Post-translational regulation of PGC-1α modulates fibrotic repair

Jennifer L Larson-Casey, Linlin Gu, Dana Davis, Guo-Qiang Cai, Qiang Ding, Chao He, A Brent Carter, Jennifer L Larson-Casey, Linlin Gu, Dana Davis, Guo-Qiang Cai, Qiang Ding, Chao He, A Brent Carter

Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease associated with mitochondrial oxidative stress. Mitochondrial reactive oxygen species (mtROS) are important for cell homeostasis by regulating mitochondrial dynamics. Here, we show that IPF BAL cells exhibited increased mitochondrial biogenesis that is, in part, due to increased nuclear expression of peroxisome proliferator-activated receptor-ɣ (PPARɣ) coactivator (PGC)-1α. Increased PPARGC1A mRNA expression directly correlated with reduced pulmonary function in IPF subjects. Oxidant-mediated activation of the p38 MAPK via Akt1 regulated PGC-1α activation to increase mitochondrial biogenesis in monocyte-derived macrophages. Demonstrating the importance of PGC-1α in fibrotic repair, mice harboring a conditional deletion of Ppargc1a in monocyte-derived macrophages or mice administered a chemical inhibitor of mitochondrial division had reduced biogenesis and increased apoptosis, and the mice were protected from pulmonary fibrosis. These observations suggest that Akt1-mediated regulation of PGC-1α maintains mitochondrial homeostasis in monocyte-derived macrophages to induce apoptosis resistance, which contributes to the pathogenesis of pulmonary fibrosis.

Keywords: PGC-1α; mitochondrial biogenesis; monocyte-derived macrophages; pulmonary fibrosis.

Conflict of interest statement

The authors declare no conflicts of interest exists.

© 2021 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.

Figures

FIGURE 1
FIGURE 1
IPF BAL cells have increased mitochondrial biogenesis. A, Nuclear immunoblot analysis and B, quantification of PGC‐1α in BAL cells from normal and IPF subjects (n = 7). mRNA expression in BAL cells from normal (n = 5‐7) or IPF subjects (n = 5‐8) for C, PPARGC1A, TFAM, CS, and 12s rRNA. D, Ratio of mtDNA to nuclear DNA in normal and IPF BAL cells (n = 5). E, mRNA analysis of PPARGC1A in IPF BALC with silencing of PGC‐1α (n = 8). Inset, PGC‐1α immunoblot analysis. F, TFAM, CS, and COX4I1 expression (n = 8) in IPF BALC with silencing of PGC‐1α. Pearson’s correlation of PPARGC1A mRNA in BAL cells and G, FVC or H, FVC % predicted in IPF subjects (n = 5). I, Nuclear immunoblot of PGC‐1α in BAL cells from saline or bleomycin‐exposed mice at 21 days. J, mRNA analysis Ppargc1a, Tfam, Cs, and Cox4i1 mRNA in BAL cells from saline or bleomycin‐exposed mice at 21 days (n = 5). *P < .05; **P < .001; ***P < .0001. Values shown as mean ± S.E.M. Two‐tailed t‐test statistical analysis was utilized for B‐F, J. Pearson’s coefficient was used for G, H
FIGURE 2
FIGURE 2
Akt1 promotes mitochondrial biogenesis. A, MitoTracker green staining of MH‐S cells transfected with empty (Emp) (n = 4) or Akt1CA (n = 4). Scale bars represent 10 μm. B, Flow cytometry of transfected THP‐1 cells (n = 3). BAL cells were isolated at indicated days after saline (S) (n = 5/time point) or bleomycin (B, bleo) exposure (n = 5/time point) from Akt1fl/fl or Akt1−/−Lyz2‐cre mice (Akt1−/−). C, Immunoblot analysis and D, quantification of p‐Akt1 (n = 5). E, Representative histology of lung sections with Masson’s trichome staining (n = 5). Scale bar = 500 μm. F, Hydroxyproline analysis of lung homogenates (n = 5). mRNA analysis of G, Ppargc1a (n = 5), H, Tfam (n = 5), and I, Cs (n = 5). J, TEM analysis of BAL cells (n = 8) from Akt1fl/fl and Akt1−/−Lyz2‐cre mice. Scale bar represents 1 μm. K, Total number of mitochondria per BAL cell (n = 8). *P < .05; **P < .001; ***P < .0001. Values shown as mean ± S.E.M. Two‐tailed t test statistical analysis was utilized for B. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized for D, F, K. Two‐way ANOVA followed by Bonferroni post‐test was utilized for G‐I. ***with open line in G‐I refer to Akt1fl/fl bleomycin vs Akt1fl/fl saline at 5, 10, 14, and 21 days and Akt1fl/fl bleomycin vs Akt1−/−Lyz2‐cre bleomycin at 5, 10, 14, and 21 days. Bracketed lines in G‐I is comparing Akt1fl/fl bleomycin at indicated timepoints
FIGURE 3
FIGURE 3
Monocyte‐derived macrophages are resistant to apoptosis. A, Caspase‐3 activity (n = 4‐6) in IPF BALC with silencing of PGC‐1α. BAL cells were isolated at indicated days after saline (n = 5/time point) or bleomycin exposure (n = 5/time point) from Akt1fl/fl or Akt1−/−Lyz2‐cre mice. B, TUNEL staining (n = 5) and C, quantification (n = 5). Scale bar represents 40 μm. BAL cells were isolated 21 days after exposure. D, Representative flow cytometry plots of monocyte‐derived macrophages (MDM, CD45+CD11b+/−Ly6G−CD64+Ly6c−Siglec Flow) and resident alveolar macrophages (RAM, CD45+CD11b+/−Ly6G−CD64+Ly6c−Siglec Fhi) from saline or bleomycin‐exposed Akt1fl/fl and Akt1−/−Lyz2‐cre mice. Total cell number of (E) MDM (n = 5), F, annexin V positive MDM (n = 5), (G) RAM (n = 5), and H, annexin V positive RAM from Akt1fl/fl and Akt1−/−Lyz2‐cre BAL (n = 5). **P < .001; ***P < .0001. Values shown as mean ± S.E.M. Two‐tailed t‐test statistical analysis was utilized for A. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized for C, E‐H
FIGURE 4
FIGURE 4
Pulmonary fibrosis does not require Akt2. Akt2fl/fl or Akt2−/−Lyz2‐cre mice were exposed to saline or bleomycin (Bleo). BAL cells were isolated 21 days later. A, TUNEL staining (n = 5), B, TUNEL quantification (n = 4‐5), and C, caspase‐3 activity (n = 5). Scale bar represent 50 μm. mRNA analysis of D, Ppargc1a (n = 5‐6), E, Tfam (n = 5‐6), F, Cs (n = 5‐6), and G, Cox4i1 expression (n = 5‐6) in BAL cells. H, Histology of lung sections with Masson’s trichrome staining (n = 5) and I, hydroxyproline analysis of homogenized lung (n = 5‐6). ***P < .0001. Values shown as mean ± S.E.M. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized
FIGURE 5
FIGURE 5
Akt1‐mediated mtROS activates PGC‐1α via phosphorylation of p38 MAPK. A, Nuclear immunoblot analysis for p‐PGC‐1α (S570) and PGC‐1α in transfected macrophages. B, PPARGC1A promoter activity in transfected macrophages (n = 8). Inset, p‐Akt1 and p‐Akt2 immunoblot. C, Nuclear immunoblot analysis in transfected macrophages. D, Schematic of Akt2 phosphorylation and mutation sites on PGC‐1α. E, NFE2L2 mRNA analysis in macrophages expressing empty (emp), Akt1C(A) or Akt2CA together with PGC‐1αWT (WT) or PGC‐1αS570A (S570A) (n = 3). Macrophages were transfected with empty or Akt1CA together with MKK6(Glu) or p38DN. F, PPARGC1A mRNA expression (n = 3), G, nuclear immunoblot analysis, H, quantification of PGC‐1α expression (n = 3); inset, Akt1 immunoblot analysis, I, NRF1 mRNA expression (n = 