PubMed: 24904418

Title
Heme on innate immunity and inflammation.
Journal
Frontiers in pharmacology
Volume
5
Issue
None
Pages
115
Date
2014-01-01
Authors
Bozza MT | Dutra FF

Evidence a51adf7c4f

Furthermore, heme induces ROS generation dependent on enzymatic reactions.

Evidence 9804c1bdc0

Heme induces ROS generation independently of TLR4.

Evidence aa56fbd3fb

Heme activates neutrophils and endothelial cells, by ROS generation.

Evidence 1f7a8628b6

Differently from biliverdin and CO, which have anti-inflammatory effects (Otterbein et al., 2000; Baranano et al., 2002), free Fe is highly oxidative and can promote free radicals generation through the Fenton reaction, which catalyzes hydroxyl radicals from the reaction of Fe with H2O2 (Fenton, 1894).

Evidence 617bc38603

Heme also induces the expression of the adhesion molecules ICAM-1, VCAM-1, and E-selectins.

Evidence b54040e125

CO inhibits Hb oxidation and subsequently heme release, thus blocking heme accumulation in serum and preventing heme from exerting its inflammatory effects in the course of malaria disease (Ferreira et al., 2011).

Evidence b7865d6247

Besides its physiological importance, heme has a potent oxidative capacity oxidizing lipids (Tappel, 1953, 1955; Vincent et al., 1988) and proteins (Aft and Mueller, 1984; Vincent, 1989), and damaging DNA (Aft and Mueller, 1983).

Evidence f13172c282

Heme-induced necroptosis is reversed by deferoxamine, a Fe chelator.

Evidence 1483ab0c31

Once intercalated into cellular plasma membranes heme amplifies cellular susceptibility to oxidative-mediated injury by oxidants such as H2O2 or those derived from activated inflammatory cells (Balla et al., 1991a,b, 1993).

Evidence 835c429aa4

These concepts challenged the idea that the cytotoxic and inflammatory effects of heme were exclusively mediated by the oxidative capability of the Fe associated with the amphipathic property of the porphyrin ring.

Evidence 100d76b39b

In fact, heme can induce neutrophil migration by acting as a chemotactic molecule (Porto et al., 2007) or by inducing the production of leukotriene B4 (LTB4) by macrophages (Monteiro et al., 2011).

Evidence 397d6c52f2

As described before, LTB4 has an important function regulating heme-induced neutrophils migration (Monteiro et al., 2011).

Evidence 9240a50013

KC (keratinocyte-derived chemokine) is a chemokine that attracts neutrophils to sites of inflammation and LTB4 is a lipid mediator that functions as a chemoattractant molecule and also activates leukocytes.

Evidence 9e5837ce24

Low-density lipoprotein is the major lipid involved in plaque formation.

Evidence 566745a966

In fact, heme-induced LDL oxidation is highly cytotoxic for endothelial cells and LDL oxidation seems to be mediated by Fe (Jeney et al., 2002; Nagy et al., 2010).

Evidence 6d0dfb19b0

mROS scavenger (Mito-TEMPO) and NADPH oxidases inhibitors (apocynin and DPI) block TNF production induced by heme.

Evidence 042edb978f

mROS scavenger (Mito-TEMPO) and NADPH oxidases inhibitors (apocynin and DPI) block heme-induced necroptosis.

Evidence 71beb31d85

Heme injection in mice leads to vascular permeability, leukocyte migration from the intravascular environment to tissues and increase of acute-phase proteins (Lyoumi et al., 1999; Wagener et al., 2001b), hallmarks of acute inflammation.

Evidence 81fefd8986

Heme activates endothelial cells inducing the expression of the adhesion molecules ICAM-1 (intercellular adhesion molecule 1), VCAM-1 (vascular cell adhesion molecule 1), E-selectin, Pselectin, and von Willebrand factor (VWF; Wagener et al., 1997; Belcher et al., 2014) and causes neutrophil migration (GraçaSouza et al., 2002; Porto et al., 2007).

Evidence 8e32f2a860

Importantly, heme b interaction with heme oxygenase (HO; Lad et al., 2003), the enzyme responsible for heme intracellular catabolism, and hemopexin (Hx; Paoli et al., 1999), a plasmatic heme scavenger, is essential for the regulation of free heme availability and Fe recycling (Kovtunovych et al., 2010; Tolosano et al., 2010).

