Mitophagy deficiency increases NLRP3 to induce brown fat dysfunction in mice

ABSTRACT Although macroautophagy/autophagy deficiency causes degenerative diseases, the deletion of essential autophagy genes in adipocytes paradoxically reduces body weight. Brown adipose tissue (BAT) plays an important role in body weight regulation and metabolic control. However, the key cellular mechanisms that maintain BAT function remain poorly understood. in this study, we showed that global or brown adipocyte-specific deletion of pink1, a Parkinson disease-related gene involved in selective mitochondrial autophagy (mitophagy), induced BAT dysfunction, and obesity-prone type in mice. Defective mitochondrial function is among the upstream signals that activate the NLRP3 inflammasome. NLRP3 was induced in brown adipocyte precursors (BAPs) from pink1 knockout (KO) mice. Unexpectedly, NLRP3 induction did not induce canonical inflammasome activity. Instead, NLRP3 induction led to the differentiation of pink1 KO BAPs into white-like adipocytes by increasing the expression of white adipocyte-specific genes and repressing the expression of brown adipocyte-specific genes. nlrp3 deletion in pink1 knockout mice reversed BAT dysfunction. Conversely, adipose tissue-specific atg7 KO mice showed significantly lower expression of Nlrp3 in their BAT. Overall, our data suggest that the role of mitophagy is different from general autophagy in regulating adipose tissue and whole-body energy metabolism. Our results uncovered a new mitochondria-NLRP3 pathway that induces BAT dysfunction. The ability of the nlrp3 knockouts to rescue BAT dysfunction suggests the transcriptional function of NLRP3 as an unexpected, but a quite specific therapeutic target for obesity-related metabolic diseases. Abbreviations: ACTB: actin, beta; BAPs: brown adipocyte precursors; BAT: brown adipose tissue; BMDMs: bone marrow-derived macrophages; CASP1: caspase 1; CEBPA: CCAAT/enhancer binding protein (C/EBP), alpha; ChIP: chromatin immunoprecipitation; EE: energy expenditure; HFD: high-fat diet; IL1B: interleukin 1 beta; ITT: insulin tolerance test; KO: knockout; LPS: lipopolysaccharide; NLRP3: NLR family, pyrin domain containing 3; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RD: regular diet; ROS: reactive oxygen species; RT: room temperature; UCP1: uncoupling protein 1 (mitochondrial, proton carrier); WT: wild-type.

[1]  J. Ting,et al.  The NLRP3 inflammasome: molecular activation and regulation to therapeutics , 2019, Nature Reviews Immunology.

[2]  G. Shulman,et al.  Mechanisms of Insulin Action and Insulin Resistance. , 2018, Physiological reviews.

[3]  H. Cai,et al.  Parkin and PINK1 mitigate STING-induced inflammation , 2018, Nature.

[4]  T. Finkel,et al.  The role of mitochondria in aging , 2018, The Journal of clinical investigation.

[5]  S. Chiu,et al.  Mitophagy is required for brown adipose tissue mitochondrial homeostasis during cold challenge , 2018, Scientific Reports.

[6]  S. Kajimura,et al.  Mitophagy controls beige adipocyte maintenance through a Parkin-dependent and UCP1-independent mechanism , 2018, Science Signaling.

[7]  D. Malide,et al.  A fluorescence-based imaging method to measure in vitro and in vivo mitophagy using mt-Keima , 2017, Nature Protocols.

[8]  R. Curi,et al.  Combination of a high-fat diet with sweetened condensed milk exacerbates inflammation and insulin resistance induced by each separately in mice , 2017, Scientific Reports.

[9]  P. Seale,et al.  Control of brown and beige fat development , 2016, Nature Reviews Molecular Cell Biology.

[10]  J. Debnath,et al.  Beige Adipocyte Maintenance Is Regulated by Autophagy-Induced Mitochondrial Clearance. , 2016, Cell metabolism.

[11]  B. Kemp,et al.  Lack of Adipocyte AMPK Exacerbates Insulin Resistance and Hepatic Steatosis through Brown and Beige Adipose Tissue Function. , 2016, Cell metabolism.

[12]  B. Ryffel,et al.  The receptor NLRP3 is a transcriptional regulator of TH2 differentiation , 2015, Nature Immunology.

[13]  V. Dixit,et al.  Drivers of age‐related inflammation and strategies for healthspan extension , 2015, Immunological reviews.

[14]  K. Reymann,et al.  Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy , 2014, Kidney international.

[15]  Yingwei Chen,et al.  Curcumin suppresses NLRP3 inflammasome activation and protects against LPS-induced septic shock. , 2013, Molecular nutrition & food research.

[16]  T. Schulz,et al.  Mechanisms of Aging-Related Impairment of Brown Adipocyte Development and Function , 2014, Gerontology.

[17]  A. Wagers,et al.  Stem cell aging: mechanisms, regulators and therapeutic opportunities , 2014, Nature Medicine.

[18]  A. Brunetti,et al.  Evaluation of Growth Patterns and Body Composition in C57Bl/6J Mice Using Dual Energy X-Ray Absorptiometry , 2014, BioMed research international.

