Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Liver Int ; 44(8): 1842-1855, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38717058

ABSTRACT

Congenital erythropoietic porphyria (CEP) is a rare autosomal recessive disease due to the deficient, but not absent, activity of uroporphyrinogen III synthase (UROS), the fourth enzyme in the heme biosynthesis pathway. Biallelic variants in the UROS gene result in decreased UROS enzymatic activity and the accumulation of non-physiologic Type I porphyrins in cells and fluids. Overproduced uroporphyrins in haematopoietic cells are released into the circulation and distributed to tissues, inducing primarily hematologic and dermatologic symptoms. The clinical manifestations vary in severity ranging from non-immune hydrops fetalis in utero to mild dermatologic manifestations in adults. Here, the biochemical, molecular and clinical features of CEP as well as current and new treatment options, including the rescue of UROS enzyme activity by chaperones, are presented.


Subject(s)
Porphyria, Erythropoietic , Uroporphyrinogen III Synthetase , Humans , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/therapy , Uroporphyrinogen III Synthetase/genetics , Uroporphyrinogen III Synthetase/metabolism , Uroporphyrins/genetics
2.
Clin Pediatr (Phila) ; 62(5): 399-403, 2023 06.
Article in English | MEDLINE | ID: mdl-36217751

ABSTRACT

Congenital erythropoietic porphyria (CEP), a rare form of porphyria, is caused by a defect in the heme biosynthesis pathway of the enzyme uroporphyrinogen III synthase (UROS). Uroporphyrinogen III synthase deficiency leads to an accumulation of nonphysiological porphyrins in bone marrow, red blood cells, skin, bones, teeth, and spleen. Consequently, the exposure to sunlight causes severe photosensitivity, long-term intravascular hemolysis, and eventually, irreversible mutilating deformities. Several supportive therapies such as strict sun avoidance, physical sunblocks, red blood cells transfusions, hydroxyurea, and splenectomy are commonly used in the management of CEP. Currently, the only available curative treatment of CEP is hematopoietic stem cell transplantation (HSCT). In this article, we present a young girl in which precocious genetic testing enabled early diagnosis and allowed curative treatment with HSCT for CEP at the age of 3 months of age, that is, the youngest reported case thus far.


Subject(s)
Hematopoietic Stem Cell Transplantation , Porphyria, Erythropoietic , Female , Humans , Infant , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/therapy , Uroporphyrinogen III Synthetase/genetics , Bone Marrow , Genetic Testing
3.
Genes (Basel) ; 12(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34828434

ABSTRACT

Congenital erythropoietic porphyria (CEP, OMIM #606938) is a severe autosomal recessive inborn error of heme biosynthesis. This rare panethnic disease is due to a deficiency of uroporphyrinogen III synthase (or cosynthase). Subsequently, its substrate, the hydroxymethylbilane is subsequently converted into uroporphyrinogen I in a non-enzymatic manner. Of note, uroporphyrinogen I cannot be metabolized into heme and its accumulation in red blood cells results in intramedullary and intravascular hemolysis. The related clinical symptoms occur most frequently during antenatal or neonatal periods but may also appear in late adulthood. The main antenatal clinical presentation is a non-immune hydrops fetalis. We report here two cases of antenatal CEP deficiency and a review of the reported cases in the literature.


Subject(s)
Hydrops Fetalis/genetics , Phenotype , Porphyria, Erythropoietic/genetics , Uroporphyrinogen III Synthetase/genetics , Adult , Female , Humans , Hydrops Fetalis/pathology , Porphyria, Erythropoietic/pathology , Pregnancy
4.
Microbiology (Reading) ; 167(10)2021 10.
Article in English | MEDLINE | ID: mdl-34661520

