Skip to main content

Advertisement

Log in

Local proliferation of macrophages in adipose tissue during obesity-induced inflammation

  • Article
  • Published:
Diabetologia Aims and scope Submit manuscript

Abstract

Aims/hypothesis

Obesity is frequently associated with low-grade inflammation of adipose tissue (AT), and the increase in adipose tissue macrophages (ATMs) is linked to an increased risk of type 2 diabetes. Macrophages have been regarded as post-mitotic, but recent observations have challenged this view. In this study, we tested the hypothesis that macrophages proliferate within AT in diet-induced obesity in mice and humans.

Methods

We studied the expression of proliferation markers by immunofluorescence, PCR and flow cytometry in three different models of mouse obesity as well as in humans (n = 239). The cell fate of dividing macrophages was assessed by live imaging of AT explants.

Results

We show that ATMs undergo mitosis within AT, predominantly within crown-like structures (CLS). We found a time-dependent increase in ATM proliferation when mice were fed a high-fat diet. Upregulation of CD206 and CD301 in proliferating ATMs indicated preferential M2 polarisation. Live imaging within AT explants from mice revealed that macrophages emigrate out of the CLS to become resident in the interstitium. In humans, we confirmed the increased expression of proliferation markers of CD68+ macrophages in CLS and demonstrated a higher mRNA expression of the proliferation marker Ki67 in AT from obese patients.

Conclusions/interpretation

Local proliferation contributes to the increase in M2 macrophages in AT. Our data confirm CLS as the primary site of proliferation and a new source of ATMs and support a model of different recruitment mechanisms for classically activated (M1) and alternatively activated (M2) macrophages in obesity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Abbreviations

AT:

Adipose tissue

ATMs:

Adipose tissue macrophages

CCR2:

C-C motif chemokine receptor 2

CLS:

Crown-like structures

CSF1R:

Colony-stimulating factor 1 receptor

eGFP:

Enhanced green fluorescent protein

EWAT:

Epididymal white adipose tissue

HFD:

High-fat diet

MCP-1:

Monocyte chemoattractant protein 1

PBST:

PBS with 0.3% Triton-X

PCNA:

Proliferating cell nuclear antigen

SWAT:

Subcutaneous white adipose tissue

References

  1. James WP (2008) WHO recognition of the global obesity epidemic. Int J Obes 32(Suppl 7):S120–S126

    Article  Google Scholar 

  2. Olshansky SJ, Passaro DJ, Hershow RC et al (2005) A potential decline in life expectancy in the United States in the 21st century. N Engl J Med 352:1138–1145

    Article  CAS  PubMed  Google Scholar 

  3. Percik R, Stumvoll M (2009) Obesity and cancer. Exp Clin Endocrinol Diabetes 117:563–566

    Article  CAS  PubMed  Google Scholar 

  4. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Investig 112:1796–1808

    Article  CAS  PubMed  Google Scholar 

  5. Nishimura S, Manabe I, Nagasaki M et al (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15:914–920

    Article  CAS  PubMed  Google Scholar 

  6. Winer DA, Winer S, Shen L et al (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17:610–617

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Liu J, Divoux A, Sun J et al (2009) Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med 15:940–945

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Elgazar-Carmon V, Rudich A, Hadad N, Levy R (2008) Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res 49:1894–1903

    Article  CAS  PubMed  Google Scholar 

  9. Strissel KJ, Stancheva Z, Miyoshi H et al (2007) Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes 56:2910–2918

    Article  CAS  PubMed  Google Scholar 

  10. Cinti S (2005) Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res 46:2347–2355

    Article  CAS  PubMed  Google Scholar 

  11. Cinti S (2009) Reversible physiological transdifferentiation in the adipose organ. Proc Nutr Soc 68:340

    Article  PubMed  Google Scholar 

  12. Fujisaka S, Usui I, Ikutani M et al (2013) Adipose tissue hypoxia induces inflammatory M1 polarity of macrophages in an HIF-1alpha-dependent and HIF-1alpha-independent manner in obese mice. Diabetologia 56:1403–1412

