Journal Information
Vol. 38. Issue. 1.January - February 2018
Pages 1-108
Visits
16817
Vol. 38. Issue. 1.January - February 2018
Pages 1-108
Review
Open Access
Adverse effects of the renal accumulation of haem proteins. Novel therapeutic approaches
Efectos adversos de la acumulación renal de hemoproteínas. Nuevas herramientas terapéuticas
Visits
16817
Melania Guerrero-Huea,, Alfonso Rubio-Navarroa,, Ángel Sevillanob,c, Claudia Yusteb,c, Eduardo Gutiérrezb,c, Alejandra Palomino-Antolína, Elena Románd, Manuel Pragab,c, Jesús Egidoa, Juan Antonio Morenoa,
Corresponding author
jamoreno@fjd.es

Corresponding author.
a Laboratorio de Nefrología Experimental, Patología Vascular y Diabetes, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
b Red de Investigación Renal (REDINREN), Madrid, Spain
c Departamento de Nefrología, Hospital 12 de Octubre, Madrid, Spain
d Departamento de Nefrología Pediátrica, Hospital La Fe, Valencia, Spain
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (3)
Show moreShow less
Tables (1)
Table 1. Clinical trials in diseases associated with the renal accumulation of haemoproteins.
Abstract

Haemoglobin and myoglobin are haem proteins that play a key role as they help transport oxygen around the body. However, because of their chemical structure, these molecules can exert harmful effects when they are released massively into the bloodstream, as reported in certain pathological conditions associated with rhabdomyolysis or intravascular haemolysis. Once in the plasma, these haem proteins can be filtered and can accumulate in the kidney, where they become cytotoxic, particularly for the tubular epithelium, inducing acute kidney failure and chronic kidney disease. In this review, we will analyse the different pathological contexts that lead to the renal accumulation of these haem proteins, their relation to both acute and chronic loss of renal function, the pathophysiological mechanisms that cause adverse effects and the defence systems that counteract such actions. Finally, we will describe the different treatments currently used and present new therapeutic options based on the identification of new cellular and molecular targets, with particular emphasis on the numerous clinical trials that are currently ongoing.

Keywords:
Haemoglobin
Myoglobin
Haemoglobinuria
Rhabdomyolysis
Haematuria
Acute kidney failure
Chronic kidney disease
Resumen

La hemoglobina y la mioglobina son hemoproteínas que juegan un papel fundamental en el organismo ya que participan en el transporte de oxígeno. Sin embargo, debido a su estructura química, estas moléculas pueden ejercer efectos deletéreos cuando se liberan al torrente sanguíneo de forma masiva, como sucede en determinadas condiciones patológicas asociadas a rabdomiólisis o hemólisis intravascular. Una vez en el plasma, estas hemoproteínas se pueden filtrar y acumular en el riñón, donde resultan citotóxicas, principalmente para el epitelio tubular, e inducen fracaso renal agudo y enfermedad renal crónica. En la presente revisión analizaremos los distintos contextos patológicos que provocan la acumulación renal de estas hemoproteínas, su relación con la pérdida de función renal a corto y largo plazo, los mecanismos fisiopatólogicos responsables de sus efectos adversos y los sistemas de defensa que contrarrestan tales acciones. Por último, describiremos los distintos tratamientos utilizados actualmente y mostraremos nuevas opciones terapéuticas basadas en la identificación de nuevas dianas celulares y moleculares, prestando especial atención a los diversos ensayos clínicos que se encuentran en marcha en la actualidad.

Palabras clave:
Hemoglobina
Mioglobina
Hemoglobinuria
Rabdomiólisis
Hematuria
Fracaso renal agudo
Enfermedad renal crónica
Full Text
Introduction

Haemoglobin (Hb) and myoglobin (Mb) are haemoproteins that play a crucial role in the body's homeostasis, by oxygenating tissues and participating in the regulation of blood pH levels. Hb has a molecular weight of 64.5kDa and is composed of four polypeptide chains known as globins.1 Each globin contains a haem group with an iron atom in its interior, which is responsible for its functional properties. Mb is a smaller protein with a molecular weight of 17kDa, which is formed by a single globin. In physiological conditions, both Hb and Mb are found inside erythrocytes and muscle cells, respectively. However, in certain pathological conditions, these molecules are released into the blood stream and may enter and accumulate in the kidneys, where they are cytotoxic, especially for the proximal tubule epithelium. In fact, the renal accumulation of haemoproteins may induce acute kidney injury (AKI) and chronic kidney disease (CKD). In recent years, new mechanisms have been identified which are involved in kidney damage linked to these molecules, which have helped to develop experimental treatments that have already yielded positive results in recently published studies or in ongoing clinical trials, as described in more detail below.

Origin of the renal accumulation of haemoproteins

Mb builds up in the kidneys as a result of severe muscular damage (rhabdomyolysis), whereas Hb accumulates due to the intravascular haemolysis of red blood cells or the rupture of red blood cells that cross the glomerular membrane in diseases with glomerular haematuria, such as IgA nephropathy (IgAN), lupus or Alport syndrome. In this review, we will focus on myoglobinuria and haemoglobinuria due to space limitations.

Myoglobinuria

Myoglobinuria is the presence of Mb in urine, for which the main cause is rhabdomyolysis or the rupture of skeletal muscle.2 Rhabdomyolysis may be caused by severe trauma, situations of prolonged ischaemia, metabolic disorders, intense physical activity, alcohol abuse and some toxic compounds of chemical or biological origin3 (Fig. 1). The incidence of rhabdomyolysis is not entirely clear, but it has been estimated that it could affect 7–10% of patients presenting with an AKI.3,4

Fig. 1.

Main causes of haemoglobinuria and rhabdomyolysis.

(0.28MB).
Haemoglobinuria

Haemoglobinuria is the presence of Hb in urine as a result of intravascular haemolysis. This causes a renal overload of Hb, especially when there is recurring exposure to free Hb.5 Some of the main aetiological causes of haemoglobinuria include hereditary conditions, such as paroxysmal nocturnal haemoglobinuria, thrombotic thrombocytopenic purpura, haemolytic-uraemic syndrome (HUS), sickle-cell anaemia (SCA), cell membrane defects (elliptocytosis, spherocytosis, etc.), enzymatic defects (glucose-6-phosphate dehydrogenase deficiency, pyruvate kinase deficiency), severe haemolytic anaemia caused by massive transfusion reactions, as well as other causes acquired from HUS and thrombotic microangiopathies of various origins6 (Fig. 1).

Haemoproteins and acute kidney injury

AKI is a common complication in patients with haemoglobinuria or rhabdomyolysis, especially if they were already suffering from kidney disease. Up to 50% of patients suffering from rhabdomyolysis develop AKI, depending on what is the causes.7,8 Therefore, rhabdomyolysis is one of the main causes of AKI (5–25%) and results in death in 2–46% of cases in the absence of dialysis.3,4 On many occasions, situations associated with intravascular haemolysis may also induce AKI.9,10

Haemoproteins and chronic kidney disease

The onset of kidney disease in patients with renal accumulation of haemoproteins is well documented. It has been reported that haemoglobinuria is an independent risk factor for the onset and progression of CKD in people suffering with SCA.11 Something similar occurs in paroxysmal nocturnal haemoglobinuria, a disease in which CKD secondary to the onset of renal vein thrombosis and haemoglobinuria is one of its most significant complications, which may affect 64% of patients and cause 18% of deaths.12 In the absence of treatment, the prognosis for atypical HUS (aHUS) is also poor, with a mortality rate during outbreak of 25%, and progression to CKD in over half of the patients the year after the diagnosis.13

Pathophysiological mechanisms involved in haemoproteins induced renal damage

The main effect of haemoproteins on the kidneys is their direct tubule cell toxicity, regardless of what causes their release (haemo- or myoglobinuria) (Fig. 2). Under normal conditions, Hb binds to haptoglobin and forms the Hb–haptoglobin complex in the plasma.14 This complex is too large to be filtered by the glomerulus, and is therefore broken down by the spleen, bone marrow and liver. However, during intravascular haemolysis, the massive release of Hb causes haptoglobin to be consumed. As a result, Hb remains in the plasma for longer periods of time and is more likely to dissociate into dimers, which are more easily filtered by the glomerulus. Unlike Hb, Mb directly crosses the glomerular filtration membrane due to its smaller molecular size.

Fig. 2.

Mechanisms of renal damage caused by haemoproteins.

(0.25MB).

Once in the lumen of the tubule, the haemoproteins can be reabsorbed by the proximal tubules through the megalin/cubilin receptors complex,14 or even break down by releasing the haem group and free iron, which also have deleterious actions such as nitric oxide neutralisation, vasoconstriction and ischaemia.15 The reduced bioavailability of nitric oxide causes the deregulation of factors that control vascular tone, such as endothelin-1, thromboxane A2, tumour necrosis factor and isoprostanes.16,17 Hb and Mb are also powerful vasoconstrictors because they also react with nitric oxide, as described in diseases associated with intravascular haemolysis and rhabdomyolysis.18–20 When present in the lumen of the tubule, both Mb and Hb can precipitate and bind to the Tamm-Horsfall protein and give rise to RBC casts, which cause intratubular obstruction in the distal nephron segments.21 This obstruction is assisted by the acidic pH found in urine, which increases the stability of the links between the haemoproteins and the Tamm-Horsfall protein.22,23

Inside the tubule cells, the haemoproteins dissociate by releasing globins and the haem group, which induces oxidative stress, cell death and the production of inflammatory cytokines and fibrosis, as discussed in more detail below.