ABSTRACT

Uroporphyrinogen III, the universal progenitor of macrocyclic, modified tetrapyrroles, is produced from aminolaevulinic acid (ALA) by a conserved pathway involving three enzymes: porphobilinogen synthase (PBGS), hydroxymethylbilane synthase (HmbS) and uroporphyrinogen III synthase (UroS). The gene encoding uroporphyrinogen III synthase has not yet been identified in Plasmodium falciparum, but it has been suggested that this activity is housed inside a bifunctional hybroxymethylbilane synthase (HmbS). Additionally, an unknown protein encoded by PF3D7_1247600 has also been predicted to possess UroS activity. In this study it is demonstrated that neither of these proteins possess UroS activity and the real UroS remains to be identified. This was demonstrated by the failure of codon-optimized genes to complement a defined Escherichia coli hemD- mutant (SASZ31) deficient in UroS activity. Furthermore, HPLC analysis of the oxidized reaction product from recombinant, purified P. falciparum HmbS showed that only uroporphyrin I could be detected (corresponding to hydroxymethylbilane production). No uroporphyrin III was detected, showing that P. falciparum HmbS does not have UroS activity and can only catalyze the formation of hydroxymethylbilane from porphobilinogen.


Subject(s)
Heme/biosynthesis , Hydroxymethylbilane Synthase/metabolism , Plasmodium falciparum/enzymology , Biosynthetic Pathways , Escherichia coli/genetics , Genetic Complementation Test , Hydroxymethylbilane Synthase/genetics , Mutation , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Uroporphyrinogen III Synthetase/genetics , Uroporphyrinogen III Synthetase/metabolism , Uroporphyrinogens/metabolism
5.
Stem Cell Reports ; 15(3): 677-693, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32795423

ABSTRACT

CRISPR/Cas9 is a promising technology for gene correction. However, the edition is often biallelic, and uncontrolled small insertions and deletions (indels) concomitant to precise correction are created. Mutation-specific guide RNAs were recently tested to correct dominant inherited diseases, sparing the wild-type allele. We tested an original approach to correct compound heterozygous recessive mutations. We compared editing efficiency and genotoxicity by biallelic guide RNA versus mutant allele-specific guide RNA in iPSCs derived from a congenital erythropoietic porphyria patient carrying compound heterozygous mutations resulting in UROS gene invalidation. We obtained UROS function rescue and metabolic correction with both guides with the potential of use for porphyria clinical intervention. However, unlike the biallelic one, the mutant allele-specific guide was free of on-target collateral damage. We recommend this design to avoid genotoxicity and to obtain on-target scarless gene correction for recessive disease with frequent cases of compound heterozygous mutations.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , Mutation/genetics , Porphyrias/genetics , Porphyrias/therapy , RNA, Guide, Kinetoplastida/metabolism , Stem Cells/metabolism , Alleles , Base Sequence , Clone Cells , Exons/genetics , Genetic Therapy , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Uroporphyrinogen III Synthetase/genetics
6.
Mol Biol (Mosk) ; 54(2): 262-266, 2020.
Article in Russian | MEDLINE | ID: mdl-32392195

ABSTRACT

rDNA genes play an important role in epigenetic regulation and in differentiation of eukaryotic cells. Using the 4C (circular chromosome conformation capture) approach and model HEK293T cells, we analyzed the rDNA-contacting gene FANK1, using anchor located inside rDNA genes. At the 5' end of the FANK1 gene we detected frequent contacts with rDNA clusters. The contact sites coincide with the border where chromatin state changes and nucleosome positioning. The adjacent genes DHX32, BCCIP and UROS are located in the active chromatin and are transcribed, but do not contact with rDNA genes, while FANK1 gene is silenced, and is located in repressed chromatin. Heat shock treatment dramatically changes the pattern of rDNA contacts in the region and induces about 4-fold increase in activation of the FANK1 gene. We conclude that rDNA contacts may be involved in repression of the FANK1 gene.