    Article  CAS  PubMed  Google Scholar 

  13. Bluher M, Bashan N, Shai I et al (2009) Activated Ask1-MKK4-p38MAPK/JNK stress signaling pathway in human omental fat tissue may link macrophage infiltration to whole-body Insulin sensitivity. J Clin Endocrinol Metab 94:2507–2515

    Article  PubMed  Google Scholar 

  14. Morris DL, Cho KW, Delproposto JL et al (2013) Adipose tissue macrophages function as antigen presenting cells and regulate adipose tissue CD4+ T cells in mice. Diabetes 62(8):2762–2772

    Article  CAS  PubMed  Google Scholar 

  15. Deng T, Lyon CJ, Minze LJ et al (2013) Class II major histocompatibility complex plays an essential role in obesity-induced adipose inflammation. Cell Metab 17:411–422

    Article  CAS  PubMed  Google Scholar 

  16. Lumeng CN, Deyoung SM, Bodzin JL, Saltiel AR (2007) Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes 56:16–23

    Article  CAS  PubMed  Google Scholar 

  17. Osborn O, Olefsky JM (2012) The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med 18:363–374

    Article  CAS  PubMed  Google Scholar 

  18. Lumeng CN, DelProposto JB, Westcott DJ, Saltiel AR (2008) Phenotypic switching of adipose tissue macrophages with obesity is generated by spatiotemporal differences in macrophage subtypes. Diabetes 57:3239–3246

    Article  CAS  PubMed  Google Scholar 

  19. Aron-Wisnewsky J, Tordjman J, Poitou C et al (2009) Human adipose tissue macrophages: m1 and m2 cell surface markers in subcutaneous and omental depots and after weight loss. J Clin Endocrinol Metab 94:4619–4623

    Article  CAS  PubMed  Google Scholar 

  20. Hotamisligil GS, Shargill NS, Spiegelman BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259:87–91

    Article  CAS  PubMed  Google Scholar 

  21. Ye J, McGuinness OP (2013) Inflammation during obesity is not all bad: evidence from animal and human studies. Am J Physiol Endocrinol Metab 304:E466–E477

    Article  CAS  PubMed  Google Scholar 

  22. Klöting N, Fasshauer M, Dietrich A et al (2010) Insulin-sensitive obesity. Am J Physiol Endocrinol Metab 299:E506–E515

    Article  PubMed  Google Scholar 

  23. Yona S, Kim KW, Wolf Y et al (2013) Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38:79–91

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Jenkins SJ, Ruckerl D, Cook PC et al (2011) Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332:1284–1288

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Xu H, Barnes GT, Yang Q et al (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112:1821–1830

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Harman-Boehm I, Bluher M, Redel H et al (2007) Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab 92:2240–2247

    Article  CAS  PubMed  Google Scholar 

  27. Chakaroun R, Raschpichler M, Kloting N et al (2012) Effects of weight loss and exercise on chemerin serum concentrations and adipose tissue expression in human obesity. Metab Clin Exp 61:706–714

    Article  CAS  PubMed  Google Scholar 

  28. Youn BS, Bang SI, Kloting N et al (2009) Serum progranulin concentrations may be associated with macrophage infiltration into omental adipose tissue. Diabetes 58:627–636

    Article  CAS  PubMed  Google Scholar 

  29. Nishimura S, Manabe I, Nagasaki M et al (2008) In vivo imaging in mice reveals local cell dynamics and inflammation in obese adipose tissue. J Clin Investig 118:710–721

    CAS  PubMed  Google Scholar 

  30. Oh DY, Morinaga H, Talukdar S, Bae EJ, Olefsky JM (2012) Increased macrophage migration into adipose tissue in obese mice. Diabetes 61:346–354

    Article  CAS  PubMed  Google Scholar 

  31. Hashimoto D, Chow A, Noizat C et al (2013) Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38:792–804

    Article  CAS  PubMed  Google Scholar 

  32. Bourlier V, Zakaroff-Girard A, Miranville A et al (2008) Remodeling phenotype of human subcutaneous adipose tissue macrophages. Circulation 117:806–815

    Article  CAS  PubMed  Google Scholar 

  33. Sasmono RT, Oceandy D, Pollard JW et al (2003) A macrophage colony-stimulating factor receptor-green fluorescent protein transgene is expressed throughout the mononuclear phagocyte system of the mouse. Blood 101:1155–1163

