Acessibilidade / Reportar erro

The role of ClpX in erythropoietic protoporphyria

ABSTRACT

Hemoglobin is an essential biological component of human physiology and its production in red blood cells relies upon proper biosynthesis of heme and globin protein. Disruption in the synthesis of these precursors accounts for a number of human blood disorders found in patients. Mutations in genes encoding heme biosynthesis enzymes are associated with a broad class of metabolic disorders called porphyrias. In particular, one subtype – erythropoietic protoporphyria – is caused by the accumulation of protoporphyrin IX. Erythropoietic protoporphyria patients suffer from photosensitivity and a higher risk of liver failure, which is the principle cause of morbidity and mortality. Approximately 90% of these patients carry loss-of-function mutations in the enzyme ferrochelatase (FECH), while 5% of cases are associated with activating mutations in the C-terminus of ALAS2. Recent work has begun to uncover novel mechanisms of heme regulation that may account for the remaining 5% of cases with previously unknown genetic basis. One erythropoietic protoporphyria family has been identified with inherited mutations in the AAA+ protease ClpXP that regulates ALAS activity. In this review article, recent findings on the role of ClpXP as both an activating unfoldase and degrading protease and its impact on heme synthesis will be discussed. This review will also highlight the role of ClpX dysfunction in erythropoietic protoporphyria.

Keywords:
Heme biosynthesis enzymes; Porphyria; Erythropoietic protoporphyria; ClpXP; ALAS gene

Introduction

Heme is a chemical moiety that is essential for all life.11 Paoli M, Marles-Wright J, Smith A. Structure–function relationships in heme-proteins. DNA Cell Biol. 2002;21(4):271-80.66 Korolnek T, Hamza I. Like iron in the blood of the people: the requirement for heme trafficking in iron metabolism. Front Pharmacol. 2014;5:126. In humans, heme is most commonly known for its role in red blood cells (RBCs) as the oxygen-binding prosthetic group in hemoglobin. Given its importance in RBC physiology, defective heme metabolism is strongly associated with hematologic diseases in humans.

With few exceptions, heme is synthesized via a highly conserved set of eight enzymatic reactions. The first committed step of heme biosynthesis involves the condensation of glycine and succinyl-CoA in the mitochondria to generate δ-aminolevulinic acid (ALA) utilizing the enzyme δ-aminolevulinic acid synthesis (ALAS1). ALA is then transferred into the cytosol where it is converted to tetrapyrrole coporphyrinogen III (CPgenIII) by four enzymes – ALA dehydratase (ALAD), hydroxymethylbilane (HMBS), uroporphyrinogen III synthase (UROS), and uroporphyrinogen decarboxylase (UROD).77 Krishnamurthy PC, Du G, Fukuda Y, Sun D, Sampath J, Mercer KE, et al. Identification of a mammalian mitochondrial porphyrin transporter. Nature. 2006;443(7111):586-9. After entering the mitochondrial inner membrane space, the enzyme coproporphyrinogen oxidase (CPOX) catalyzes the conversion of CPgenIII to protoporphyrinogen IX (PPgenIX).88 Helias V, Saison C, Ballif BA, Peyrard T, Takahashi J, Takahashi H, et al. ABCB6 is dispensable for erythropoiesis and specifies the new blood group system Langereis. Nat Genet. 2012;44(2):170-3. PPgenIX is then transported into the mitochondrial matrix where it is the reactant for protoporphyrinogen oxidase (PPOX), to produce protoporphyrin IX (PPIX). The penultimate enzyme in the cascade, ferrochelatase (FECH), is responsible for the final insertion of iron into PPIX to form heme (Figure 1).99 Oborník M, Green BR. Mosaic origin of the heme biosynthesis pathway in photosynthetic eukaryotes. Mol Biol Evol. 2005;22(12):2343-53.

Figure 1
Highly conserved heme/protoporphyrin IX (PPIX) synthesis in eukaryotic cells. Precursors, glycine and succinyl-CoA, undergo 8 enzymatic reactions with δ-aminolevulinic acid synthesis (ALAS), ALA dehydratase (ALAD), hydroxymethylbilane (HMBS), uroporphyrinogen III synthase (UROS), uroporphyrinogen decarboxylase (UROD), coproporphyrinogen oxidase (CPOX), protoporphyrinogen oxidase (PPIOX), and ferrochelatase (FECH) in both the mitochondria and cytoplasm to form heme.

Figure illustration courtesy of Johannes G. Wittig (Technische-Universität-Dresden, Germany). Reprinted with permission from Yien et al.3737 Yien YY, Ringel AR, Paw BH. Mitochondrial transport of protoporphyrinogen IX in erythroid cells. Oncotarget. 2015;6(25):20742-3. and Oncotarget (Impact Journals, LLC).


Erythroid heme production must meet the vast production of hemoglobin and accounts for roughly 85% of the daily heme synthesis.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. All heme biosynthesis genes are upregulated during RBC differentiation.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. However, RBCs also have unique mechanisms in addition to the conical heme biosynthetic pathway. For example, the first step of heme synthesis, involves an erythroid-specific isoform of ALAS (ALAS2) that becomes transcriptionally induced during RBC maturation. ALAS2 differs from ALAS1 through an iron responsive element (IRE) in ALAS2 that can interact with iron responsive proteins (IRPs) allowing for the association of ALAS2 to iron availability.1010 Chiabrando D, Mercurio S, Tolosano E. Heme and erythropoieis: more than a structural role. Haematologica. 2014;99(6):973-83.

