World Health Organization Chronic Disease Definition

Iron and anemia of chronic disease. Anemia of chronic disease (ACD) is the most frequent anemia found in hospitalized patients, often occurring in subjects suffering from chronic inflammatory disorders. The underlying diversion of iron traffic leads to a withdrawal of the metal from the sites of erythropoiesis and the circulation to the storage compartment in the reticuloendothelial system, thus resulting, at the same time, in hypoferremia and hyperferritinemia. Proinflammatory and anti-inflammatory cytokines, acute-phase proteins, and radicals are prominently involved in causing these disturbances of iron homeostasis. The role of these factors, as well as the pathophysiological reasons for the development of ACD, is discussed in this review.

Keywords

  • iron homeostasis
  • inflammation
  • hypoferremia
  • erythropoiesis

APPENDIX

ACD

anemia of chronic disease

e-ALAS

erythroid amino-levulinate synthase

IFN-γ

interferon-gamma

IL

interleukin

iNOS

inducible nitric oxide synthase

IRE

iron responsive elements

IRPs

iron regulatory proteins

NO

nitric oxide

TfR

serum transferrin receptor

Th-1

T-helper type-1 cells

TNF-β

tumor necrosis factor-β

Iron is a Janus-faced molecule that, on one hand, is an essential cofactor for enzymes in the mitochondrial respiration chain, in the citric acid cycle or for DNA synthesis, as well as a central molecule for binding and transporting oxygen by hemoglobin and myoglobin

. On the other hand, cellular accumulation of metabolically active iron may be detrimental to cells and surrounding tissues because iron is also able to catalyze the formation of highly toxic hydroxyl radicals by the "Haber-Weiss" reaction

,

. Therefore, tight regulation of iron homeostasis is an absolute requirement for maintaining essential cellular functions while avoiding cellular damage. Thus far, many proteins, molecules, hormones and, last but not least, iron itself have been demonstrated to affect iron metabolism by various mechanisms at different regulatory levels

, which is also detailed elsewhere in this issue.

Some of these factors may become dominant over others in cases of chronic inflammatory diseases, thus leading to disturbances of iron metabolism. Therefore, patients suffering from a chronic infectious or tumor disease or an autoimmune disorder frequently develop a moderate anemia, which is termed "anemia of chronic disease" (ACD). This normochromic/microcytic anemia is clinically characterized by reduced serum iron concentrations and transferrin saturation, whereas iron stores, as reflected by ferritin levels, are often increased

,

4.

  • Konjin A.
  • Hershko C.

The anaemia of inflammation and chronic disease,.

  • Google Scholar

,

5.

  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Dierich M.P.
  • Wachter H.

Immune activation and the anaemia associated with chronic inflammatory disorders.

  • Crossref
  • PubMed
  • Scopus (113)
  • Google Scholar

,

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

. Transferrin concentrations and reticulocyte counts are normal or somewhat reduced, whereas serum transferrin receptor (TfR) levels are slightly enhanced

,

4.

  • Konjin A.
  • Hershko C.

The anaemia of inflammation and chronic disease,.

  • Google Scholar

,

5.

  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Dierich M.P.
  • Wachter H.

Immune activation and the anaemia associated with chronic inflammatory disorders.

  • Crossref
  • PubMed
  • Scopus (113)
  • Google Scholar

,

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

,

7.

  • Kuiper-Kramer E.P.
  • Huisman C.M.
  • vanRaan J.
  • vanEijk H.G.

Analytical and clinical implications of soluble transferrin receptor in serum.

  • PubMed
  • Google Scholar

.

Although ACD is probably the most frequent anemia in hospitalized patients, the underlying mechanisms are still elusive. In addition to various other factors such as repression of erythropoiesis, a blunted response to erythropoietin, and erythrophagocytosis, disturbances of iron homeostasis appear to be prominently involved in the development of anemia under chronic inflammatory conditions

,

4.

  • Konjin A.
  • Hershko C.

The anaemia of inflammation and chronic disease,.

  • Google Scholar

,

5.

  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Dierich M.P.
  • Wachter H.

Immune activation and the anaemia associated with chronic inflammatory disorders.

  • Crossref
  • PubMed
  • Scopus (113)
  • Google Scholar

,

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

,

. This review focuses on mechanisms that contribute to the diversion of iron traffic underlying ACD, leading to the withdrawal of iron from the sites of erythropoiesis and from serum to the iron storage sites in the reticuloendothelial system.

THE TH-1/TH-2 PARADIGM AND IRON HOMEOSTASIS

In prior years, evidence has accumulated for the existence of two CD 4+ T-helper cell subsets in humans, each of which produce a typical set of cytokines that regulate different immune effector functions and cross-react with each other

,

,

. T-helper cells type 1 (Th-1) produce interferon (IFN)-γ, interleukin (IL)-2, and lymphotoxin [tumor necrosis factor (TNF)-β]. These cytokines activate macrophages, thus contributing to the formation of proinflammatory cytokines, such as TNF-α, IL-1, or IL-6, and the induction of cytotoxic immune effector mechanisms of macrophages. By contrast, Th-2 cells produce IL-4, IL-5, IL-10, and IL-13, which induce a strong antibody response but also inhibit various macrophage functions

,

,

.

T-helper cells type 1 (Th-1)–derived cytokines have been demonstrated to also affect iron homeostasis by different mechanisms, although the data available so far are conflicting.

Interleukin-1 and TNF-α are able to induce hypoferremia by modulating macrophage iron metabolism

12.

  • Alvarez-Hernandez X.
  • Licega J.
  • McKay I.
  • Brock J.H.

Induction of hypoferremia and modulation of macrophage iron metabolism by tumor necrosis factor.

  • PubMed
  • Google Scholar

,

13.

  • Brock J.H.
  • Alvarez-Hernandez X.

Modulation of macrophage iron metabolism by tumour necrosis factor and interleukin-1.

  • Crossref
  • Google Scholar

. This is primarily exerted by an effect of the cytokines on the expression of the iron storage protein ferritin

. IL-1 and TNF-α increase the expression of L-chain and H-chain ferritin by an as yet not fully elucidated transcriptional mechanism

15.

  • Torti S.V.
  • Kwak E.L.
  • Miller S.C.
  • Miller L.L.
  • Ringold G.M.
  • Myambo K.B.
  • Young A.P.
  • Torti F.M.

The molecular cloning and characterisation of ferritin heavy chain, a tumor necrosis factor inducible gene.

  • Abstract
  • Full Text PDF
  • PubMed
  • Google Scholar

,

,

. In addition, IL-1 and IL-6 increase L- and H-ferritin expression in hepatoma cells by a translational mechanism

18.

  • Rogers J.T.
  • Bridges K.B.
  • Durmowics G.
  • Glass J.
  • Auron P.R.
  • Munro H.N.

Translational control during the acute phase response: Ferritin synthesis in response to interleukin 1.

  • Abstract
  • Full Text PDF
  • PubMed
  • Google Scholar

,

19.

  • Rogers J.T.

Ferritin translation by interleukin-1 and interleu-kin-6: The role of sequences upstream of the start codons of the heavy and light subunit genes.