Oxidative stress

Haemoproteins present various redox forms and are an endogenous source of reactive oxygen species.24 When haemoproteins are captured by tubule cells, the haem group is oxidised from Fe2+ to Fe3+ and produces hydroxyl radicals.25 In the presence of peroxides, Fe3+ oxidises to Fe4+ and generates hydroperoxyl radicals, which are highly reactive and contribute to the formation of new reactive oxygen species in the kidneys.26,27 All of these radicals promote the lipid peroxidation of plasma membranes and generate malondialdehyde, which intervenes in the oxidation of proteins and genetic material.4,28,29 This process leads to the production of isoprostanes, proinflammatory cytokines and the expression of adhesion molecules, which increases inflammatory response.30

Inflammation

The haem group acts as a TLR-4 agonist and induces inflammatory response by activating the transcription factor NF-kB.31,32 After binding to the pattern recognition receptor, Hb promotes the activation of several signal transduction pathways such as c-Jun N-terminal, p38 and MAP kinases.33 Another involved pathway is mediated by the activation of the NLRP3 (nitrogen permease regulator-like 3) inflammasome, which is responsible for releasing different cytokines and chemokines involved in the monocyte/macrophage recruitment.34 The presence of proinflammatory macrophages (M1) has been reported in early phases in experimental models of AKI due to the accumulation of haemoproteins, which differ from anti-inflammatory macrophages (M2) in later phases.35,36 These M2 macrophages are found in renal biopsies of patients suffering from rhabdomyolysis, favism, paroxysmal nocturnal haemoglobinuria and outbreaks of macroscopic haematuria associated with IgAN.37–39

Cell death

There have been reports of several types of cell death in the epithelial tubule of patients and in experimental models associated with the accumulation of haemoproteins.4,39–43 Necrosis and apoptosis are the types of death that have been studied at a greater depth.34,44–46 The molecular mechanisms causing death by apoptosis are associated with mitochondrial dysfunction and an increase in pro-apoptotic proteins (BAX and BAD), as well as the activation of caspase-3, the main effector caspase,34,47 and endoplasmic reticulum stress proteins.48 Other types of cell death have been described in these diseases, such as pyroptosis (cell death mediated by caspase-1 which leads to DNA fragmentation and cell lysis) and ferroptosis (iron-dependent cell death). Caspase-1 activation has been observed in experimental rat models of rhabdomyolysis,34 whereas the use of ferroptosis inhibitors in these rats reduced cell death of proximal tubules.49 Lastly, the accumulation of haemoproteins and their derivatives may induce autophagy as a defence mechanism.46,50,51

Fibrosis

Renal fibrosis is another mechanism involved in renal damage caused by haemoproteins. In fact, patients with SCA present with renal fibrosis and increased TGF-β in urine, which is one of the main profibrotic mediators.52 Even though fibroblasts and tubule cells play a very important role in the production of extracellular matrix proteins, recent studies show that macrophages may increase profibrotic response due to the production of mediators such as CTGF and TGF-β during rhabdomyolysis.35,36

Renal tubules are considered as the main sites of Hb toxicity. However, the presence of proteinuria has been reported in experimental models of recurring exposure to haemoproteins.53 There have also been reports of the presence of focal segmental glomerulosclerosis in experimental models of SCA54 and in patients with chronic and recurring haemolysis, such as paroxysmal nocturnal haemoglobinuria, HUS and SCA.55 These patients develop proteinuria56 and suffer from a chronic reduction of glomerular filtration.12,57 These data suggest that there is a link between intravascular haemolysis and glomerular dysfunction. The physiopathological mechanisms, however, are not clear. There are indications that the haemodynamic changes linked to this disease may be responsible for proteinuria and progressive renal damage; however, there is no definitive proof for this theory.58 Given that focal segmental glomerulosclerosis entails a loss of podocytes, these cells may also suffer from haemoprotein-mediated injury. In this sense, unpublished data from our group show that podocytes are capable of capturing Hb, which induces oxidative stress and causes these cells to die, as well as a loss of proteins involved in the glomerular filtration process such as synaptopodin and nephrin.

Defence mechanisms against the renal toxicity of haemoproteins

There are two types of defence mechanisms that work against the harmful effects of haemoproteins: direct and indirect. The direct mechanisms promote the catabolism of haemoproteins and by-products, whereas the indirect mechanisms reduce oxidative stress resulting from the presence of these molecules, thus eliminating the reactive oxygen species or repairing the possible damage caused (Fig. 3). Below is an analysis of each of these defence mechanisms relating to renal damage caused by haemoproteins.

Fig. 3.

Main defence mechanisms and adverse effects of the renal accumulation of haemoproteins.

(0.11MB).
Direct mechanismsHaptoglobin

Haptoglobin (Hp) is a glycoprotein found in high concentrations in plasma (0.3–3g/l) and is mainly secreted by hepatocytes, although it is also synthesised in other tissues such as kidneys. Hp irreversibly binds to Hb and impedes its filtration in the kidneys59 and its translocation to the endothelium,60 which counteracts its harmful effects.61 Hp can also bind to Mb, but with less affinity that it does to Hb.62 The Hb–Hp binding promotes the interaction and subsequent internalisation of this complex through the CD163 receptor of the membrane, which is present in monocytes and macrophages.63 Hp levels are highly reduced in patients with chronic haemolysis, such as SCA,64 because Hp breaks down after being endocited.65 The importance of this protein for Hp has been reported in gene knockout studies in rats, which are more sensitive to damage from haemolysis.59 Studies in animal models of SCA and rhabdomyolysis have shown that the administration of Hp reduces vaso-occlusion,31 oxidative stress66 and renal damage.60,67,68

CD163

CD163 is a receptor found on the surface of circulating monocytes and macrophages, whose main function is Hb clearance in tissue.63 CD163 has a high affinity for Hb–Hp complexes, although it can also bind to free Hb.69 The macrophages that express CD163 have reduced hydrogen peroxide release and important anti-inflammatory functions through the production of IL-10 and HO-1 stimulation.70 Our group has observed an increase in the macrophages expressing CD163 in renal biopsies of patients with massive haemolysis, such as paroxysmal nocturnal haemoglobinuria37 and favism.39 The renal expression of CD163 was higher in areas where iron had accumulated and where oxidative stress markers were found. We have recently written about the presence of CD163 in the kidneys of patients and experimental models of rhabdomyolysis.35 Since the anti-inflammatory and antioxidant functions of CD163 are well known, these data suggest that CD163 could play a nephroprotective role in response to the renal accumulation of haemoproteins.

Haem oxygenase

Haem oxygenase (HO) is one of the main defence mechanisms in situations of renal overload of Mb and Hb. HO is the enzyme responsible for breaking down the haem group, and thus releasing biliverdin, Fe2+ and carbon monoxide,71 which are powerful anti-inflammatory and antioxidant molecules that enhance the beneficial effects of HO.72,73 There are three isoforms of HO (HO-1, HO-2 and HO-3) which differ in their tissue distribution, regulation and function. Unlike other isoforms, the expression of HO-1 is induced in conditions of oxidative stress, and is expressed in many tissues, including the kidneys.74 The renal expression of HO-1 is increased in experimental models of haemoglobinuria and rhabdomyolysis, as well as in patients with intravascular haemolysis.66,75,76 The deficiency of this enzyme in patients with intravascular haemolysis increases tubular and glomerular damage.77 Similarly, animals used in gene knockout studies for HO-1 have shown greater sensitivity to rhabdomyolysis, higher levels of creatinine and higher mortality rates.78

Transferrin

Transferrin (Tf) is a glycoprotein that is mainly secreted by the liver, and which binds to free iron to mitigate its adverse effects.79 Depending on the iron concentration, the tubule cells express the Tf receptor (TfR1), which plays an essential role in the metabolism of this molecule in the kidneys.80 Its expression is regulated by the iron-regulatory proteins 1 and 2, which are highly expressed in the proximal tubules and which act as sensors of iron levels.81–83 Under normal conditions, approximately 30% of the Tf iron-binding sites are saturated. However, these levels increase in the presence of iron accumulation disorders,84 such as severe haemochromatosis, in which case Tf saturation exceeds 60%.85,86 In addition, patients or experimental models of hypotransferrinaemia have low levels of Tf, which promotes renal overload of iron.87,88

Haemopexin

Haemopexin (Hx) is a plasma protein that complexes to the haem group for its subsequent internalisation and hepatic clearance through its binding to the LDL receptor-related protein-1 (LRP1) receptor.89–91 In haemolysis, the Hb oxidises and releases the haem group into the bloodstream to later bind to the serum albumin, which transfers the haem group to the Hx and releases the complex in the liver. Once in hepatocytes, the Hx–haem complex breaks down in lysosomes, although a small amount of Hx is recycled and returned to the bloodstream. Therefore, in patients with haemolytic events, the plasma concentrations of serum Hx are reduced92–95 as it builds up in the renal cortex and increases its levels in urine.96 Hx plays a protective role against the harmful effects of the haem group.31,32,97 Rats used in gene knockout studies for Hx have a poor recovery of renal function after suffering an intravascular haemolysis event, because they have a greater renal accumulation of iron and, therefore, higher oxidative stress levels.98,99

Ferritin

Ferritin is a protein consisting of 24 subunits, forming a hollow spherical structure.100 Its primary function is to store iron, so it has a protective capacity against the toxicity caused by iron and haemoproteins. After the HO-1-catalysed reaction, the iron is released from the haem group and is stored inside the ferritin.101 The expression of ferritin is regulated by the concentration of iron and the HO-1 activity.102 This protein plays a crucial role in diseases related to haemoproteins, as ferritin-deficient rats show notable renal damage,103 and it is a good serum marker for SCA.104 Plasma levels of Tf are also higher in ferritin-deficient rats that are subjected to rhabdomyolysis.103

Nrf2

Nrf2 is a transcription factor that controls the expression of several antioxidant genes such as HO-1 and ferritin.105,106 Under normal conditions, Nrf2 is found in the cytoplasm bound to its repressor Keap1, which is susceptible to changes in the redox state and is subjected to proteolytic degradation through the proteasome. In the presence of oxidative stress, Nrf2 is released from Keap1 and translocates to the nucleus, where it activates the expression of antioxidant genes.107–112 The activation of Nrf2 has a positive effect against renal damage linked to the accumulation of haemoproteins in experimental models and patients with haemolytic anaemia.113–115

Indirect mechanisms

This second group is composed of antioxidant molecules and various antioxidant enzymes.

Non-enzymatic mechanisms

There are many molecules in the body that have an antioxidant action, such as vitamins, melatonin and bilirubin. These molecules neutralise free radicals and are involved in the protection against renal damage caused by haemoproteins.

