Correlations of Calf Muscle Macrophage Content With Muscle Properties and Walking Performance in Peripheral Artery Disease

Kate Kosmac, Marta Gonzalez-Freire, Mary M McDermott, Sarah H White, R Grace Walton, Robert L Sufit, Lu Tian, Lingyu Li, Melina R Kibbe, Michael H Criqui, Jack M Guralnik, Tamar S Polonsky, Christiaan Leeuwenburgh, Luigi Ferrucci, Charlotte A Peterson, Kate Kosmac, Marta Gonzalez-Freire, Mary M McDermott, Sarah H White, R Grace Walton, Robert L Sufit, Lu Tian, Lingyu Li, Melina R Kibbe, Michael H Criqui, Jack M Guralnik, Tamar S Polonsky, Christiaan Leeuwenburgh, Luigi Ferrucci, Charlotte A Peterson

Abstract

Background Peripheral artery disease (PAD) is a manifestation of atherosclerosis characterized by reduced blood flow to the lower extremities and mobility loss. Preliminary evidence suggests PAD damages skeletal muscle, resulting in muscle impairments that contribute to functional decline. We sought to determine whether PAD is associated with an altered macrophage profile in gastrocnemius muscles and whether muscle macrophage populations are associated with impaired muscle phenotype and walking performance in patients with PAD. Methods and Results Macrophages, satellite cells, and extracellular matrix in gastrocnemius muscles from 25 patients with PAD and 7 patients without PAD were quantified using immunohistochemistry. Among patients with PAD, both the absolute number and percentage of cluster of differentiation (CD) 11b+CD206+ M2-like macrophages positively correlated to satellite cell number (r=0.461 [P=0.023] and r=0.416 [P=0.042], respectively) but not capillary density or extracellular matrix. The number of CD11b+CD206- macrophages negatively correlated to 4-meter walk tests at normal (r=-0.447, P=0.036) and fast pace (r=-0.510, P=0.014). Extracellular matrix occupied more muscle area in PAD compared with non-PAD (8.72±2.19% versus 5.30±1.03%, P<0.001) and positively correlated with capillary density (r=0.656, P<0.001). Conclusions Among people with PAD, higher CD206+ M2-like macrophage abundance was associated with greater satellite cell numbers and muscle fiber size. Lower CD206- macrophage abundance was associated with better walking performance. Further study is needed to determine whether CD206+ macrophages are associated with ongoing reparative processes enabling skeletal muscle adaptation to damage with PAD. Registration URL: https://www.clini​caltr​ials.gov; Unique identifiers: NCT00693940, NCT01408901, NCT0224660.

Keywords: macrophage; peripheral artery disease; skeletal muscle; walking performance.

Figures

Figure 1. Representative images of total cluster…
Figure 1. Representative images of total cluster of differentiation (CD) 11b+, CD206−, and CD206+ macrophage abundance in the gastrocnemius muscle of patients with peripheral artery disease (PAD) compared with patients without PAD.
A, Total muscle macrophages in patients without PAD (left) and patients with PAD (right) visualized by immunohistochemistry with an antibody against the pan‐macrophage marker CD11b (green). Scale bar=200 µm. B, Higher magnification images of macrophages from boxed regions in panel A, showing CD11b+ (green) macrophages, CD206+ (red) macrophages, and the merged images, also overlaid with DAPI staining of nuclei (blue). Whereas the majority of macrophages are CD11b+CD206+, white arrows indicate CD11b+CD206− macrophages. Scale bar=200 µm.
Figure 2. Quantification of macrophage content in…
Figure 2. Quantification of macrophage content in gastrocnemius muscle cross‐sections. A through D, Non‐peripheral artery disease (non‐PAD, n=6), peripheral artery disease (PAD) (n=25).
Data are expressed as mean±SE; P values adjusted for race, *significance P≤0.05. A, Total cluster of differentiation (CD) 11b+ macrophages in patients with PAD compared with patients without PAD. B, CD11b+CD206− macrophages in patients without PAD and patients with PAD. C, CD11b+CD206+ macrophages in patients without PAD and patients with PAD. D, Relative abundance (% of total) of CD206− and CD206+ macrophages in patients with PAD and patients without PAD. E, Association of the M2c macrophage marker CD163 with the number of CD206+ macrophages, supporting CD206+ macrophages as M2‐like. Non‐PAD (n=5), PAD (n=18). Association determined by Pearson product moment correlation, r=partial correlation coefficient.
Figure 3. Representative images of muscle satellite…
Figure 3. Representative images of muscle satellite cells with laminin‐demarcated muscle fibers in gastrocnemius from peripheral artery disease (PAD).