  • Crossref
  • PubMed
  • Google Scholar

. A limitation of some of these studies is due to the fact that the described observations were made in different cell lines (like myoblasts, adipocytes, fibroblasts), which is of interest because iron metabolism appears to be differently regulated, for example, between erythroid cells, macrophages, or hepatocytes

. For example, it was demonstrated that IL-6 enhances TfR expression on hepatocytes, whereas it had no effect in Kupffer cells

, and similarly IL-1 and TNF-α affect iron uptake by increasing TfR expression in fibroblasts

, whereas just the opposite is true for monocytic cells

17.

  • Fahmy M.
  • Young S.P.

Modulation of iron metabolism in monocyte cell line U937 by inflammatory cytokines: Changes in transferrin uptake, iron handling and ferritin mRNA.

  • Crossref
  • PubMed
  • Scopus (123)
  • Google Scholar

.

In contrast, IFN-γ may rather depress TfR expression by transcriptional and post-transcriptional mechanisms

,

23.

  • Byrd T.
  • Horwitz M.A.

Regulation of transferrin receptor expression and ferritin content in human mononuclear macrophages: Coordinate upregulation by iron transferrin and downregulation by interferon gamma.

  • Crossref
  • PubMed
  • Scopus (130)
  • Google Scholar

,

,

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

. However, the regulation of TfR expression in activated macrophages appears to be very complex and sensitive to experimental differences in the activation, differentiation, and in vitro stimulation of the macrophages investigated by different groups

,

,

23.

  • Byrd T.
  • Horwitz M.A.

Regulation of transferrin receptor expression and ferritin content in human mononuclear macrophages: Coordinate upregulation by iron transferrin and downregulation by interferon gamma.

  • Crossref
  • PubMed
  • Scopus (130)
  • Google Scholar

,

,

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

,

,

27.

  • Testa U.
  • Conti L.
  • Sposi N.M.
  • Varano B.
  • Tritarelli E.
  • Malorni W.
  • Samoggia P.
  • Rainalidi G.
  • Peschle C.
  • Beladelli F.
  • Gessani S.

IFN-β selectively downregulates transferrin receptor expression in human peripheral blood macrophages by a posttranslational mechanism.

  • PubMed
  • Google Scholar

. Moreover, in human and murine macrophages, ferritin mRNA expression is induced by IFN-γ

,

28.

  • Weiss G.
  • Goossen B.
  • Doppler W.
  • Fuchs D.
  • Pantopoulos K.
  • Werner-Felmayer G.
  • Wachter H.
  • Hentze M.W.

Translational regulation via iron-responsive elements by the nitric oxide/NO-synthase pathway.

  • Crossref
  • PubMed
  • Scopus (333)
  • Google Scholar

, whereas at the same time, a translational repression occurs because of the formation of the short-lived radical nitric oxide (NO) via stimulation of the interaction of iron regulatory proteins (IRPs) with an RNA stem loop structure, iron responsive elements (IRE), as detailed later here

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

,

28.

  • Weiss G.
  • Goossen B.
  • Doppler W.
  • Fuchs D.
  • Pantopoulos K.
  • Werner-Felmayer G.
  • Wachter H.
  • Hentze M.W.

Translational regulation via iron-responsive elements by the nitric oxide/NO-synthase pathway.

  • Crossref
  • PubMed
  • Scopus (333)
  • Google Scholar

,

29.

  • Pantopoulos K.
  • Hentze M.W.

Nitric oxide signaling to iron-regulatory protein: Direct control of ferritin mRNA translation and transferrin receptor mRNA stability in transfected fibroblasts.

  • Crossref
  • PubMed
  • Scopus (199)
  • Google Scholar

. This is compatible with observations made in human macrophages in which IFN-γ decreases intracellular ferritin content

23.

  • Byrd T.
  • Horwitz M.A.

Regulation of transferrin receptor expression and ferritin content in human mononuclear macrophages: Coordinate upregulation by iron transferrin and downregulation by interferon gamma.

  • Crossref
  • PubMed
  • Scopus (130)
  • Google Scholar

.

In summary, proinflammatory cytokines appear to cause hypoferremia via the induction of ferritin synthesis and iron storage in macrophages and hepatocytes, whereas IFN-γ tries to keep iron out of macrophages. This makes sense, given the inhibitory effects of iron on IFN-γ–mediated pathways (discussed later in this article).

Interestingly, thus far, hardly any attention has been paid to the role of Th-2–derived cytokines on iron homeostasis. In activated murine macrophages, IL-4 and IL-13 have recently been demonstrated to modulate iron metabolism by two different pathways

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

: (1) by opposing IFN-γ–mediated IRP activation via NO formation, thus increasing ferritin translation; and (2) by an augmentation of TfR mRNA expression, which is most likely due to reversing the inhibitory effect of IFN-γ on TfR mRNA transcription. Therefore, Th-2–derived cytokines are able to increase iron uptake and storage in activated macrophages.

From this point of view, it appears reasonable that Th-1– and Th-2–derived cytokines may collaborate in inducing hypoferremia/hyperferretinemia in a way that Th-2–derived cytokines primarily enhance TfR-mediated iron uptake in activated macrophages, which is then stored in ferritin that has been effectively produced following stimulation with proinflammatory cytokines.

SHORT-LIVED RADICALS

Recently, it has been shown that short-lived radicals such as hydrogen peroxide (H2O2) or nitric oxide (NO)—both of which are produced during inflammatory responses associated with ACD—are able to influence cellular iron homeostasis. NO and H2O2 stimulate the binding affinity of cytoplasmic proteins (IRPs) to RNA stem loop structures (IRE)

28.

  • Weiss G.
  • Goossen B.
  • Doppler W.
  • Fuchs D.
  • Pantopoulos K.
  • Werner-Felmayer G.
  • Wachter H.
  • Hentze M.W.

Translational regulation via iron-responsive elements by the nitric oxide/NO-synthase pathway.

  • Crossref
  • PubMed
  • Scopus (333)
  • Google Scholar

,

29.

  • Pantopoulos K.
  • Hentze M.W.

Nitric oxide signaling to iron-regulatory protein: Direct control of ferritin mRNA translation and transferrin receptor mRNA stability in transfected fibroblasts.

  • Crossref
  • PubMed
  • Scopus (199)
  • Google Scholar

,

,

. One IRE is present within the 5′ untranslated regions of L-ferritin, H-ferritin mRNA, and erythroid amino-levulinate synthase (e-ALAS) mRNA, whereas five IREs have been detected within the 3′ untranslated region of TfR mRNA

,

. Binding of IRP-1 and IRP-2 to IREs within the 5′ untranslated region of target mRNAs results in a translational repression of their expression, whereas interaction with the IREs in the 3′ region enhances TfR mRNA stability by protecting it from digestion by a specific RNase. Therefore, circumstances that stimulate IRE-binding affinity of IRPs, such as NO formation, oxidative stress, or iron deprivation states, enhance iron uptake, whereas iron consumption and iron storage are reduced. In contrast, increased cellular iron concentrations as well as inhibition of NO or H2O2 formation result in loss of IRE-binding function of IRPs, thus leading to derepression of ferritin and eALAS mRNA translation while the TfR mRNA half-life is reduced. This, in turn, leads to iron consumption and storage while iron uptake is blocked

,

.

