Haldar M, Kohyama M, So AY, et al.
Heme-mediated SPI-C induction promotes monocyte differentiation into iron-recycling macrophages.
Cell.
2014;156:1223-1234.

During their development, circulation in blood, and demise from senescence, erythrocytes have key interactions with a specific subset of macrophages in bone marrow, splenic red pulp, and hepatic sinusoids (Kupffer cells). Some marrow macrophages are central macrophages of the erythroblastic islands where erythroid precursors complete differentiation into reticulocytes prior to entering circulation. Splenic red pulp macrophages form sheaths through which erythrocytes pass with the physically damaged, immunologically recognized, or senescent erythrocytes being culled by phagocytosis. The normal daily phagocytosis of the most senescent one percent of erythrocytes (about 200 billion cells) and subsequent hemoglobin degradation (with heme being catabolized to iron, carbon monoxide, biliverdin, and bilirubin) make red pulp macrophages the major recyclers of iron, which is required for sufficient hemoglobin production to supply the one percent of reticulocytes that replace the senescent erythrocytes daily. Erythroblastic island-central macrophages phagocytose extruded nuclei with associated hemoglobin (pyrenocytes) that are created as erythroblasts enucleate to become reticulocytes.1  The reticulocytes, and to a much lesser degree mature erythrocytes, shed hemoglobin-filled vesicles, which have membrane changes similar to senescent erythrocytes that target them for macrophage-mediated phagocytosis, mostly in the liver and bone marrow.2  Free hemoglobin and heme are bound to plasma proteins, haptoglobin and hemopexin, respectively, that in turn bind to the CD163 and CD91 receptors on liver and spleen macrophages leading to endocytosis with subsequent heme degradation.3  Thus, specific macrophages in marrow, splenic red pulp, and liver sinusoids receive and catabolize large amounts of heme while recycling the iron via transferrin to developing erythroblasts, the major consumers of iron. However, even these specialized macrophages can accumulate toxic levels of heme that result in oxidative damage and apoptosis, such as when the inducible enzyme, heme oxygenase-1, that metabolizes heme, is deficient4  or when the amount of hemoglobin turnover is massive,5  as in severe hemolytic anemia.

The marrow, spleen, and liver macrophages that catabolize heme and recycle iron require the expression of Spi-C, a transcription factor related to PU.1, the major mediator of myeloid differentiation in early-stage hematopoietic progenitor cells.6 Using transgenic mice engineered to co-express Spic and green fluorescent protein, Dr. Malay Haldar in the lab of Dr. Kenneth Murphy at Washington University in St. Louis identified the location and characteristics of cells expressing Spic. Such cells were found in the splenic red pulp, the bone marrow, and the liver and were characterized by expression of F4/80, VCAM-1, CD169 (Siglec-1), and CD68, markers of macrophage differentiation, including central macrophages of erythroblastic islands. Subsequent in vitro studies showed that heme directly induces Spicexpression in bone marrow macrophages and monocytes. The spleen and marrow of transgenic mice lost their Spic-expressing macrophages as a consequence of heme toxicity when exogenous heme administration was combined with either genetic deficiency or chemical inhibition of heme oxygenase, or with phenylhydrazine-induced hemolysis. As theSpic-expressing macrophages were killed by heme toxicity, a population of cells with the myeloid marker CD11b and low-level expression of Spic appeared in both of these organs. This myeloid cell population was an intermediate stage of differentiation between monocytes that do not express Spic and the prominently Spic-expressing macrophages. Further investigation demonstrated that, under severe hemolytic stress, monocytes responded to heme by generating the intermediate stage cells that in turn completed differentiation into macrophages, thereby replacing those macrophages previously lost to heme toxicity. The mechanism for this hemolysis-mediated induction of Spicexpression was shown to be heme-mediated enhancement of proteasome-dependent degradation of Bach1, a transcriptional repressor of Spic.

Some hemolytic disorders may be sufficiently severe such that they cause excessive accumulation of cytotoxic heme that is lethal for the splenic, marrow, and hepatic macrophages responsible for the normal, steady-state metabolism of heme and iron due to erythrocyte turnover. Under these pathological conditions, the excess heme mediates differentiation of monocytes into macrophages, restoring the capacity of the spleen, marrow, and liver to metabolize the excess hemoglobin and recycle the iron needed for the increased erythropoiesis that accompanies such hemolysis. This induction of macrophages that specialize in hemoglobin degradation and iron recycling will interest those studying monocyte-macrophage differentiation, transcription factors, and heme and iron metabolism. In the clinical setting, a heme-mediated increase in splenic macrophage numbers helps explain the splenomegaly that arises during active hemolysis in malaria and autoimmune hemolytic anemia. In terms of therapy, splenectomy reduces the numbers of erythrophagocytic macrophages in severe cases of immune-mediated hemolytic anemia and hereditary spherocytosis, but suppression of heme-mediated differentiation of macrophages in such situations could potentially reduce the hemolytic rate and, thereby, obviate splenectomy.

1.
1.Toda S, Segawa K, Nagata S.
MerTK-mediated engulfment of pyrenocytes by central macrophages in erythroblastic islands.
Blood.
2014;123:3963-3971.
http://www.ncbi.nlm.nih.gov/pubmed/24659633
2.
2.Willekens FL, Werre JM, Kruijt JK, et al.
Liver Kupffer cells rapidly remove red blood cell-derived vesicles from the circulation by scavenger receptors.
Blood.
2005;105:2141-2145.
http://www.ncbi.nlm.nih.gov/pubmed/15550489
3.
3.Nielsen MJ, Møller HJ, Moestrup SK.
Hemoglobin and heme scavenger receptors.
Antioxid Redox Signal.
2010;12:261-273.
http://www.ncbi.nlm.nih.gov/pubmed/19659436
4.
4.Kovtunovych G, Eckhaus MA, Ghosh MC, et al.
Dysfunction of the heme recycling system in heme oxygenase 1-deficient mice: effects on macrophage viability and tissue iron distribution.
Blood.
2010;116:6054-6062.
http://www.ncbi.nlm.nih.gov/pubmed/20844238
5.
Cambos M, Scorza T.
Robust erythrophagocytosis leads to macrophage apoptosis via a hemin-mediated redox imbalance: role in hemolytic disorders.
J Leukoc Biol.
2011;89:159-171.
http://www.ncbi.nlm.nih.gov/pubmed/20884648
6.
6.Kohyama M, Ise W, Edelson BT, et al.
Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis.
Nature.
2009;457:318-321.
http://www.ncbi.nlm.nih.gov/pubmed/19037245

Competing Interests

Dr. Koury declares no relevant conflicts of interest.