A, Paired box 7+ (Pax7+) (green) staining overlapping cell nuclei (DAPI, blue) identifies satellite cells. Scale bar=200 µm. B, Laminin demarcating muscle fiber borders (red). Scale bar=200 µm. C, Higher magnification showing satellite cells located beneath the laminin‐stained basal lamina. White arrows point to satellite cell nuclei; Pax7 (green), nuclei (DAPI, blue), laminin (red). Scale bar=100 µm.
Figure 4. Relative abundance of cluster of…
Figure 4. Relative abundance of cluster of differentiation (CD) 206+ macrophages is positively associated with satellite cell number and fiber size in the gastrocnemius muscle from patients with peripheral artery disease (PAD).
Scatterplots showing correlations between the relative abundance of CD206+ macrophages and (A) satellite cell content and (B) fiber size, measured by minimum feret diameter. C, Correlation between satellite cells and fiber size. For all measures, relationships determined by Pearson product moment correlation adjusting for race, r=partial correlation coefficient, PAD (n=25). D, Representative images showing a satellite cell (paired box 7 [Pax7]+, green) and a CD206+ macrophage (red) in close proximity. DAPI (blue) stains nuclei. Scale bar=20 µm.
Figure 5. Peripheral artery disease ( PAD…
Figure 5. Peripheral artery disease (PAD) gastrocnemius muscles show higher extracellular matrix (ECM) content compared with non‐PAD.
A, Representative images of gastrocnemius muscle showing fluorescently labeled α‐wheat germ agglutinin (WGA) staining of glycosaminoglycans in the ECM between muscle fibers in patients without PAD and patients with PAD, quantified in (B). Scale bar=200 µm. Non‐PAD (n=7), PAD (n=24). Data are expressed as mean±SD; P values adjusted for race, *significance P≤0.05. C, Correlation of ECM content with capillary density (the number of lectin+ capillaries/fiber) in patients with PAD (n=24), determined by Pearson product moment correlation adjusting for race, r=partial correlation coefficient. No relationship between ECM content and capillary density was observed in non‐PAD samples (data not shown).
Figure 6. Peripheral artery disease ( PAD…
Figure 6. Peripheral artery disease (PAD) gastrocnemius muscles show higher collagen content compared with non‐PAD.
A, Representative images of gastrocnemius muscle showing Sirius Red (SR) staining of collagens between muscle fibers in patients without PAD and patients with PAD, quantified in (B). Scale bar=200 μm. Non‐PAD (n=7), PAD (n=24). Data are expressed as mean±SD; P values adjusted for race, *significance P≤0.05. C, Correlation of collagen content (SR+ area) with extracellular matrix (ECM) content (α‐wheat germ agglutinin [WGA]+ area) in all patients (n=31), determined by Pearson product moment correlation, r=partial correlation coefficient.
Figure 7. Cluster of differentiation (CD) 206−…
Figure 7. Cluster of differentiation (CD) 206− macrophages in gastrocnemius muscle are associated with poor walking performance in patients with peripheral artery disease (PAD).
Scatterplots showing correlations between the abundance of CD206− macrophages and functional performance measured with (A) 4‐meter normal pace test (95% CI of r=−0.726 to −0.021), (B) 4‐meter fast pace test (95% CI of r=−0.762 to −0.101) For all measures, relationships determined by Pearson product moment correlation adjusting for race, r=partial correlation coefficient, PAD (n=23).

References

    1. Fowkes FG, Aboyans V, Fowkes FJ, McDermott MM, Sampson UK, Criqui MH. Peripheral artery disease: epidemiology and global perspectives. Nat Rev Cardiol. 2017;14:156–170.
    1. McDermott MM, Greenland P, Liu K, Guralnik JM, Criqui MH, Dolan NC, Chan C, Celic L, Pearce WH, Schneider JR, et al. Leg symptoms in peripheral arterial disease: associated clinical characteristics and functional impairment. JAMA. 2001;286:1599–1606.
    1. McDermott MM, Liu K, Greenland P, Guralnik JM, Criqui MH, Chan C, Pearce WH, Schneider JR, Ferrucci L, Celic L, et al. Functional decline in peripheral arterial disease: associations with the ankle brachial index and leg symptoms. JAMA. 2004;292:453–461.
    1. McDermott MM. Functional impairment in peripheral artery disease and how to improve it in 2013. Curr Cardiol Rep. 2013;15:347.
    1. McDermott MM, Greenland P, Liu K, Guralnik JM, Celic L, Criqui MH, Chan C, Martin GJ, Schneider J, Pearce WH, et al. The ankle brachial index is associated with leg function and physical activity: the Walking and Leg Circulation Study. Ann Intern Med. 2002;136:​873–883.