It is worth noting that the effects of NO and H2O2 on IRP activation are of a different nature. While H2O2 activates IRP-1 but not IRP-2

by a poorly understood fast-reacting mechanism involving kinase/phosphatase pathways, NO stimulates IRE-binding activity of both IRPs at least in macrophages with an albeit slower kinetic, which is comparable to that observed after pharmacological induction of iron deprivation by means of iron chelators

. This suggests that NO and H2O2 may be involved in different regulatory properties of iron homeostasis during inflammatory conditions

,

. Moreover, induction of oxidative stress on addition of a glutathione-depleting drug to hepatic cells was also shown to stimulate ferritin synthesis by a transcriptional mechanisms

35.

  • Cairo G.
  • Tacchini L.
  • Pogliaghi G.
  • Anzon E.
  • Tomasi A.
  • Bernelli-Zazzera A.

Induction of ferritin synthesis by oxidative stress: Transcriptional and post-transcriptional regulation by expansion of the free iron pool.

  • Crossref
  • PubMed
  • Scopus (287)
  • Google Scholar

.

However, cytokines may overcome the regulatory effects of these radicals. Apart from other mechanisms, oxygen radicals are formed by macrophages stimulated with IFN-γ, TNF-α and/or lipopolysaccharide (LPS), and murine macrophages or human hepatocytes also generate NO after treatment with the same stimuli, due to the induction of a cytokine-inducible form of NO synthase (NOS type II). Nevertheless, as stated earlier here, these cytokines have also direct effects on iron homeostasis, acting sometimes proximally from regulation by the IRE/IRP system. In activated murine macrophages and hepatocytes producing high amounts of NO after stimulation with IFN-γ and LPS, ferritin expression is inhibited translationally due to NO-mediated activation of IRPs, whereas NO is not able to stabilize TfR mRNA under such conditions

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

,

28.

  • Weiss G.
  • Goossen B.
  • Doppler W.
  • Fuchs D.
  • Pantopoulos K.
  • Werner-Felmayer G.
  • Wachter H.
  • Hentze M.W.

Translational regulation via iron-responsive elements by the nitric oxide/NO-synthase pathway.

  • Crossref
  • PubMed
  • Scopus (333)
  • Google Scholar

,

29.

  • Pantopoulos K.
  • Hentze M.W.

Nitric oxide signaling to iron-regulatory protein: Direct control of ferritin mRNA translation and transferrin receptor mRNA stability in transfected fibroblasts.

  • Crossref
  • PubMed
  • Scopus (199)
  • Google Scholar

,

36.

  • Phillips J.D.
  • Kinikini D.V.
  • Yu Y.
  • Guo B.
  • Leibold E.A.

Differential regulation of IRP1 and IRP2 by nitric oxide in rat hepatoma cells.

  • Crossref
  • PubMed
  • Google Scholar

. The latter observation is due to the fact that IFN-γ and LPS act proximally from the cytoplasmic regulation by the IRE/IRP system to inhibit TfR mRNA expression, probably within the nucleus.

However, the question remains of how radicals may be involved in the development of ACD. The observation that iron by itself also modulates the expression of NOS II by transcriptional mechanisms provided evidence for the existence of an autoregulatory loop in macrophages that links the maintenance of iron homeostasis with the formation of NO

37.

  • Weiss G.
  • Werner-Felmayer G.
  • Werner E.R.
  • Grünewald K.
  • Wachter H.
  • Hentze M.W.

Iron regulates nitric oxide synthase activity by controlling nuclear transcription.

  • Crossref
  • PubMed
  • Scopus (351)
  • Google Scholar

, which is centrally involved in host defense

. This loop could serve as a suitable mechanism for macrophages to acquire and store iron under inflammatory conditions, leading to the characteristic diversion of iron underlying ACD

37.

  • Weiss G.
  • Werner-Felmayer G.
  • Werner E.R.
  • Grünewald K.
  • Wachter H.
  • Hentze M.W.

Iron regulates nitric oxide synthase activity by controlling nuclear transcription.

  • Crossref
  • PubMed
  • Scopus (351)
  • Google Scholar

.

Besides its effect on cellular iron homeostasis, NO may contribute to the development of ACD by other mechanisms. First, NO is able to inhibit heme biosynthesis. This may be achieved on one hand by NO-mediated stimulation of high-affinity binding of IRP-1 to the IRE within the 5′ untranslated region of e-ALAS mRNA, thus resulting in translational repression of e-ALAS mRNA expression

39.

  • Rafferty S.P.
  • Domachowske J.B.
  • Malech H.L.

Inhibition of hemoglobin expression by heterologous production of nitric oxide synthase in the K562 erythroleukemic cell line.

  • Crossref
  • PubMed
  • Google Scholar

. On the other hand, NO is able to inhibit ferrochelatase activity in part by targeting its central iron sulfur cluster

40.

  • Furukawa T.
  • Kohno H.
  • Tokynaga R.
  • Taketani S.

Nitric oxide mediated inactivation of mammalian ferrochelatase in vivo and in vitro: Possible involvement of the iron-sulphur cluster of the enzyme.

  • Crossref
  • PubMed
  • Scopus (55)
  • Google Scholar

, which may be another mechanism contributing to abnormal heme biosynthesis in inflammation

41.

  • Houston T.
  • Moore M.
  • Porter D.
  • Sturrock R.
  • Fitzsimons E.

Abnormal haem biosynthesis in the chronic anemia of rheumatoid arthritis.

  • Crossref
  • Scopus (9)
  • Google Scholar

. Second, NO can directly block erythropoiesis by inhibiting the proliferation of erythroid progenitor cells in the bone marrow

,

43.

  • Ponka P.

Tissue specific regulation of iron metabolism and heme synthesis: Distinct control mechanisms in erythroid cells.

  • Crossref
  • PubMed
  • Google Scholar

.

Whether H2O2 may be involved in similar regulatory loops or have similar effects on e-ALAS expression or erythropoiesis remains to be shown.

Currently, ongoing studies with NOS II knock-out mice will hopefully contribute to the definition of the part NO plays among all other factors orchestrating iron metabolism under chronic inflammatory conditions.

ACUTE PHASE PROTEINS AND HORMONES

Some of the activating effects exerted by cytokines such as IL-1 or IL-6 on ferritin mRNA expression have been proposed to occur as part of the acute phase response in the liver in order to contribute to detoxification of iron by promoting its storage, thus preventing the catalyzation of toxic radical reactions by the metal

,

19.

  • Rogers J.T.

Ferritin translation by interleukin-1 and interleu-kin-6: The role of sequences upstream of the start codons of the heavy and light subunit genes.

  • Crossref
  • PubMed
  • Google Scholar

,

. Moreover, other acute phase proteins, such as α-1 antitrypsin or α-2 macroglobulin, have been demonstrated to inhibit TfR-mediated iron uptake into erythroid cells and hepatocytes, but not into monocytic cells, by direct competitive interaction with the TfR

45.