Subject(s)
Chromatin/genetics , DNA, Ribosomal/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Transcription Factors/genetics , Calcium-Binding Proteins , Cell Cycle Proteins , Chromatin Assembly and Disassembly , DEAD-box RNA Helicases , HEK293 Cells , Humans , Nuclear Proteins , Uroporphyrinogen III Synthetase
7.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32457103

ABSTRACT

The human intestinal anaerobic commensal and opportunistic pathogen Bacteroides fragilis does not synthesize the tetrapyrrole protoporphyrin IX in order to form heme that is required for growth stimulation and survival in vivo Consequently, B. fragilis acquires essential heme from host tissues during extraintestinal infection. The absence of several genes necessary for de novo heme biosynthesis is a common characteristic of many anaerobic bacteria; however, the uroS gene, encoding a uroporphyrinogen III synthase for an early step of heme biosynthesis, is conserved among the heme-requiring Bacteroidales that inhabit the mammalian gastrointestinal tract. In this study, we show that the ability of B. fragilis to utilize heme or protoporphyrin IX for growth was greatly reduced in a ΔuroS mutant. This growth defect appears to be linked to the suppression of reverse chelatase and ferrochelatase activities in the absence of uroS In addition, this ΔuroS suppressive effect was enhanced by the deletion of the yifB gene, which encodes an Mg2+-chelatase protein belonging to the ATPases associated with various cellular activities (AAA+) superfamily of proteins. Furthermore, the ΔuroS mutant and the ΔuroS ΔyifB double mutant had a severe survival defect compared to the parent strain in competitive infection assays using animal models of intra-abdominal infection and intestinal colonization. This shows that the presence of the uroS and yifB genes in B. fragilis seems to be linked to pathophysiological and nutritional competitive fitness for survival in host tissues. Genetic complementation studies and enzyme kinetics assays indicate that B. fragilis UroS is functionally different from canonical bacterial UroS proteins. Taken together, these findings show that heme assimilation and metabolism in the anaerobe B. fragilis have diverged from those of aerobic and facultative anaerobic pathogenic bacteria.


Subject(s)
Bacterial Proteins/genetics , Bacteroides Infections/microbiology , Bacteroides fragilis/genetics , Bacteroides fragilis/pathogenicity , Ferrochelatase/genetics , Heme/metabolism , Uroporphyrinogen III Synthetase/genetics , Animals , Bacterial Proteins/immunology , Bacteroides Infections/immunology , Bacteroides Infections/metabolism , Bacteroides Infections/pathology , Bacteroides fragilis/immunology , Binding, Competitive , Biological Transport , Ferrochelatase/immunology , Gene Deletion , Gene Expression Regulation , Genetic Complementation Test , Heme/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Intraabdominal Infections/immunology , Intraabdominal Infections/metabolism , Intraabdominal Infections/microbiology , Intraabdominal Infections/pathology , Male , Mice , Mice, Inbred C57BL , Protein Binding , Rats, Sprague-Dawley , Uroporphyrinogen III Synthetase/immunology , Virulence
8.
Biol Blood Marrow Transplant ; 26(4): 704-711, 2020 04.
Article in English | MEDLINE | ID: mdl-31843562

ABSTRACT

Congenital erythropoietic porphyria (CEP) is a rare disease characterized by erosive photosensitivity and chronic hemolysis due to a defect of the enzyme uroporphyrinogen-III-synthase (UROS). To date, hematopoietic stem cell transplantation (HSCT) is the only curative therapy for the devastating early and severe form of the disease. We describe 6 patients with CEP treated with HSCT (3 of them twice after failure of a first graft) between 1994 and 2016 in our center, including 2 of the very first living patients treated more than 20 years ago. Four patients are doing well at 6 to 25 years post-HSCT, with near-normal biochemical parameters of porphyrin metabolism without the cutaneous or hematologic features of CEP. One patient died within the first year after HSCT from severe graft-versus-host disease (GVHD), and 1 child died of unexplained acute hepatic failure at 1 year after HSCT, despite full donor chimerism. Retrospectively, it appears that all but 1 child had increased transaminase activity with onset from the early postnatal period, which was significantly more marked in the child who died of liver failure. In contrast, liver function values progressively normalized after engraftment in all other children. Liver pathology before HSCT for 3 patients revealed varying degrees of portal, centrilobular, and perisinusoidal fibrosis; clarification of hepatocytes; and cytosolic porphyrin deposits. The liver porphyrin content in biopsy specimens was >60 times the normal values. Despite difficult engraftment, the long-term efficacy of HSCT in CEP appears to be favorable and reinforces its benefits for the severe form of CEP. Hepatic involvement requires careful evaluation before and after HSCT and further investigation into its pathophysiology and care.