The final enzyme, FECH, is also regulated in a red cell specific manner. FECH is an iron-sulfur cluster protein located on the inner mitochondrial membrane. In RBCs, FECH is post-translationally stabilized through the formation of a complex with mitoferrin1 and ABCB10.1111 Chen W, Dailey HA, Paw BH. Ferrochelatase forms an oligomeric complex with mitoferrin-1 and Abcb10 for erythroid heme biosynthesis. Blood. 2010;116(4):628-30.1313 Seguin A, Takahashi-Makise N, Yien YY, Huston NC, Whitman JC, Musso G, et al. Reductions in the mitochondrial ABC transporter Abcb10 affect the transcriptional profile of heme biosynthesis genes. J Biol Chem. 2017;292(39):16284-99. Recent work has also found that erythropoietin (EPO) signaling together with the GATA-1 transcriptional program regulates the phosphorylation and activation of FECH in RBCs.1010 Chiabrando D, Mercurio S, Tolosano E. Heme and erythropoieis: more than a structural role. Haematologica. 2014;99(6):973-83.1212 Chen W, Paradkar PN, Li L, Pierce EL, Langer NB, Takahashi-Makise N, et al. Abcb10 Physically interacts with mitoferrin-1 (slc25a37) to enhance its stability and function in the erythroid mitochondria. Proc Natl Acad Sci U S A. 2009;106(38):16263-8. Based on the number of regulatory pathways converging on FECH in red cells, it is not surprising that FECH is a rate-limiting enzyme in erythroid heme biosynthesis.1010 Chiabrando D, Mercurio S, Tolosano E. Heme and erythropoieis: more than a structural role. Haematologica. 2014;99(6):973-83.

Porphyrias

The intermediates produced by each enzyme in heme synthesis are cytotoxic and their accumulation can have deleterious effects. Clinically, the accumulation of heme intermediates cause porphyrias – a diverse group of metabolic disorders. Patients with porphyria suffer from a variety of symptoms including cutaneous photosensitivity, behavioral changes, restlessness, insomnia, seizures, abnormal liver function, and several other life threatening concerns.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.,1414 Puy H, Gouya L, Deybach JC. Porphyrias. Lancet. 2010;375(9718):924-37. The complex genotype-phenotype correlations in combination with environmental factors have given rise to a broad range of disease presentation. Mutations in nine genes are currently known to be associated with porphyrias. Of the nine, defects in seven genes are associated with hepatic porphyrias, such as porphyria cutanea tarda (PCT), and characterized by the accumulation of heme precursors in the liver.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. In contrast, defects in the remaining two genes – ALAS2 and FECH – encode enzymes particularly important in RBC biology and give rise to erythropoietic porphyrias with the distinctive feature of accumulating heme precursors in the bone marrow.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.

FECH is the gene most commonly associated with erythropoietic protoporphyria (EPP), accounting for 90% of diagnosed cases.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. These patients have reduced FECH activity.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.ALAS2-associated EPP mutations are activating and result in increased ALA production. Ultimately, both FECH and ALAS2 mutations lead to an accumulation of photoreactive PPIX. In EPP patients, accumulated PPIX reacts with sunlight resulting in the production of cytotoxic reactive oxygen species (ROS).44 Sachar M, Anderson KE, Ma X. Protoporphyrin IX: the good, the bad, and the ugly. J Pharmacol Exp Ther. 2016;356(2):267-75.,1515 Lim HW. Mechanisms of phototoxicity in porphyria cutanea tarda and erythropoietic protoporphyria. Immunol Ser. 1989;46:671-85. For some patients, even a small amount of light exposure can trigger skin lesions, while severe sun exposure can result in edema, erythema, and wax-like scarring. Increased amounts of free PPIX can also cause patients to suffer from more life threating concerns such as gallstones, liver disease, and liver failure.1414 Puy H, Gouya L, Deybach JC. Porphyrias. Lancet. 2010;375(9718):924-37.