Vitamins are an important antioxidant group. One such example is vitamin C, which reacts with the superoxide anion and lipid peroxides, thus reducing the oxidative stress induced by the in vitro116 and in vivo117 RBC lysis. Similarly, treatment with vitamin C was effective in a context of AKI caused by haemoglobinaemia in a patient with glucose-6-phosphate dehydrogenase deficiency.118 The levels of vitamin C decrease after the development of rhabdomyolysis, and its administration has partially reduced histological disorders and renal function in experimental models of rhabdomyolysis.119 Vitamin E is another important vitamin because it plays a significant role in maintaining the redox balance and the integrity of cell membranes, acting on peroxyl and hydroperoxyl radicals. The administration of vitamin E inhibited the RBC lysis of patients suffering from paroxysmal nocturnal haemoglobinuria, which suggests that this vitamin is an effective treatment for these patients.120,121 Vitamin E has not been as effective as vitamin C, however, in the treatment of rhabdomyolysis.122

Melatonin is a hormone secreted by the pineal gland which has several antioxidant properties, as it neutralises free radicals such as hydrogen peroxide, the hydroxyl radical, peroxynitrite and the superoxide anion. This molecule also stimulates the expression of other antioxidant molecules, such as superoxide dismutase, glutathione peroxidase and glutathione reductase. Several studies have shown that this hormone plays a protective role in models of AKI caused by rhabdomyolysis or intravascular haemolysis by reducing tubular necrosis and lipid peroxidation associated with these conditions.117,123

Glutathione, in its reduced state (GSH), is a powerful cell antioxidant that can be oxidised to glutathione disulfide (GSSG) through several enzymatic reactions. Several experimental models of myoglobinuria and haemoglobinuria, as well as studies in patients with HUS and SCA, show decreased levels of GSH in the kidneys.117,123–128 The depletion of GSH increases toxicity mediated by oxidative stress in these diseases, because, by restoring the GSH levels, treatment with N-acetylcysteine reduces histological disorders and inhibits cell death associated with these conditions.129,130

Enzymatic mechanisms

This group includes molecules with enzymatic activity that reduce the content of intracellular reactive oxygen species and, therefore, protect cells from oxidative damage. Superoxide dismutase (SOD) is able to dismutate O2− in O2 and H2O2. For the elimination of H2O2, there are other enzymes with peroxidase activity. These include catalase, glutathione peroxidase (GPx) and reduced thioredoxin (Trx). Catalase is an oxidoreductase that catalyses the decomposition reaction of H2O2 in O2 and water. GPx catalyses the decomposition of H2O2 through the oxidation of GSH to GSSG and water. The GPx, catalase and SOD activity is reduced in experimental models of intravascular haemolysis117 and rhabdomyolysis.125,128,131–134 Furthermore, the plasma levels of GPx and SOD show a negative correlation with albuminuria in patients with SCA.135 Lastly, Trx protects from renal damage associated with rhabdomyolysis through the reduction of oxidative stress and inflammation.136

Treatments

There is currently no specific treatment for preventing the damage induced by ferroportins in their different forms of clinical presentation. The alkalisation of urine may be beneficial by reducing the dissociation of the iron found in haemoproteins. This alkalisation can be carried out with oral bicarbonate, while monitoring the urine and serum pH levels. However, no clear benefits have been reliably proven. The use of calcium channel blockers in experimental models has shown an increase in the urinary excretion of iron by mechanisms that remain unknown, which results in a decrease in the renal accumulation of iron.137

The use of iron-chelating agents in diseases associated with the accumulation of this molecule reduces oxidative damage138 and also avoids iron deposition.139 Prophylactically administered deferoxamine reduces oxidative stress resulting from the presence of Hb.140 Iron-chelating agents reduce the toxicity produced by the massive deposition of iron in multitransfused patients,141 although recent studies question their nephroprotective capacity in haemoglobin-induced AKI.142 It should be noted that certain iron-chelating agents, such as deferasirox143 and deferoxamine,144 are potential nephrotoxic agents, requiring strict monitoring during their use.145

Prophylactic treatment with antioxidants such as N-acetylcysteine has yielded positive results in preventing tubular damage secondary to myoglobinuria or haemoglobinuria.146 Other antioxidants, such as acetaminophen, were also effective.27,147 The possible protective role of vitamin E148 and vitamin C,27 in addition to polyphenols,149 flavonoids128,150–152 and l-carnitine,153 has also been investigated, yielding disparate results, as mentioned earlier. Recent studies have also addressed the use of stem cells and have produced positive results, especially in models of rhabdomyolysis.154

Clinical trials

Ongoing clinical trials for the treatment of disorders caused by haemoproteins are focused on two aspects. First, treating the underlying disease to prevent the release of haemoproteins into the plasma, and second, mitigating the damage potentially caused by haemoproteins once they are released. As such, in diseases such as aHUS and paroxysmal nocturnal haemoglobinuria, the majority of trials test drugs that act on the complement system, mainly eculizumab, or even new molecules that act in other ways. These include: CCX168 (C5aR antagonist); conversin (protein that prevents action on its C5 convertase); TT30 (ALXN1102 and ALXN1103; recombinant proteins containing Factor H domains 1–5 and which reduce the complement's convertase activity and activate Factor I); LFG316 (anti-C5 monoclonal antibody); APL-2 (C3 inhibitor) and ALN-CC5 (hepatic inhibitor of C5 synthesis) (Table 1). In aHUS, antibodies are also being developed that work against MASP-2, known as OMS 723. In SCA there are trials involving drugs such as SCD 101 and ICA-17043 (which stop red blood cells from turning into falciform cells); decitabine, vorinostat and panobinostat (to increase foetal haemoglobin); and statins, sodium nitrate and ambrisentan (type A-selective endothelin receptor antagonist) to improve endothelial dysfunction, maintain good tissue perfusion during crises and avoid the rupture of red blood cells. SCD 101 has also been used in beta thalassaemia.

Table 1.

Clinical trials in diseases associated with the renal accumulation of haemoproteins.

  Disease  Mode of action  Trial treatment  Clinical trial number 
Treatment of underlying diseaseHUSInhibition of complementEculizumab  NCT00838513 
CCX168  NCT02464891 
Antibody against MASP-2  OMS 721  NCT02222545 
Paroxysmal nocturnal haemoglobinuriaInhibition of complementConversin  NCT02591862 
TT30  NCT01335165 
LFG316  NCT02534909 
APL2  NCT02588833 
ALN-CC5  NCT02352493 
Sickle-cell anaemiaCovalent modifiers of haemoglobinSCD-101  NCT02380079 
ICA-17043  NCT00294541 
Increase in foetal haemoglobin productionDecitabine  NCT01375608 
Vorinostat  NCT01000155 
Panobinostat  NCT01245179 
Improvement of endothelium functionSimvastatin  NCT00508027 
Sodium nitrate  NCT00095472 
Ambrisentan  NCT02712346 
Prevention of damage caused by haemoproteinsRhabdomyolysisElimination of myoglobin through renal replacement therapiesContinuous therapies  NCT00391911 
High cut-off HicoRhabdo filters  NCT01467180 
Immunoabsorption (CytoSorb®)  NCT02111018 
Decreased oxidation  N-acetylcysteine  NCT00391911 
Malaria  Decreased oxidation  Paracetamol  NCT01641289 
Beta thalassaemiaIron-chelating agentsDeferasirox  NCT00560820 
Exjade-desferal  NCT00901199 

Once haemoproteins are released into the plasma, the trials focus on two strategies. The first strategy is to try to clear these molecules from the plasma. This has been done in several trials in patients suffering from rhabdomyolysis who require renal replacement therapy, in which the effects of continuous techniques, high cut-off haemofilters and immunoabsorption techniques (CytoSorb®) have been analysed in order to remove Mb from plasma as quickly as possible. The second strategy is based on reducing their toxic effect. To do this, efforts are being made to prevent haemoglobinuria-induced oxidation in malaria using paracetamol, and myoglobinuria-induced oxidation using N-acetylcysteine. Trials are also under way that use iron-chelating agents (deferasirox or a combination of exjade and desferal) in thalassaemia to prevent iron deposition in target organs.

Conclusion

The accumulation of haemoproteins in the kidneys is nephrotoxic. There is evidence showing the short-term adverse effects and the chronic loss of renal function. Even though several adverse effects have been reported about these molecules, we must continue to determine the pathogenic mechanisms of haemoproteins to identify new therapeutic targets and prevent their adverse effects. In this sense, podocytes may constitute new cellular targets of the harmful effects of haemoproteins. From a therapeutic perspective, the data currently available are primarily based on studies in animal models. Therapeutic measures aimed at reducing myoglobinuria or haemoglobinuria will be hugely important to prevent renal damage caused by these molecules.

Funding

This study was funded using grants from: the Spanish Society of Nephrology (Sociedad Española de Nefrología), the Spanish Health Research Fund (Fondo de Investigaciones Sanitarias, FIS) (ISCIII/FEDER) (Miguel Servet Programme: CP10/00479 and CPII16/00017; PI13/00802 and PI14/00883) and Fundación Renal Íñigo Álvarez de Toledo (FRIAT) (grants given to JAM); Fundación Conchita Rábago (grant given to MGH); REDinREN (RD012/0021), FIS PI13/02502 and ICI14/00350 (grants given to MP); and FIS/FEDER PI14/00386 and Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas asociadas, CIBERDEM) (grants given to JE).

Conflicts of interest

The authors declare that they have no conflicts of interest.