    Article  CAS  PubMed  Google Scholar 

  34. Crisan M, Yap S, Casteilla L et al (2008) A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 3:301–313

    Article  CAS  PubMed  Google Scholar 

  35. Berry R, Rodeheffer MS (2013) Characterization of the adipocyte cellular lineage in vivo. Nat Cell Biol 15:302–308

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Prodinger C, Bunse J, Kruger M et al (2011) CD11c-expressing cells reside in the juxtavascular parenchyma and extend processes into the glia limitans of the mouse nervous system. Acta Neuropathol 121:445–458

    Article  CAS  PubMed  Google Scholar 

  37. Handel-Fernandez ME, Lopez DM (2000) Isolation of macrophages from tissues, fluids, and immune response sites. In: Paulnock DM (ed) Macrophages. Oxford University Press, Oxford

    Google Scholar 

  38. Feng B, Jiao P, Nie Y et al (2011) Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS ONE 6:e24358

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Patsouris D, Li P-P, Thapar D, Chapman J, Olefsky JM, Neels JG (2008) Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab 8:301–309

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Gutierrez DA, Kennedy A, Orr JS et al (2011) Aberrant accumulation of undifferentiated myeloid cells in the adipose tissue of CCR2-deficient mice delays improvements in insulin sensitivity. Diabetes 60:2820–2829

    Article  CAS  PubMed  Google Scholar 

  41. Kanda H, Tateya S, Tamori Y et al (2006) MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest 116:1494–1505

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Kamei N, Tobe K, Suzuki R et al (2006) Overexpression of monocyte chemoattractant protein-1 in adipose tissues causes macrophage recruitment and insulin resistance. J Biol Chem 281:26602–26614

    Article  CAS  PubMed  Google Scholar 

  43. Nara N, Nakayama Y, Okamoto S et al (2007) Disruption of CXC motif chemokine ligand-14 in mice ameliorates obesity-induced insulin resistance. J Biol Chem 282:30794–30803

    Article  CAS  PubMed  Google Scholar 

  44. Chavey C, Lazennec G, Lagarrigue S et al (2009) CXC ligand 5 is an adipose-tissue derived factor that links obesity to insulin resistance. Cell Metab 9:339–349

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  45. Maumus M, Sengenes C, Decaunes P et al (2008) Evidence of in situ proliferation of adult adipose tissue-derived progenitor cells: influence of fat mass microenvironment and growth. J Clin Endocrinol Metab 93:4098–4106

    Article  CAS  PubMed  Google Scholar 

  46. Schulz C, Gomez Perdiguero E, Chorro L et al (2012) A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336:86–90

    Article  CAS  PubMed  Google Scholar 

  47. Ji Y, Sun S, Xia S, Yang L, Li X, Qi L (2012) Short term high fat diet challenge promotes alternative macrophage polarization in adipose tissue via natural killer T cells and interleukin-4. J Biol Chem 287:24378–24386

    Article  CAS  PubMed  Google Scholar 

  48. El-Wakkad A, Hassan Nel M, Sibaii H, El-Zayat SR (2013) Proinflammatory, anti-inflammatory cytokines and adiponkines in students with central obesity. Cytokine 61:682–687

    Article  CAS  PubMed  Google Scholar 

  49. Zeyda M, Farmer D, Todoric J et al (2007) Human adipose tissue macrophages are of an anti-inflammatory phenotype but capable of excessive pro-inflammatory mediator production. Int J Obes (Lond) 31:1420–1428

    Article  CAS  Google Scholar 

  50. Wentworth JM, Naselli G, Brown WA et al (2010) Pro-inflammatory CD11c+CD206+ adipose tissue macrophages are associated with insulin resistance in human obesity. Diabetes 59:1648–1656

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful for the excellent technical assistance of C. Hobusch, A. Ehrlich and C. Merkwitz. We also thank M. Krüger for helpful discussions and K. Jäger and A. Lösche from the FACS core unit (all Leipzig University).

Funding

This work was in part supported by the Kompetenznetz Adipositas (Competence Network for Obesity) funded by the Federal Ministry of Education and Research (German Obesity Biomaterial Bank; FKZ 01GI1128 and FKZ 01EO1001), a grant from the Deutsche Forschungsgemeinschaft DFG-SFB 1052/1: ‘Obesity mechanisms’ (projects A04, B01, B04) and by the Helmholtz Alliance ‘Imaging and Curing Environmental Metabolic Disease’ through the Initiative and Networking Fund of the Helmholtz Association. MG received a start-up grant from the Medical Faculty of Leipzig University.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

JH planned and performed morphological analyses of murine and human AT samples. UW and JH analysed ATMs by flow cytometry. MG and JE designed, performed and evaluated live imaging experiments of AT explants. KI planned, established and analysed murine macrophage cultures. NK and MB analysed gene expression of human AT samples. IB and MG designed the study. JH and MG wrote the paper. All authors participated in drafting and revising this article. All authors approved the final version of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Martin Gericke.

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM Fig. 1

ATMs incorporate BrdU as a sign of cell replication. (A) Representative FACS plots are shown for chow controls (left) and mice after 10 weeks of HFD (right). F4/80+ ATMs were analyzed for BrdU incorporation (S phase) and the DNA content, measured by 7-AAD. Thus, ATMs in G1 phase (single DNA content) and G2 phase (double DNA content) can be distinguished. (B) Bar graph summarizing the percentage of resting (G0/G1) and proliferating (S/G2) ATMs after 1, 4 and 10 weeks of HFD. (PDF 97 kb)

ESM Fig. 2

ATMs proliferate in AT of ob/ob mice. (A-C) Triple immunofluorescence staining of the macrophage marker F4/80 (red), the proliferation marker Ki67 (green) and DAPI (blue) is shown. (A and B) Representative images of either ob/ob (n = 4) or ob/+ mice (n = 4). (C) Higher magnification and single color channels of the area depicted in (A) reveal a co-staining. Scale bar represents 20 μm in (A) and (B) or 10 μm in (C). (D) Quantification of Ki67+ ATMs in ob/+ and ob/ob animals. Data are shown for subcutaneous (SWAT) and epididymal (EWAT) AT. (E) Percentage of ATMs located within CLS for ob/+ and ob/ob mice (n.d. = not detected). (F) Percentage of Ki67+ ATMs in ob/ob mice not associated to CLS (interstitium) or associated to CLS (CLS). Data are represented as means ± SEM and tested for statistical significance by the Student-Newman-Keuls multiple comparison test. *** p ≤ 0.001 (PDF 52 kb)

ESM Fig. 3

ATMs proliferate in AT of db/db mice. (A-C) Triple immunofluorescence staining of the macrophage marker F4/80 (red), the proliferation marker Ki67 (green) and DAPI (blue) is shown. (A and B) Representative images of either db/db (n = 4) or db/+ mice (n = 3). (C) Higher magnification and single color channels of the area delineated in (A) reveal Ki67+ ATMs. Scale bar represents 20 μm in (A) and (B) or 10 μm in (C). (D) Bar graph summarizing the percentage of Ki67+ ATMs in db/+ and db/db mice. Data are shown for subcutaneous (SWAT) and epididymal (EWAT) AT. (E) Percentage of ATMs located within CLS for db/+ and db/db mice (n.d. = not detected). (F) Percentage of Ki67+ ATMs of db/db mice not associated to CLS (interstitium) or within CLS (CLS). Data are represented as means ± SEM and tested for statistical significance by the Student-Newman-Keuls multiple comparison test. ** p ≤ 0.01 and *** p ≤ 0.001 (PDF 63 kb)

ESM Video 1

ATMs migrate out of the CLS, rather than towards the CLS. Live-imaging of living AT explants was performed by using MacGreen mice (CSF-1R eGFP) to visualize ATMs in situ. Adipocytes were stained using Bodipy598 (blue), whereas ATMs are shown in green. Time is presented in h:min. (AVI 600 kb)

ESM Table 1

(PDF 21 kb)

ESM Table 2

(PDF 14 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Haase, J., Weyer, U., Immig, K. et al. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia 57, 562–571 (2014). https://doi.org/10.1007/s00125-013-3139-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00125-013-3139-y

Keywords

Navigation