Genetic basis of EPP

EPP mutations can be inherited in an autosomal dominant, recessive, or X-linked fashion.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. The genetics of EPP have been extensively studied. EPP cases with FECH mutations most commonly exhibit an autosomal dominant pattern of inheritance in which the hypomorphic IVS3-48C FECH allele is found in all affected family members.1616 Schneider-Yin X, Gouya L, Dorsey M, Rüfenacht U, Deybach JC, Ferreira GC. Mutations in the iron-sulfur cluster ligands of the human ferrochelatase lead to erythropoietic protoporphyria. Blood. 2000;96(4):1545-9.1818 Gouya L, Schneider-Yin X, Rüfenacht U, Herrero C, Lecha M, Mascaro JM, et al. Mutations in the ferrochelatase gene of four Spanish patients with erythropoietic protoporphyria. J Invest Dermatol. 1998;111(3):406-9. When the other FECH allele is mutated, FECH enzymatic activity is substantially reduced to 15–25% of its normal activity level.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.,1717 Whatley SD, Mason NG, Holme SA, Anstey AV, Elder GH, Badminton MN. Gene dosage analysis identifies large deletions of the FECH gene in 10% of families with erythropoietic protoporphyria. J Invest Dermatol. 2007;127(12):2790-4.,1919 Gouya L, Martin-Schmitt C, Robreau AM, Austerlitz F, Da Silva V, Brun P, et al. Contribution of a common single-nucleotide polymorphism to the genetic predisposition for erythropoietic protoporphyria. Am J Hum Genet. 2006;78(1):2-14. Over 60 different FECH mutations including partial deletions, frame shifts, and substitutions, have been identified to be associated with the IVS3-48C allele and found to cause decreased FECH activity.1616 Schneider-Yin X, Gouya L, Dorsey M, Rüfenacht U, Deybach JC, Ferreira GC. Mutations in the iron-sulfur cluster ligands of the human ferrochelatase lead to erythropoietic protoporphyria. Blood. 2000;96(4):1545-9.1818 Gouya L, Schneider-Yin X, Rüfenacht U, Herrero C, Lecha M, Mascaro JM, et al. Mutations in the ferrochelatase gene of four Spanish patients with erythropoietic protoporphyria. J Invest Dermatol. 1998;111(3):406-9. The most common mutations found in this region associated with EPP involve partial deletions. Partial deletions within the FECH allele, varying among patients, have been found to alter the structure, activation regions, or stability of FECH resulting in reduced enzyme acitivity.1717 Whatley SD, Mason NG, Holme SA, Anstey AV, Elder GH, Badminton MN. Gene dosage analysis identifies large deletions of the FECH gene in 10% of families with erythropoietic protoporphyria. J Invest Dermatol. 2007;127(12):2790-4. Patients suffering from frame shift mutations result in either a defective protein or a premature stop codon.1818 Gouya L, Schneider-Yin X, Rüfenacht U, Herrero C, Lecha M, Mascaro JM, et al. Mutations in the ferrochelatase gene of four Spanish patients with erythropoietic protoporphyria. J Invest Dermatol. 1998;111(3):406-9. These changes disrupt the secondary structure of FECH, leading to a decrease in the effectiveness of the enzyme.1818 Gouya L, Schneider-Yin X, Rüfenacht U, Herrero C, Lecha M, Mascaro JM, et al. Mutations in the ferrochelatase gene of four Spanish patients with erythropoietic protoporphyria. J Invest Dermatol. 1998;111(3):406-9. Research preformed by Schneider-Yin et al. also found that FECH substitution mutations also cause EPP through changes of the structural components of the [2Fe-2S] cluster of FECH.1616 Schneider-Yin X, Gouya L, Dorsey M, Rüfenacht U, Deybach JC, Ferreira GC. Mutations in the iron-sulfur cluster ligands of the human ferrochelatase lead to erythropoietic protoporphyria. Blood. 2000;96(4):1545-9. The changes in the [2Fe-2S] cluster, found to play a structural role in FECH, destabilize the FECH enzyme structure resulting in a reduced level of enzymatic activity.1616 Schneider-Yin X, Gouya L, Dorsey M, Rüfenacht U, Deybach JC, Ferreira GC. Mutations in the iron-sulfur cluster ligands of the human ferrochelatase lead to erythropoietic protoporphyria. Blood. 2000;96(4):1545-9. Even though the mechanisms through which FECH activity is disrupted are diverse, they all hinder heme production and result in high PPIX.

Although the majority of EPP cases arising from FECH mutations are inherited in an autosomal dominant fashion, there are also a few FECH-associated EPP cases that present as autosomal recessive.2020 Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6. In one family, two healthy parents, each containing a different mutation to the FECH allele, developed a minor increase in PPIX levels, but no other symptoms.2020 Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6. When their children inherited both mutant alleles, both children developed severe EPP and presented with liver failure and extreme photosensitivity.2020 Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6. In this particular case, the two mutations were a substitution and an insertion that reduced FECH activity to 57% and 64%, respectively.2020 Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6. Although each mutation alone only partially hindered FECH activity, in combination, the mutations led to an extreme depletion of FECH activity, 13–25%.2020 Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6. Even though these double mutations are rare, they can result in severe EPP.

Another 5% of EPP cases result from gain-of-function mutations in ALAS2, which presents as an X-linked disorder.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. These mutations cluster in the inhibitory C-terminal region of ALAS2 and cause either the elongation or deletion of the C-terminus on ALAS2.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.,2121 Whatley SD, Ducamp S, Gouya L, Grandchamp B, Beaumont C, Badminton MN, et al. C-terminal deletions in the ALAS2 gene lead to gain of function and cause X-linked dominant protoporphyria without anemia or iron overload. Am J Hum Genet. 2008;83(3):408-14. Alterations to the C-terminus of ALAS2 triggers increased ALAS activity resulting in abnormally high PPIX levels.2121 Whatley SD, Ducamp S, Gouya L, Grandchamp B, Beaumont C, Badminton MN, et al. C-terminal deletions in the ALAS2 gene lead to gain of function and cause X-linked dominant protoporphyria without anemia or iron overload. Am J Hum Genet. 2008;83(3):408-14.,2222 Ducamp S, Schneider-Yin X, de Rooij F, Clayton J, Fratz EJ, Rudd A, et al. Molecular and functional analysis of the C-terminal region of human erythroid-specific 5-aminolevulinic synthase associated with X-linked dominant protoporphyria (XLDPP). Hum Mol Genet. 2013;22(7):1280-8. However, the remaining 5% of EPP patients do not harbor FECH or ALAS2 mutations, suggesting additional genetic modifiers still exist.