3). J, Schematic of p38MAPK phosphorylation and mutation sites on PGC‐1α. K, NFE2L2 mRNA analysis of macrophages expressing empty or Akt1CA together with PGC‐1αWT (WT), PGC‐1αT262A (T262A), PGC‐1αS265A (S265A), PGC‐1αT298A (T298A) (n = 3). L, Immunoblot analysis of BAL cells from saline or bleomycin‐exposed Akt1fl/fl and Akt1−/−Lyz2‐cre mice. Macrophages were transfected with scramble or PGC‐1α siRNA and empty or Akt1CA. M, Mitochondrial DNA content (n = 3) and N, Tfam mRNA expression (n = 3). Inset, PGC‐1α mRNA expression. O, Nuclear immunoblot analysis in macrophages expressing empty or Akt1CA and treated with vehicle or mitoTEMPO (10 µM, overnight). **P < .001; ***, P < .0001. Values shown as mean ± S.E.M. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized. CPD = Cdc4 phosphodegron motifs
FIGURE 6
FIGURE 6
Mitochondrial division inhibitor prevents bleomycin‐induced pulmonary fibrosis. A, Immunoblot analysis and B, quantification of p‐Drp1 (S616) expression in BAL cells from normal (n = 5) or IPF subjects (n = 4). C, TUNEL staining (n = 5) and D, TUNEL quantification (n = 5) of IPF BAL cells treated with vehicle or mdivi‐1 (20 µM, overnight). Scale bar represents 50 μm. C57BL/6J WT mice were exposed to saline or bleomycin (bleo), 10 days after exposure mice were administered daily ip injections of vehicle or mdivi‐1. BAL cells were isolated 21 days later. E, Immunoblot analysis in BAL cells. F, TUNEL staining (n = 5), G, TUNEL quantification (n = 5), and H, Caspase‐3 activity (n = 6), Scale bar represent 50 μm. mRNA analysis of I, Ppargc1a (n = 5), J, Tfam (n = 5), K, Cs (n = 5), and L, Cox4i1 expression (n = 5). M, Masson’s trichrome staining of lung sections (n = 5) and N, hydroxyproline analysis (n = 5). **P < .001; ***P < .0001. Values shown as mean ± S.E.M. Two‐tailed t‐test statistical analysis was utilized for B and D. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized for G‐L, N
FIGURE 7
FIGURE 7
Deletion of PGC‐1α in monocyte‐derived macrophages protects against pulmonary fibrosis. Ppargc1afl/fl or Ppargc1a−/−Csf1rMeriCreMer mice were administered tamoxifen (TAM) or regular (Reg) chow and exposed to saline or bleomycin (bleo). BAL cells were isolated 21 days later. A, mRNA analysis of PGC‐1α in BAL cells (n = 5‐6). B, TUNEL staining (n = 5‐6), and C, TUNEL quantification (n = 5‐6). Scale bar represents 50 μm. D, Tfam (n = 5), E, Cs (n = 5‐6), and F, Cox4i1 mRNA expression (n = 5‐6). G, mtDNA/nDNA in BAL cells (n = 4‐5). H, Masson’s trichrome staining of lung sections (n = 5‐6) and I, hydroxyproline analysis (n = 5). BAL cells were isolated from IPF patients by BAL. **P < .001; ***P < .0001. Values shown as mean ± S.E.M. One‐way ANOVA followed by Tukey’s multiple comparison test was utilized
FIGURE 8
FIGURE 8
PGC‐1α regulates fibrotic progression in monocyte‐derived macrophages. Lung injury promotes Akt1 activation and the generation of the mitochondrial ROS (mtROS), which induces p38 MAPK. The p38‐mediated increase of PGC‐1α activation occurs via Akt1 and augments the transcription of mitochondrial DNA genes, including TFAM and CS (citrate synthase) to induce apoptosis resistance in monocyte‐derived macrophages and fibrotic progression

References

    1. Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G. Incidence and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2006;174:810‐816.