References
[1]
M.F. Perutz, M.G. Rossmann, A.F. Cullis, H. Muirhead, G. Will, A.C. North.
Structure of haemoglobin: a three-dimensional Fourier synthesis at 5.5-Å resolution, obtained by X-ray analysis.
Nature [Internet], 185 (2016), pp. 416-422
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18990801
[2]
Y. Hamel, A. Mamoune, F.-X. Mauvais, F. Habarou, L. Lallement, N.B. Romero, et al.
Acute rhabdomyolysis and inflammation.
J Inherit Metab Dis [Internet], 38 (2015), pp. 621-628
Available in: http://link.springer.com/10.1007/s10545-015-9827-7
[3]
W.H. Bagley, H. Yang, K.H. Shah.
Rhabdomyolysis.
Intern Emerg Med [Internet], 2 (2007), pp. 210-218
Available in: http://link.springer.com/10.1007/s11739-007-0060-8
[4]
N. Panizo, A. Rubio-Navarro, J.M. Amaro-Villalobos, J. Egido, J.A. Moreno.
Molecular mechanisms and novel therapeutic approaches to rhabdomyolysis-induced acute kidney injury.
Kidney Blood Press Res [Internet], 40 (2015), pp. 520-532
Available in: http://www.karger.com/?doi=10.1159/000368528
[5]
J.A. Moreno, C. Martin-Cleary, E. Gutierrez, O. Toldos, L.M. Blanco-Colio, M. Praga, et al.
AKI associated with macroscopic glomerular hematuria: clinical and pathophysiologic consequences.
Clin J Am Soc Nephrol [Internet], 7 (2012), pp. 175-184
Available in: http://cjasn.asnjournals.org/cgi/doi/10.2215/CJN.01970211
[6]
R.P. Rother, L. Bell, P. Hillmen, M.T. Gladwin.
The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin.
JAMA [Internet], 293 (2005), pp. 1653
Available in: http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.293.13.1653
[7]
G. Melli, V. Chaudhry, D.R. Cornblath.
Rhabdomyolysis: an evaluation of 475 hospitalized patients.
Medicine (Baltimore) [Internet], 84 (2005), pp. 377-385
Available in: http://www.ncbi.nlm.nih.gov/pubmed/16267412
[8]
S.G. Holt, K.P. Moore.
Pathogenesis and treatment of renal dysfunction in rhabdomyolysis.
Intensive Care Med [Internet], 27 (2001), pp. 803-811
Available in: http://www.ncbi.nlm.nih.gov/pubmed/11430535
[9]
J.M. Campistol, M. Arias, G. Ariceta, M. Blasco, L. Espinosa, M. Espinosa, et al.
An update for atypical haemolytic uraemic syndrome: diagnosis and treatment. A consensus document.
Nefrologia [Internet], 35 (2015), pp. 421-447
Available in: http://www.ncbi.nlm.nih.gov/pubmed/26456110
[10]
T.T. Trang, N.H. Phu, H. Vinh, T.T. Hien, B.M. Cuong, T.T. Chau, et al.
Acute renal failure in patients with severe falciparum malaria.
Clin Infect Dis [Internet], 15 (1992), pp. 874-880
Available in: http://www.ncbi.nlm.nih.gov/pubmed/1445988
[11]
S.L. Saraf, X. Zhang, T. Kanias, J.P. Lash, R.E. Molokie, B. Oza, et al.
Haemoglobinuria is associated with chronic kidney disease and its progression in patients with sickle cell anaemia.
Br J Haematol [Internet], 164 (2014), pp. 729-739
Available in: http://doi.wiley.com/10.1111/bjh.12690
[12]
P. Hillmen, M. Elebute, R. Kelly, A. Urbano-Ispizua, A. Hill, R.P. Rother, et al.
Long-term effect of the complement inhibitor eculizumab on kidney function in patients with paroxysmal nocturnal hemoglobinuria.
Am J Hematol [Internet], 85 (2010), pp. 553-559
Available in: http://doi.wiley.com/10.1002/ajh.21757
[13]
B.S. Kaplan, K.E. Meyers, S.L. Schulman.
The pathogenesis and treatment of hemolytic uremic syndrome.
J Am Soc Nephrol [Internet], 9 (1998), pp. 1126-1133
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9621299
[14]
M.J. Tracz, J. Alam, K.A. Nath.
Physiology and pathophysiology of heme: implications for kidney disease.
J Am Soc Nephrol [Internet], 18 (2007), pp. 414-420
Available in: http://www.jasn.org/cgi/doi/10.1681/ASN.2006080894
[15]
S.N. Heyman, M. Brezis.
Acute renal failure in glomerular bleeding: a puzzling phenomenon.
Nephrol Dial Transplant [Internet], 10 (1995), pp. 591-593
Available in: http://www.ncbi.nlm.nih.gov/pubmed/7566565
[16]
B.J. Reeder, D.A. Svistunenko, C.E. Cooper, M.T. Wilson.
The radical and redox chemistry of myoglobin and hemoglobin: from in vitro studies to human pathology.
Antioxid Redox Signal [Internet], 6 (2004), pp. 954-966
Available in: http://www.liebertonline.com/doi/abs/10.1089/ars.2004.6.954
[17]
J.V. Bonventre, J.M. Weinberg.
Recent advances in the pathophysiology of ischemic acute renal failure.
J Am Soc Nephrol [Internet], 14 (2003), pp. 2199-2210
Available in: http://www.ncbi.nlm.nih.gov/pubmed/12874476
[18]
C.D. Reiter, X. Wang, J.E. Tanus-Santos, N. Hogg, R.O. Cannon, A.N. Schechter, et al.
Cell-free hemoglobin limits nitric oxide bioavailability in sickle-cell disease.
Nat Med [Internet], 8 (2002), pp. 1383-1389
Available in: http://www.nature.com/doifinder/10.1038/nm799
[19]
J. Villagra, S. Shiva, L.A. Hunter, R.F. Machado, M.T. Gladwin, G.J. Kato.
Platelet activation in patients with sickle disease, hemolysis-associated pulmonary hypertension, and nitric oxide scavenging by cell-free hemoglobin.
Blood [Internet], 110 (2007), pp. 2166-2172
Available in: http://www.bloodjournal.org/cgi/doi/10.1182/blood-2006-12-061697
[20]
C. Donadee, N.J.H. Raat, T. Kanias, J. Tejero, J.S. Lee, E.E. Kelley, et al.
Nitric oxide scavenging by red blood cell microparticles and cell-free hemoglobin as a mechanism for the red cell storage lesion.
Circulation [Internet], 124 (2011), pp. 465-476
Available in: http://circ.ahajournals.org/cgi/doi/10.1161/CIRCULATIONAHA.110.008698
[21]
M.A. Khalighi, K.J. Henriksen, A. Chang, S.M. Meehan.
Intratubular hemoglobin casts in hemolysis-associated acute kidney injury.
Am J Kidney Dis [Internet], 65 (2015), pp. 337-341
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0272638614011901
[22]
R.A. Zager.
Rhabdomyolysis and myohemoglobinuric acute renal failure.
Kidney Int [Internet], 49 (1996), pp. 314-326
Available in: http://www.ncbi.nlm.nih.gov/pubmed/8821813
[23]
A. Krouzecký, M. Mat¿jovic, R. Rokyta, I. Novák.
[Rhabdomyolysis – development, causes, sequelae and therapy] [article in Czech].
Vnitr Lek [Internet], 49 (2003), pp. 668-672
Available in: http://www.ncbi.nlm.nih.gov/pubmed/14518093
[24]
P.W. Buehler, A.I. Alayash.
Redox biology of blood revisited: the role of red blood cells in maintaining circulatory reductive capacity.
Antioxid Redox Signal [Internet], 7 (2005), pp. 1755-1760
Available in: http://www.liebertonline.com/doi/abs/10.1089/ars.2005.7.1755
[25]
C.P. Baron, H.J. Andersen.
Myoglobin-induced lipid oxidation. A review.
J Agric Food Chem [Internet], 50 (2002), pp. 3887-3897
Available in: http://pubs.acs.org/doi/abs/10.1021/jf011394w
[26]
J.M. Gutteridge.
Iron promoters of the Fenton reaction and lipid peroxidation can be released from haemoglobin by peroxides.
FEBS Lett [Internet], 201 (1986), pp. 291-295
Available in: http://www.ncbi.nlm.nih.gov/pubmed/2423372
[27]
O. Boutaud, L.J. Roberts.
Mechanism-based therapeutic approaches to rhabdomyolysis-induced renal failure.
Free Radic Biol Med [Internet], 51 (2011), pp. 1062-1067
Available in: http://www.ncbi.nlm.nih.gov/pubmed/21034813
[28]
Y. Jia, P.W. Buehler, R.A. Boykins, R.M. Venable, A.I. Alayash.
Structural basis of peroxide-mediated changes in human hemoglobin: a novel oxidative pathway.
J Biol Chem [Internet], 282 (2007), pp. 4894-4907
Available in: http://www.jbc.org/cgi/doi/10.1074/jbc.M609955200
[29]
P.W. Buehler, F. D’Agnillo, D.J. Schaer.
Hemoglobin-based oxygen carriers: from mechanisms of toxicity and clearance to rational drug design.
Trends Mol Med [Internet], 16 (2010), pp. 447-457
Available in: http://linkinghub.elsevier.com/retrieve/pii/S1471491410001097
[30]
K.P. Moore, S.G. Holt, R.P. Patel, D.A. Svistunenko, W. Zackert, D. Goodier, et al.
A causative role for redox cycling of myoglobin and its inhibition by alkalinization in the pathogenesis and treatment of rhabdomyolysis-induced renal failure.
J Biol Chem [Internet], 273 (1998), pp. 31731-31737
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9822635
[31]
J.D. Belcher, C. Chen, J. Nguyen, L. Milbauer, F. Abdulla, A.I. Alayash, et al.
Heme triggers TLR4 signaling leading to endothelial cell activation and vaso-occlusion in murine sickle cell disease.
Blood [Internet], 123 (2014), pp. 377-390
Available in: http://www.ncbi.nlm.nih.gov/pubmed/24277079
[32]
T. Lin, F. Sammy, H. Yang, S. Thundivalappil, J. Hellman, K.J. Tracey, et al.
Identification of hemopexin as an anti-inflammatory factor that inhibits synergy of hemoglobin with HMGB1 in sterile and infectious inflammation.
J Immunol [Internet], 189 (2012), pp. 2017-2022
Available in: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.1103623
[33]
R. Medzhitov.
Origin and physiological roles of inflammation.
Nature [Internet], 454 (2008), pp. 428-435
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18650913
[34]