ClpX – a novel regulator of heme production

ClpX is a mitochondrial AAA+ ATPase found in tandem with the mitochondrial tetradecameric peptidase hexamer, ClpP, to form the AAA+ protease ClpXP.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. Through in vitro studies using the model substrate casein, a model for ClpXP function has been developed.2424 Kang SG, Ortega J, Singh SK, Wang N, Huang NN, Steven AC. Functional proteolytic complexes of the human mitochondrial ATP-dependent protease HClpXP. J Biol Chem. 2002;277(23):21095-102.2626 Farmer WH, Yuan ZY. A continuous fluorescent assay for measuring protease activity using natural protein substrate. Anal Biochem. 1991;197(2):347-52. ClpX is involved in substrate binding, unfolding, and translocation to ClpP, which mediates protein degradation (Figure 2).2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. Independent of ClpP, ClpX is still able to unfold proteins without degradation.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. While substrate recognition by ClpX requires only ATP binding, ATP hydrolysis is required for the unfoldase and translocase activities of ClpX.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. Although the biochemical function of ClpXP has been known for some time, physiologic substrates of ClpXP in mammalian cells are largely unknown.

Figure 2
ClpXP protease model of substrate recognition, unfolding, and degradation function. The peptide tag binds to the axial pore of ClpX. Subsequently, ClpX unfolds the substrate, with energy provided by ATP, and translocates the unfolded polypeptide to ClpP, which mediates proteolysis.

Reprinted with permission from Baker et al.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. and Biochim. Biophys. Acta (Elsevier, Inc.).


This has recently changed with a series of elegant studies using both yeast (S. cerevisiae) as well as vertebrate models.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. Yeasts lack ClpP and, thus, they are an excellent system to study ClpX unfoldase function without substrate degradation. Researchers, first, found that the yeast equivalent of ClpX, MCX1, promoted the activity of HEM1, which is the yeast homolog of ALAS1.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. Similarly, Kardon et al. discovered that mammalian ClpX, independent of ClpP, activated ALAS2 in vitro by facilitating PLP insertion. Based on these results, the authors proposed that ClpX activates ALAS2 in mammalian cells. In support, expression of ClpX but not ClpP was induced in maturing murine erythroid cells.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. Introduction of mammalian ClpP led to a decrease in ALAS activity, suggesting that active ALAS2 is a substrate for ClpXP degradation.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. Together these findings led the authors to propose that ClpXP plays a dual role in heme biosynthesis with ClpX as an activator of ALAS function and ClpP as a repressor. To further confirm ClpX as an activator, Kardon et al. showed that ClpX was required for efficient erythroid heme synthesis in zebrafish (Danio rerio) embryos. Morpholino knockdown of the zebrafish homolog, clpxa, in zebrafish embryos (termed morphants) resulted in a substantial decrease in hemoglobinization and the number of erythroid cells in vivo.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29. The anemia found in ClpX morphants was rescued by ALA supplementation that by-passes the block in ALAS activity. These results suggest that the activation of ALAS by ClpX is a conserved pathway and represents an important step in heme synthesis.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29.

Further work by Kubota et al. has shed light into how ClpXP functions in both capacities and when it is important to do so.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29. Using the human HepG2 liver cell line, the authors found that siRNA-mediated knockdown of ClpX led to a robust accumulation of ALAS1. This is consistent with the idea that ClpX is required not only for substrate binding and unfolding, but also shuttling to ClpP for degradation.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29. Interestingly, similar inhibition of ClpP expression with siRNA did not result in ALAS1 accumulation. ALAS1 levels were only increased following prolonged knockdown of ClpP (ten days). This raises the possibility that additional proteases may be responsible for ClpX-dependent turnover of ALAS1. Alternatively, the mechanisms through which ClpXP regulates ALAS1 may differ from ALAS2.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.,2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. It is also important to note that the authors did not measure ALAS1 activity in ClpP-deficient cells.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29. There is evidence to indicate that ClpP inhibits the ATPase activity of ClpX, which is crucial for ClpX unfoldase function.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. Presumably, in this fashion, ClpP would inhibit ALAS1 activity by blocking PLP insertion without affecting steady state ALAS1 expression. Nevertheless, based on these findings, Kubota et al. propose that ClpXP constitutes part of the negative feedback loop in which excess heme triggers ALAS1 degradation.