  • Graziadei I.
  • Gaggl S.
  • Kaserbacher R.
  • Braunsteiner H.
  • Vogel W.

The acute phase protein alpha-1 antitrypsin inhibits growth and proliferation of human early erythroid progenitor cells and of human erythroleukemic cells by interfering with transferrin iron uptake.

  • Crossref
  • PubMed
  • Google Scholar

,

46.

  • Weiss G.
  • Graziadei I.
  • Urbanek M.
  • Grünewald K.
  • Vogel W.

Divergent effects of α1-antitrypsin on the regulation of iron metabolism in human erythroleukemic (K562) and myelomonocytic (THP-1) cells.

  • Crossref
  • PubMed
  • Scopus (17)
  • Google Scholar

. This may also contribute to the ACD-typical diversion of iron traffic by favoring iron uptake by monocytic cells while the metal is withdrawn from the erythron, thus blocking the proliferation of erythroid progenitors

45.

  • Graziadei I.
  • Gaggl S.
  • Kaserbacher R.
  • Braunsteiner H.
  • Vogel W.

The acute phase protein alpha-1 antitrypsin inhibits growth and proliferation of human early erythroid progenitor cells and of human erythroleukemic cells by interfering with transferrin iron uptake.

  • Crossref
  • PubMed
  • Google Scholar

.

Most recently, thyroid hormones have been shown to modulate hepatic iron metabolism translationally via stimulation of ferritin synthesis by inducing alterations of IRE/IRP interactions

,

48.

  • Leedman P.J.
  • Stein A.R.
  • Chin W.W.
  • Rogers J.T.

Thyroid hormone modulates the interaction between iron regulatory proteins and the ferritin mRNA iron-responsive element.

  • Crossref
  • PubMed
  • Scopus (72)
  • Google Scholar

.

Because circulating amounts of stress hormones may be increased during inflammatory processes, it will be interesting to see to which extent thyroid hormones may participate in alterations of iron homeostasis occurring in ACD.

WHY ANEMIA OF CHRONIC DISEASE?

Because ACD is very frequently occurring under chronic inflammatory conditions, it is tempting to speculate about what could be the body's idea causing these diversions of iron traffic that underlie this disease.

Iron and immunity

Iron has multiple effects on cell-mediated immunity. On one hand, it differently modulates the proliferation and differentiation of lymphocyte subsets, whereas, on the other hand, it strongly affects the immune potential of macrophages

,

49.

  • De Sousa M.
  • Reimao R.
  • Porto G.
  • Grady R.W.
  • Hilgartner M.W.
  • Giardina P.

Iron and lymphocytes: Reciprocal regulatory interactions.

  • Crossref
  • Google Scholar

,

50.

  • Brock J.H.

Iron in infection, immunity, inflammation and neoplasia,.

  • Google Scholar

. The latter effects are due in part to an iron-mediated inhibition of IFN-γ–directed immune response pathways in macrophages

51.

  • Weiss G.
  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Wachter H.

Iron modulates interferon-gamma effects in the human myelomonocytic cell line THP-1.

  • PubMed
  • Google Scholar

. Therefore, iron-loaded macrophages exhibit reduced IFN-γ responsiveness, TNF-α production, and the formation of NO

,

,

50.

  • Brock J.H.

Iron in infection, immunity, inflammation and neoplasia,.

  • Google Scholar

,

51.

  • Weiss G.
  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Wachter H.

Iron modulates interferon-gamma effects in the human myelomonocytic cell line THP-1.

  • PubMed
  • Google Scholar

,

52.

  • Gordeuk V.R.
  • Ballou S.
  • Lozanski G.
  • Brittenham G.M.

Decreased concentrations of tumor necrosis factor-alpha in supernatants of monocytes from homozygotes for hereditary hemochromatosis.

  • PubMed
  • Google Scholar

, and the immune defense against various intracellular pathogens and viruses is impaired under such conditions

,

54.

  • Byrd T.F.
  • Horwitz M.A.

Lactoferrin inhibits or promotes Legionella pneumophila intracellular multiplication in nonactivated and interferon-gamma activated human monocytes depending upon its degree of iron saturation: Iron-lactoferrin and nonphysiologic iron chelates reverse monocyte activation against Legionella pneumophila.

  • Crossref
  • PubMed
  • Scopus (97)
  • Google Scholar

,

55.

  • Barnewell R.E.
  • Rikihias Y.

Abrogation of gamma interferon-induced inhibition of Ehrlichia chaffeensis infection in human monocytes with transferrin iron.

  • Google Scholar

,

,

. From this point of view, withdrawal of metabolically active iron from the circulation and storage of the metal, as it occurs in ACD, would contribute to strengthening the immune response via stimulation of IFN-γ–mediated immune effector mechanisms.

On the contrary, IL-4 and IL-13 may reduce Th-1–mediated immune effector function by increasing intracellular iron content via stimulation of TfR expression, which may be part of their anti-inflammatory and macrophage-deactivating function

25.

  • Weiss G.
  • Bogdan C.
  • Hentze M.W.

Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

  • PubMed
  • Google Scholar

.

Iron withholding

Because of the multiple functions of iron in metabolism outlined in the beginning of this article, this metal is an essential growth factor for proliferating tissues and microorganisms. Therefore, withholding iron from invading pathogens and tumor cells would limit their growth by affecting their energy metabolism or DNA synthesis

,

. Furthermore, decreased formation of hemoglobin by withholding iron from the erythron and inhibition of erythropoiesis by cytokines reduces the oxygen transport capacity of the blood, thus decreasing the overall oxygen supply that may primarily affect rapid proliferating (malignant) tissues and microorganisms.

Other mechanisms

Besides the underlying mechanism of iron perturbations in ACD, other factors not discussed in this review may also contribute to ACD. Bacterial toxins and cytokines, such as TNF-α, have been proposed to decrease erythrocyte survival and to cause an increased phagocytosis of these cells by splenic macrophages. This is compatible with the observation that during inflammatory states an increase of erythrocyte-derived iron in splenic macrophages and Kupffer cells takes place

,

60.

  • Fillet G.
  • Beguin Y.
  • Baldelli L.

Model of reticuloendothelial iron metabolism in human: Abnormal behaviour in idiopathic hemochromatosis and in inflammation.

  • Crossref
  • PubMed
  • Google Scholar

,

61.

  • Kitagawa S.
  • Yuo A.
  • Yagisawa M.
  • Azuma E.
  • Yoshida M.
  • Furukawa Y.
  • Takahashi M.
  • Masuyama J.
  • Takaku F.

Activation of human monocyte functions by tumor necrosis factor: Rapid priming for enhanced release of superoxide and erythrophagocytosis, but no direct triggering of superoxide release.

  • PubMed
  • Google Scholar

. Nevertheless, to date there is no evidence on the extent to which erythrophagocytosis may play a role in ACD and the associated disturbances of iron homeostasis, although the release of iron from phagocytosed erythrocytes may have distinct, but not yet elucidated, effects on the regulation of iron metabolism in macrophages.

