associated with eGFR (B) (n = 35). Representative images of satellite cell and capillary staining (C). Satellite cell activation is numerically, but not statistically, elevated in patients with stage 3 CKD only (D) and not associated with eGFR (E) (n = 34). Representative images of activated satellite cell staining (F). The distance between satellite cells and nearest capillary is elevated in subjects with CKD (G) and negatively associated with eGFR (H) (n = 32). CFPE, an index of capillary density and blood-muscle exchange, is lower in subjects with CKD (I) and positively associated with eGFR (J) (n = 27). Comparisons made using 2-tailed t tests or Wilcoxon rank-sum tests. Spearman coefficients calculated to test correlations. Scale bar: 100 μm. *P < 0.05, control compared with CKD.
Figure 4. Transcriptomic differences in skeletal muscle…
Figure 4. Transcriptomic differences in skeletal muscle of patients with CKD reveal alteration of genes within angiogenic and fibrotic pathways and rescue of Myc and interferon pathways postdialysis.
Volcano plot highlighting global gene expression differences in CKD versus control (A). Heatmap with the top 100 differentially expressed genes in CKD (B). Orange bar, CKD; purple bar, control. Genes involved in VEGF signaling, ECM organization, and collagen formation are downregulated in CKD (C). Heatmaps of genes belonging to VEGF (D), ECM organization (E), and collagen formation (F) pathways that are differentially expressed in CKD versus control (n = 14). Orange bar, CKD; purple bar, control. GSEA plots reveal differential regulation of Myc, interferon-α, and interferon-γ pathways in CKD and rescue (change in regulation toward control values) after dialysis (G) (n = 3).
Figure 5. Dialysis rescues pathological phenotype in…
Figure 5. Dialysis rescues pathological phenotype in skeletal muscle associated with CKD.
ECM collagen content (A) (n = 32), densely packed collagen (B) (n = 31), loosely packed collagen (C) (n = 31), and total muscle collagen assayed biochemically (D) (n = 29) are lower in subjects who have undergone dialysis compared with patients with advanced CKD. Total satellite cell abundance (E) (n = 33), the distance between satellite cells and their nearest capillary (F) (n = 31), and the CFPE ratio (G) (n = 27) are altered in patients who have undergone dialysis compared with those with advanced CKD, with a restoration toward control values. Data in AG include only control, CKD stage 4/5, and dialysis patients. CKD stages 4/5 represent a subset of total CKD patients as presented in Figures 1 and 2 who have an eGFR < 30 mL/min/1.73 m2. Comparisons made using mixed effects models. Satellite cell abundance is negatively associated with densely packed collagen content (H) (n = 37). Spearman coefficient calculated to test correlation. *P < 0.05 compared with CKD stages 4/5. #P < 0.05 compared with control.

References

    1. Bikbov B, et al. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–733. doi: 10.1016/S0140-6736(20)30045-3.
    1. Bao Y, et al. Frailty, dialysis initiation, and mortality in end-stage renal disease. Arch Intern Med. 2012;172(14):1071–1077.
    1. O’Hare AM, et al. Decreased survival among sedentary patients undergoing dialysis: results from the dialysis morbidity and mortality study wave 2. Am J Kidney Dis. 2003;41(2):447–454. doi: 10.1053/ajkd.2003.50055.
    1. Painter P, Roshanravan B. The association of physical activity and physical function with clinical outcomes in adults with chronic kidney disease. Curr Opin Nephrol Hypertens. 2013;22(6):615–623. doi: 10.1097/MNH.0b013e328365b43a.
    1. Reese PP, et al. Physical performance and frailty in chronic kidney disease. Am J Nephrol. 2013;38(4):307–315. doi: 10.1159/000355568.
    1. Roshanravan B, et al. Association between physical performance and all-cause mortality in CKD. J Am Soc Nephrol. 2013;24(5):822–830. doi: 10.1681/ASN.2012070702.
    1. Wesson DE, et al. Long-term safety and efficacy of veverimer in patients with metabolic acidosis in chronic kidney disease: a multicentre, randomised, blinded, placebo-controlled, 40-week extension. Lancet. 2019;394(10196):396–406. doi: 10.1016/S0140-6736(19)31388-1.
