4.7 Article

Macrophage-to-Myofibroblast Transition Contributes to Interstitial Fibrosis in Chronic Renal Allograft Injury

Journal

JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY
Volume 28, Issue 7, Pages 2053-2067

Publisher

AMER SOC NEPHROLOGY
DOI: 10.1681/ASN.2016050573

Keywords

-

Funding

  1. Research Grants Council of Hong Kong [GRF 468711, CUHK3/CRF/12R]
  2. Focused Investment Scheme A from the Chinese University of Hong Kong
  3. Major State Basic Research Development Program of China (973 program) [2012CB517705]
  4. National Nature Science Foundation of China [2011BAI10B07, 2012CB517603, 2012AA02A512, 2014KYA057]
  5. National Fund Committee of China [81470938]
  6. National Health Medical Research Council of Australia

Ask authors/readers for more resources

Interstitial fibrosis is an important contributor to graft loss in chronic renal allograft injury. Inflammatory macrophages are associated with fibrosis in renal allografts, but how these cells contribute to this damaging response is not clearly understood. Here, we investigated the role of macrophage-to-myofibroblast transition in interstitial fibrosis in human and experimental chronic renal allograft injury. In biopsy specimens from patients with active chronic allograft rejection, we identified cells undergoingmacrophage-to-myofibroblast transition by the coexpression of macrophage (CD68) and myofibroblast (alpha-smooth muscle actin [alpha-SMA]) markers. CD68(+)/alpha-SMA(+) cells accounted for approximately 50% of the myofibroblast population, and the number of these cells correlated with allograft function andthe severityof interstitialfibrosis. Similarly, in C57BL/6J micewith aBALB/c renal allograft, cells coexpressing macrophage markers (CD68 or F4/80) and alpha-SMA composed a significant population in the interstitium of allografts undergoing chronic rejection. Fate-mapping in Lyz2-Cre/Rosa26-Tomato mice showed that approximately half of alpha-SMA(+) myofibroblasts in renal allografts originated from recipient bone marrow-derived macrophages. Knockout of Smad3 protected against interstitial fibrosis in renal allografts and substantially reduced the number of macrophage-to-myofibroblast transition cells. Furthermore, the majority of macrophage- to-myofibroblast transition cells in human and experimental renal allograft rejection coexpressed the M2-type macrophage marker CD206, and this expression was considerably reduced in Smad3-knockout recipients. In conclusion, our studies indicate that macrophage-to-myofibroblast transition contributes to interstitial fibrosis in chronic renal allograft injury. Moreover, the transition of bone marrow-derived M2type macrophages to myofibroblasts in the renal allograft is regulated via a Smad3-dependent mechanism.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available