T. Komada, F. Usui, A. Kawashima, H. Kimura, T. Karasawa, Y. Inoue, et al.
Role of NLRP3 inflammasomes for rhabdomyolysis-induced acute kidney injury.
Sci Rep [Internet], 5 (2015), pp. 10901
Available in: http://www.nature.com/articles/srep10901
[35]
A. Rubio-Navarro, M. Carril, D. Padro, M. Guerrero-Hue, C. Tarín, R. Samaniego, et al.
CD163-macrophages are involved in rhabdomyolysis-induced kidney injury and may be detected by MRI with targeted gold-coated iron oxide nanoparticles.
Theranostics [Internet], 6 (2016), pp. 896-914
Available in: http://www.thno.org/v06p0896.htm
[36]
J. Belliere, A. Casemayou, L. Ducasse, A. Zakaroff-Girard, F. Martins, J.S. Iacovoni, et al.
Specific macrophage subtypes influence the progression of rhabdomyolysis-induced kidney injury.
J Am Soc Nephrol [Internet], 26 (2015), pp. 1363-1377
Available in: http://www.jasn.org/cgi/doi/10.1681/ASN.2014040320
[37]
J. Ballarín, Y. Arce, R. Torra Balcells, M. Diaz Encarnación, F. Manzarbeitia, A. Ortiz, et al.
Acute renal failure associated to paroxysmal nocturnal haemoglobinuria leads to intratubular haemosiderin accumulation and CD163 expression.
Nephrol Dial Transplant [Internet], 26 (2011), pp. 3408-3411
Available in: http://www.ncbi.nlm.nih.gov/pubmed/21771756
[38]
E. Gutiérrez, J. Egido, A. Rubio-Navarro, I. Buendía, L.M. Blanco Colio, O. Toldos, et al.
Oxidative stress macrophage infiltration and CD163 expression are determinants of long-term renal outcome in macrohematuria-induced acute kidney injury of IgA nephropathy.
Nephron Clin Pract [Internet], 121 (2012), pp. c42-c53
Available in: http://www.karger.com/doi/10.1159/000342385
[39]
R.M. García-Camín, M. Goma, R.G. Osuna, A. Rubio-Navarro, I. Buendía, A. Ortiz, et al.
Molecular mediators of favism-induced acute kidney injury.
Clin Nephrol [Internet], 81 (2014), pp. 203-209
Available in: http://www.dustri.com/article_response_page.html?artId=10009&doi=10.5414/CN107520&L=0
[40]
M.N. Bae, D.H. Kwak, S.J. Park, B.S. Choi, C.W. Park, Y.J. Choi, et al.
Acute kidney injury induced by thrombotic microangiopathy in a patient with hemophagocytic lymphohistiocytosis.
BMC Nephrol [Internet], 17 (2016), pp. 4
Available in: http://bmcnephrol.biomedcentral.com/articles/10.1186/s12882-015-0217-z
[41]
C.-W. Tsai, V.-C. Wu, W.-C. Lin, J.-W. Huang, M.-S. Wu.
Acute renal failure in a patient with paroxysmal nocturnal hemoglobinuria.
Kidney Int [Internet], 71 (2007), pp. 1187
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0085253815522709
[42]
S. Hussain, A. Qureshi, J. Kazi.
Renal involvement in paroxysmal nocturnal hemoglobinuria.
Nephron Clin Pract [Internet], 123 (2013), pp. 28-35
Available in: http://www.karger.com?doi=10.1159/000351345
[43]
W.C. Lye, S.O. Leong, E.J. Lee.
Rhabdomyolysis with acute tubular necrosis – a nonglomerular cause of dysmorphic hematuria.
N Engl J Med [Internet], 327 (1992), pp. 570-571
Available in: http://www.ncbi.nlm.nih.gov/pubmed/1635582
[44]
J.H. Baek, F. D’Agnillo, F. Vallelian, C.P. Pereira, M.C. Williams, Y. Jia, et al.
Hemoglobin-driven pathophysiology is an in vivo consequence of the red blood cell storage lesion that can be attenuated in guinea pigs by haptoglobin therapy.
J Clin Invest [Internet], 122 (2012), pp. 1444-1458
Available in: http://www.jci.org/articles/view/59770
[45]
L. Gonzalez-Michaca.
Heme: a determinant of life and death in renal tubular epithelial cells.
AJP Ren Physiol [Internet], 286 (2004), pp. 370F-377
Available in: http://ajprenal.physiology.org/cgi/doi/10.1152/ajprenal.00300.2003
[46]
M. Fahling, S. Mathia, A. Paliege, R. Koesters, R. Mrowka, H. Peters, et al.
Tubular von Hippel-Lindau knockout protects against rhabdomyolysis-induced AKI.
J Am Soc Nephrol [Internet], 24 (2013), pp. 1806-1819
Available in: http://www.jasn.org/cgi/doi/10.1681/ASN.2013030281
[47]
J.H. Kim, S.S. Lee, M.H. Jung, H.D. Yeo, H.J. Kim, J.I. Yang, et al.
N-acetylcysteine attenuates glycerol-induced acute kidney injury by regulating MAPKs and Bcl-2 family proteins.
Nephrol Dial Transplant [Internet], 25 (2010), pp. 1435-1443
Available in: http://ndt.oxfordjournals.org/cgi/doi/10.1093/ndt/gfp659
[48]
J.W. Deuel, C.A. Schaer, F.S. Boretti, L. Opitz, I. Garcia-Rubio, J.H. Baek, et al.
Hemoglobinuria-related acute kidney injury is driven by intrarenal oxidative reactions triggering a heme toxicity response.
Cell Death Dis [Internet], 7 (2016), pp. e2064
Available in: http://www.nature.com/doifinder/10.1038/cddis.2015.392
[49]
R. Skouta, S.J. Dixon, J. Wang, D.E. Dunn, M. Orman, K. Shimada, et al.
Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models.
J Am Chem Soc [Internet], 136 (2014), pp. 4551-4556
Available in: http://pubs.acs.org/doi/abs/10.1021/ja411006a
[50]
J.A. Funk, R.G. Schnellmann.
Persistent disruption of mitochondrial homeostasis after acute kidney injury.
AJP Ren Physiol [Internet], 302 (2012), pp. F853-F864
Available in: http://ajprenal.physiology.org/cgi/doi/10.1152/ajprenal.00035.2011
[51]
A.N. Higdon, G.A. Benavides, B.K. Chacko, X. Ouyang, M.S. Johnson, A. Landar, et al.
Hemin causes mitochondrial dysfunction in endothelial cells through promoting lipid peroxidation: the protective role of autophagy.
AJP Hear Circ Physiol [Internet], 302 (2012), pp. H1394-H1409
Available in: http://ajpheart.physiology.org/cgi/doi/10.1152/ajpheart.00584.2011
[52]
E.E. Ghobrial, H.A. Abdel-Aziz, A.M. Kaddah, N.A. Mubarak.
Urinary transforming growth factor β-1 as a marker of renal dysfunction in sickle cell disease.
Pediatr Neonatol [Internet], 57 (2016), pp. 174-180
Available in: http://linkinghub.elsevier.com/retrieve/pii/S1875957215001229
[53]
K.A. Nath, A.J. Croatt, J.J. Haggard, J.P. Grande.
Renal response to repetitive exposure to heme proteins: chronic injury induced by an acute insult.
Kidney Int [Internet], 57 (2000), pp. 2423-2433
Available in: http://www.ncbi.nlm.nih.gov/pubmed/10844611
[54]
M.E. De Paepe, M. Trudel.
The transgenic SAD mouse: a model of human sickle cell glomerulopathy.
Kidney Int [Internet], 46 (1994), pp. 1337-1345
Available in: http://www.ncbi.nlm.nih.gov/pubmed/7853792
[55]
M.G. Caletti, G. Gallo, C.A. Gianantonio.
Development of focal segmental sclerosis and hyalinosis in hemolytic uremic syndrome.
Pediatr Nephrol [Internet], 10 (1996), pp. 687-692
Available in: http://www.ncbi.nlm.nih.gov/pubmed/8971879
[56]
M. Noris, C. Mele, G. Remuzzi.
Podocyte dysfunction in atypical haemolytic uraemic syndrome.
Nat Rev Nephrol [Internet], 11 (2015), pp. 245-252
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25599621
[57]
R.P. Naik, V.K. Derebail, M.E. Grams, N. Franceschini, P.L. Auer, G.M. Peloso, et al.
Association of sickle cell trait with chronic kidney disease and albuminuria in African Americans.
JAMA [Internet], 312 (2014), pp. 2115-2125
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25393378
[58]
F. Schmitt, F. Martinez, G. Brillet, I. Giatras, G. Choukroun, R. Girot, et al.
Early glomerular dysfunction in patients with sickle cell anemia.
Am J Kidney Dis [Internet], 32 (1998), pp. 208-214
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9708603
[59]
S.K. Lim, H. Kim, S.K. Lim, A. bin Ali, Y.K. Lim, Y. Wang, et al.
Increased susceptibility in Hp knockout mice during acute hemolysis.
Blood [Internet], 92 (1998), pp. 1870-1877
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9731043
[60]
M. Lipiski, J.W. Deuel, J.H. Baek, W.R. Engelsberger, P.W. Buehler, D.J. Schaer.
Human Hp 1-1 and Hp 2-2 phenotype-specific haptoglobin therapeutics are both effective in vitro and in guinea pigs to attenuate hemoglobin toxicity.
Antioxid Redox Signal [Internet], 19 (2013), pp. 1619-1633
Available in: http://www.ncbi.nlm.nih.gov/pubmed/23418677
[61]
A.R. Andersen, S.G. Hasselbalch, O.B. Paulson, N.A. Lassen, K. Kristensen, R.D. Neirinckx.
Evaluation of a 99Tcm bound brain scanning agent for single photon emission computed tomography.
Acta Radiol Suppl [Internet], 369 (1986), pp. 456-459
Available in: http://www.ncbi.nlm.nih.gov/pubmed/2980525
[62]
S. Sakata, N. Yoshioka, M.Z. Atassi.
Human haptoglobin binds to human myoglobin.
Biochim Biophys Acta [Internet], 873 (1986), pp. 312-315
Available in: http://www.ncbi.nlm.nih.gov/pubmed/3756181
[63]
M. Kristiansen, J.H. Graversen, C. Jacobsen, O. Sonne, H.J. Hoffman, S.K. Law, et al.
Identification of the haemoglobin scavenger receptor.
Nature [Internet], 409 (2001), pp. 198-201
Available in: http://www.ncbi.nlm.nih.gov/pubmed/11196644
[64]
U. Muller-Eberhard, J. Javid, H.H. Liem, A. Hanstein, M. Hanna.
Plasma concentrations of hemopexin, haptoglobin and heme in patients with various hemolytic diseases.
Blood [Internet], 32 (1968), pp. 811-815
Available in: http://www.ncbi.nlm.nih.gov/pubmed/5687939