    1. McDermott MM, Kibbe MR. Improving lower extremity functioning in peripheral artery disease: exercise, endovascular revascularization, or both? JAMA. 2017;317:689–690.
    1. Hamburg NM, Creager MA. Pathophysiology of intermittent claudication in peripheral artery disease. Circ J. 2017;81:281–289.
    1. Gillani S, Cao J, Suzuki T, Hak DJ. The effect of ischemia reperfusion injury on skeletal muscle. Injury. 2012;43:670–675.
    1. Koutakis P, Ismaeel A, Farmer P, Purcell S, Smith RS, Eidson JL, Bohannon WT. Oxidative stress and antioxidant treatment in patients with peripheral artery disease. Physiol Rep. 2018;6:e13650.
    1. Paradis S, Charles AL, Meyer A, Lejay A, Scholey JW, Chakfe N, Zoll J, Geny B. Chronology of mitochondrial and cellular events during skeletal muscle ischemia‐reperfusion. Am J Physiol Cell Physiol. 2016;310:C968–C982.
    1. Bunte MC, Shishehbor MH. Next generation endovascular therapies in peripheral artery disease. Prog Cardiovasc Dis. 2018;60:593–599.
    1. Pandey A, Banerjee S, Ngo C, Mody P, Marso SP, Brilakis ES, Armstrong EJ, Giri J, Bonaca MP, Pradhan A, et al. Comparative efficacy of endovascular revascularization versus supervised exercise training in patients with intermittent claudication: meta‐analysis of randomized controlled trials. JACC Cardiovasc Interv. 2017;10:712–724.
    1. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER, Cohen DJ, Reynolds MR, Massaro JM, Lewis BA, Cerezo J, Oldenburg NC, et al. Supervised exercise versus primary stenting for claudication resulting from aortoiliac peripheral artery disease: six‐month outcomes from the claudication: exercise versus endoluminal revascularization (CLEVER) study. Circulation. 2012;125:130–139.
    1. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER III, Cohen DJ, Reynolds MR, Massaro JM, Lewis BA, Cerezo J, Oldenburg NC, et al. Supervised exercise, stent revascularization, or medical therapy for claudication due to aortoiliac peripheral artery disease: the CLEVER study. J Am Coll Cardiol. 2015;65:999–1009.
    1. Olin JW, White CJ, Armstrong EJ, Kadian‐Dodov D, Hiatt WR. Peripheral artery disease: evolving role of exercise, medical therapy, and endovascular options. J Am Coll Cardiol. 2016;67:1338–1357.
    1. Chazaud B, Brigitte M, Yacoub‐Youssef H, Arnold L, Gherardi R, Sonnet C, Lafuste P, Chretien F. Dual and beneficial roles of macrophages during skeletal muscle regeneration. Exerc Sport Sci Rev. 2009;37:18–22.
    1. Kharraz Y, Guerra J, Mann CJ, Serrano AL, Munoz‐Canoves P. Macrophage plasticity and the role of inflammation in skeletal muscle repair. Mediators Inflamm. 2013;2013:491497.
    1. Saclier M, Cuvellier S, Magnan M, Mounier R, Chazaud B. Monocyte/macrophage interactions with myogenic precursor cells during skeletal muscle regeneration. FEBS J. 2013;280:4118–4130.
    1. Sciorati C, Rigamonti E, Manfredi AA, Rovere‐Querini P. Cell death, clearance and immunity in the skeletal muscle. Cell Death Differ. 2016;23:927–937.
    1. Tidball JG, Dorshkind K, Wehling‐Henricks M. Shared signaling systems in myeloid cell‐mediated muscle regeneration. Development. 2014;141:1184–1196.
    1. Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1173–R1187.
    1. Mackey AL, Kjaer M. The breaking and making of healthy adult human skeletal muscle in vivo. Skelet Muscle. 2017;7:24.
    1. Tidball JG. Inflammatory processes in muscle injury and repair. Am J Physiol Regul Integr Comp Physiol. 2005;288:R345–R353.
    1. Arnold L, Henry A, Poron F, Baba‐Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204:1057–1069.
    1. Beneke A, Guentsch A, Hillemann A, Zieseniss A, Swain L, Katschinski DM. Loss of PHD3 in myeloid cells dampens the inflammatory response and fibrosis after hind‐limb ischemia. Cell Death Dis. 2017;8:e2976.
    1. Braga TT, Agudelo JS, Camara NO. Macrophages during the fibrotic process: M2 as friend and foe. Front Immunol. 2015;6:602.
    1. Mann CJ, Perdiguero E, Kharraz Y, Aguilar S, Pessina P, Serrano AL, Munoz‐Canoves P. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle. 2011;1:21.