    1. Padilla J, et al. Physical functioning in patients with chronic kidney disease. J Nephrol. 2008;21(4):550–559.
    1. Kestenbaum B, et al. Impaired skeletal muscle mitochondrial bioenergetics and physical performance in chronic kidney disease. JCI Insight. 2020;5(5):133289.
    1. Garibotto G, et al. Insulin sensitivity of muscle protein metabolism is altered in patients with chronic kidney disease and metabolic acidosis. Kidney Int. 2015;88(6):1419–1426. doi: 10.1038/ki.2015.247.
    1. Zilles M, et al. How to prevent renal cachexia? A clinical randomized pilot study testing oral supplemental nutrition in hemodialysis patients with and without human immunodeficiency virus infection. J Ren Nutr. 2018;28(1):37–44. doi: 10.1053/j.jrn.2017.07.003.
    1. Macdonald JH, et al. Nandrolone decanoate as anabolic therapy in chronic kidney disease: a randomized phase II dose-finding study. Nephron Clin Pract. 2007;106(3):c125–c135. doi: 10.1159/000103000.
    1. Jeong JH, et al. Results from the randomized controlled IHOPE trial suggest no effects of oral protein supplementation and exercise training on physical function in hemodialysis patients. Kidney Int. 2019;96(3):777–786. doi: 10.1016/j.kint.2019.03.018.
    1. Abramowitz MK, et al. Skeletal muscle fibrosis is associated with decreased muscle inflammation and weakness in patients with chronic kidney disease. Am J Physiol Renal Physiol. 2018;315(6):F1658–F1669. doi: 10.1152/ajprenal.00314.2018.
    1. Peng H, et al. Myokine mediated muscle-kidney crosstalk suppresses metabolic reprogramming and fibrosis in damaged kidneys. Nat Commun. 2017;8(1):1493. doi: 10.1038/s41467-017-01646-6.
    1. Dong J, et al. The pathway to muscle fibrosis depends on myostatin stimulating the differentiation of fibro/adipogenic progenitor cells in chronic kidney disease. Kidney Int. 2017;91(1):119–128. doi: 10.1016/j.kint.2016.07.029.
    1. Zhang L, et al. Satellite cell dysfunction and impaired IGF-1 signaling cause CKD-induced muscle atrophy. J Am Soc Nephrol. 2010;21(3):419–427. doi: 10.1681/ASN.2009060571.
    1. Fry CS, et al. Regulation of the muscle fiber microenvironment by activated satellite cells during hypertrophy. FASEB J. 2014;28(4):1654–1665. doi: 10.1096/fj.13-239426.
    1. Gillies AR, Lieber RL. Structure and function of the skeletal muscle extracellular matrix. Muscle Nerve. 2011;44(3):318–331. doi: 10.1002/mus.22094.
    1. Lieber RL, et al. Inferior mechanical properties of spastic muscle bundles due to hypertrophic but compromised extracellular matrix material. Muscle Nerve. 2003;28(4):464–471. doi: 10.1002/mus.10446.
    1. Lund DK, Cornelison DD. Enter the matrix: shape, signal and superhighway. FEBS J. 2013;280(17):4089–4099. doi: 10.1111/febs.12171.
    1. Alexakis C, et al. Implication of the satellite cell in dystrophic muscle fibrosis: a self-perpetuating mechanism of collagen overproduction. Am J Physiol Cell Physiol. 2007;293(2):C661–C669. doi: 10.1152/ajpcell.00061.2007.
    1. Brack AS, Rando TA. Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Rev. 2007;3(3):226–237. doi: 10.1007/s12015-007-9000-2.
    1. Mann CJ, et al. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle. 2011;1(1):21. doi: 10.1186/2044-5040-1-21.
    1. Labat-Robert J. Age-dependent remodeling of connective tissue: role of fibronectin and laminin. Pathol Biol (Paris) 2003;51(10):563–568. doi: 10.1016/j.patbio.2003.09.006.
    1. Gilbert PM, et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science. 2010;329(5995):1078–1081. doi: 10.1126/science.1191035.
    1. Urciuolo A, et al. Collagen VI regulates satellite cell self-renewal and muscle regeneration. Nat Commun. 2013;4:1964. doi: 10.1038/ncomms2964.
    1. Fry CS, et al. Myogenic progenitor cells control extracellular matrix production by fibroblasts during skeletal muscle hypertrophy. Cell Stem Cell. 2017;20(1):56–69. doi: 10.1016/j.stem.2016.09.010.