ClpX is a novel EPP gene

Recently, a family was identified with a history of EPP but no mutations in FECH or ALAS2 that was inherited in an autosomal dominant fashion. Additional analysis found a common heterozygote ClpX missense mutation in all affected family members. This substitution of glycine with aspartic acid at position 298 of ClpX is located in the conserved Walker A sequence motif involved in ATP-binding.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28. This ATP binding region is thought to be absolutely essential for the formation of the ClpXP complex.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. The alteration of an amino acid at the ATP binding region, according to models found by Baker et al. and Kardon et al., can cause an alteration in ClpX-ClpP interaction resulting in a variation in the translocation and degradation of both ALAS1 and ALAS2 producing altered heme levels.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67.

When the G298D ClpX mutant protein was exogenously overexpressed in murine erythroid cells, ALAS activity was increased with concomitantly higher PPIX levels.2828 Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52. Interestingly, in vitro studies showed that the G298D mutation severely compromised ATPase activity of mutant ClpX. When mixed together in vitro with wild-type ClpX, mutant ClpX was also able to inhibit wild-type ATPase activity.2828 Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52. Since ATP hydrolysis is required to form the ClpXP complex, the authors propose that mutant ClpX is unable to shuttle ALAS2 to ClpP for degradation; but retains the ability to unfold ALAS2 to allow PLP insertion (Figure 3). These results are significant for several reasons. First, the ability of mutant ClpX to inhibit wild-type function suggests that it is a dominant negative and provides a mechanistic explanation to the observed pattern of inheritance. Second, high ALAS activity even with diminished ATPase function suggests that ATP hydrolysis is not required for the unfoldase properties of ClpX or PLP insertion. It would be interesting to test whether this aspect of mutant ClpX also holds true with model substrates in vitro or if this is an effect specific for ALAS2. Furthermore, if ClpP is, indeed, a negative regulator, it is tempting to speculate that it too would be a candidate gene mutated in EPP. To date, the phenotype of ClpP knockdown or deletion remains unexplored.2323 Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.,2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29.,2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67.,2828 Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52. The potential role of ClpP in EPP and heme biosynthesis remains an exciting avenue of future research.

Figure 3
A model for how ClpXP regulates ALAS function and heme production. Under normal conditions (left), ClpX unfoldase activity is needed for PLP insertion and activation of ALAS proteins. ClpP mediated degradation constitutes negative feedback regulation in response to high heme levels.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. In EPP patients with the heterozygous G298D ClpX dominant negative mutation, active ALAS2 is unable to be shuttled to ClpP for degradation, resulting in excess production of intermediates (middle).2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67. In contrast, the complete absence of ClpX not only reduces degradation but also fails to activate ALAS. This causes a profound loss of heme production and anemia (right).2626 Farmer WH, Yuan ZY. A continuous fluorescent assay for measuring protease activity using natural protein substrate. Anal Biochem. 1991;197(2):347-52.

Reprinted with permission from and Yien et al. and Proceedings of the National Academy of Sciences (PNAS).


Another remaining issue is whether ALAS2 gain-of-function mutations in EPP affect its interaction with ClpX. This is even more important since it has been proposed that ClpXP is a critical mediator of heme-dependent negative feedback regulation on ALAS1 activity.2525 Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29. Additional studies are necessary to better understand the interplay between ClpX and mutant ALAS2. It is also tempting to speculate that the ClpX EPP mutations have disrupted heme feedback regulation. Nevertheless, ClpX mutations are now being recognized as a third variant of EPP. Whether ClpX mutations account for the entirety of the remaining cases of EPP with no known genetic cause is still unknown. Additional genetic studies are needed to determine the complete prevalence of ClpX mutations in EPP.

Current EPP treatments

Current treatment options for patients are very limited. These options include avoiding sunlight exposure, reducing effect of sunlight exposure with oral β-carotene, administering porphyrin absorbents, such as cholestyramine and activated charcoal, transfusions, and in extreme cases liver or bone marrow transplant.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.,2929 Thapar M, Bonkovsky HL. The diagnosis and management of erythropoietic protoporphyria. Gastroenterol Hepatol. 2008;4(8):561-6. Unfortunately, almost all current treatment options only alleviate the symptoms rather than addressing the pathogenesis of the disease. Only a bone marrow transplant has been found to cure a patient with EPP, but finding an efficient treatment plan and the stage to intervene has still not been fully determined.2929 Thapar M, Bonkovsky HL. The diagnosis and management of erythropoietic protoporphyria. Gastroenterol Hepatol. 2008;4(8):561-6.

A proposed treatment for EPP involves the administration of ALADs to decrease intermediates upstream of PPIX. While this treatment has shown some promise in mice trials, ALADs has been shown to cause anemia if prescribed in excess.3030 Ademuyiwa O, Ugbaja RN, Ojo DA, Owoigbe AO, Adeokun SE. Reversal of aminolevulinic acid dehydratase (ALAD) inhibition and reduction of erythrocyte protoporphyrin levels by Vitamin C in occupational lead exposure in Abeokuta Nigeria. Environ Toxicol Pharmacol. 2005;20(3):404-11. Therefore, this regimen must be closely monitored for adverse side effects.