Subject(s)
Hematopoietic Stem Cell Transplantation , Liver Diseases , Porphyria, Erythropoietic , Bone Marrow Transplantation , Child , Humans , Porphyria, Erythropoietic/therapy , Retrospective Studies , Uroporphyrinogen III Synthetase
9.
Biochem Biophys Res Commun ; 520(2): 297-303, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31601421

ABSTRACT

Clinical severity is heterogeneous among patients suffering from congenital erythropoietic porphyria (CEP) suggesting a modulation of the disease (UROS deficiency) by environmental factors and modifier genes. A KI model of CEP due to a missense mutation of UROS gene present in human has been developed on 3 congenic mouse strains (BALB/c, C57BL/6, and 129/Sv) in order to study the impact of genetic background on disease severity. To detect putative modifiers of disease expression in congenic mice, hematologic data, iron parameters, porphyrin content and tissue samples were collected. Regenerative hemolytic anemia, a consequence of porphyrin excess in RBCs, had various expressions: 129/Sv mice were more hemolytic, BALB/c had more regenerative response to anemia, C57BL/6 were less affected. Iron status and hemolysis level were directly related: C57BL/6 and BALB/c had moderate hemolysis and active erythropoiesis able to reduce iron overload in the liver, while, 129/Sv showed an imbalance between iron release due to hemolysis and erythroid use. The negative control of hepcidin on the ferroportin iron exporter appeared strain specific in the CEP mice models tested. Full repression of hepcidin was observed in BALB/c and 129/Sv mice, favoring parenchymal iron overload in the liver. Unchanged hepcidin levels in C57BL/6 resulted in retention of iron predominantly in reticuloendothelial tissues. These findings open the field for potential therapeutic applications in the human disease, of hepcidin agonists and iron depletion in chronic hemolytic anemia.


Subject(s)
Hepcidins/metabolism , Iron/metabolism , Porphyria, Erythropoietic/genetics , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Disease Models, Animal , Female , Hemolysis , Hepcidins/genetics , Iron Overload/genetics , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Porphyria, Erythropoietic/etiology , Porphyria, Erythropoietic/metabolism , Porphyrins/metabolism , Uroporphyrinogen III Synthetase/genetics
10.
Nat Commun ; 10(1): 1136, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850590

ABSTRACT

CRISPR-Cas9 is a promising technology for genome editing. Here we use Cas9 nuclease-induced double-strand break DNA (DSB) at the UROS locus to model and correct congenital erythropoietic porphyria. We demonstrate that homology-directed repair is rare compared with NHEJ pathway leading to on-target indels and causing unwanted dysfunctional protein. Moreover, we describe unexpected chromosomal truncations resulting from only one Cas9 nuclease-induced DSB in cell lines and primary cells by a p53-dependent mechanism. Altogether, these side effects may limit the promising perspectives of the CRISPR-Cas9 nuclease system for disease modeling and gene therapy. We show that the single nickase approach could be safer since it prevents on- and off-target indels and chromosomal truncations. These results demonstrate that the single nickase and not the nuclease approach is preferable, not only for modeling disease but also and more importantly for the safe management of future CRISPR-Cas9-mediated gene therapies.