    1. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30:245–257.
    1. Lolmede K, Campana L, Vezzoli M, Bosurgi L, Tonlorenzi R, Clementi E, Bianchi ME, Cossu G, Manfredi AA, Brunelli S, et al. Inflammatory and alternatively activated human macrophages attract vessel‐associated stem cells, relying on separate HMGB1‐ and MMP‐9‐dependent pathways. J Leukoc Biol. 2009;85:779–787.
    1. Lu H, Huang D, Saederup N, Charo IF, Ransohoff RM, Zhou L. Macrophages recruited via CCR2 produce insulin‐like growth factor‐1 to repair acute skeletal muscle injury. FASEB J. 2011;25:358–369.
    1. Tonkin J, Temmerman L, Sampson RD, Gallego‐Colon E, Barberi L, Bilbao D, Schneider MD, Musaro A, Rosenthal N. Monocyte/macrophage‐derived IGF‐1 orchestrates murine skeletal muscle regeneration and modulates autocrine polarization. Mol Ther. 2015;23:1189–1200.
    1. Dumont N, Frenette J. Macrophages protect against muscle atrophy and promote muscle recovery in vivo and in vitro: a mechanism partly dependent on the insulin‐like growth factor‐1 signaling molecule. Am J Pathol. 2010;176:2228–2235.
    1. Brack AS, Rando TA. Tissue‐specific stem cells: lessons from the skeletal muscle satellite cell. Cell Stem Cell. 2012;10:504–514.
    1. Lepper C, Partridge TA, Fan CM. An absolute requirement for Pax7‐positive satellite cells in acute injury‐induced skeletal muscle regeneration. Development. 2011;138:3639–3646.
    1. Saclier M, Yacoub‐Youssef H, Mackey AL, Arnold L, Ardjoune H, Magnan M, Sailhan F, Chelly J, Pavlath GK, Mounier R, et al. Differentially activated macrophages orchestrate myogenic precursor cell fate during human skeletal muscle regeneration. Stem Cells. 2013;31:384–396.
    1. Pande RL, Brown J, Buck S, Redline W, Doyle J, Plutzky J, Creager MA. Association of monocyte tumor necrosis factor alpha expression and serum inflammatory biomarkers with walking impairment in peripheral artery disease. J Vasc Surg. 2015;61:155–161.
    1. Brevetti G, Giugliano G, Brevetti L, Hiatt WR. Inflammation in peripheral artery disease. Circulation. 2010;122:1862–1875.
    1. Tzoulaki I, Murray GD, Lee AJ, Rumley A, Lowe GD, Fowkes FG. C‐reactive protein, interleukin‐6, and soluble adhesion molecules as predictors of progressive peripheral atherosclerosis in the general population: Edinburgh Artery Study. Circulation. 2005;112:976–983.
    1. Wildman RP, Muntner P, Chen J, Sutton‐Tyrrell K, He J. Relation of inflammation to peripheral arterial disease in the National Health and Nutrition Examination Survey, 1999–2002. Am J Cardiol. 2005;96:1579–1583.
    1. Tzoulaki I, Murray GD, Lee AJ, Rumley A, Lowe GD, Fowkes FG. Inflammatory, haemostatic, and rheological markers for incident peripheral arterial disease: Edinburgh Artery Study. Eur Heart J. 2007;28:354–362.
    1. McDermott MM, Guralnik JM, Corsi A, Albay M, Macchi C, Bandinelli S, Ferrucci L. Patterns of inflammation associated with peripheral arterial disease: the INCHIANTI study. Am Heart J. 2005;150:276–281.
    1. McDermott MM, Green D, Greenland P, Liu K, Criqui MH, Chan C, Guralnik JM, Pearce WH, Ridker PM, Taylor L, et al. Relation of levels of hemostatic factors and inflammatory markers to the ankle brachial index. Am J Cardiol. 2003;92:194–199.
    1. McDermott MM, Liu K, Ferrucci L, Tian L, Guralnik JM, Tao H, Ridker PM, Criqui MH. Relation of interleukin‐6 and vascular cellular adhesion molecule‐1 levels to functional decline in patients with lower extremity peripheral arterial disease. Am J Cardiol. 2011;107:1392–1398.
    1. Ozkaramanli Gur D, Guzel S, Akyuz A, Alpsoy S, Guler N. The role of novel cytokines in inflammation: defining peripheral artery disease among patients with coronary artery disease. Vasc Med. 2018;23:428–436. DOI: 10.1177/1358863X18763096
    1. McDermott MM, Ferrucci L, Guralnik JM, Tian L, Green D, Liu K, Tan J, Liao Y, Pearce WH, Schneider JR, et al. Elevated levels of inflammation, d‐dimer, and homocysteine are associated with adverse calf muscle characteristics and reduced calf strength in peripheral arterial disease. J Am Coll Cardiol. 2007;50:897–905.