[65]
M.J. Nielsen, H.J. Møller, S.K. Moestrup.
Hemoglobin and heme scavenger receptors.
Antioxid Redox Signal [Internet], 12 (2010), pp. 261-273
Available in: http://www.ncbi.nlm.nih.gov/pubmed/19659436
[66]
N.R. Chintagari, J. Nguyen, J.D. Belcher, G.M. Vercellotti, A.I. Alayash.
Haptoglobin attenuates hemoglobin-induced heme oxygenase-1 in renal proximal tubule cells and kidneys of a mouse model of sickle cell disease.
Blood Cells Mol Dis [Internet], 54 (2015), pp. 302-306
Available in: http://linkinghub.elsevier.com/retrieve/pii/S1079979614001569
[67]
F.S. Boretti, P.W. Buehler, F. D’Agnillo, K. Kluge, T. Glaus, O.I. Butt, et al.
Sequestration of extracellular hemoglobin within a haptoglobin complex decreases its hypertensive and oxidative effects in dogs and guinea pigs.
J Clin Invest [Internet], 119 (2009), pp. 2271-2280
Available in: http://www.ncbi.nlm.nih.gov/pubmed/19620788
[68]
N.R. Chintagari, J. Nguyen, J.D. Belcher, G.M. Vercellotti, A.I. Alayash.
Haptoglobin attenuates hemoglobin-induced heme oxygenase-1 in renal proximal tubule cells and kidneys of a mouse model of sickle cell disease.
Blood Cells Mol Dis [Internet], 54 (2015), pp. 302-306
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25582460
[69]
D.J. Schaer, C.A. Schaer, P.W. Buehler, R.A. Boykins, G. Schoedon, A.I. Alayash, et al.
CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin.
Blood [Internet], 107 (2006), pp. 373-380
Available in: http://www.ncbi.nlm.nih.gov/pubmed/16189277
[70]
J.J. Boyle.
Heme and haemoglobin direct macrophage Mhem phenotype and counter foam cell formation in areas of intraplaque haemorrhage.
Curr Opin Lipidol [Internet], 23 (2012), pp. 453-461
Available in: http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00041433-201210000-00008
[71]
T.S. Banerjee, A.K. Giri.
Effects of sorbic acid and sorbic acid-nitrite in vivo on bone marrow chromosomes of mice.
Toxicol Lett [Internet], 31 (1986), pp. 101-106
Available in: http://www.ncbi.nlm.nih.gov/pubmed/3715920
[72]
T. Jansen, A. Daiber.
Direct antioxidant properties of bilirubin and biliverdin is there a role for biliverdin reductase?.
Front Pharmacol [Internet], 3 (2012),
Available in: http://journal.frontiersin.org/article/10.3389/fphar.2012.00030/Abstract
[73]
J.D. Beckman, J.D. Belcher, J.V. Vineyard, C. Chen, J. Nguyen, M.O. Nwaneri, et al.
Inhaled carbon monoxide reduces leukocytosis in a murine model of sickle cell disease.
AJP Hear Circ Physiol [Internet], 297 (2009), pp. H1243-H1253
Available in: http://ajpheart.physiology.org/cgi/doi/10.1152/ajpheart.00327.2009
[74]
K.A. Nath.
Heme oxygenase-1 and acute kidney injury.
Curr Opin Nephrol Hypertens [Internet], 23 (2014), pp. 17-24
Available in: http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00041552-201401000-00004
[75]
L.D. Zorova, I.B. Pevzner, A.A. Chupyrkina, S.D. Zorov, D.N. Silachev, E.Y. Plotnikov, et al.
The role of myoglobin degradation in nephrotoxicity after rhabdomyolysis.
Chem Biol Interact [Internet], 256 (2016), pp. 64-70
Available in: http://www.ncbi.nlm.nih.gov/pubmed/27329933
[76]
R. Boddu, T.D. Hull, S. Bolisetty, X. Hu, M.S. Moehle, J.P.L. Daher, et al.
Leucine-rich repeat kinase 2 deficiency is protective in rhabdomyolysis-induced kidney injury.
Hum Mol Genet [Internet], 24 (2015), pp. 4078-4093
Available in: https://academic.oup.com/hmg/article-lookup/doi/10.1093/hmg/ddv147
[77]
K. Ohta, A. Yachie, K. Fujimoto, H. Kaneda, T. Wada, T. Toma, et al.
Tubular injury as a cardinal pathologic feature in human heme oxygenase-1 deficiency.
Am J Kidney Dis [Internet], 35 (2000), pp. 863-870
Available in: http://www.ncbi.nlm.nih.gov/pubmed/10793020
[78]
K.A. Nath, J.J. Haggard, A.J. Croatt, J.P. Grande, K.D. Poss, J. Alam.
The indispensability of heme oxygenase-1 in protecting against acute heme protein-induced toxicity in vivo.
Am J Pathol [Internet], 156 (2000), pp. 1527-1535
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0002944010650249
[79]
P. Aisen.
Transferrin, the transferrin receptor, and the uptake of iron by cells.
Met Ions Biol Syst [Internet], 35 (1998), pp. 585-631
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9444770
[80]
H. Kawabata, R.E. Fleming, D. Gui, S.Y. Moon, T. Saitoh, J. O’Kelly, et al.
Expression of hepcidin is down-regulated in TfR2 mutant mice manifesting a phenotype of hereditary hemochromatosis.
Blood [Internet], 105 (2005), pp. 376-381
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15345587
[81]
Z. Gdaniec, H. Sierzputowska-Gracz, E.C. Theil.
Iron regulatory element and internal loop/bulge structure for ferritin mRNA studied by cobalt(III) hexammine binding, molecular modeling, and NMR spectroscopy.
Biochemistry [Internet], 37 (1998), pp. 1505-1512
Available in: http://www.ncbi.nlm.nih.gov/pubmed/9484220
[82]
K.J. Addess, J.P. Basilion, R.D. Klausner, T.A. Rouault, A. Pardi.
Structure and dynamics of the iron responsive element RNA: implications for binding of the RNA by iron regulatory binding proteins.
J Mol Biol [Internet], 274 (1997), pp. 72-83
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0022283697913772
[83]
E.G. Meyron-Holtz, M.C. Ghosh, K. Iwai, T. LaVaute, X. Brazzolotto, U.V. Berger, et al.
Genetic ablations of iron regulatory proteins 1 and 2 reveal why iron regulatory protein 2 dominates iron homeostasis.
EMBO J [Internet], 23 (2004), pp. 386-395
Available in: http://emboj.embopress.org/cgi/doi/10.1038/sj.emboj.7600041
[84]
M.W. Hentze, M.U. Muckenthaler, B. Galy, C. Camaschella.
Two to tango: regulation of mammalian iron metabolism.
Cell [Internet], 142 (2010), pp. 24-38
Available in: http://www.ncbi.nlm.nih.gov/pubmed/20603012
[85]
T.L. Wright, P. Brissot, W.L. Ma, R.A. Weisiger.
Characterization of non-transferrin-bound iron clearance by rat liver.
J Biol Chem [Internet], 261 (1986), pp. 10909-10914
Available in: http://www.ncbi.nlm.nih.gov/pubmed/3733737
[86]
K. Gkouvatsos, G. Papanikolaou, K. Pantopoulos.
Regulation of iron transport and the role of transferrin.
Biochim Biophys Acta [Internet], 1820 (2012), pp. 188-202
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0304416511002674
[87]
C.C. Trenor, D.R. Campagna, V.M. Sellers, N.C. Andrews, M.D. Fleming.
The molecular defect in hypotransferrinemic mice.
Blood [Internet], 96 (2000), pp. 1113-1118
Available in: http://www.ncbi.nlm.nih.gov/pubmed/10910930
[88]
T.B. Bartnikas.
Known and potential roles of transferrin in iron biology.
Biometals [Internet], 25 (2012), pp. 677-686
Available in: http://www.ncbi.nlm.nih.gov/pubmed/22294463
[89]
P. Boucher, M. Gotthardt, W.-P. Li, R.G. Anderson, J. Herz.
LRP: Role in vascular wall integrity and protection from atherosclerosis.
Science [Internet], 300 (2003), pp. 329-332
Available in: http://www.ncbi.nlm.nih.gov/pubmed/12690199
[90]
V. Hvidberg, M.B. Maniecki, C. Jacobsen, P. Højrup, H.J. Møller, S.K. Moestrup.
Identification of the receptor scavenging hemopexin–heme complexes.
Blood [Internet], 106 (2005), pp. 2572-2579
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15947085
[91]
A.P. Lillis, I. Mikhailenko, D.K. Strickland.
Beyond endocytosis: LRP function in cell migration, proliferation and vascular permeability.
J Thromb Haemost [Internet], 3 (2005), pp. 1884-1893
Available in: http://www.ncbi.nlm.nih.gov/pubmed/16102056
[92]
U. Muller-Eberhard.
Hemopexin.
N Engl J Med [Internet], 283 (1970), pp. 1090-1094
Available in: http://www.ncbi.nlm.nih.gov/pubmed/4921465
[93]
D.J. Schaer, F. Vinchi, G. Ingoglia, E. Tolosano, P.W. Buehler.
Haptoglobin, hemopexin, and related defense pathways-basic science, clinical perspectives, and drug development.
Front Physiol [Internet], 5 (2014), pp. 415
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25389409
[94]
B.T. Adornato, L.J. Kagen, F.A. Garver, W.K. Engel.
Depletion of serum hemopexin in fulminant rhabdomyolysis: evidence for an interaction of hemopexin with myoglobin-derived heme.
Arch Neurol [Internet], 35 (1978), pp. 547-548
Available in: http://www.ncbi.nlm.nih.gov/pubmed/666618
[95]
R. Engler, M.F. Jayle.
[Clinical value of the immunochemical determination of the plasma proteins] [article in French].
Sem Hop [Internet], 52 (1976), pp. 2481-2484
Available in: http://www.ncbi.nlm.nih.gov/pubmed/188189
[96]
R.A. Zager, A.C. Johnson, K. Becker.
Renal cortical hemopexin accumulation in response to acute kidney injury.
Am J Physiol Renal Physiol [Internet], 303 (2012), pp. F1460-F1472
Available in: http://www.ncbi.nlm.nih.gov/pubmed/22993068
[97]
F. Vinchi, L. De Franceschi, A. Ghigo, T. Townes, J. Cimino, L. Silengo, et al.
Hemopexin therapy improves cardiovascular function by preventing heme-induced endothelial toxicity in mouse models of hemolytic diseases.