Subject(s)
CRISPR-Cas Systems , Chromosomes, Human, Pair 10 , DNA Breaks, Double-Stranded , Deoxyribonuclease I/genetics , Gene Editing/methods , Genetic Therapy/methods , Uroporphyrinogen III Synthetase/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Chromosome Deletion , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , Deoxyribonuclease I/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Genome, Human , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , K562 Cells , Models, Biological , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Porphyria, Erythropoietic/pathology , Porphyria, Erythropoietic/therapy , Primary Cell Culture , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Recombinational DNA Repair , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Uroporphyrinogen III Synthetase/metabolism
11.
Mol Genet Metab ; 128(3): 358-362, 2019 11.
Article in English | MEDLINE | ID: mdl-30454868

ABSTRACT

The erythropoietic porphyrias are inborn errors of heme biosynthesis with prominent cutaneous manifestations. They include autosomal recessive Congenital Erythropoietic Porphyria (CEP) due to loss-of-function (LOF) mutations in the Uroporphyrinogen III Synthase (UROS) gene, Erythropoietic Protoporphyria (EPP) due to LOF mutations in the ferrochelatase (FECH) gene, and X-Linked Protoporphyria (XLP) due to gain-of-function mutations in the terminal exon of the Aminolevulinic Acid Synthase 2 (ALAS2) gene. During the 11-year period from 01/01/2007 through 12/31/2017, the Mount Sinai Porphyrias Diagnostic Laboratory provided molecular diagnostic testing for one or more of these disorders in 628 individuals, including 413 unrelated individuals. Of these 628, 120 patients were tested for CEP, 483 for EPP, and 331 for XLP, for a total of 934 tests. For CEP, 24 of 78 (31%) unrelated individuals tested had UROS mutations, including seven novel mutations. For EPP, 239 of 362 (66%) unrelated individuals tested had pathogenic FECH mutations, including twenty novel mutations. The IVS3-48 T > C low-expression allele was present in 231 (97%) of 239 mutation-positive EPP probands with a pathogenic FECH mutation. In the remaining 3%, three patients with two different FECH mutations in trans were identified. For XLP, 24 of 250 (10%) unrelated individuals tested had ALAS2 exon 11 mutations. No novel ALAS2 mutations were identified. Among family members referred for testing, 33 of 42 (79%) CEP, 62 of 121 (51%) EPP, and 31 of 81 (38%) XLP family members had the respective family mutation. Mutation-positive CEP, EPP, and XLP patients who had been biochemically tested had marked elevations of the disease-appropriate porphyrin intermediates. These results expand the molecular heterogeneity of the erythropoietic porphyrias by adding a total of 27 novel mutations. The results document the usefulness of molecular testing to confirm the positive biochemical findings in these patients and to identify heterozygous family members.


Subject(s)
Ferrochelatase/genetics , Genetic Heterogeneity , Mutation , Porphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/genetics , Uroporphyrinogen III Synthetase/genetics , Family , Female , Genetic Carrier Screening , Heme/biosynthesis , Humans , Male , Molecular Diagnostic Techniques , Photosensitivity Disorders/etiology , Protoporphyria, Erythropoietic/diagnosis
12.
Sci Rep ; 8(1): 16145, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385858

ABSTRACT

Blood platelets can interact with bacteria, possibly leading to platelet activation, cytokine and microparticle release and immune signalling. Besides, bacteria can also affect the platelet RNA content. We investigated the impact of non-pathogenic K12 and pathogenic O18:K1 Escherichia (E.) coli strains on platelet activation, RNA expression patterns, and selected proteins. Depending on bacteria concentration, contact of platelets with E. coli K12 lead to an increase of P-selectin (24-51.3%), CD63 (15.9-24.3%), PAC-1 (3.8-14.9%) and bound fibrinogen (22.4-39%) on the surface. E. coli O18:K1 did not affect these markers. Sequencing analysis of total RNA showed that E. coli K12 caused a significant concentration change of 103 spliced mRNAs, of which 74 decreased. For the RNAs of HMBS (logFC = +5.73), ATP2C1 (logFC = -3.13) and LRCH4 (logFC = -4.07) changes were detectable by thromboSeq and Tuxedo pipelines. By Western blot we observed the conversion of HMBS protein from a 47 kDA to 40 kDa product by E. coli K12, O18:K1 and by purified lipopolysaccharide. While ATP2C1 protein was released from platelets, E. coli either reduced the secretion or broke down the released protein making it undetectable by antibodies. Our results demonstrate that different E. coli strains influence activation, RNA and protein levels differently which may affect platelet-bacteria crosstalk.