    1. McDermott MM, Ferrucci L, Liu K, Criqui MH, Greenland P, Green D, Guralnik JM, Ridker PM, Taylor LM, Rifai N, et al. D‐dimer and inflammatory markers as predictors of functional decline in men and women with and without peripheral arterial disease. J Am Geriatr Soc. 2005;53:1688–1696.
    1. McDermott MM, Greenland P, Green D, Guralnik JM, Criqui MH, Liu K, Chan C, Pearce WH, Taylor L, Ridker PM, et al. D‐dimer, inflammatory markers, and lower extremity functioning in patients with and without peripheral arterial disease. Circulation. 2003;107:3191–3198.
    1. Ganta VC, Choi M, Farber CR, Annex BH. Antiangiogenic VEGF165b regulates macrophage polarization via S100A8/S100A9 in peripheral artery disease. Circulation. 2019;139:226–242.
    1. Patel AS, Smith A, Nucera S, Biziato D, Saha P, Attia RQ, Humphries J, Mattock K, Grover SP, Lyons OT, et al. TIE2‐expressing monocytes/macrophages regulate revascularization of the ischemic limb. EMBO Mol Med. 2013;5:858–869.
    1. Ganta VC, Choi MH, Kutateladze A, Fox TE, Farber CR, Annex BH. A microRNA93‐interferon regulatory factor‐9‐immunoresponsive gene‐1‐itaconic acid pathway modulates M2‐like macrophage polarization to revascularize ischemic muscle. Circulation. 2017;135:2403–2425.
    1. Pellegrin M, Bouzourene K, Poitry‐Yamate C, Mlynarik V, Feihl F, Aubert JF, Gruetter R, Mazzolai L. Experimental peripheral arterial disease: new insights into muscle glucose uptake, macrophage, and T‐cell polarization during early and late stages. Physiol Rep. 2014;2:e00234.
    1. Liu Y, Chen L, Shen Y, Tan T, Xie N, Luo M, Li Z, Xie X. Curcumin ameliorates ischemia‐induced limb injury through immunomodulation. Med Sci Monit. 2016;22:2035–2042.
    1. Hsieh PL, Rybalko V, Baker AB, Suggs LJ, Farrar RP. Recruitment and therapeutic application of macrophages in skeletal muscles after hind limb ischemia. J Vasc Surg. 2018;67:1908–1920.e1901.
    1. Shireman PK. The chemokine system in arteriogenesis and hind limb ischemia. J Vasc Surg. 2007;45(suppl A):A48–A56.
    1. Jetten N, Donners MM, Wagenaar A, Cleutjens JP, van Rooijen N, de Winther MP, Post MJ. Local delivery of polarized macrophages improves reperfusion recovery in a mouse hind limb ischemia model. PLoS One. 2013;8:e68811.
    1. Zhu H, Zhang M, Liu Z, Xing J, Moriasi C, Dai X, Zou MH. AMP‐activated protein kinase alpha1 in macrophages promotes collateral remodeling and arteriogenesis in mice in vivo. Arterioscler Thromb Vasc Biol. 2016;36:1868–1878.
    1. Corona BT, Rathbone CR. Accelerated functional recovery after skeletal muscle ischemia‐reperfusion injury using freshly isolated bone marrow cells. J Surg Res. 2014;188:100–109.
    1. Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MP, Donners MM. Anti‐inflammatory M2, but not pro‐inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 2014;17:109–118.
    1. Kosmac K, Peck BD, Walton RG, Mula J, Kern PA, Bamman MM, Dennis RA, Jacobs CA, Lattermann C, Johnson DL, et al. Immunohistochemical identification of human skeletal muscle macrophages. Bio Protoc. 2018;8:e2883.
    1. Walton RG, Kosmac K, Mula J, Fry CS, Peck BD, Groshong JS, Finlin BS, Zhu B, Kern PA, Peterson CA. Human skeletal muscle macrophages increase following cycle training and are associated with adaptations that may facilitate growth. Sci Rep. 2019;9:969.
    1. McDermott MM, Liu K, Carr J, Criqui MH, Tian L, Li D, Ferrucci L, Guralnik JM, Kramer CM, Yuan C, et al. Superficial femoral artery plaque, the ankle‐brachial index, and leg symptoms in peripheral arterial disease: the Walking and Leg Circulation Study (WALCS) III. Circ Cardiovasc Imaging. 2011;4:246–252.