    1. Thomas R, et al. Chronic kidney disease and its complications. Prim Care. 2008;35(2):329–344. doi: 10.1016/j.pop.2008.01.008.
    1. Brashear SE, et al. Passive stiffness of fibrotic skeletal muscle in mdx mice relates to collagen architecture. J Pathol. 2021;599(3):943–962.
    1. Lepper C, et al. An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration. Development. 2011;138(17):3639–3646. doi: 10.1242/dev.067595.
    1. Murach KA, et al. Starring or supporting role? Satellite cells and skeletal muscle fiber size regulation. Physiology (Bethesda) 2018;33(1):26–38. doi: 10.1152/physiol.00019.2017.
    1. Arentson-Lantz EJ, et al. Fourteen days of bed rest induces a decline in satellite cell content and robust atrophy of skeletal muscle fibers in middle-aged adults. J Appl Physiol (1985) 2016;120(8):965–975. doi: 10.1152/japplphysiol.00799.2015.
    1. Moro T, et al. Low skeletal muscle capillarization limits muscle adaptation to resistance exercise training in older adults. Exp Gerontol. 2019;127:110723. doi: 10.1016/j.exger.2019.110723.
    1. Christov C, et al. Muscle satellite cells and endothelial cells: close neighbors and privileged partners. Mol Biol Cell. 2007;18(4):1397–1409. doi: 10.1091/mbc.e06-08-0693.
    1. Hepple RT. A new measurement of tissue capillarity: the capillary-to-fibre perimeter exchange index. Can J Appl Physiol. 1997;22(1):11–22. doi: 10.1139/h97-002.
    1. Dreyer HC, et al. Essential amino acid supplementation in patients following total knee arthroplasty. J Clin Invest. 2013;123(11):4654–4666. doi: 10.1172/JCI70160.
    1. Muyskens JB, et al. Cellular and morphological changes with EAA supplementation before and after total knee arthroplasty. J Appl Physiol (1985) 2019;127(2):531–545. doi: 10.1152/japplphysiol.00869.2018.
    1. Gao Y, et al. Age-related changes in the mechanical properties of the epimysium in skeletal muscles of rats. J Biomech. 2008;41(2):465–469. doi: 10.1016/j.jbiomech.2007.09.021.
    1. Ahonen RE. Striated muscle ultrastructure in uremic patients and in renal transplant recipients. Acta Neuropathol. 1980;50(2):163–166. doi: 10.1007/BF00692869.
    1. Diesel W, et al. Morphologic features of the myopathy associated with chronic renal failure. Am J Kidney Dis. 1993;22(5):677–684. doi: 10.1016/S0272-6386(12)80430-6.
    1. Lewis MI, et al. Metabolic and morphometric profile of muscle fibers in chronic hemodialysis patients. J Appl Physiol (1985) 2012;112(1):72–78. doi: 10.1152/japplphysiol.00556.2011.
    1. Shah AJ, et al. Muscle in chronic uremia--a histochemical and morphometric study of human quadriceps muscle biopsies. Clin Neuropathol. 1983;2(2):83–89.
    1. Wilkinson TJ, et al. Quality over quantity? Association of skeletal muscle myosteatosis and myofibrosis on physical function in chronic kidney disease. Nephrol Dial Transplant. 2019;34(8):1344–1353. doi: 10.1093/ndt/gfy139.
    1. Lehmann J, et al. Collagen type IV remodelling gender-specifically predicts mortality in decompensated cirrhosis. Liver Int. 2019;39(5):885–893. doi: 10.1111/liv.14070.
    1. Rasmussen DGK, et al. Collagen turnover profiles in chronic kidney disease. Sci Rep. 2019;9(1):16062. doi: 10.1038/s41598-019-51905-3.
    1. Dumont NA, et al. Intrinsic and extrinsic mechanisms regulating satellite cell function. Development. 2015;142(9):1572–1581. doi: 10.1242/dev.114223.
    1. Lacraz G, et al. Increased stiffness in aged skeletal muscle impairs muscle progenitor cell proliferative activity. PLoS One. 2015;10(8):e0136217. doi: 10.1371/journal.pone.0136217.
    1. Gueugneau M, et al. Lower skeletal muscle capillarization in hypertensive elderly men. Exp Gerontol. 2016;76:80–88. doi: 10.1016/j.exger.2016.01.013.