    1. Hutchinson JP, McKeever TM, Fogarty AW, Navaratnam V, Hubbard RB. Increasing global mortality from idiopathic pulmonary fibrosis in the twenty‐first century. Ann Am Thorac Soc. 2014;11:1176‐1185.
    1. Raghu G, Collard HR, Egan JJ, et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence‐based guidelines for diagnosis and management. Am J Respir Crit Care Med. 2011;183:788‐824.
    1. Hutchinson J, Fogarty A, Hubbard R, McKeever T. Global incidence and mortality of idiopathic pulmonary fibrosis: a systematic review. Eur Respir J. 2015;46:795‐806.
    1. American Cancer Society Cancer Facts & Figures 2015. Atlanta, GA: American Cancer Society; 2015.
    1. King TE Jr, Bradford WZ, Castro‐Bernardini S et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083‐2092.
    1. Richeldi L, du Bois RM, Raghu G, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2071‐2082.
    1. He C, Ryan AJ, Murthy S, Carter AB. Accelerated development of pulmonary fibrosis via Cu, Zn‐superoxide dismutase‐induced alternative activation of macrophages. J Biol Chem. 2013;288:20745‐20757.
    1. Murthy S, Larson‐Casey JL, Ryan AJ, He C, Kobzik L, Carter AB. Alternative activation of macrophages and pulmonary fibrosis are modulated by scavenger receptor, macrophage receptor with collagenous structure. FASEB J. 2015;29:3527‐3536.
    1. Misharin AV, Morales‐Nebreda L, Reyfman PA, et al. Monocyte‐derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med. 2017;214:2387‐2404.
    1. McCubbrey AL, Barthel L, Mohning MP, et al. Deletion of c‐FLIP from CD11b(hi) macrophages prevents development of bleomycin‐induced lung fibrosis. Am J Respir Cell Mol Biol. 2018;58:66‐78.
    1. He C, Larson‐Casey JL, Davis D, et al. NOX4 modulates macrophage phenotype and mitochondrial biogenesis in asbestosis. JCI Insight. 2019;4(16). 10.1172/jci.insight.126551
    1. Larson‐Casey JL, Vaid M, Gu L, et al. Increased flux through the mevalonate pathway mediates fibrotic repair without injury. J Clin Invest. 2019;129:4962‐4978.
    1. Larson‐Casey JL, Deshane JS, Ryan AJ, Thannickal VJ, Carter AB. Macrophage Akt1 kinase‐mediated mitophagy modulates apoptosis resistance and pulmonary fibrosis. Immunity. 2016;44:582‐596.
    1. Twig G, Elorza A, Molina AJ, et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 2008;27:433‐446.
    1. Suliman HB, Carraway MS, Ali AS, Reynolds CM, Welty‐Wolf KE, Piantadosi CA. The CO/HO system reverses inhibition of mitochondrial biogenesis and prevents murine doxorubicin cardiomyopathy. J Clin Invest. 2007;117:3730‐3741.
    1. Ikeda Y, Shirakabe A, Maejima Y, et al. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res. 2015;116:264‐278.
    1. Palikaras K, Lionaki E, Tavernarakis N. Coordination of mitophagy and mitochondrial biogenesis during ageing in C. elegans . Nature. 2015;521:525‐528.
    1. Mannam P, Shinn AS, Srivastava A, et al. MKK3 regulates mitochondrial biogenesis and mitophagy in sepsis‐induced lung injury. Am J Physiol Lung Cell Mol Physiol. 2014;306:L604‐L619.
    1. Piantadosi CA, Carraway MS, Babiker A, Suliman HB. Heme oxygenase‐1 regulates cardiac mitochondrial biogenesis via Nrf2‐mediated transcriptional control of nuclear respiratory factor‐1. Circ Res. 2008;103:1232‐1240.
    1. Larsson NG, Wang J, Wilhelmsson H, et al. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat Genet. 1998;18:231‐236.
    1. St‐Pierre J, Drori S, Uldry M, et al. Suppression of reactive oxygen species and neurodegeneration by the PGC‐1 transcriptional coactivators. Cell. 2006;127:397‐408.