Developing new treatments for EPP patients

For the majority of EPP patients with defective FECH, research needs to focus on boosting residual FECH activity. Emerging evidence indicates that FECH in RBCs is activated by EPO signaling. EPO receptor signaling triggers activation of protein kinase A (PKA) leading to phosphorylation and activation of FECH.3131 Chung J, Wittig JG, Ghamari A, Maeda M, Dailey TA, Bergonia H, et al. Erythropoietin signaling regulates heme biosynthesis. Elife. 2017:6. Since EPP patients with FECH mutations retain minor FECH activity, increasing the activity of FECH through EPO administration could help lower PPIX levels.22 Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04. However, researchers also found that if a mutation is altering the position of the FECH gene that is required for phosphorylation by PKA there is no increased activity.3131 Chung J, Wittig JG, Ghamari A, Maeda M, Dailey TA, Bergonia H, et al. Erythropoietin signaling regulates heme biosynthesis. Elife. 2017:6. Therefore, this strategy may only be effective if the phosphorylation site is intact.

In addition to the treatment plans discussed earlier, EPP patients, without a FECH mutation, have found beneficial results when treated with iron supplements.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67.,2828 Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52.,3232 Barman-Aksoezen J, Schneider-Yin X, Minder EI. Iron in erythropoietic protoporphyrias: Dr Jekyll or Mr. Hyde? J Rare Dis Res Treat. 2017;2(4):1-5. Iron supplements help clear excess PPIX by driving the metalation reaction catalyzed by FECH.2727 Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67.,2828 Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52.,3232 Barman-Aksoezen J, Schneider-Yin X, Minder EI. Iron in erythropoietic protoporphyrias: Dr Jekyll or Mr. Hyde? J Rare Dis Res Treat. 2017;2(4):1-5. However, long-term effects of continual iron supplementation are still being investigated.3232 Barman-Aksoezen J, Schneider-Yin X, Minder EI. Iron in erythropoietic protoporphyrias: Dr Jekyll or Mr. Hyde? J Rare Dis Res Treat. 2017;2(4):1-5. In addition, treating with iron supplements has only been met with limited success. This is likely due to feedback inhibition of iron uptake via the bone morphogenetic protein (BMP) signaling pathway. Excess circulating iron leads to BMP-mediated stimulation of hepcidin production that, in turn, blocks cellular iron uptake into cells.3333 Babitt JL, Huang FW, Wrighting DM, Xia Y, Sidis Y, Samad TA, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet. 2006;38(5):531-9. Thus, although exogenous iron supplements place more iron in circulation, homeostatic mechanisms limit the extent of iron entering the cell. As a result, BMP inhibitors, such as inhibin and BMP-3, have been a major focus for research.3434 Rosen V. BMP and BMP inhibitors in bone. Ann N Y Acad Sci. 2006;1068:19-25. Current efforts have failed to find a BMP inhibitor that is both potent and has a high enough selectivity to be effective in patients.3535 Hopkins CR. Inhibitors of the Bone Morphogenetic Protein (BMP) signaling pathway: a patent review (2008–2015). Expert Opin Ther Pat. 2016;:1-14.

Another approach is to use small molecules to increase iron uptake. In a recent small molecule screen, hinokitiol, found naturally in the leaves of a Japanese cypress tree, was found to bind and transport iron in and out of the cell.3636 Grillo AS, SantaMaria AM, Kafina MD, Cioffi AG, Huston NC, Han M, et al. Restored iron transport by a small molecule promotes absorption and hemoglobinization in animals. Science. 2017;356(6338):608-16. Hinokitiol has been shown to rescue zebrafish mutants with defects in cellular iron uptake as well as intracellular iron transport.3636 Grillo AS, SantaMaria AM, Kafina MD, Cioffi AG, Huston NC, Han M, et al. Restored iron transport by a small molecule promotes absorption and hemoglobinization in animals. Science. 2017;356(6338):608-16. Although its effectiveness has not yet been tested for treating EPP, this study displays the possibility of using iron carriers in hematologic diseases and paves the way for the discovery of other small molecules.

  • a
    Posthumously (see Appendix A Appendix A. Posthumously Barry biographical note Barry Paw, MD, PhD, was a pediatric oncologist at the Dana-Farber/Boston Children's Cancer and Blood Disorders Center and principal investigator at Brigham and Women's Hospital, Harvard Medical School. Dr. Paw received his undergraduate degree in Biochemistry from University of California, Berkeley and his doctorate from University of California, Los Angeles School of Medicine. He had a highly distinguished career and was a recipient of several awards, including the William Randolph Hearst Young Investigator Award in 2002, the Basil O’Connor Scholar Award from the March of Dimes Birth Defects Foundation in 2004, the Young Investigator President's Award from the International BioIron Society in 2005, and was elected into the American Society for Clinical Investigation in 2008. Dr. Paw was a true pioneer in the field of heme and iron metabolism, combining his expertise of zebrafish genetics and biochemistry to study red cell development. His research has resulted in the identification of several novel genes involved in red cell development and disease. Dr. Paw will be remembered for his contributions to the scientific community. However, even more importantly, he will be forever cherished as a caring mentor, a good friend, and an impeccable person unmatched in his strength of character and love of science. ).