Evidence 42cf7c55fb

Heme amplifies cytokines induced by cell surface receptors (TLR2, TLR4, TLR5), endosome receptors (TLR3, TLR9), and cytosolic receptors (NOD1 and NOD2).

Evidence c8297bb808

During intravascular hemolysis the serum proteins responsible for removing heme get saturated and heme can exert its inflammatory effects.

Evidence 511a096672

The consequences of heme toxicity can be appreciated in hemolytic diseases such as β-thalassemia, sickle-cell disease (SCD), ischemia-reperfusion (IR), and malaria (Katori et al., 2002; Pamplona et al., 2007;Vinchi et al., 2013).

Evidence eb22825170

. Heme can destabilize biological membranes increasing its permeability and the chance of lysis (Schmitt et al., 1993), as it happens with erythrocytes (Chiu and Lubin, 1989).

Evidence cd926ecce7

Alternatively, heme induced formation of radical species relies on the conversion of low-reactive organic hydroperoxides (ROOH) into highly reactive alkoxyl (RO•) and peroxyl (ROO•) radicals (Tappel, 1953, 1955; Van der Zee et al., 1996).

Evidence edd7a64175

In endothelial cells, heme induces TLR4-dependent degranulation of Weibel–Palade bodies and P-selectins and VWF release.

Evidence 839b9742e2

A seminal study demonstrated that the ability of heme to activate neutrophils depend on protein kinase C (PKC) activation and ROS generation, inducing the expression of adhesion molecules and modifying actin cytoskeleton dynamics, necessary features for neutrophils migration (Graça-Souza et al., 2002).

Evidence 1d1bedb6ec

Heme-induced neutrophils activation leads to extracellular traps (NETs) release through a mechanism dependent on ROS generation (Chen et al., 2014).

Evidence e3cc49aef0

In this context, heme inhibits neutrophils apoptosis, increasing their longevity, and possibly enhancing harmful stimuli from these cells (Arruda et al., 2004, 2006).

Evidence 794ab79dee

. Heme activates macrophages inducing the production of TNF, KC (Figueiredo et al., 2007), IL-1β (unpublished), and LTB4 (Monteiro et al., 2011).

Evidence cab881ef54

On the other hand, TNF secretion induced by heme is essential for the activation of the programed necrotic cell death pathway, which is denominated necroptosis (Fortes et al., 2012).

Evidence 2ec6832e25

Moreover, heme amplifies MyD88- (TNF and IL-6) and TRIF-dependent (IP-10) cytokines.

Evidence 71a3ce3866

Highly purified heme free of endotoxin contamination was used, as well as polymyxin B, anti-TLR4/MD2, and lipid A antagonist, all of which inhibited the effects of LPS but did not interfere with the induction of TNF by heme.

Evidence e0abab0ce9

The TLR4 activates two distinct pathways: MyD88 and TRIF. In macrophages, heme induces a biased MyD88 activation and the secretion of the pro-inflammatory cytokines TNF and KC.

Evidence 325827d071

Moreover, human embryonic kidney (HEK) cells transfected with human TLR4 secretes IL-8 upon stimulation with heme (Piazza et al., 2011).

Evidence cafbc832fe

In fact, TLR4 is involved in intracerebral hemorrhage (ICH) induced by heme (Lin et al., 2012).

Evidence 27aa6e93af

Moreover, HO-1 has a protective effect during heme-induced necroptosis.

Evidence 77a82ec4e4

Therefore, it is clear that heme triggers ROS-induced sensitization of macrophages to TNFR-mediated cell death through RIP1 and RIP3 activation to promote necroptosis (Figure 2).

Evidence bb07b03d17

Heme induces Syk phosphorylation in macrophages.

Evidence 713921ad7c

Moreover, heme induces apoptosis in human brain vascular endothelial cells (HBVEC) by STAT3 (signal transducer and activator of transcription 3)-dependent activation of matrix metallopeptidase 3 (MMP3; Liu et al., 2013).

Evidence aa6c77f118

These radicals may initiate further lipid peroxidation forming alkyl radicals that in the presence of O2 form more peroxyl radicals leading to a facile propagation of free radical reactions.

Evidence 6741361833

. In the presence of reactive oxygen species (ROS), Hb is oxidized to methemoglobin (MetHb; Balla et al., 1993), characterized by the change in the oxidative state of the Fe present in the heme molecule from ferrous (Fe+2) to ferric (Fe+3).

Evidence a6509d38a2

Although neutrophils have important functions controlling infection, these cells can promote vascular and tissue injury by generating ROS, secreting proteases, and releasing extracellular chromatin (NETs; reviewed in Mócsai, 2013).

Evidence adf4df1ed4

However, ROS is necessary to induce TNF secretion and MAPK activation.

Evidence 0edc9041ae

Because of its large molecular size, this complex is maintained in the intravascular space, preventing the association of otherwise free Hb with nitric oxide (NO; Reiter et al., 2002) and inhibiting the release of free heme (Melamed-Frank et al., 2001).

Evidence ff7477a312

Stimulation of astrocytes with heme, as a model of hemorrhagic injury, causes cell death with characteristics of programed necrosis including the loss of plasma membrane integrity, reversion by necrostatin-1, a selective inhibitor of RIP1, and by antioxidants (Laird et al., 2008).

Evidence 7e1308411c

Hemoglobin is used as a nutrient by Plasmodium, the protozoan parasite that causes malaria, during its replication stage inside erythrocytes in the course of the infection in mammals (Francis et al., 1997).

Evidence 9d997d347e

Similarly to heme, ferrylHb activates endothelial cells through NFκB activation (Silva et al., 2009).

Evidence 2363dd2ca3

Moreover, ferrylHb is unable to induce cytokine secretion by endothelial cells (Silva et al., 2009), another difference to heme which induces IL-8 production (Natarajan et al., 2007).

Evidence 7e9d9e5b3d

Like MetHb, ferrylHb is unstable and releases free heme to further increase oxLDL formation (Potor et al., 2013).

Evidence a739dbe4b7

Similarly to heme, Hz also induces the production of several inflammatory mediators by macrophages such as cytokines and chemokines, and induces leukocytes migration (reviewed by Olivier et al., 2014).

Evidence bed13a28eb

Atypical hemolytic uremic syndrome (aHUS) is characterized by an over activation of the complement alternative pathway (CAP) due to genetic and acquired abnormalities (Noris and Remuzzi, 2009).

Evidence 8406bd752d

Once inside the cells, heme is catabolized by HO enzymes, generating equimolar amounts of biliverdin, carbon monoxide (CO), and Fe (Tenhunen et al., 1968).

Evidence 1e0df1d9e3

LXR-β protects against lipid overload by activating a lipid exportation program regulated by proteins such as LXR-α and ATP binding cassette transporter A1 (ABCA1), preventing foam cells formation.

Evidence a15354fafe

Hemolysis increases the concentration of Hb which, under oxidative stress, releases free heme.

Evidence 5d2ad9e63c

In vivo, injection of heme and LPS induces a significant increase in the concentrations of TNF and IL-6 when compared to the challenge with LPS alone (Fernandez et al., 2010).

Evidence 50dc1b63c3

Hx inhibits the oxidative property of heme (Eskew et al., 1999) and mediates heme transportation to intracellular compartments through the macrophage receptor CD91 (Hvidberg et al., 2005), a critical step on heme catabolism.

Evidence c0996ece7c

In fact, hemolysis or heme injection in Hx−/− mice cause increased inflammation and severe renal damage compared to wild type (WT) mice (Tolosano et al., 1999; Vinchi et al., 2008).

Evidence f7c8c219c6

TLR4 activation leads to MAPKs and NFκB activation, which are necessary to TNF secretion.

Evidence d6a9e8a0db

In fact, Tlr4-/- or anti-TLR4 treatment suppresses heme-induced neuroinflammation, edema, and neurologic deficit.

Evidence b0c62e824e

Indeed, TLRs and NLRP3 have been associated with atherosclerosis development.

Evidence b028f5e382

After hemolysis, sustained interaction between Hb and ROS can lead to ferrylhemoglobin (ferrylHb) formation, which is characterized by an increase in the Fe oxidative state to Fe+4 (Harel and Kanner, 1988; Patel et al., 1996).

Evidence a02c41a7ae

Hb synergizes with LPS enhancing the production of pro-inflammatory cytokines by macrophages (Yang et al., 2002).

Evidence 55072721ac

WhileMAPK8 increases ROS generation, TNF induces RIP1–RIP3 necrosome which triggers necroptosis.

Evidence 532a56f2e2

Cells deficient on FtH are more susceptible to oxidative damage, while increased amounts of FtH protects cells from death induced by challenges such as Fe, tumor necrosis factor (TNF), heme, heme plus TNF, or oxidized low-density lipoprotein (LDL; Juckett et al., 1995; Pham et al., 2004; Gozzelino et al., 2012).

Evidence 8e95e5f192

Importantly, increased expression of FtH also protects different cell types from the cytotoxic effects of heme, TNF or heme and TNF (Balla et al., 1992; Berberat et al., 2003; Cozzi et al., 2003; Gozzelino et al., 2012).

Evidence dbe9854215

The antioxidant property of FtH blocks TNF-induced JNK activation, reducing cell death (Pham et al., 2004; Kamata et al., 2005; Gozzelino et al., 2012).

Evidence dc50f5637d

Interestingly, a 6-year-old patient with HO-1 deficiency experienced a severe atherosclerotic pathology (Yachie et al., 1999).

Evidence 890137ee47

During the resolution phase of inflammation HO-1 expression in leukocytes reduces adhesion molecules expression and leukocytes migration, thus contributing to wound healing (Wagener et al., 2003a).

Evidence 7c11771873

Moreover, IL-1β and TNF modifies the hypothalamus threshold of the body temperature causing fever.

Evidence 95ecd1a4ee

Although KC and IL-1β functions were not investigated during heme-induced inflammatory effects, TNF and LTB4 were described as essential inflammatory mediators during inflammatory events induced by heme.

Evidence a817c40ae5

The MyD88-dependent pathway leads to the activation of mitogen-activated protein kinases (MAPKs) and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) transcription factors inducing the expression of inflammatory cytokines such as TNF, IL-6, IL-1β, and KC (Takeuchi and Akira, 2010)

Evidence cf750bb70c

. Interestingly, NLRP3 knockdown and mROS inhibitors reduce brain edema and improve neurological functions (Ma et al., 2014).

Evidence fac6eaa740

The VWF is prothrombotic and can increase the adhesion of erythrocytes to the endothelium.

Evidence 55500014f3

Mice lacking HO-1 (Hmox1−/−) are highly susceptible to pathologic conditions associated with increased serum heme concentration.

Evidence df6bc0c14d

For instance, Hmox−/− mice develops acute renal failure and marked mortality when submitted to rhabdomyolysis, a pathological condition that increases serum myoglobin which can be oxidized and release heme (Nath et al., 2000).

Evidence 8a130e6db7

Furthermore, Hmox1−/− mice are susceptible to liver IR which is characterized by tissue damage in sites that are reperfused after ischemia injury and hemolysis (Devey et al., 2009).

Evidence c2a9696c59

Depending on the extension of the vaso-occlusion, some tissues may experience hypoxia and damage.

Evidence 83bf50cd4e

Vaso-occlusion of the lung microvasculature may result in the development of the ACS through the infarction of the lung parenchyma.

Evidence a7d79d8bf4

SCD and β-thalassemia are genetic diseases associated to erythrocytes that are prone to lysis due to defective Hb production (Heinle and Read, 1948; Pauling et al., 1949; Ingram, 1957; discussed later).

Evidence 8ba8d986de

Hemolysis can happen due to ischemia/reperfusion, SCD or β-thalassemia.

About

BEL Commons is developed and maintained in an academic capacity by Charles Tapley Hoyt and Daniel Domingo-Fernández at the Fraunhofer SCAI Department of Bioinformatics with support from the IMI project, AETIONOMY. It is built on top of PyBEL, an open source project. Please feel free to contact us here to give us feedback or report any issues. Also, see our Publishing Notes and Data Protection information.

If you find BEL Commons useful in your work, please consider citing: Hoyt, C. T., Domingo-Fernández, D., & Hofmann-Apitius, M. (2018). BEL Commons: an environment for exploration and analysis of networks encoded in Biological Expression Language. Database, 2018(3), 1–11.