    1. He C, Murthy S, McCormick ML, Spitz DR, Ryan AJ, Carter AB. Mitochondrial Cu, Zn‐superoxide dismutase mediates pulmonary fibrosis by augmenting H2O2 generation. J Biol Chem. 2011;286:15597‐15607.
    1. Raingeaud J, Whitmarsh AJ, Barrett T, Derijard B, Davis RJ. MKK3‐ and MKK6‐regulated gene expression is mediated by the p38 mitogen‐activated protein kinase signal transduction pathway. Mol Cell Biol. 1996;16:1247‐1255.
    1. Larson‐Casey JL, Murthy S, Ryan AJ, Carter AB. Modulation of the mevalonate pathway by akt regulates macrophage survival and development of pulmonary fibrosis. J Biol Chem. 2014;289:36204‐36219.
    1. Handschin C, Rhee J, Lin J, Tarr PT, Spiegelman BM. An autoregulatory loop controls peroxisome proliferator‐activated receptor gamma coactivator 1alpha expression in muscle. Proc Natl Acad Sci USA. 2003;100:7111‐7116.
    1. Ichida M, Nemoto S, Finkel T. Identification of a specific molecular repressor of the peroxisome proliferator‐activated receptor gamma Coactivator‐1 alpha (PGC‐1alpha). J Biol Chem. 2002;277:50991‐50995.
    1. Gu L, Larson‐Casey JL, Carter AB. Macrophages utilize the mitochondrial calcium uniporter for profibrotic polarization. FASEB J. 2017;31:3072‐3083.
    1. Carter AB, Hunninghake GW. A constitutive active MEK ‐> ERK pathway negatively regulates NF‐kappa B‐dependent gene expression by modulating TATA‐binding protein phosphorylation. J Biol Chem. 2000;275:27858‐27864.
    1. Gu L, Larson Casey JL, Andrabi SA, et al. Mitochondrial calcium uniporter regulates PGC‐1alpha expression to mediate metabolic reprogramming in pulmonary fibrosis. Redox Biol. 2019;26:101307.
    1. Li X, Monks B, Ge Q, Birnbaum MJ. Akt/PKB regulates hepatic metabolism by directly inhibiting PGC‐1alpha transcription coactivator. Nature. 2007;447:1012‐1016.
    1. Wu Z, Puigserver P, Andersson U, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC‐1. Cell. 1999;98:115‐124.
    1. Knutti D, Kressler D, Kralli A. Regulation of the transcriptional coactivator PGC‐1 via MAPK‐sensitive interaction with a repressor. Proc Natl Acad Sci USA. 2001;98:9713‐9718.
    1. Bueno M, Lai Y‐C, Romero Y, et al. PINK1 deficiency impairs mitochondrial homeostasis and promotes lung fibrosis. J. Clin. Investig. 2015;125(2):521‐538. 10.1172/jci74942
    1. Qian BZ, Li J, Zhang H, et al. CCL2 recruits inflammatory monocytes to facilitate breast‐tumour metastasis. Nature. 2011;475:222‐225.
    1. Hughes R, Qian BZ, Rowan C, et al. Perivascular M2 macrophages stimulate tumor relapse after chemotherapy. Cancer Res. 2015;75:3479‐3491.
    1. Sheikh AQ, Saddouk FZ, Ntokou A, Mazurek R, Greif DM. Cell autonomous and non‐cell autonomous regulation of SMC progenitors in pulmonary hypertension. Cell Rep. 2018;23:1152‐1165.
    1. Scarpulla RC. Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene. 2002;286:81‐89.
    1. Zhao C, Tan YC, Wong WC, et al. The CD14(+/low)CD16(+) monocyte subset is more susceptible to spontaneous and oxidant‐induced apoptosis than the CD14(+)CD16(−) subset. Cell Death Dis. 2010;1:e95.
    1. Alzaid F, Lagadec F, Albuquerque M, et al. IRF5 governs liver macrophage activation that promotes hepatic fibrosis in mice and humans. JCI Insight. 2016;1:e88689.