Acknowledgments

We would like to thank members of our lab for insightful discussions. We would like to dedicate this review to Dr. Barry H. Paw, who was a tremendous mentor and role model. This work was supported by the Diamond-Blackfan Anemia Foundation (BHP) and the National Institutes of Health Research (P01 HL032262, BHP and JC).

Appendix A. Posthumously Barry biographical note

Barry Paw, MD, PhD, was a pediatric oncologist at the Dana-Farber/Boston Children's Cancer and Blood Disorders Center and principal investigator at Brigham and Women's Hospital, Harvard Medical School. Dr. Paw received his undergraduate degree in Biochemistry from University of California, Berkeley and his doctorate from University of California, Los Angeles School of Medicine. He had a highly distinguished career and was a recipient of several awards, including the William Randolph Hearst Young Investigator Award in 2002, the Basil O’Connor Scholar Award from the March of Dimes Birth Defects Foundation in 2004, the Young Investigator President's Award from the International BioIron Society in 2005, and was elected into the American Society for Clinical Investigation in 2008. Dr. Paw was a true pioneer in the field of heme and iron metabolism, combining his expertise of zebrafish genetics and biochemistry to study red cell development. His research has resulted in the identification of several novel genes involved in red cell development and disease. Dr. Paw will be remembered for his contributions to the scientific community. However, even more importantly, he will be forever cherished as a caring mentor, a good friend, and an impeccable person unmatched in his strength of character and love of science.