    1. Valle-Tenney R, et al. Role of hypoxia in skeletal muscle fibrosis: synergism between hypoxia and TGF-β signaling upregulates CCN2/CTGF expression specifically in muscle fibers. Matrix Biol. 2020;87:48–65. doi: 10.1016/j.matbio.2019.09.003.
    1. Provenzano PP, et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418–429. doi: 10.1016/j.ccr.2012.01.007.
    1. Prommer H-U, et al. Chronic kidney disease induces a systemic microangiopathy, tissue hypoxia and dysfunctional angiogenesis. Sci Rep. 2018;8(1):5317. doi: 10.1038/s41598-018-23663-1.
    1. Campistol JM. Uremic myopathy. Kidney Int. 2002;62(5):1901–1913. doi: 10.1046/j.1523-1755.2002.00614.x.
    1. Flisiński M, et al. Decreased hypoxia-inducible factor-1α in gastrocnemius muscle in rats with chronic kidney disease. Kidney Blood Press Res. 2012;35(6):608–618. doi: 10.1159/000339706.
    1. Qian F-Y, et al. Hypoxia-inducible factor-prolyl hydroxylase inhibitor ameliorates myopathy in a mouse model of chronic kidney disease. Am J Physiol Renal Physiol. 2019;317(5):F1265–F1273. doi: 10.1152/ajprenal.00260.2019.
    1. Flisiński M, et al. Influence of different stages of experimental chronic kidney disease on rats locomotor and postural skeletal muscles microcirculation. Ren Fail. 2008;30(4):443–451. doi: 10.1080/08860220801985694.
    1. Burkhardt D, et al. Reduced microvascular density in omental biopsies of children with chronic kidney disease. PLoS One. 2016;11(11):e0166050. doi: 10.1371/journal.pone.0166050.
    1. Yang X, et al. The hypoxia-inducible factors HIF1α and HIF2α are dispensable for embryonic muscle development but essential for postnatal muscle regeneration. J Biol Chem. 2017;292(14):5981–5991. doi: 10.1074/jbc.M116.756312.
    1. Conboy IM, et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433(7027):760–764. doi: 10.1038/nature03260.
    1. Nederveen JP, et al. The influence of capillarization on satellite cell pool expansion and activation following exercise-induced muscle damage in healthy young men. J Physiol. 2018;596(6):1063–1078. doi: 10.1113/JP275155.
    1. Verma M, et al. Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and Notch signaling. Cell Stem Cell. 2018;23(4):530–543. doi: 10.1016/j.stem.2018.09.007.
    1. Zeisberg M, Kalluri R. Cellular mechanisms of tissue fibrosis. 1. Common and organ-specific mechanisms associated with tissue fibrosis. Am J Physiol Cell Physiol. 2013;304(3):C216–C225. doi: 10.1152/ajpcell.00328.2012.
    1. Conboy IM, et al. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302(5650):1575–1577. doi: 10.1126/science.1087573.
    1. Brack AS, et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science. 2007;317(5839):807–810. doi: 10.1126/science.1144090.
    1. Meyer TW, Hostetter TH. Uremia. N Engl J Med. 2007;357(13):1316–1325. doi: 10.1056/NEJMra071313.
    1. Abramowitz MK, et al. The serum anion gap is altered in early kidney disease and associates with mortality. Kidney Int. 2012;82(6):701–709. doi: 10.1038/ki.2012.196.
    1. Wang K, Kestenbaum B. Proximal tubular secretory clearance: a neglected partner of kidney function. Clin J Am Soc Nephrol. 2018;13(8):1291–1296. doi: 10.2215/CJN.12001017.
    1. Alcalde-Estévez E, et al. Uraemic toxins impair skeletal muscle regeneration by inhibiting myoblast proliferation, reducing myogenic differentiation, and promoting muscular fibrosis. Sci Rep. 2021;11(1):512. doi: 10.1038/s41598-020-79186-1.
    1. Stenvinkel P, Larsson TE. Chronic kidney disease: a clinical model of premature aging. Am J Kidney Dis. 2013;62(2):339–351. doi: 10.1053/j.ajkd.2012.11.051.
    1. Eilers M, Eisenman RN. Myc’s broad reach. Genes Dev. 2008;22(20):2755–2766. doi: 10.1101/gad.1712408.
    1. Hofmann JW, et al. Reduced expression of MYC increases longevity and enhances healthspan. Cell. 2015;160(3):477–488. doi: 10.1016/j.cell.2014.12.016.