    1. Narula J, Haider N, Virmani R, et al. Apoptosis in myocytes in end‐stage heart failure. N Engl J Med. 1996;335:1182‐1189.
    1. Qiu W, Wu B, Wang X, et al. PUMA‐mediated intestinal epithelial apoptosis contributes to ulcerative colitis in humans and mice. J Clin Invest. 2011;121:1722‐1732.
    1. Kaufmann T, Jost PJ, Pellegrini M, et al. Fatal hepatitis mediated by tumor necrosis factor TNFalpha requires caspase‐8 and involves the BH3‐only proteins Bid and Bim. Immunity. 2009;30:56‐66.
    1. Han L, Zhang EB, Yin DD, et al. Low expression of long noncoding RNA PANDAR predicts a poor prognosis of non‐small cell lung cancer and affects cell apoptosis by regulating Bcl‐2. Cell Death Dis. 2015;6:e1665.
    1. Shearn AI, Deswaerte V, Gautier EL, et al. Bcl‐x inactivation in macrophages accelerates progression of advanced atherosclerotic lesions in Apoe(−/−) mice. Arterioscler Thromb Vasc Biol. 2012;32:1142‐1149.
    1. Li MO, Sarkisian MR, Mehal WZ, Rakic P, Flavell RA. Phosphatidylserine receptor is required for clearance of apoptotic cells. Science. 2003;302:1560‐1563.
    1. Breunig C, Pahl J, Kublbeck M, et al. MicroRNA‐519a‐3p mediates apoptosis resistance in breast cancer cells and their escape from recognition by natural killer cells. Cell Death Dis. 2017;8:e2973.
    1. Atreya R, Mudter J, Finotto S, et al. Blockade of interleukin 6 trans signaling suppresses T‐cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat Med. 2000;6:583‐588.
    1. Meinecke I, Cinski A, Baier A, et al. Modification of nuclear PML protein by SUMO‐1 regulates Fas‐induced apoptosis in rheumatoid arthritis synovial fibroblasts. Proc Natl Acad Sci USA. 2007;104:5073‐5078.
    1. Redente EF, Keith RC, Janssen W, et al. Tumor necrosis factor‐alpha accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages. Am J Respir Cell Mol Biol. 2014;50:825‐837.
    1. Ginhoux F, Schultze JL, Murray PJ, Ochando J, Biswas SK. New insights into the multidimensional concept of macrophage ontogeny, activation and function. Nat Immunol. 2016;17:34‐40.
    1. Duffield JS, Forbes SJ, Constandinou CM, et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 2005;115:56‐65.
    1. Odegaard JI, Chawla A. Alternative macrophage activation and metabolism. Annu Rev Pathol. 2011;6:275‐297.
    1. Ichijo H, Nishida E, Irie K, et al. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. 1997;275:90‐94.
    1. Okamoto S, Krainc D, Sherman K, Lipton SA. Antiapoptotic role of the p38 mitogen‐activated protein kinase‐myocyte enhancer factor 2 transcription factor pathway during neuronal differentiation. Proc Natl Acad Sci USA. 2000;97:7561‐7566.
    1. Park JM, Greten FR, Li ZW, Karin M. Macrophage apoptosis by anthrax lethal factor through p38 MAP kinase inhibition. Science. 2002;297:2048‐2051.
    1. Seimon TA, Wang Y, Han S, et al. Macrophage deficiency of p38alpha MAPK promotes apoptosis and plaque necrosis in advanced atherosclerotic lesions in mice. J Clin Invest. 2009;119:886‐898.
    1. Luyendyk JP, Schabbauer GA, Tencati M, Holscher T, Pawlinski R, Mackman N. Genetic analysis of the role of the PI3K‐Akt pathway in lipopolysaccharide‐induced cytokine and tissue factor gene expression in monocytes/macrophages. J Immunol. 2008;180:4218‐4226.
    1. Puigserver P, Rhee J, Lin J, et al. Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator‐1. Mol Cell. 2001;8:971‐982.