[19]  Antonio Vidal-Puig,et al.  The different shades of fat , 2014, Nature.

[20]  K. Walsh,et al.  Vascular rarefaction mediates whitening of brown fat in obesity. , 2014, The Journal of clinical investigation.

[21]  Y. Tseng,et al.  Brown fat fuel utilization and thermogenesis , 2014, Trends in Endocrinology & Metabolism.

[22]  A. Wree,et al.  NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice , 2014, Hepatology.

[23]  P. Seale,et al.  Brown and beige fat: development, function and therapeutic potential , 2013, Nature Medicine.

[24]  R. Gottlieb,et al.  MitoTimer probe reveals the impact of autophagy, fusion, and motility on subcellular distribution of young and old mitochondrial protein and on relative mitochondrial protein age , 2013, Autophagy.

[25]  C. Chu,et al.  After the banquet , 2013, Autophagy.

[26]  M. Mori,et al.  Anatomical Localization, Gene Expression Profiling, and Functional Characterization of Adult Human Neck Brown Fat , 2013, Nature Medicine.

[27]  Yan Zeng,et al.  Nitric oxide suppresses NLRP3 inflammasome activation and protects against LPS-induced septic shock , 2013, Cell Research.

[28]  Han-Woong Lee,et al.  Knockout mice created by TALEN-mediated gene targeting , 2013, Nature Biotechnology.

[29]  Richard A. Flavell,et al.  Inflammasomes in health and disease , 2012, Nature.

[30]  D. Green,et al.  Mitochondria and the Autophagy–Inflammation–Cell Death Axis in Organismal Aging , 2011, Science.

[31]  Haitao Wen,et al.  The inflammasome NLRs in immunity, inflammation, and associated diseases. , 2011, Annual review of immunology.

[32]  S. Ryter,et al.  Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. , 2011, Nature immunology.

[33]  J. Tschopp,et al.  A role for mitochondria in NLRP3 inflammasome activation , 2011, Nature.

[34]  L. Joosten,et al.  The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. , 2010, Cell metabolism.

[35]  B. Spiegelman,et al.  Transcriptional control of brown fat development. , 2010, Cell metabolism.

[36]  Kate Schroder,et al.  The NLRP3 Inflammasome: A Sensor for Metabolic Danger? , 2010, Science.

[37]  R. Youle,et al.  Mechanisms of mitophagy , 2010, Nature Reviews Molecular Cell Biology.

[38]  M. Komatsu,et al.  Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis , 2009, Proceedings of the National Academy of Sciences.

[39]  M. Czaja,et al.  Autophagy regulates adipose mass and differentiation in mice. , 2009, The Journal of clinical investigation.

[40]  R. Friedline,et al.  Interleukin-10 Prevents Diet-Induced Insulin Resistance by Attenuating Macrophage and Cytokine Response in Skeletal Muscle , 2009, Diabetes.

[41]  E. Palmer,et al.  Identification and importance of brown adipose tissue in adult humans. , 2009, The New England journal of medicine.

[42]  Wei Jiang,et al.  Parkin, PINK1, and DJ-1 form a ubiquitin E3 ligase complex promoting unfolded protein degradation. , 2009, The Journal of clinical investigation.

[43]  B. Cannon,et al.  UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality. , 2009, Cell metabolism.

[44]  C. Wahlestedt,et al.  Genomic variants at the PINK1 locus are associated with transcript abundance and plasma nonesterified fatty acid concentrations in European whites , 2008, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[45]  Jie Shen,et al.  Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress , 2008, Proceedings of the National Academy of Sciences.

[46]  D. Tenen,et al.  Metabolic Response of Mice to a Postnatal Ablation of CCAAT/Enhancer-binding Protein α* , 2005, Journal of Biological Chemistry.

[47]  C. Darimont,et al.  C/EBPalpha regulates human adiponectin gene transcription through an intronic enhancer. , 2005, Diabetes.

[48]  B. Spiegelman,et al.  C/EBPalpha induces adipogenesis through PPARgamma: a unified pathway. , 2002, Genes & development.

[49]  H. Linhart,et al.  C/EBPα is required for differentiation of white, but not brown, adipose tissue , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[50]  H. Linhart,et al.  C/EBPalpha is required for differentiation of white, but not brown, adipose tissue. , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[51]  B. Cannon,et al.  Cultures of adipose precursor cells from brown adipose tissue and of clonal brown-adipocyte-like cell lines. , 2001, Methods in molecular biology.

[52]  J. Gimble,et al.  Modulation of the murine peroxisome proliferator-activated receptor gamma 2 promoter activity by CCAAT/enhancer-binding proteins. , 2000, The Journal of biological chemistry.

[53]  Bruce M. Spiegelman,et al.  Towards a molecular understanding of adaptive thermogenesis , 2000, Nature.

[54]  B. Spiegelman,et al.  Cross-Regulation of C/EBPα and PPARγ Controls the Transcriptional Pathway of Adipogenesis and Insulin Sensitivity , 1999 .

[55]  B. Spiegelman,et al.  Cross-regulation of C/EBP alpha and PPAR gamma controls the transcriptional pathway of adipogenesis and insulin sensitivity. , 1999, Molecular cell.