Subject(s)
Blood Platelets/metabolism , Calcium-Transporting ATPases/genetics , Escherichia coli K12/genetics , Nerve Tissue Proteins/genetics , Uroporphyrinogen III Synthetase/genetics , Antigens, Bacterial/genetics , Calcium-Transporting ATPases/blood , Escherichia coli Infections/blood , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli K12/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Humans , Lipopolysaccharides/genetics , P-Selectin/genetics , Platelet Activation/genetics , RNA/blood , RNA/genetics , Sequence Analysis, RNA , Tetraspanin 30/genetics
13.
Sci Transl Med ; 10(459)2018 09 19.
Article in English | MEDLINE | ID: mdl-30232228

ABSTRACT

Congenital erythropoietic porphyria is a rare autosomal recessive disease produced by deficient activity of uroporphyrinogen III synthase, the fourth enzyme in the heme biosynthetic pathway. The disease affects many organs, can be life-threatening, and currently lacks curative treatments. Inherited mutations most commonly reduce the enzyme's stability, altering its homeostasis and ultimately blunting intracellular heme production. This results in uroporphyrin by-product accumulation in the body, aggravating associated pathological symptoms such as skin photosensitivity and disfiguring phototoxic cutaneous lesions. We demonstrated that the synthetic marketed antifungal ciclopirox binds to the enzyme, stabilizing it. Ciclopirox targeted the enzyme at an allosteric site distant from the active center and did not affect the enzyme's catalytic role. The drug restored enzymatic activity in vitro and ex vivo and was able to alleviate most clinical symptoms of congenital erythropoietic porphyria in a genetic mouse model of the disease at subtoxic concentrations. Our findings establish a possible line of therapeutic intervention against congenital erythropoietic porphyria, which is potentially applicable to most of deleterious missense mutations causing this devastating disease.


Subject(s)
Ciclopirox/therapeutic use , Drug Repositioning , Porphyria, Erythropoietic/drug therapy , Allosteric Site , Animals , Biophysical Phenomena , Cell Line , Ciclopirox/pharmacokinetics , Disease Models, Animal , Homeostasis , Mice , Phenotype , Porphyria, Erythropoietic/enzymology , Porphyria, Erythropoietic/pathology , Uroporphyrinogen III Synthetase/antagonists & inhibitors , Uroporphyrinogen III Synthetase/chemistry , Uroporphyrinogen III Synthetase/metabolism
14.
Structure ; 26(4): 565-571.e3, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29551288

ABSTRACT

There are numerous applications that use the structures of protein-ligand complexes from the PDB, such as 3D pharmacophore identification, virtual screening, and fragment-based drug design. The structures underlying these applications are potentially much more informative if they contain biologically relevant bound ligands, with high similarity to the cognate ligands. We present a study of ligand-enzyme complexes that compares the similarity of bound and cognate ligands, enabling the best matches to be identified. We calculate the molecular similarity scores using a method called PARITY (proportion of atoms residing in identical topology), which can conveniently be combined to give a similarity score for all cognate reactants or products in the reaction. Thus, we generate a rank-ordered list of related PDB structures, according to the biological similarity of the ligands bound in the structures.