    1. McDermott MM, Liu K, Carroll TJ, Kibbe M, Ferrucci L, Guralnik JM, Morasch M, Pearce W, Carr J, Yuan C, et al. Plaque characteristics in the superficial femoral artery correlate with walking impairment questionnaire scores in peripheral arterial disease: the Walking and Leg Circulation Study (WALCS) III. J Surg Radiol. 2012;3:148–157.
    1. McDermott MM, Liu K, Carroll TJ, Tian L, Ferrucci L, Li D, Carr J, Guralnik JM, Kibbe M, Pearce WH, et al. Superficial femoral artery plaque and functional performance in peripheral arterial disease: Walking and Leg Circulation Study (WALCS III). JACC Cardiovasc Imaging. 2011;4:730–739.
    1. McDermott MM, Greenland P, Liu K, Tian L, Green D, Shah SJ, Huffman M, Wilkins J, Kibbe M, Liao Y, et al. Vulnerable blood in high risk vascular patients: study design and methods. Contemp Clin Trials. 2014;38:121–129.
    1. McDermott MM, Liu K, Green D, Greenland P, Tian L, Kibbe M, Tracy R, Shah SJ, Wilkins J, Huffman M, et al. Changes in D‐dimer and inflammatory biomarkers before ischemic events in patients with peripheral artery disease: the BRAVO study. Vasc Med. 2016;21:12–20.
    1. McDermott MM, Liu K, Guralnik JM, Criqui MH, Spring B, Tian L, Domanchuk K, Ferrucci L, Lloyd‐Jones D, Kibbe M, et al. Home‐based walking exercise intervention in peripheral artery disease: a randomized clinical trial. JAMA. 2013;310:57–65.
    1. McDermott MM, Domanchuk K, Liu K, Guralnik JM, Tian L, Criqui MH, Ferrucci L, Kibbe M, Jones DL, Pearce W, et al. The Group Oriented Arterial Leg Study (GOALS) to improve walking performance in patients with peripheral artery disease. Contemp Clin Trials. 2012;33:1311–1320.
    1. Domanchuk K, Ferrucci L, Guralnik JM, Criqui MH, Tian L, Liu K, Losordo D, Stein J, Green D, Kibbe M, et al. Progenitor cell release plus exercise to improve functional performance in peripheral artery disease: the PROPEL Study. Contemp Clin Trials. 2013;36:502–509.
    1. White SH, McDermott MM, Sufit RL, Kosmac K, Bugg AW, Gonzalez‐Freire M, Ferrucci L, Tian L, Zhao L, Gao Y, et al. Walking performance is positively correlated to calf muscle fiber size in peripheral artery disease subjects, but fibers show aberrant mitophagy: an observational study. J Transl Med. 2016;14:284.
    1. Criqui MH, Denenberg JO, Bird CE, Fronek A, Klauber MR, Langer RD. The correlation between symptoms and non‐invasive test results in patients referred for peripheral arterial disease testing. Vasc Med. 1996;1:65–71.
    1. McDermott MM, Criqui MH, Liu K, Guralnik JM, Greenland P, Martin GJ, Pearce W. Lower ankle/brachial index, as calculated by averaging the dorsalis pedis and posterior tibial arterial pressures, and association with leg functioning in peripheral arterial disease. J Vasc Surg. 2000;32:1164–1171.
    1. Shadman R, Criqui MH, Bundens WP, Fronek A, Denenberg JO, Gamst AC, McDermott MM. Subclavian artery stenosis: prevalence, risk factors, and association with cardiovascular diseases. J Am Coll Cardiol. 2004;44:618–623.
    1. McDermott MM, Tian L, Liu K, Guralnik JM, Ferrucci L, Tan J, Pearce WH, Schneider JR, Criqui MH. Prognostic value of functional performance for mortality in patients with peripheral artery disease. J Am Coll Cardiol. 2008;51:1482–1489.
    1. McDermott MM, Guralnik JM, Tian L, Ferrucci L, Liu K, Liao Y, Criqui MH. Baseline functional performance predicts the rate of mobility loss in persons with peripheral arterial disease. J Am Coll Cardiol. 2007;50:974–982.
    1. Guralnik JM, Ferrucci L, Simonsick EM, Salive ME, Wallace RB. Lower‐extremity function in persons over the age of 70 years as a predictor of subsequent disability. N Engl J Med. 1995;332:556–561.
    1. Guralnik JM, Ferrucci L, Pieper CF, Leveille SG, Markides KS, Ostir GV, Studenski S, Berkman LF, Wallace RB. Lower extremity function and subsequent disability: consistency across studies, predictive models, and value of gait speed alone compared with the short physical performance battery. J Gerontol A Biol Sci Med Sci. 2000;55:M221–M231.
    1. Emde B, Heinen A, Godecke A, Bottermann K. Wheat germ agglutinin staining as a suitable method for detection and quantification of fibrosis in cardiac tissue after myocardial infarction. Eur J Histochem. 2014;58:2448.
    1. Crawford F, Welch K, Andras A, Chappell FM. Ankle brachial index for the diagnosis of lower limb peripheral arterial disease. Cochrane Database Syst Rev. 2016;9:CD010680.
    1. Kirkeby S, Mandel U, Vedtofte P. Identification of capillaries in sections from skeletal muscle by use of lectins and monoclonal antibodies reacting with histo‐blood group ABH antigens. Glycoconj J. 1993;10:181–188.
    1. McDermott MM, Polonsky TS, Kibbe MR, Tian L, Zhao L, Pearce WH, Gao Y, Guralnik JM. Racial differences in functional decline in peripheral artery disease and associations with socioeconomic status and education. J Vasc Surg. 2017;66:826–834.
    1. McDermott MM, Polonsky TS, Guralnik JM, Ferrucci L, Tian L, Zhao L, Stein J, Domanchuk K, Criqui MH, Taylor DA, et al. Racial differences in the effect of granulocyte macrophage colony‐stimulating factor on improved walking distance in peripheral artery disease: the PROPEL randomized clinical trial. J Am Heart Assoc. 2019;8:e011001 DOI: 10.1161/JAHA.118.011001
    1. Ceafalan LC, Fertig TE, Popescu AC, Popescu BO, Hinescu ME, Gherghiceanu M. Skeletal muscle regeneration involves macrophage‐myoblast bonding. Cell Adh Migr. 2018;12:228–235.
    1. Du H, Shih CH, Wosczyna MN, Mueller AA, Cho J, Aggarwal A, Rando TA, Feldman BJ. Macrophage‐released ADAMTS1 promotes muscle stem cell activation. Nat Commun. 2017;8:669.
    1. Capote J, Kramerova I, Martinez L, Vetrone S, Barton ER, Sweeney HL, Miceli MC, Spencer MJ. Osteopontin ablation ameliorates muscular dystrophy by shifting macrophages to a pro‐regenerative phenotype. J Cell Biol. 2016;213:275–288.
    1. Madaro L, Torcinaro A, De Bardi M, Contino FF, Pelizzola M, Diaferia GR, Imeneo G, Bouche M, Puri PL, De Santa F. Macrophages fine tune satellite cell fate in dystrophic skeletal muscle of mdx mice. PLoS Genet. 2019;15:e1008408.
    1. Wehling‐Henricks M, Welc SS, Samengo G, Rinaldi C, Lindsey C, Wang Y, Lee J, Kuro OM, Tidball JG. Macrophages escape Klotho gene silencing in the mdx mouse model of Duchenne muscular dystrophy and promote muscle growth and increase satellite cell numbers through a Klotho‐mediated pathway. Hum Mol Genet. 2018;27:14–29.
    1. Mansour Z, Bouitbir J, Charles AL, Talha S, Kindo M, Pottecher J, Zoll J, Geny B. Remote and local ischemic preconditioning equivalently protects rat skeletal muscle mitochondrial function during experimental aortic cross‐clamping. J Vasc Surg. 2012;55:497–505.e491.
    1. Kocman EA, Ozatik O, Sahin A, Guney T, Kose AA, Dag I, Alatas O, Cetin C. Effects of ischemic preconditioning protocols on skeletal muscle ischemia‐reperfusion injury. J Surg Res. 2015;193:942–952.
    1. Thaveau F, Zoll J, Rouyer O, Chafke N, Kretz JG, Piquard F, Geny B. Ischemic preconditioning specifically restores complexes I and II activities of the mitochondrial respiratory chain in ischemic skeletal muscle. J Vasc Surg. 2007;46:541–547; discussion 547.
    1. Tang GL, Chang DS, Sarkar R, Wang R, Messina LM. The effect of gradual or acute arterial occlusion on skeletal muscle blood flow, arteriogenesis, and inflammation in rat hindlimb ischemia. J Vasc Surg. 2005;41:312–320.
    1. Yang Y, Tang G, Yan J, Park B, Hoffman A, Tie G, Wang R, Messina LM. Cellular and molecular mechanism regulating blood flow recovery in acute versus gradual femoral artery occlusion are distinct in the mouse. J Vasc Surg. 2008;48:1546–1558.
    1. McDermott MM, Hoff F, Ferrucci L, Pearce WH, Guralnik JM, Tian L, Liu K, Schneider JR, Sharma L, Tan J, et al. Lower extremity ischemia, calf skeletal muscle characteristics, and functional impairment in peripheral arterial disease. J Am Geriatr Soc. 2007;55:400–406.
    1. Baum O, Torchetti E, Malik C, Hoier B, Walker M, Walker PJ, Odriozola A, Graber F, Tschanz SA, Bangsbo J, et al. Capillary ultrastructure and mitochondrial volume density in skeletal muscle in relation to reduced exercise capacity of patients with intermittent claudication. Am J Physiol Regul Integr Comp Physiol. 2016;310:R943–R951.
    1. Ho TK, Rajkumar V, Black CM, Abraham DJ, Baker DM. Increased angiogenic response but deficient arteriolization and abnormal microvessel ultrastructure in critical leg ischaemia. Br J Surg. 2006;93:1368–1376.
    1. Keeling AN, Carroll TJ, McDermott MM, Liu K, Liao Y, Farrelly CT, Pearce WH, Carr J. Clinical correlates of size and number of collateral vessels in peripheral artery disease. Vasc Med. 2012;17:223–230.
    1. McGuigan MR, Bronks R, Newton RU, Sharman MJ, Graham JC, Cody DV, Kraemer WJ. Muscle fiber characteristics in patients with peripheral arterial disease. Med Sci Sports Exerc. 2001;33:2016–2021.
    1. Askew CD, Green S, Walker PJ, Kerr GK, Green AA, Williams AD, Febbraio MA. Skeletal muscle phenotype is associated with exercise tolerance in patients with peripheral arterial disease. J Vasc Surg. 2005;41:802–807.
    1. Jones WS, Duscha BD, Robbins JL, Duggan NN, Regensteiner JG, Kraus WE, Hiatt WR, Dokun AO, Annex BH. Alteration in angiogenic and anti‐angiogenic forms of vascular endothelial growth factor‐A in skeletal muscle of patients with intermittent claudication following exercise training. Vasc Med. 2012;17:94–100.
    1. Robbins JL, Jones WS, Duscha BD, Allen JD, Kraus WE, Regensteiner JG, Hiatt WR, Annex BH. Relationship between leg muscle capillary density and peak hyperemic blood flow with endurance capacity in peripheral artery disease. J Appl Physiol (1985). 2011;111:​81–86.
    1. Ha DM, Carpenter LC, Koutakis P, Swanson SA, Zhu Z, Hanna M, DeSpiegelaere HK, Pipinos II, Casale GP. Transforming growth factor‐beta 1 produced by vascular smooth muscle cells predicts fibrosis in the gastrocnemius of patients with peripheral artery disease. J Transl Med. 2016;14:39.
    1. Baum O, Djonov V, Ganster M, Widmer M, Baumgartner I. Arteriolization of capillaries and FGF‐2 upregulation in skeletal muscles of patients with chronic peripheral arterial disease. Microcirculation. 2005;12:527–537.
    1. He S, Gleason J, Fik‐Rymarkiewicz E, DiFiglia A, Bharathan M, Morschauser A, Djuretic I, Xu Y, Krakovsky M, Jankovic V, et al. Human placenta‐derived mesenchymal stromal‐like cells enhance angiogenesis via T cell‐dependent reprogramming of macrophage differentiation. Stem Cells. 2017;35:1603–1613.
    1. Tepekoylu C, Lobenwein D, Urbschat A, Graber M, Pechriggl EJ, Fritsch H, Paulus P, Grimm M, Holfeld J. Shock wave treatment after hindlimb ischaemia results in increased perfusion and M2 macrophage presence. J Tissue Eng Regen Med. 2018;12:e486–e494.
    1. Takeda Y, Costa S, Delamarre E, Roncal C, Leite de Oliveira R, Squadrito ML, Finisguerra V, Deschoemaeker S, Bruyere F, Wenes M, et al. Macrophage skewing by Phd2 haplodeficiency prevents ischaemia by inducing arteriogenesis. Nature. 2011;479:122–126.
    1. Brechot N, Gomez E, Bignon M, Khallou‐Laschet J, Dussiot M, Cazes A, Alanio‐Brechot C, Durand M, Philippe J, Silvestre JS, et al. Modulation of macrophage activation state protects tissue from necrosis during critical limb ischemia in thrombospondin‐1‐deficient mice. PLoS One. 2008;3:e3950.
    1. Krishnasamy K, Limbourg A, Kapanadze T, Gamrekelashvili J, Beger C, Hager C, Lozanovski VJ, Falk CS, Napp LC, Bauersachs J, et al. Blood vessel control of macrophage maturation promotes arteriogenesis in ischemia. Nat Commun. 2017;8:952.

Source: PubMed

3
Tilaa