Circulation [Internet], 127 (2013), pp. 1317-1329
Available in: http://www.ncbi.nlm.nih.gov/pubmed/23446829
[98]
E. Tolosano, E. Hirsch, E. Patrucco, C. Camaschella, R. Navone, L. Silengo, et al.
Defective recovery and severe renal damage after acute hemolysis in hemopexin-deficient mice.
Blood [Internet], 94 (1999), pp. 3906-3914
Available in: http://www.ncbi.nlm.nih.gov/pubmed/10572107
[99]
F. Vinchi, S. Gastaldi, L. Silengo, F. Altruda, E. Tolosano.
Hemopexin prevents endothelial damage and liver congestion in a mouse model of heme overload.
Am J Pathol [Internet], 173 (2008), pp. 289-299
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18556779
[100]
P. Arosio, F. Carmona, R. Gozzelino, F. Maccarinelli, M. Poli.
The importance of eukaryotic ferritins in iron handling and cytoprotection.
Biochem J [Internet], 472 (2015), pp. 1-15
Available in: http://www.ncbi.nlm.nih.gov/pubmed/26518749
[101]
S. Bolisetty, A. Zarjou, T.D. Hull, A.M. Traylor, A. Perianayagam, R. Joseph, et al.
Macrophage and epithelial cell H-ferritin expression regulates renal inflammation.
Kidney Int [Internet], 88 (2015), pp. 95-108
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25874599
[102]
F.C. Fervenza, A.J. Croatt, C.M. Bittar, D.W. Rosenthal, D.J. Lager, N. Leung, et al.
Induction of heme oxygenase-1 and ferritin in the kidney in warm antibody hemolytic anemia.
Am J Kidney Dis [Internet], 52 (2008), pp. 972-977
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18805612
[103]
A. Zarjou, S. Bolisetty, R. Joseph, A. Traylor, E.O. Apostolov, P. Arosio, et al.
Proximal tubule H-ferritin mediates iron trafficking in acute kidney injury.
J Clin Invest [Internet], 123 (2013), pp. 4423-4434
Available in: http://www.ncbi.nlm.nih.gov/pubmed/24018561
[104]
J. Porter, M. Garbowski.
Consequences and management of iron overload in sickle cell disease.
Hematol Am Soc Hematol Educ Prog [Internet], (2013), pp. 447-456
Available in: http://www.ncbi.nlm.nih.gov/pubmed/24319218
[105]
R. Venugopal, A.K. Jaiswal.
Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H: quinone oxidoreductase1 gene.
Proc Natl Acad Sci U S A [Internet], 93 (1996), pp. 14960-14965
Available in: http://www.ncbi.nlm.nih.gov/pubmed/8962164
[106]
A.K. Jaiswal.
Nrf2 signaling in coordinated activation of antioxidant gene expression.
Free Radic Biol Med [Internet], 36 (2004), pp. 1199-1207
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15110384
[107]
T. Nguyen, P.J. Sherratt, H.-C. Huang, C.S. Yang, C.B. Pickett.
Increased protein stability as a mechanism that enhances Nrf2-mediated transcriptional activation of the antioxidant response elemen: degradation of Nrf2 by the 26 S proteasome.
J Biol Chem [Internet], 278 (2003), pp. 4536-4541
Available in: http://www.ncbi.nlm.nih.gov/pubmed/12446695
[108]
D. Stewart, E. Killeen, R. Naquin, S. Alam, J. Alam.
Degradation of transcription factor Nrf2 via the ubiquitin–proteasome pathway and stabilization by cadmium.
J Biol Chem [Internet], 278 (2003), pp. 2396-2402
Available in: http://www.ncbi.nlm.nih.gov/pubmed/12441344
[109]
Y. Katoh, K. Iida, M.-I. Kang, A. Kobayashi, M. Mizukami, K.I. Tong, et al.
Evolutionary conserved N-terminal domain of Nrf2 is essential for the Keap1-mediated degradation of the protein by proteasome.
Arch Biochem Biophys [Internet], 433 (2005), pp. 342-350
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15581590
[110]
M.-I. Kang, A. Kobayashi, N. Wakabayashi, S.-G. Kim, M. Yamamoto.
Scaffolding of Keap1 to the actin cytoskeleton controls the function of Nrf2 as key regulator of cytoprotective phase 2 genes.
Proc Natl Acad Sci U S A [Internet], 101 (2004), pp. 2046-2051
Available in: http://www.ncbi.nlm.nih.gov/pubmed/14764898
[111]
M.-K. Kwak, N. Wakabayashi, J.L. Greenlaw, M. Yamamoto, T.W. Kensler.
Antioxidants enhance mammalian proteasome expression through the Keap1-Nrf2 signaling pathway.
Mol Cell Biol [Internet], 23 (2003), pp. 8786-8794
Available in: http://www.ncbi.nlm.nih.gov/pubmed/14612418
[112]
D.D. Zhang, M. Hannink.
Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress.
Mol Cell Biol [Internet], 23 (2003), pp. 8137-8151
Available in: http://www.ncbi.nlm.nih.gov/pubmed/14585973
[113]
J.F. Doss, J.C. Jonassaint, M.E. Garrett, A.E. Ashley-Koch, M.J. Telen, J.T. Chi.
Phase 1 study of a sulforaphane-containing broccoli sprout homogenate for sickle cell disease.
PLoS One [Internet], 11 (2016),
e0152895. Available in: http://www.ncbi.nlm.nih.gov/pubmed/27071063
[114]
N. Keleku-Lukwete, M. Suzuki, A. Otsuki, K. Tsuchida, S. Katayama, M. Hayashi, et al.
Amelioration of inflammation and tissue damage in sickle cell model mice by Nrf2 activation.
Proc Natl Acad Sci U S A [Internet], 112 (2015), pp. 12169-12174
Available in: http://www.ncbi.nlm.nih.gov/pubmed/26371321
[115]
J.D. Belcher, C. Chen, J. Nguyen, P. Zhang, F. Abdulla, P. Nguyen, et al.
Control of oxidative stress and inflammation in sickle cell disease with the Nrf2 activator dimethyl fumarate.
Antioxid Redox Signal [Internet], (2016),
Available in: http://www.ncbi.nlm.nih.gov/pubmed/26914345
[116]
L.M. Claro, M.S.S. Leonart, S.R. Comar, A.J. do Nascimento.
Effect of vitamins C and E on oxidative processes in human erythrocytes.
Cell Biochem Funct [Internet], 24 (2006), pp. 531-535
Available in: http://doi.wiley.com/10.1002/cbf.1255
[117]
T.O. Ajibade, A.A. Oyagbemi, L.A. Durotoye, T.O. Omóbòwálé, E.R. Asenuga, F.O. Olayemi.
Modulatory effects of melatonin and vitamin C on oxidative stress-mediated haemolytic anaemia and associated cardiovascular dysfunctions in rats.
J Complement Integ Med [Internet], 14 (2017),
Available in: http://www.degruyter.com/view/j/jcim.2017.14.issue-1/jcim-2015-0082/jcim-2015-0082.xml
[118]
D.J. Reeves, L.M. Saum, R. Birhiray.
I.V. ascorbic acid for treatment of apparent rasburicase-induced methemoglobinemia in a patient with acute kidney injury and assumed glucose-6-phosphate dehydrogenase deficiency.
Am J Health Syst Pharm [Internet], 73 (2016), pp. e238-e242
Available in: http://www.ajhp.org/cgi/doi/10.2146/ajhp150591
[119]
S. Ustundag, O. Yalcin, S. Sen, Z. Cukur, S. Ciftci, B. Demirkan.
Experimental myoglobinuric acute renal failure: the effect of vitamin C.
Ren Fail [Internet], 30 (2008), pp. 727-735
Available in: http://www.tandfonline.com/doi/full/10.1080/08860220802212965
[120]
J. Amer, O. Zelig, E. Fibach.
Oxidative status of red blood cells, neutrophils, and platelets in paroxysmal nocturnal hemoglobinuria.
Exp Hematol [Internet], 36 (2008), pp. 369-377
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0301472X07006935
[121]
E.D. Gomperts, S.S. Zail, D. Christensen, J. Metz.
The effect of vitamin E on haemolysis in paroxysmal nocturnal haemoglobinuria: In vitro and in vivo studies.
Scand J Haematol [Internet], 14 (1975), pp. 81-85
Available in: http://www.ncbi.nlm.nih.gov/pubmed/1145115
[122]
H.B. Kim, A. Shanu, S. Wood, S.N. Parry, M. Collet, A. McMahon, et al.
Phenolic antioxidants tert-butyl-bisphenol and vitamin E decrease oxidative stress and enhance vascular function in an animal model of rhabdomyolysis yet do not improve acute renal dysfunction.
Free Radic Res [Internet], 45 (2011), pp. 1000-1012
Available in: http://www.tandfonline.com/doi/full/10.3109/10715762.2011.590137
[123]
F.F. Ferraz, A.G. Kos, P. Janino, E. Homsi.
Effects of melatonin administration to rats with glycerol-induced acute renal failure.
Ren Fail [Internet], 24 (2002), pp. 735-746
Available in: http://www.ncbi.nlm.nih.gov/pubmed/12472196
[124]
S.R. Abul-Ezz, P.D. Walker, S.V. Shah.
Role of glutathione in an animal model of myoglobinuric acute renal failure.
Proc Natl Acad Sci U S A [Internet], 88 (1991), pp. 9833-9837
Available in: http://www.ncbi.nlm.nih.gov/pubmed/1946409
[125]
R. Manikandan, M. Beulaja, R. Thiagarajan, M. Pandi, C. Arulvasu, N.M. Prabhu, et al.
Ameliorative effect of ferulic acid against renal injuries mediated by nuclear factor-kappaB during glycerol-induced nephrotoxicity in Wistar rats.
Ren Fail [Internet], 36 (2014), pp. 154-165
Available in: http://www.tandfonline.com/doi/full/10.3109/0886022X.2013.835223
[126]
S.A. Gomez, M.J. Abrey-Recalde, C.A. Panek, N.F. Ferrarotti, M.G. Repetto, M.P. Mejías, et al.
The oxidative stress induced in vivo by Shiga toxin-2 contributes to the pathogenicity of haemolytic uraemic syndrome.
Clin Exp Immunol [Internet], 173 (2013), pp. 463-472
Available in: http://doi.wiley.com/10.1111/cei.12124
[127]
T. Dasgupta, M.E. Fabry, D.K. Kaul.
Antisickling property of fetal hemoglobin enhances nitric oxide bioavailability and ameliorates organ oxidative stress in transgenic-knockout sickle mice.