Subject(s)
Acetylcholine/chemistry , Acetylcholinesterase/chemistry , Biosimilar Pharmaceuticals/chemistry , Uroporphyrinogen III Synthetase/chemistry , Uroporphyrinogens/chemistry , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Binding Sites , Biosimilar Pharmaceuticals/metabolism , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Substrate Specificity , Uroporphyrinogen III Synthetase/metabolism , Uroporphyrinogens/metabolism
15.
Hum Mol Genet ; 26(8): 1565-1576, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28334762

ABSTRACT

Congenital erythropoietic porphyria (CEP) is an inborn error of heme biosynthesis characterized by uroporphyrinogen III synthase (UROS) deficiency resulting in deleterious porphyrin accumulation in blood cells responsible for hemolytic anemia and cutaneous photosensitivity. We analyzed here the molecular basis of UROS impairment associated with twenty nine UROS missense mutations actually described in CEP patients. Using a computational and biophysical joint approach we predicted that most disease-causing mutations would affect UROS folding and stability. Through the analysis of enhanced green fluorescent protein-tagged versions of UROS enzyme we experimentally confirmed these data and showed that thermodynamic instability and premature protein degradation is a major mechanism accounting for the enzymatic deficiency associated with twenty UROS mutants in human cells. Since the intracellular loss in protein homeostasis is in excellent agreement with the in vitro destabilization, we used molecular dynamic simulation to rely structural 3D modification with UROS disability. We found that destabilizing mutations could be clustered within three types of mechanism according to side chain rearrangements or contact alterations within the pathogenic UROS enzyme so that the severity degree correlated with cellular protein instability. Furthermore, proteasome inhibition using bortezomib, a clinically available drug, significantly enhanced proteostasis of each unstable UROS mutant. Finally, we show evidence that abnormal protein homeostasis is a prevalent mechanism responsible for UROS deficiency and that modulators of UROS proteolysis such as proteasome inhibitors or chemical chaperones may represent an attractive therapeutic option to reduce porphyrin accumulation and prevent skin photosensitivity in CEP patients when the genotype includes a missense variant.


Subject(s)
Mutation, Missense/genetics , Porphyria, Erythropoietic/genetics , Structure-Activity Relationship , Uroporphyrinogen III Synthetase/genetics , Computational Biology , Homeostasis , Humans , Porphyria, Erythropoietic/metabolism , Porphyria, Erythropoietic/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/therapeutic use , Protein Folding , Uroporphyrinogen III Synthetase/chemistry
16.
Proteins ; 85(1): 46-53, 2017 01.
Article in English | MEDLINE | ID: mdl-27756106

ABSTRACT

In an earlier study, we showed that two-domain segment-swapped proteins can evolve by domain swapping and fusion, resulting in a protein with two linkers connecting its domains. We proposed that a potential evolutionary advantage of this topology may be the restriction of interdomain motions, which may facilitate domain closure by a hinge-like movement, crucial for the function of many enzymes. Here, we test this hypothesis computationally on uroporphyrinogen III synthase, a two-domain segment-swapped enzyme essential in porphyrin metabolism. To compare the interdomain flexibility between the wild-type, segment-swapped enzyme (having two interdomain linkers) and circular permutants of the same enzyme having only one interdomain linker, we performed geometric and molecular dynamics simulations for these species in their ligand-free and ligand-bound forms. We find that in the ligand-free form, interdomain motions in the wild-type enzyme are significantly more restricted than they would be with only one interdomain linker, while the flexibility difference is negligible in the ligand-bound form. We also estimated the entropy costs of ligand binding associated with the interdomain motions, and find that the change in domain connectivity due to segment swapping results in a reduction of this entropy cost, corresponding to ∼20% of the total ligand binding free energy. In addition, the restriction of interdomain motions may also help the functional domain-closure motion required for catalysis. This suggests that the evolution of the segment-swapped topology facilitated the evolution of enzyme function for this protein by influencing its dynamic properties. Proteins 2016; 85:46-53. © 2016 Wiley Periodicals, Inc.