REFERENCES

  • 1
    Paoli M, Marles-Wright J, Smith A. Structure–function relationships in heme-proteins. DNA Cell Biol. 2002;21(4):271-80.
  • 2
    Balwani M, Desnick RJ. The porphyrias: advances in diagnosis and treatment. Blood. 2012;120(23):4496-04.
  • 3
    Li T, Bonkovsky HL, Guo J. Structural analysis of heme proteins: implications for design and prediction. BMC Struct Biol. 2011;11:13.
  • 4
    Sachar M, Anderson KE, Ma X. Protoporphyrin IX: the good, the bad, and the ugly. J Pharmacol Exp Ther. 2016;356(2):267-75.
  • 5
    Hamza I, Dailey HA. One ring to rule them all: trafficking of heme and heme synthesis intermediates in the metazoans. Biochim Biophys Acta. 2012;1823(9):1617-32.
  • 6
    Korolnek T, Hamza I. Like iron in the blood of the people: the requirement for heme trafficking in iron metabolism. Front Pharmacol. 2014;5:126.
  • 7
    Krishnamurthy PC, Du G, Fukuda Y, Sun D, Sampath J, Mercer KE, et al. Identification of a mammalian mitochondrial porphyrin transporter. Nature. 2006;443(7111):586-9.
  • 8
    Helias V, Saison C, Ballif BA, Peyrard T, Takahashi J, Takahashi H, et al. ABCB6 is dispensable for erythropoiesis and specifies the new blood group system Langereis. Nat Genet. 2012;44(2):170-3.
  • 9
    Oborník M, Green BR. Mosaic origin of the heme biosynthesis pathway in photosynthetic eukaryotes. Mol Biol Evol. 2005;22(12):2343-53.
  • 10
    Chiabrando D, Mercurio S, Tolosano E. Heme and erythropoieis: more than a structural role. Haematologica. 2014;99(6):973-83.
  • 11
    Chen W, Dailey HA, Paw BH. Ferrochelatase forms an oligomeric complex with mitoferrin-1 and Abcb10 for erythroid heme biosynthesis. Blood. 2010;116(4):628-30.
  • 12
    Chen W, Paradkar PN, Li L, Pierce EL, Langer NB, Takahashi-Makise N, et al. Abcb10 Physically interacts with mitoferrin-1 (slc25a37) to enhance its stability and function in the erythroid mitochondria. Proc Natl Acad Sci U S A. 2009;106(38):16263-8.
  • 13
    Seguin A, Takahashi-Makise N, Yien YY, Huston NC, Whitman JC, Musso G, et al. Reductions in the mitochondrial ABC transporter Abcb10 affect the transcriptional profile of heme biosynthesis genes. J Biol Chem. 2017;292(39):16284-99.
  • 14
    Puy H, Gouya L, Deybach JC. Porphyrias. Lancet. 2010;375(9718):924-37.
  • 15
    Lim HW. Mechanisms of phototoxicity in porphyria cutanea tarda and erythropoietic protoporphyria. Immunol Ser. 1989;46:671-85.
  • 16
    Schneider-Yin X, Gouya L, Dorsey M, Rüfenacht U, Deybach JC, Ferreira GC. Mutations in the iron-sulfur cluster ligands of the human ferrochelatase lead to erythropoietic protoporphyria. Blood. 2000;96(4):1545-9.
  • 17
    Whatley SD, Mason NG, Holme SA, Anstey AV, Elder GH, Badminton MN. Gene dosage analysis identifies large deletions of the FECH gene in 10% of families with erythropoietic protoporphyria. J Invest Dermatol. 2007;127(12):2790-4.
  • 18
    Gouya L, Schneider-Yin X, Rüfenacht U, Herrero C, Lecha M, Mascaro JM, et al. Mutations in the ferrochelatase gene of four Spanish patients with erythropoietic protoporphyria. J Invest Dermatol. 1998;111(3):406-9.
  • 19
    Gouya L, Martin-Schmitt C, Robreau AM, Austerlitz F, Da Silva V, Brun P, et al. Contribution of a common single-nucleotide polymorphism to the genetic predisposition for erythropoietic protoporphyria. Am J Hum Genet. 2006;78(1):2-14.
  • 20
    Sarkany RP, Alexander GJ, Cox TM. Recessive inheritance of erythropoietic protoporphyria with liver failure. Lancet. 1994;343(8910):1394-6.
  • 21
    Whatley SD, Ducamp S, Gouya L, Grandchamp B, Beaumont C, Badminton MN, et al. C-terminal deletions in the ALAS2 gene lead to gain of function and cause X-linked dominant protoporphyria without anemia or iron overload. Am J Hum Genet. 2008;83(3):408-14.
  • 22
    Ducamp S, Schneider-Yin X, de Rooij F, Clayton J, Fratz EJ, Rudd A, et al. Molecular and functional analysis of the C-terminal region of human erythroid-specific 5-aminolevulinic synthase associated with X-linked dominant protoporphyria (XLDPP). Hum Mol Genet. 2013;22(7):1280-8.
  • 23
    Baker TA, Sauer RT. ClpXP, an ATP-powered unfolding and protein-degradation machine. Biochim Biophys Acta. 2012;1823(1):15-28.
  • 24
    Kang SG, Ortega J, Singh SK, Wang N, Huang NN, Steven AC. Functional proteolytic complexes of the human mitochondrial ATP-dependent protease HClpXP. J Biol Chem. 2002;277(23):21095-102.
  • 25
    Kubota Y, Nomura K, Katoh Y, Yamashita R, Kaneko K, Furuyama K. Novel mechanisms for heme-dependent degradation of ALAS1 protein as a component of negative feedback regulation of heme biosynthesis. J Biol Chem. 2016;291(39):20516-29.
  • 26
    Farmer WH, Yuan ZY. A continuous fluorescent assay for measuring protease activity using natural protein substrate. Anal Biochem. 1991;197(2):347-52.
  • 27
    Kardon JR, Yien YY, Huston NC, Branco DS, Hildick-Smith GJ, Rhee KY, et al. Mitochondrial ClpX activates a key enzyme for heme biosynthesis and erythropoiesis. Cell. 2015;161(4):858-67.
  • 28
    Yien YY, Ducamp S, van der Vorm LN, Kardon JR, Manceau H, Kannengiesser C, et al. Mutation in human CLPX elevates levels of δ-aminolevulinate synthase and protoporphyrin IX to promote erythropoietic protoporphyria. Proc Natl Acad Sci U S A. 2017;114(38):E8045-52.
  • 29
    Thapar M, Bonkovsky HL. The diagnosis and management of erythropoietic protoporphyria. Gastroenterol Hepatol. 2008;4(8):561-6.
  • 30
    Ademuyiwa O, Ugbaja RN, Ojo DA, Owoigbe AO, Adeokun SE. Reversal of aminolevulinic acid dehydratase (ALAD) inhibition and reduction of erythrocyte protoporphyrin levels by Vitamin C in occupational lead exposure in Abeokuta Nigeria. Environ Toxicol Pharmacol. 2005;20(3):404-11.
  • 31
    Chung J, Wittig JG, Ghamari A, Maeda M, Dailey TA, Bergonia H, et al. Erythropoietin signaling regulates heme biosynthesis. Elife. 2017:6.
  • 32
    Barman-Aksoezen J, Schneider-Yin X, Minder EI. Iron in erythropoietic protoporphyrias: Dr Jekyll or Mr. Hyde? J Rare Dis Res Treat. 2017;2(4):1-5.
  • 33
    Babitt JL, Huang FW, Wrighting DM, Xia Y, Sidis Y, Samad TA, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet. 2006;38(5):531-9.
  • 34
    Rosen V. BMP and BMP inhibitors in bone. Ann N Y Acad Sci. 2006;1068:19-25.
  • 35
    Hopkins CR. Inhibitors of the Bone Morphogenetic Protein (BMP) signaling pathway: a patent review (2008–2015). Expert Opin Ther Pat. 2016;:1-14.
  • 36
    Grillo AS, SantaMaria AM, Kafina MD, Cioffi AG, Huston NC, Han M, et al. Restored iron transport by a small molecule promotes absorption and hemoglobinization in animals. Science. 2017;356(6338):608-16.
  • 37
    Yien YY, Ringel AR, Paw BH. Mitochondrial transport of protoporphyrinogen IX in erythroid cells. Oncotarget. 2015;6(25):20742-3.

Publication Dates

  • Publication in this collection
    Apr-Jun 2018

History

  • Received
    28 Feb 2018
  • Accepted
    2 Mar 2018
Associação Brasileira de Hematologia, Hemoterapia e Terapia Celular (ABHH) R. Dr. Diogo de Faria, 775 cj 133, 04037-002, São Paulo / SP - Brasil - São Paulo - SP - Brazil
E-mail: htct@abhh.org.br