Alternatively, ACD may just be seen as a disease occurring as a side effect of an ongoing immune response. Cytokines not only affect iron metabolism, as shown herein, but they also exert inhibitory effects on erythropoiesis by blocking proliferation and differentiation of erythroid progenitor cells, affecting their ability to respond to erythropoietin and causing deficiency in the production of erythropoietin

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

,

62.

  • Greendyke R.M.
  • Sharma K.
  • Gifford F.R.

Serum levels of erythropoietin and selected cytokines in patients with anemia of chronic disease.

  • PubMed
  • Google Scholar

,

. Suppression of erythropoiesis can also be exerted by invading pathogens itself, as shown for human immunodeficiency virus (HIV) or malaria plasmodia

64.

  • Boelaert J.R.
  • Weinberg G.A.
  • Weinberg E.D.

Altered iron metabolism in HIV infection: Mechanisms, possible consequences and proposals for management.

  • PubMed
  • Google Scholar

,

65.

  • Yap G.S.
  • Stevenson M.M.

Inhibition of in vitro erythropoiesis by soluble mediators of Plasmodium chabaudi AS malaria: Lack of a major role of interleukin 1, tumor necrosis factor alpha, and gamma interferon.

  • PubMed
  • Google Scholar

. Finally, antiproliferative effects toward erythropoiesis have also been described for ferritin

66.

  • Broxmeyer H.E.

H-ferritin: A regulatory cytokine that down-modulates cell proliferation.

  • PubMed
  • Google Scholar

. This may refer to an as yet not fully understood mechanism by which extracellular ferritin causes the limitation of transferrin-mediated iron uptake into erythroid progenitor cells

66.

  • Broxmeyer H.E.

H-ferritin: A regulatory cytokine that down-modulates cell proliferation.

  • PubMed
  • Google Scholar

. Because cytokines are able to block hematopoiesis and, at the same time, induce ferritin expression, it appears reasonable that some of the antiproliferative actions of cytokines can be attributed to their interference with iron metabolism

,

,

.

TREATMENT OF ANEMIA OF CHRONIC DISEASE

Finally, the question remains of how ACD should be treated. There is a wide consensus in literature that the superior treatment of ACD is the cure of the underlying disease. The supply of iron to such patients is controversial

,

4.

  • Konjin A.
  • Hershko C.

The anaemia of inflammation and chronic disease,.

  • Google Scholar

,

5.

  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Dierich M.P.
  • Wachter H.

Immune activation and the anaemia associated with chronic inflammatory disorders.

  • Crossref
  • PubMed
  • Scopus (113)
  • Google Scholar

,

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

,

,

,

,

60.

  • Fillet G.
  • Beguin Y.
  • Baldelli L.

Model of reticuloendothelial iron metabolism in human: Abnormal behaviour in idiopathic hemochromatosis and in inflammation.

  • Crossref
  • PubMed
  • Google Scholar

, and should be avoided (at least in my opinion) in patients with chronic infectious or tumor diseases for several reasons. On one hand, supplementation of iron would promote growth and proliferation of tumor cells and microorganisms underlying the chronic inflammatory disease causing ACD

,

; on the other hand, cell-mediated immune effector mechanisms may be weakened by the action of iron

,

,

50.

  • Brock J.H.

Iron in infection, immunity, inflammation and neoplasia,.

  • Google Scholar

,

51.

  • Weiss G.
  • Fuchs D.
  • Hausen A.
  • Reibnegger G.
  • Werner E.R.
  • Werner-Felmayer G.
  • Wachter H.

Iron modulates interferon-gamma effects in the human myelomonocytic cell line THP-1.

  • PubMed
  • Google Scholar

,

52.

  • Gordeuk V.R.
  • Ballou S.
  • Lozanski G.
  • Brittenham G.M.

Decreased concentrations of tumor necrosis factor-alpha in supernatants of monocytes from homozygotes for hereditary hemochromatosis.

  • PubMed
  • Google Scholar

,

,

54.

  • Byrd T.F.
  • Horwitz M.A.

Lactoferrin inhibits or promotes Legionella pneumophila intracellular multiplication in nonactivated and interferon-gamma activated human monocytes depending upon its degree of iron saturation: Iron-lactoferrin and nonphysiologic iron chelates reverse monocyte activation against Legionella pneumophila.

  • Crossref
  • PubMed
  • Scopus (97)
  • Google Scholar

,

55.

  • Barnewell R.E.
  • Rikihias Y.

Abrogation of gamma interferon-induced inhibition of Ehrlichia chaffeensis infection in human monocytes with transferrin iron.

  • Google Scholar

,

,

. Moreover, it is questionable how much iron may pass over into the erythron due to the withholding mechanism outlined in this review. On the contrary, supplementation of iron to ACD patients with an autoimmune disorder may be rather beneficial

67.

  • Cazzola M.
  • Ponchio L.
  • deBenedetti F.
  • Ravelli A.
  • Rosti V.
  • Beguin Y.
  • Invernizzi R.
  • Barosi G.
  • Martini A.

Defective iron supply for erythropoiesis and adequate endogenous erythropoietin production in anemia associated with systemic onset juvenile chronic arthritis.

  • PubMed
  • Google Scholar

,

68.

  • Mutane J.
  • Piug-Parellada P.
  • Mitjavila M.T.

Iron metabolism and oxidative stress during acute and chronic phases of experimental inflammation: Effect of iron dextran and deferoxamine.

  • Google Scholar

, most likely because of the described inhibitory actions of iron on cytokine actions and macrophage-mediated cytotoxicity, although the contribution of the metal to the formation of toxic radicals has to be taken into account

68.

  • Mutane J.
  • Piug-Parellada P.
  • Mitjavila M.T.

Iron metabolism and oxidative stress during acute and chronic phases of experimental inflammation: Effect of iron dextran and deferoxamine.

  • Google Scholar

. Interestingly, recent reports have also suggested using iron chelation with desferrioxamine for the treatment of ACD despite low serum iron concentrations. Such a therapy was associated with a rise in hemoglobin levels, which was attributed to stimulation of erythropoietin production

69.

  • Salvarani C.
  • Baricchi R.
  • Lasagni D.
  • Boiardi L.
  • Piccinini R.
  • Brunati C.
  • Macchioni P.
  • Portiolo I.

Effects of desferrioxamine therapy on chronic disease anemia associated with rheumatoid arthritis.

  • Crossref
  • PubMed
  • Scopus (18)
  • Google Scholar

via induction of the binding of hypoxia regulatory factors to the erythropoietin promoter.

Recombinant erythropoietin is a widely used therapeutic approach for ACD (discussed in this issue)

6.

  • Means R.T.
  • Krantz S.B.

Progress in understanding the pathogenesis of the anemia of chronic disease.

  • PubMed
  • Google Scholar

,

62.

  • Greendyke R.M.
  • Sharma K.
  • Gifford F.R.

Serum levels of erythropoietin and selected cytokines in patients with anemia of chronic disease.