Subject(s)
Bacterial Proteins/chemistry , Thermus thermophilus/chemistry , Uroporphyrinogen III Synthetase/chemistry , Uroporphyrinogens/chemistry , Biocatalysis , Entropy , Evolution, Molecular , Ligands , Molecular Dynamics Simulation , Motion , Protein Binding , Protein Domains , Protein Structure, Secondary , Thermus thermophilus/enzymology
18.
Br J Dermatol ; 175(6): 1346-1350, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27086902

ABSTRACT

Deficiency of uroporphyrinogen III synthase (UROS) causes congenital erythropoietic porphyria (CEP). The disease, originating from the inheritance of mutations within the UROS gene, presents a recessive form of transmission. In a few patients, a late-onset CEP-like phenotype without UROS mutations appears to be associated with a myelodysplastic syndrome. We report a 60-year-old man with late-onset signs of cutaneous porphyria and accumulation in urine, plasma and faeces of type I porphyrin isomers characteristic of CEP. Analysis of DNA from peripheral leucocytes, skin and bone marrow aspirate showed that he was a heterozygous carrier of a Cys73Arg (c.217 T>C) mutation within UROS. Sequencing of cDNA from peripheral blood confirmed heterozygosity and expression of the normal allele. Measurement of UROS enzymatic activity in erythrocytes showed values ~70% of normal, indirectly indicating expression of the normal allele. Differently from other cases of late-onset uroporphyria, the patient did not present thrombocytopenia or any evidence of a myelodysplastic syndrome. Five years of clinical follow-up showed persistence of skin signs and increased excretion of porphyrins, independently of lifestyle factors or changes in medication regimes. We hypothesize acquired mosaicism (in the bone marrow) affecting the UROS gene. Thus, unstable cellular clones initiated overproduction of isomer I porphyrins leading to a CEP phenotype. This could be explained either by a clonal expansion of the porphyric (Cys73Arg) allele or by loss of function of the normal allele. Cellular turnover would facilitate release of uroporphyrins into circulation and subsequent skin lesions. This is the first case of a CEP heterozygous carrier presenting clinical manifestations.


Subject(s)
Hand Dermatoses/genetics , Late Onset Disorders/genetics , Mutation, Missense/genetics , Porphyrias/genetics , Uroporphyrinogen III Synthetase/genetics , Heterozygote , Humans , Male , Middle Aged , Porphyrins/metabolism
19.
Br J Haematol ; 173(3): 365-79, 2016 05.
Article in English | MEDLINE | ID: mdl-26969896

ABSTRACT

Congenital erythropoietic porphyria (CEP) is a rare genetic disease resulting from the remarkable deficient activity of uroporphyrinogen III synthase, the fourth enzyme of the haem biosynthetic pathway. This enzyme defect results in overproduction of the non-physiological and pathogenic porphyrin isomers, uroporphyrin I and coproporphyrin I. The predominant clinical characteristics of CEP include bullous cutaneous photosensitivity to visible light from early infancy, progressive photomutilation and chronic haemolytic anaemia. The severity of clinical manifestations is markedly heterogeneous among patients; and interdependence between disease severity and porphyrin amount in the tissues has been pointed out. A more pronounced endogenous production of porphyrins concomitant to activation of ALAS2, the first and rate-limiting of the haem synthesis enzymes in erythroid cells, has also been reported. CEP is inherited as autosomal recessive or X-linked trait due to mutations in UROS or GATA1 genes; however an involvement of other causative or modifier genes cannot be ruled out.


Subject(s)
Porphyria, Erythropoietic/pathology , GATA1 Transcription Factor/genetics , Heme/biosynthesis , Humans , Mutation , Phenotype , Porphyria, Erythropoietic/etiology , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Porphyrins/biosynthesis , Porphyrins/metabolism , Uroporphyrinogen III Synthetase
SELECTION OF CITATIONS
SEARCH DETAIL
...