    1. Cao W, Medvedev AV, Daniel KW, Collins S. beta‐Adrenergic activation of p38 MAP kinase in adipocytes: cAMP induction of the uncoupling protein 1 (UCP1) gene requires p38 MAP kinase. J Biol Chem. 2001;276:27077‐27082.
    1. Akimoto T, Pohnert SC, Li P, et al. Exercise stimulates Pgc‐1alpha transcription in skeletal muscle through activation of the p38 MAPK pathway. J Biol Chem. 2005;280:19587‐19593.
    1. Ptasinska A, Wang S, Zhang J, Wesley RA, Danner RL. Nitric oxide activation of peroxisome proliferator‐activated receptor gamma through a p38 MAPK signaling pathway. FASEB J. 2007;21:950‐961.
    1. Aquilano K, Vigilanza P, Baldelli S, Pagliei B, Rotilio G, Ciriolo MR. Peroxisome proliferator‐activated receptor gamma co‐activator 1alpha (PGC‐1alpha) and sirtuin 1 (SIRT1) reside in mitochondria: possible direct function in mitochondrial biogenesis. J Biol Chem. 2010;285:21590‐21599.
    1. Eschbach J, Schwalenstocker B, Soyal SM, et al. PGC‐1alpha is a male‐specific disease modifier of human and experimental amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22:3477‐3484.
    1. Arany Z, He H, Lin J, et al. Transcriptional coactivator PGC‐1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab. 2005;1:259‐271.
    1. Patti ME, Butte AJ, Crunkhorn S, et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc Natl Acad Sci USA. 2003;100:8466‐8471.
    1. Lin J, Wu PH, Tarr PT, et al. Defects in adaptive energy metabolism with CNS‐linked hyperactivity in PGC‐1alpha null mice. Cell. 2004;119:121‐135.
    1. Huss JM, Imahashi K, Dufour CR, et al. The nuclear receptor ERRalpha is required for the bioenergetic and functional adaptation to cardiac pressure overload. Cell Metab. 2007;6:25‐37.
    1. Han SH, Wu MY, Nam BY, et al. PGC‐1alpha protects from notch‐induced kidney fibrosis development. J Am Soc Nephrol. 2017;28:3312‐3322.
    1. Zhang L, Liu J, Zhou F, Wang W, Chen N. PGC‐1alpha ameliorates kidney fibrosis in mice with diabetic kidney disease through an antioxidative mechanism. Mol Med Rep. 2018;17:4490‐4498.
    1. Yu G, Tzouvelekis A, Wang R, et al. Thyroid hormone inhibits lung fibrosis in mice by improving epithelial mitochondrial function. Nat Med. 2018;24:39‐49.
    1. Bauer Y, White ES, de Bernard S, et al. MMP‐7 is a predictive biomarker of disease progression in patients with idiopathic pulmonary fibrosis. ERJ Open Research. 2017;3(1):00074‐2016. 10.1183/23120541.00074-2016
    1. Richards TJ, Kaminski N, Baribaud F, et al. Peripheral blood proteins predict mortality in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2012;185:67‐76.
    1. Abram CL, Roberge GL, Hu Y, Lowell CA. Comparative analysis of the efficiency and specificity of myeloid‐Cre deleting strains using ROSA‐EYFP reporter mice. J Immunol Methods. 2014;408:89‐100.
    1. Jakubzick C, Bogunovic M, Bonito AJ, Kuan EL, Merad M, Randolph GJ. Lymph‐migrating, tissue‐derived dendritic cells are minor constituents within steady‐state lymph nodes. J Exp Med. 2008;205:2839‐2850.
    1. Geng T, Li P, Yin X, Yan Z. PGC‐1alpha promotes nitric oxide antioxidant defenses and inhibits FOXO signaling against cardiac cachexia in mice. Am J Pathol. 2011;178:1738‐1748.
    1. Besse‐Patin A, Jeromson S, Levesque‐Damphousse P, Secco B, Laplante M, Estall JL. PGC1A regulates the IRS1:IRS2 ratio during fasting to influence hepatic metabolism downstream of insulin. Proc Natl Acad Sci USA. 2019;116:4285‐4290.

Source: PubMed

3
Subskrybuj