  • PubMed
  • Google Scholar

,

. However, response rates to the therapy with erythropoietin are sometimes low. This may refer, in part, to the fact that a prominently stimulated immune activation will inhibit erythropoiesis very efficiently via the action of cytokines, whereas, on the other hand, iron is withdrawn from the erythron and shifted into the reticuloendothelial system by the mechanisms described earlier in this article. However, erythropoietin is able to up-regulate TfR expression on erythroid progenitor cells via transcriptional and post-transcriptional mechanisms and therefore affects iron balance

43.

  • Ponka P.

Tissue specific regulation of iron metabolism and heme synthesis: Distinct control mechanisms in erythroid cells.

  • Crossref
  • PubMed
  • Google Scholar

,

70.

  • Weiss G.
  • Houston T.
  • Kastner S.
  • Grünewald K.
  • Brock J.

Regulation of cellular iron metabolism by erythropoietin: Activation of iron regulatory protein and up-regulation of transferrin-receptor in erythroid cells.

  • Crossref
  • PubMed
  • Google Scholar

,

71.

  • Adamson J.W.

The relationship of erythropoietin and iron metabolism to red blood cell production in humans.

  • PubMed
  • Google Scholar

,

72.

  • Pootrakul P.
  • Kitcharoen K.
  • Yansukon P.
  • Wasi P.
  • Fucharoen S.
  • Charoenlarp P.
  • Brittenham G.
  • Pippard M.J.
  • Finch C.A.

The effect of erythroid hyperplasia on iron balance.

  • PubMed
  • Google Scholar

. It remains to be seen whether sequential administration of erythropoietin and iron (such as 48 hr later) will favor a more efficient uptake of iron into erythroid progenitors and consecutive stimulation of erythropoiesis, so that the detrimental effects of this metal on the growth of pathogens and down-regulation of immune effector function can be minimized.

ACKNOWLEDGMENTS

Support by grant FWF 12186 from the Austrian Research Funds is gratefully acknowledged. I thank Dr. Jeremy Brock for critical reading of the manuscript.

References

    • Brock J.H.
    • Halliday J.W.
    • Pippard M.J.
    • Powell L.W.
    Iron Metabolism in Health and Disease. W.B. Saunders, London 1994
    • Rosen G.M.
    • Pou S.
    • Ramos C.L.
    • Cohen M.S.
    • Britigan B.E.

    Free radicals and phagocytic cells.

    FASEB J. 1995; 9 : 200-209
    • Cartwright G.E.

    The anemia of chronic disorders.

    Semin Hematol. 1966; 3 : 351-368
    • Konjin A.
    • Hershko C.

    The anaemia of inflammation and chronic disease,.

    in: DeSousa M. Brock J.H. Iron in Immunity, Cancer and Inflammation. Wiley and Sons, Chichester 1989: 111-143
    • Fuchs D.
    • Hausen A.
    • Reibnegger G.
    • Werner E.R.
    • Werner-Felmayer G.
    • Dierich M.P.
    • Wachter H.

    Immune activation and the anaemia associated with chronic inflammatory disorders.

    Eur J Haematol. 1991; 46 : 65-70
    • Means R.T.
    • Krantz S.B.

    Progress in understanding the pathogenesis of the anemia of chronic disease.

    Blood. 1992; 80 : 1639-1647
    • Kuiper-Kramer E.P.
    • Huisman C.M.
    • vanRaan J.
    • vanEijk H.G.

    Analytical and clinical implications of soluble transferrin receptor in serum.

    Eur J Clin Chem Clin Biochem. 1996; 34 : 645-649
    • Weiss G.
    • Wachter H.
    • Fuchs D.

    Linkage of cell mediated immunity to iron metabolism.

    Immunol Today. 1995; 16 : 495-500
    • Mosmann T.R.
    • Coffman R.L.

    Th1 and Th2 cells: Different patterns of lymphokine secretion lead to different functional properties.

    Annu Rev Immunol. 1989; 7 : 145-173
    • Seder R.A.
    • Paul W.E.

    Acquisition of lymphokine-producing phenotype by CD4+ cells.

    Ann Rev Immunol. 1994; 12 : 635-673
    • Romagnani S.

    The Th1/Th2 paradigm.

    Immunol Today. 1997; 18 : 263-266
    • Alvarez-Hernandez X.
    • Licega J.
    • McKay I.
    • Brock J.H.

    Induction of hypoferremia and modulation of macrophage iron metabolism by tumor necrosis factor.

    Lab Invest. 1989; 61 : 319-322
    • Brock J.H.
    • Alvarez-Hernandez X.

    Modulation of macrophage iron metabolism by tumour necrosis factor and interleukin-1.

    FEMS Microbiol Immunol. 1989; 47 : 309-310
    • Konjin A.M.
    • Carmel N.
    • Levy R.
    • Hershko C.

    Ferritin synthesis in inflammation. Mechanism of increased ferritin synthesis.

    Br J Haematol. 1981; 49 : 361-368
    • Torti S.V.
    • Kwak E.L.
    • Miller S.C.
    • Miller L.L.
    • Ringold G.M.
    • Myambo K.B.
    • Young A.P.
    • Torti F.M.

    The molecular cloning and characterisation of ferritin heavy chain, a tumor necrosis factor inducible gene.

    J Biol Chem. 1988; 263 : 12638-12644
    • Wei Y.S.
    • Miller S.C.
    • Tsuji Y.
    • Torti S.
    • Torti F.M.

    Interleukin 1 induces ferritin heavy chain in human muscle cells.

    Biochem Biophys Res Commun. 1989; 169 : 289-296
    • Fahmy M.
    • Young S.P.

    Modulation of iron metabolism in monocyte cell line U937 by inflammatory cytokines: Changes in transferrin uptake, iron handling and ferritin mRNA.

    Biochem J. 1993; 296 : 175-181
    • Rogers J.T.
    • Bridges K.B.
    • Durmowics G.
    • Glass J.
    • Auron P.R.
    • Munro H.N.

    Translational control during the acute phase response: Ferritin synthesis in response to interleukin 1.

    J Biol Chem. 1990; 265 : 14572-14578
    • Rogers J.T.

    Ferritin translation by interleukin-1 and interleu-kin-6: The role of sequences upstream of the start codons of the heavy and light subunit genes.

    Blood. 1996; 87 : 2525-2537
    • Kobune M.
    • Kohgo Y.
    • Kato J.
    • Miyazaki E.
    • Niitsu Y.

    Interleu-kin-6 enhances hepatic transferrin uptake and ferritin expression in rats.

    Hepatology. 1994; 19 : 1468-1475
    • Tsuji Y.
    • Miller L.L.
    • Miller S.C.
    • Torit S.V.
    • Torti F.M.

    Tumor necrosis factor-α and interleukin 1-α regulate transferrin receptor in human diploid fibroblasts.

    J Biol Chem. 1991; 266 : 7257-7261
    • Bourgeade M.F.
    • Silbermann F.
    • Kühn L.
    • Testa U.
    • Peschle C.
    • Memet S.
    • Thang M.N.
    • Besancon F.

    Post-transcriptional regulation of transferrin receptor mRNA by IFN-γ.

    Nucleic Acids Res. 1992; 20 : 2997-3003
    • Byrd T.
    • Horwitz M.A.