Am J Physiol Regul Integ Comp Physiol [Internet], 298 (2010), pp. R394-R402
Available in: http://ajpregu.physiology.org/cgi/doi/10.1152/ajpregu.00611.2009
[128]
D. Singh, V. Chander, K. Chopra.
Protective effect of naringin, a bioflavonoid on glycerol-induced acute renal failure in rat kidney.
Toxicology [Internet], 201 (2004), pp. 143-151
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0300483X04002628
[129]
F.J. Polo-Romero, A. Fernández-Fúnez, L. Broseta Viana, M.P. Atienza, F. Sánchez Gascón.
Effect of N-acetylcysteine on antioxidant status in glycerol-induced acute renal failure in rats.
Ren Fail [Internet], 26 (2004), pp. 613-618
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15600251
[130]
J.H. Kim, S.S. Lee, M.H. Jung, H.D. Yeo, H.-J. Kim, J.I. Yang, et al.
N-acetylcysteine attenuates glycerol-induced acute kidney injury by regulating MAPKs and Bcl-2 family proteins.
Nephrol Dial Transplant [Internet], 25 (2010), pp. 1435-1443
Available in: https://academic.oup.com/ndt/article-lookup/doi/10.1093/ndt/gfp659
[131]
N. Aydogdu, G. Atmaca, O. Yalcin, K. Batcioglu, K. Kaymak.
Effects of caffeic acid phenethyl ester on glycerol-induced acute renal failure in rats.
Clin Exp Pharmacol Physiol [Internet], 31 (2004), pp. 575-579
Available in: http://doi.wiley.com/10.1111/j. 1440-1681.2004.04050.x
[132]
Y. Shi, L. Xu, J. Tang, L. Fang, S. Ma, X. Ma, et al.
Inhibition of HDAC6 protects against rhabdomyolysis-induced acute kidney injury.
Am J Physiol Renal Physiol [Internet], 312 (2017), pp. F502-F515
Available in: http://ajprenal.physiology.org/lookup/doi/10.1152/ajprenal.00546.2016
[133]
M. Ibrahim, C. Luchese, S. Pinton, S.S. Roman, W. Hassan, C.W. Nogueira, et al.
Involvement of catalase in the protective effect of binaphthyl diselenide against renal damage induced by glycerol.
Exp Toxicol Pathol [Internet], 63 (2011), pp. 331-335
Available in: http://linkinghub.elsevier.com/retrieve/pii/S0940299310000242
[134]
R. Brandão, C.I. Acker, M.R. Leite, N.B. Barbosa, C.W. Nogueira.
Diphenyl diselenide protects against glycerol-induced renal damage in rats.
J Appl Toxicol [Internet], 29 (2009), pp. 612-618
Available in: http://doi.wiley.com/10.1002/jat.1449
[135]
K.E. Itokua, J.R. Makulo, F.B. Lepira, M.N. Aloni, P.M. Ekulu, E.K. Sumaili, et al.
Albuminuria, serum antioxidant enzyme levels and markers of hemolysis and inflammation in steady state children with sickle cell anemia.
BMC Nephrol [Internet], 17 (2016), pp. 178
Available in: http://bmcnephrol.biomedcentral.com/articles/10.1186/s12882-016-0398-0
[136]
K. Nishida, H. Watanabe, S. Ogaki, A. Kodama, R. Tanaka, T. Imafuku, et al.
Renoprotective effect of long acting thioredoxin by modulating oxidative stress and macrophage migration inhibitory factor against rhabdomyolysis-associated acute kidney injury.
Sci Rep [Internet], 5 (2015), pp. 14471
Available in: http://www.nature.com/articles/srep14471
[137]
S. Ludwiczek, I. Theurl, M.U. Muckenthaler, M. Jakab, S.M. Mair, M. Theurl, et al.
Ca2+ channel blockers reverse iron overload by a new mechanism via divalent metal transporter-1.
Nat Med [Internet], 13 (2007), pp. 448-454
Available in: http://www.ncbi.nlm.nih.gov/pubmed/17293870
[138]
L.J. Roberts.
Inhibition of heme protein redox cycling: reduction of ferryl heme by iron chelators and the role of a novel through-protein electron transfer pathway.
Free Radic Biol Med [Internet], 44 (2008), pp. 257-260
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18067870
[139]
N.A. Sheikh, T.R. Desai, P.R. Tirgar.
Investigation into iron chelating and antioxidant potential of Melilotus officinalis in iron dextran induced iron overloaded sprague dawley rat model.
Drug Res (Stuttg) [Internet], (2016),
Available in: http://www.ncbi.nlm.nih.gov/pubmed/27626608
[140]
B.J. Reeder, R.C. Hider, M.T. Wilson.
Iron chelators can protect against oxidative stress through ferryl heme reduction.
Free Radic Biol Med [Internet], 44 (2008), pp. 264-273
Available in: http://www.ncbi.nlm.nih.gov/pubmed/18215735
[141]
T.D. Coates.
Physiology and pathophysiology of iron in hemoglobin-associated diseases.
Free Radic Biol Med [Internet], 72 (2014), pp. 23-40
Available in: http://www.ncbi.nlm.nih.gov/pubmed/24726864
[142]
I.C. Moura, M. Arcos-Fajardo, A. Gdoura, V. Leroy, C. Sadaka, N. Mahlaoui, et al.
Engagement of transferrin receptor by polymeric IgA1: evidence for a positive feedback loop involving increased receptor expression and mesangial cell proliferation in IgA nephropathy.
J Am Soc Nephrol [Internet], 16 (2005), pp. 2667-2676
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15987753
[143]
P.D. Sánchez-González, F.J. López-Hernandez, A.I. Morales, J.F. Macías-Nuñez, J.M. López-Novoa.
Effects of deferasirox on renal function and renal epithelial cell death.
Toxicol Lett [Internet], 203 (2011), pp. 154-161
Available in: http://www.ncbi.nlm.nih.gov/pubmed/21439361
[144]
L.K. Groebler, J. Liu, A. Shanu, R. Codd, P.K. Witting.
Comparing the potential renal protective activity of desferrioxamine B and the novel chelator desferrioxamine B-N-(3-hydroxyadamant-1-yl) carboxamide in a cell model of myoglobinuria.
Biochem J [Internet], 435 (2011), pp. 669-677
Available in: http://www.ncbi.nlm.nih.gov/pubmed/21320071
[145]
A. Piga, S. Fracchia, M.E. Lai, M.D. Cappellini, R. Hirschberg, D. Habr, et al.
Deferasirox effect on renal haemodynamic parameters in patients with transfusion-dependent β thalassaemia.
Br J Haematol [Internet], 168 (2015), pp. 882-890
Available in: http://www.ncbi.nlm.nih.gov/pubmed/25402221
[146]
K. Ware, Z. Qamri, A. Ozcan, A.A. Satoskar, G. Nadasdy, B.H. Rovin, et al.
N-acetylcysteine ameliorates acute kidney injury but not glomerular hemorrhage in an animal model of warfarin-related nephropathy.
Am J Physiol Renal Physiol [Internet], 304 (2013), pp. F1421-F1427
Available in: http://www.ncbi.nlm.nih.gov/pubmed/23576637
[147]
O. Boutaud, K.P. Moore, B.J. Reeder, D. Harry, A.J. Howie, S. Wang, et al.
Acetaminophen inhibits hemoprotein-catalyzed lipid peroxidation and attenuates rhabdomyolysis-induced renal failure.
Proc Natl Acad Sci U S A [Internet], 107 (2010), pp. 2699-2704
Available in: http://www.ncbi.nlm.nih.gov/pubmed/20133658
[148]
A.L. Huerta-Alardín, J. Varon, P.E. Marik.
Bench-to-bedside review: rhabdomyolysis – an overview for clinicians.
Crit Care [Internet], 9 (2005), pp. 158-169
Available in: http://www.ncbi.nlm.nih.gov/pubmed/15774072
[149]
L.K. Groebler, X.S. Wang, H.B. Kim, A. Shanu, F. Hossain, A.C. McMahon, et al.
Cosupplementation with a synthetic, lipid-soluble polyphenol and vitamin C inhibits oxidative damage and improves vascular function yet does not inhibit acute renal injury in an animal model of rhabdomyolysis.
Free Radic Biol Med [Internet], 52 (2012), pp. 1918-1928
Available in: http://www.ncbi.nlm.nih.gov/pubmed/22343418
[150]
R. Rodrigo, C. Bosco, P. Herrera, G. Rivera.
Amelioration of myoglobinuric renal damage in rats by chronic exposure to flavonol-rich red wine.
Nephrol Dial Transplant [Internet], 19 (2004), pp. 2237-2244
Available in: https://academic.oup.com/ndt/article-lookup/doi/10.1093/ndt/gfh369
[151]
V. Chander, D. Singh, K. Chopra.
Reversal of experimental myoglobinuric acute renal failure in rats by quercetin, a bioflavonoid.
Pharmacology [Internet], 73 (2005), pp. 49-56
Available in: http://www.karger.com/?doi=10.1159/000081074
[152]
V. Avramovic, P. Vlahovic, D. Mihailovic, V. Stefanovic.
Protective effect of a bioflavonoid proanthocyanidin-BP1 in glycerol-induced acute renal failure in the rat: renal stereological study.
Ren Fail [Internet], 21 (1999), pp. 627-634
Available in: http://www.ncbi.nlm.nih.gov/pubmed/10586425
[153]
N. Aydogdu, G. Atmaca, O. Yalcin, R. Taskiran, E. Tastekin, K. Kaymak.
Protective effects of L-carnitine on myoglobinuric acute renal failure in rats.
Clin Exp Pharmacol Physiol [Internet], 33 (2006), pp. 119-124
Available in: http://doi.wiley.com/10.1111/j. 1440-1681.2006.04336.x
[154]
S. Bruno, C. Grange, M.C. Deregibus, R.A. Calogero, S. Saviozzi, F. Collino, et al.
Mesenchymal stem cell-derived microvesicles protect against acute tubular injury.
J Am Soc Nephrol [Internet], 20 (2009), pp. 1053-1067
Available in: http://www.ncbi.nlm.nih.gov/pubmed/19389847

Both authors share authorship as first authors.

Please cite this article as: Guerrero-Hue M, Rubio-Navarro A, Sevillano A, Yuste C, Gutiérrez E, Palomino-Antolín A, et al. Efectos adversos de la acumulación renal de hemoproteínas. Nuevas herramientas terapéuticas. Nefrologia. 2018;38:13–26.

Copyright © 2017. Sociedad Española de Nefrología
Idiomas
Nefrología (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?