    1. Cui CY, et al. Skewed macrophage polarization in aging skeletal muscle. Aging Cell. 2019;18(6):e13032.
    1. Przybyla B, et al. Aging alters macrophage properties in human skeletal muscle both at rest and in response to acute resistance exercise. Exp Gerontol. 2006;41(3):320–327. doi: 10.1016/j.exger.2005.12.007.
    1. Zhang C, et al. Age-related decline of interferon-gamma responses in macrophage impairs satellite cell proliferation and regeneration. J Cachexia Sarcopenia Muscle. 2020;11(5):1291–1305. doi: 10.1002/jcsm.12584.
    1. Cheng M, et al. Endogenous interferon-gamma is required for efficient skeletal muscle regeneration. Am J Physiol Cell Physiol. 2008;294(5):C1183–C1191. doi: 10.1152/ajpcell.00568.2007.
    1. Foster W, et al. Gamma interferon as an antifibrosis agent in skeletal muscle. J Orthop Res. 2003;21(5):798–804. doi: 10.1016/S0736-0266(03)00059-7.
    1. Caglar K, et al. Inflammatory signals associated with hemodialysis. Kidney Int. 2002;62(4):1408–1416. doi: 10.1111/j.1523-1755.2002.kid556.x.
    1. Deger SM, et al. Systemic inflammation is associated with exaggerated skeletal muscle protein catabolism in maintenance hemodialysis patients. JCI Insight. 2017;2(22):95185.
    1. Mansouri L, et al. Hemodialysis patients display a declined proportion of Th2 and regulatory T cells in parallel with a high interferon-γ profile. Nephron. 2017;136(3):254–260. doi: 10.1159/000471814.
    1. Himmelfarb J. Uremic toxicity, oxidative stress, and hemodialysis as renal replacement therapy. Semin Dial. 2009;22(6):636–643. doi: 10.1111/j.1525-139X.2009.00659.x.
    1. Avin KG, et al. Skeletal muscle regeneration and oxidative stress are altered in chronic kidney disease. PLoS One. 2016;11(8):e0159411. doi: 10.1371/journal.pone.0159411.
    1. Gamboa JL, et al. Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease. Physiol Rep. 2016;4(9):e12780. doi: 10.14814/phy2.12780.
    1. Qaisar R, et al. Oxidative stress-induced dysregulation of excitation-contraction coupling contributes to muscle weakness. J Cachexia Sarcopenia Muscle. 2018;9(5):1003–1017. doi: 10.1002/jcsm.12339.
    1. Jang YC, et al. Increased superoxide in vivo accelerates age-associated muscle atrophy through mitochondrial dysfunction and neuromuscular junction degeneration. FASEB J. 2010;24(5):1376–1390. doi: 10.1096/fj.09-146308.
    1. Handelman GJ, et al. Elevated plasma F2-isoprostanes in patients on long-term hemodialysis. Kidney Int. 2001;59(5):1960–1966. doi: 10.1046/j.1523-1755.2001.0590051960.x.
    1. Gamboa JL, et al. Skeletal muscle mitochondrial dysfunction is present in patients with CKD before initiation of maintenance hemodialysis. Clin J Am Soc Nephrol. 2020;15(7):926–936. doi: 10.2215/CJN.10320819.
    1. Raj DS, et al. Skeletal muscle, cytokines, and oxidative stress in end-stage renal disease. Kidney Int. 2005;68(5):2338–2344. doi: 10.1111/j.1523-1755.2005.00695.x.
    1. Johansen KL, et al. Muscle atrophy in patients receiving hemodialysis: effects on muscle strength, muscle quality, and physical function. Kidney Int. 2003;63(1):291–297. doi: 10.1046/j.1523-1755.2003.00704.x.
    1. McIntyre CW, et al. Patients receiving maintenance dialysis have more severe functionally significant skeletal muscle wasting than patients with dialysis-independent chronic kidney disease. Nephrol Dial Transplant. 2006;21(8):2210–2216. doi: 10.1093/ndt/gfl064.
    1. Fry CS, et al. Inducible depletion of satellite cells in adult, sedentary mice impairs muscle regenerative capacity without affecting sarcopenia. Nat Med. 2015;21(1):76–80. doi: 10.1038/nm.3710.
    1. Cordova G, et al. Combined therapies for Duchenne muscular dystrophy to optimize treatment efficacy. Front Genet. 2018;9:114. doi: 10.3389/fgene.2018.00114.
    1. Rivara MB, et al. Changes in symptom burden and physical performance with initiation of dialysis in patients with chronic kidney disease. Hemodial Int. 2015;19(1):147–150. doi: 10.1111/hdi.12244.
    1. John SG, et al. Natural history of skeletal muscle mass changes in chronic kidney disease stage 4 and 5 patients: an observational study. PLoS One. 2013;8(5):e65372. doi: 10.1371/journal.pone.0065372.
    1. Molsted S, et al. Fiber type-specific response of skeletal muscle satellite cells to high-intensity resistance training in dialysis patients. Muscle Nerve. 2015;52(5):736–745. doi: 10.1002/mus.24633.
    1. Cooper BA, et al. A randomized, controlled trial of early versus late initiation of dialysis. N Engl J Med. 2010;363(7):609–619. doi: 10.1056/NEJMoa1000552.
    1. KDIGO. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Accessed November 16, 2021.
    1. National Kidney Foundation. KDOQI clinical practice guideline for hemodialysis adequacy: 2015 update. Am J Kidney Dis. 2015;66(5):884–930. doi: 10.1053/j.ajkd.2015.07.015.
    1. Gura V, et al. A wearable artificial kidney for patients with end-stage renal disease. JCI Insight. 2016;1(8):86397.
    1. Hojs N, et al. Ambulatory hemodialysis-technology landscape and potential for patient-centered treatment. Clin J Am Soc Nephrol. 2020;15(1):152–159. doi: 10.2215/CJN.01970219.
    1. Abramowitz MK, et al. The Pathophysiology of Uremia. In: Himmelfarb J, Sayegh MH, eds. Chronic Kidney Disease, Dialysis, and Transplantation: Companion to Brenner & Rector’s The Kidney. Saunders; 2011:251–264.
    1. Flythe JE, Hostetter TH. Assessing clinical relevance of uremic toxins. Clin J Am Soc Nephrol. 2019;14(2):182–183. doi: 10.2215/CJN.14931218.
    1. Sirich TL, et al. Numerous protein-bound solutes are cleared by the kidney with high efficiency. Kidney Int. 2013;84(3):585–590. doi: 10.1038/ki.2013.154.
    1. Bucar Pajek M, et al. Six-minute walk test in renal failure patients: representative results, performance analysis and perceived dyspnea predictors. PLoS One. 2016;11(3):e0150414. doi: 10.1371/journal.pone.0150414.
    1. Kurella Tamura M, et al. Functional status of elderly adults before and after initiation of dialysis. N Engl J Med. 2009;361(16):1539–1547. doi: 10.1056/NEJMoa0904655.
    1. Moore GE, et al. Determinants of VO2peak in patients with end-stage renal disease: on and off dialysis. Med Sci Sports Exerc. 1993;25(1):18–23. doi: 10.1249/00005768-199301000-00004.
    1. Jassal SV, et al. Loss of independence in patients starting dialysis at 80 years of age or older. N Engl J Med. 2009;361(16):1612–1613. doi: 10.1056/NEJMc0905289.
    1. Painter P, et al. Exercise capacity in hemodialysis, CAPD, and renal transplant patients. Nephron. 1986;42(1):47–51. doi: 10.1159/000183632.
    1. Melamed ML, et al. Effects of sodium bicarbonate in CKD stages 3 and 4: a randomized, placebo-controlled, multicenter clinical trial. Am J Kidney Dis. 2020;75(2):225–234. doi: 10.1053/j.ajkd.2019.07.016.
    1. Kalantar-Zadeh K, et al. Design and development of a dialysis food frequency questionnaire. J Ren Nutr. 2011;21(3):257–262. doi: 10.1053/j.jrn.2010.05.013.
    1. Block G, et al. Nutrient sources in the American diet: quantitative data from the NHANES II survey. II. Macronutrients and fats. Am J Epidemiol. 1985;122(1):27–40. doi: 10.1093/oxfordjournals.aje.a114084.
    1. Feskanich D, et al. Computerized collection and analysis of dietary intake information. Comput Methods Programs Biomed. 1989;30(1):47–57. doi: 10.1016/0169-2607(89)90122-3.
    1. Maroni BJ, et al. A method for estimating nitrogen intake of patients with chronic renal failure. Kidney Int. 1985;27(1):58–65. doi: 10.1038/ki.1985.10.
    1. Reuben DB, et al. Motor assessment using the NIH Toolbox. Neurology. 2013;80(11 suppl 3):S65–S75.
    1. Bohannon RW, et al. Comparison of walking performance over the first 2 minutes and the full 6 minutes of the six-minute walk test. BMC Res Notes. 2014;7:269. doi: 10.1186/1756-0500-7-269.
    1. Guralnik JM, et al. A short physical performance battery assessing lower extremity function: association with self-reported disability and prediction of mortality and nursing home admission. J Gerontol. 1994;49(2):M85–M94. doi: 10.1093/geronj/49.2.M85.
    1. Troiano RP, et al. Physical activity in the United States measured by accelerometer. Med Sci Sports Exerc. 2008;40(1):181–188. doi: 10.1249/mss.0b013e31815a51b3.
    1. Matthews CE, et al. Amount of time spent in sedentary behaviors in the United States, 2003-2004. Am J Epidemiol. 2008;167(7):875–881. doi: 10.1093/aje/kwm390.
    1. Freedson PS, et al. Calibration of the Computer Science and Applications, Inc. accelerometer. Med Sci Sports Exerc. 1998;30(5):777–781. doi: 10.1097/00005768-199805000-00021.
    1. Levey AS, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–612. doi: 10.7326/0003-4819-150-9-200905050-00006.
    1. Harris PA, et al. Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42(2):377–381. doi: 10.1016/j.jbi.2008.08.010.
    1. Fry CS, et al. ACL injury reduces satellite cell abundance and promotes fibrogenic cell expansion within skeletal muscle. J Orthop Res. 2017;35(9):1876–1885. doi: 10.1002/jor.23502.
    1. Holthofer H, et al. Ulex europaeus I lectin as a marker for vascular endothelium in human tissues. Lab Invest. 1982;47(1):60–66.
    1. Junqueira LC, et al. Picrosirius staining plus polarization microscopy, a specific method for collagen detection in tissue sections. Histochem J. 1979;11(4):447–455. doi: 10.1007/BF01002772.
    1. Lattouf R, et al. Picrosirius red staining: a useful tool to appraise collagen networks in normal and pathological tissues. J Histochem Cytochem. 2014;62(10):751–758. doi: 10.1369/0022155414545787.
    1. Brightwell CR, et al. Moderate-intensity aerobic exercise improves skeletal muscle quality in older adults. Transl Sports Med. 2019;2(3):109–119. doi: 10.1002/tsm2.70.
    1. Hepple RT, Mathieu-Costello O. Estimating the size of the capillary-to-fiber interface in skeletal muscle: a comparison of methods. J Appl Physiol (1985) 2001;91(5):2150–2156. doi: 10.1152/jappl.2001.91.5.2150.
    1. Wen Y, et al. MyoVision: software for automated high-content analysis of skeletal muscle immunohistochemistry. J Appl Physiol (1985) 2018;124(1):40–51. doi: 10.1152/japplphysiol.00762.2017.
    1. Brightwell CR, et al. Thermal injury initiates pervasive fibrogenesis in skeletal muscle. Am J Physiol Cell Physiol. 2020;319(2):C277–C287. doi: 10.1152/ajpcell.00337.2019.
    1. Andrews S. FastQC: a quality control tool for high throughput sequence data. Accessed November 16, 2021.
    1. Ewels P, et al. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 2016;32(19):3047–3048. doi: 10.1093/bioinformatics/btw354.
    1. Chen S, et al. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–i890. doi: 10.1093/bioinformatics/bty560.
    1. Harrow J, et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 2012;22(9):1760–1774. doi: 10.1101/gr.135350.111.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. doi: 10.1093/bioinformatics/bts635.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323. doi: 10.1186/1471-2105-12-323.
    1. Law CW, et al. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15(2):R29. doi: 10.1186/gb-2014-15-2-r29.
    1. Herwig R, et al. Analyzing and interpreting genome data at the network level with ConsensusPathDB. Nat Protoc. 2016;11(10):1889–1907. doi: 10.1038/nprot.2016.117.
    1. Wu D, Smyth GK. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 2012;40(17):e133. doi: 10.1093/nar/gks461.

Source: PubMed

3
S'abonner