    Regulation of transferrin receptor expression and ferritin content in human mononuclear macrophages: Coordinate upregulation by iron transferrin and downregulation by interferon gamma.

    J Clin Invest. 1993; 91 : 969-976
    • Hamilton T.
    • Gray P.
    • Adams D.

    Expression of the transferrin receptor on murine peritoneal macrophages is modulated by in vitro treatment with IFN-γ.

    Cell Immunol. 1984; 89 : 478-485
    • Weiss G.
    • Bogdan C.
    • Hentze M.W.

    Pathways for the regulation of macrophage iron metabolism by the anti-inflammatory cytokines IL-4 and IL-13.

    J Immunol. 1997; 158 : 420-425
    • Taetle R.
    • Honeysett J.M.

    γ-Interferon modulates human monocyte/macrophage transferrin receptor expression.

    Blood. 1988; 71 : 1590-1595
    • Testa U.
    • Conti L.
    • Sposi N.M.
    • Varano B.
    • Tritarelli E.
    • Malorni W.
    • Samoggia P.
    • Rainalidi G.
    • Peschle C.
    • Beladelli F.
    • Gessani S.

    IFN-β selectively downregulates transferrin receptor expression in human peripheral blood macrophages by a posttranslational mechanism.

    J Immunol. 1995; 155 : 427-435
    • Weiss G.
    • Goossen B.
    • Doppler W.
    • Fuchs D.
    • Pantopoulos K.
    • Werner-Felmayer G.
    • Wachter H.
    • Hentze M.W.

    Translational regulation via iron-responsive elements by the nitric oxide/NO-synthase pathway.

    EMBO J. 1993; 12 : 3651-3657
    • Pantopoulos K.
    • Hentze M.W.

    Nitric oxide signaling to iron-regulatory protein: Direct control of ferritin mRNA translation and transferrin receptor mRNA stability in transfected fibroblasts.

    Proc Natl Acad Sci USA. 1995; 92 : 1267-1271
    • Drapier J.C.
    • Hirling H.
    • Wietzerbin H.
    • Kaldy P.
    • Kühn L.C.

    Biosynthesis of nitric oxide activates iron regulatory factor in macrophages.

    EMBO J. 1993; 12 : 3643-3650
    • Pantopoulos K.
    • Hentze M.W.

    Rapid responses to oxidative stress mediated by iron regulatory protein.

    EMBO J. 1995; 14 : 2917-2924
    • Hentze M.W.
    • Kühn L.C.

    Molecular control of vertebrate iron metabolism: mRNA-based regulatory circuits operated by iron, nitric oxide, and oxidative stress.

    Proc Natl Acad Sci USA. 1996; 93 : 8175-8182
    • Klausner R.
    • Rouault T.A.
    • Harford J.B.

    Regulating the fate of mRNA. The control of cellular iron metabolism.

    Cell. 1993; 72 : 19-22
    • Pantopoulos K.
    • Weiss G.
    • Hentze M.W.

    Nitric oxide and oxidative stress control mammalian iron metabolism by different pathways.

    Mol Cell Biol. 1996; 16 : 3871-3878
    • Cairo G.
    • Tacchini L.
    • Pogliaghi G.
    • Anzon E.
    • Tomasi A.
    • Bernelli-Zazzera A.

    Induction of ferritin synthesis by oxidative stress: Transcriptional and post-transcriptional regulation by expansion of the free iron pool.

    J Biol Chem. 1995; 270 : 700-703
    • Phillips J.D.
    • Kinikini D.V.
    • Yu Y.
    • Guo B.
    • Leibold E.A.

    Differential regulation of IRP1 and IRP2 by nitric oxide in rat hepatoma cells.

    Blood. 1996; 87 : 2983-2992
    • Weiss G.
    • Werner-Felmayer G.
    • Werner E.R.
    • Grünewald K.
    • Wachter H.
    • Hentze M.W.

    Iron regulates nitric oxide synthase activity by controlling nuclear transcription.

    J Exp Med. 1994; 180 : 969-976
    • Macmicking J.
    • Xie Q.-W.
    • Nathan C.

    Nitric oxide and macrophage function.

    Annu Rev Immunol. 1997; 15 : 323-350
    • Rafferty S.P.
    • Domachowske J.B.
    • Malech H.L.

    Inhibition of hemoglobin expression by heterologous production of nitric oxide synthase in the K562 erythroleukemic cell line.

    Blood. 1996; 88 : 1070-1078
    • Furukawa T.
    • Kohno H.
    • Tokynaga R.
    • Taketani S.

    Nitric oxide mediated inactivation of mammalian ferrochelatase in vivo and in vitro: Possible involvement of the iron-sulphur cluster of the enzyme.

    Biochem J. 1995; 310 : 533-538
    • Houston T.
    • Moore M.
    • Porter D.
    • Sturrock R.
    • Fitzsimons E.

    Abnormal haem biosynthesis in the chronic anemia of rheumatoid arthritis.

    Ann Rheum Dis. 1994; 53 : 167-170
    • Maciejewski J.P.
    • Selleri C.
    • Sato T.
    • Cho H.J.
    • Keefer L.K.
    • Nathan C.F.
    • Young N.S.

    Nitric oxide suppression of human hematopoiesis in vitro.

    J Clin Invest. 1995; 96 : 1085-1092
    • Ponka P.

    Tissue specific regulation of iron metabolism and heme synthesis: Distinct control mechanisms in erythroid cells.

    Blood. 1997; 89 : 1-25
    • Bomford A.B.
    • Munro H.N.

    Ferritin gene expression in health and malignancy.

    Pathobiology. 1992; 60 : 10-18
    • Graziadei I.
    • Gaggl S.
    • Kaserbacher R.
    • Braunsteiner H.
    • Vogel W.

    The acute phase protein alpha-1 antitrypsin inhibits growth and proliferation of human early erythroid progenitor cells and of human erythroleukemic cells by interfering with transferrin iron uptake.

    Blood. 1994; 83 : 260-268
    • Weiss G.
    • Graziadei I.
    • Urbanek M.
    • Grünewald K.
    • Vogel W.

    Divergent effects of α1-antitrypsin on the regulation of iron metabolism in human erythroleukemic (K562) and myelomonocytic (THP-1) cells.

    Biochem J. 1996; 319 : 897-902
    • Kubota K.
    • Tamura J.
    • Kurabayashi H.
    • Shirakura T.
    • Kobayashi I.

    Evaluation of increased serum ferritin levels in patients with hyperthyroidism.

    Clin Invest. 1993; 72 : 26-29
    • Leedman P.J.
    • Stein A.R.
    • Chin W.W.
    • Rogers J.T.

    Thyroid hormone modulates the interaction between iron regulatory proteins and the ferritin mRNA iron-responsive element.

    J Biol Chem. 1996; 271 : 12017-12023
    • De Sousa M.
    • Reimao R.
    • Porto G.
    • Grady R.W.
    • Hilgartner M.W.
    • Giardina P.

    Iron and lymphocytes: Reciprocal regulatory interactions.

    Curr Stud Hematol Blood Transfus. 1992; 58 : 171-177
    • Brock J.H.

    Iron in infection, immunity, inflammation and neoplasia,.

    in: Brock J.H. Halliday J.W. Pippard M.J. Powell L.W. Iron Metabolism in Health and Disease. W.B. Saunders, London 1994: 353-389
    • Weiss G.
    • Fuchs D.
    • Hausen A.
    • Reibnegger G.
    • Werner E.R.
    • Werner-Felmayer G.
    • Wachter H.

    Iron modulates interferon-gamma effects in the human myelomonocytic cell line THP-1.

    Exp Hematol. 1992; 20 : 605-610
    • Gordeuk V.R.
    • Ballou S.
    • Lozanski G.
    • Brittenham G.M.

    Decreased concentrations of tumor necrosis factor-alpha in supernatants of monocytes from homozygotes for hereditary hemochromatosis.

    Blood. 1992; 79 : 1855-1860
    • Alford C.E.
    • King T.E.
    • Campell P.A.

    Role of transferrin, transferrin receptors, and iron in macrophage listericidal activity.

    J Exp Med. 1991; 174 : 459-466
    • Byrd T.F.
    • Horwitz M.A.

    Lactoferrin inhibits or promotes Legionella pneumophila intracellular multiplication in nonactivated and interferon-gamma activated human monocytes depending upon its degree of iron saturation: Iron-lactoferrin and nonphysiologic iron chelates reverse monocyte activation against Legionella pneumophila.

    J Clin Invest. 1991; 88 : 1103-1112
    • Barnewell R.E.
    • Rikihias Y.

    Abrogation of gamma interferon-induced inhibition of Ehrlichia chaffeensis infection in human monocytes with transferrin iron.

    Infect Immun. 1994; 82 : 4804-4810
    • Karupiah G.
    • Harris N.

    Inhibition of viral replication by nitric oxide and its reversal by ferrous sulfate and tricarboxylic acid cycle metabolites.

    J Exp Med. 1995; 181 : 2171-2180
    • Menacci A.
    • Cenci E.
    • Boelaert J.R.
    • Bucci P.
    • Mosci P.
    • Fe'd'Ostiani C.
    • Bistoni F.
    • Romani L.

    Iron overload alters T helper cell responses to Candida albicans in mice.

    J Infect Dis. 1997; 175 : 1467-1476
    • Weinberg E.D.

    Iron withholding: A defense against infection and neoplasia.

    Physiol Rev. 1984; 64 : 65-102
    • Weinberg E.D.
    • Weinberg G.A.

    The role of iron in infection.

    Curr Opin Infect Dis. 1995; 8 : 164-169
    • Fillet G.
    • Beguin Y.
    • Baldelli L.

    Model of reticuloendothelial iron metabolism in human: Abnormal behaviour in idiopathic hemochromatosis and in inflammation.

    Blood. 1989; 74 : 844-851
    • Kitagawa S.
    • Yuo A.
    • Yagisawa M.
    • Azuma E.
    • Yoshida M.
    • Furukawa Y.
    • Takahashi M.
    • Masuyama J.
    • Takaku F.

    Activation of human monocyte functions by tumor necrosis factor: Rapid priming for enhanced release of superoxide and erythrophagocytosis, but no direct triggering of superoxide release.

    Exp Hematol. 1996; 24 : 559-567
    • Greendyke R.M.
    • Sharma K.
    • Gifford F.R.

    Serum levels of erythropoietin and selected cytokines in patients with anemia of chronic disease.

    Am J Clin Pathol. 1994; 101 : 338-341
    • Means R.T.

    Pathogenesis of the anemia of chronic disease: A cytokine mediated anemia.

    Stem Cells (Dayt). 1995; 13 : 32-37
    • Boelaert J.R.
    • Weinberg G.A.
    • Weinberg E.D.

    Altered iron metabolism in HIV infection: Mechanisms, possible consequences and proposals for management.

    Infect Dis Agents. 1996; 5 : 36-46
    • Yap G.S.
    • Stevenson M.M.

    Inhibition of in vitro erythropoiesis by soluble mediators of Plasmodium chabaudi AS malaria: Lack of a major role of interleukin 1, tumor necrosis factor alpha, and gamma interferon.

    Infect Immun. 1994; 62 : 357-362
    • Broxmeyer H.E.

    H-ferritin: A regulatory cytokine that down-modulates cell proliferation.

    J Lab Clin Med. 1992; 120 : 367-370
    • Cazzola M.
    • Ponchio L.
    • deBenedetti F.
    • Ravelli A.
    • Rosti V.
    • Beguin Y.
    • Invernizzi R.
    • Barosi G.
    • Martini A.

    Defective iron supply for erythropoiesis and adequate endogenous erythropoietin production in anemia associated with systemic onset juvenile chronic arthritis.

    Blood. 1996; 87 : 4824-4830
    • Mutane J.
    • Piug-Parellada P.
    • Mitjavila M.T.

    Iron metabolism and oxidative stress during acute and chronic phases of experimental inflammation: Effect of iron dextran and deferoxamine.

    J Lab Clin Med. 1995; 126 : 435-443
    • Salvarani C.
    • Baricchi R.
    • Lasagni D.
    • Boiardi L.
    • Piccinini R.
    • Brunati C.
    • Macchioni P.
    • Portiolo I.

    Effects of desferrioxamine therapy on chronic disease anemia associated with rheumatoid arthritis.

    Rheumatol Int. 1996; 16 : 45-48
    • Weiss G.
    • Houston T.
    • Kastner S.
    • Grünewald K.
    • Brock J.

    Regulation of cellular iron metabolism by erythropoietin: Activation of iron regulatory protein and up-regulation of transferrin-receptor in erythroid cells.

    Blood. 1997; 89 : 680-687
    • Adamson J.W.

    The relationship of erythropoietin and iron metabolism to red blood cell production in humans.

    Semin Oncol. 1994; 21 : 9-15
    • Pootrakul P.
    • Kitcharoen K.
    • Yansukon P.
    • Wasi P.
    • Fucharoen S.
    • Charoenlarp P.
    • Brittenham G.
    • Pippard M.J.
    • Finch C.A.

    The effect of erythroid hyperplasia on iron balance.

    Blood. 1988; 71 : 1124-1129

Article Info

Identification

DOI: https://doi.org/10.1046/j.1523-1755.1999.055Suppl.69012.x

Copyright

© 1999 International Society of Nephrology. Published by Elsevier Inc.

User License

Elsevier user license |

How you can reuse Information Icon

Permitted

For non-commercial purposes:

  • Read, print & download
  • Text & data mine
  • Translate the article

Not Permitted

  • Reuse portions or extracts from the article in other works
  • Redistribute or republish the final article
  • Sell or re-use for commercial purposes

Elsevier's open access license policy

ScienceDirect

Access this article on ScienceDirect
  • View Large Image
  • Download Hi-res image
  • Download .PPT

World Health Organization Chronic Disease Definition

Source: https://www.kidney-international.org/article/S0085-2538(15)46231-3/fulltext

0 Response to "World Health Organization Chronic Disease Definition"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel