Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
J Med Case Rep ; 17(1): 472, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37957719

ABSTRACT

BACKGROUND: Congenital erythropoietic porphyria (CEP), also known as pink tooth or Gunther disease, is a rare hereditary disorder caused by an enzyme mutation in the heme biosynthesis pathway, which leads to the accumulation of immature and non-physiological protoporphyrin rings in various tissues. CEP is characterized by sun-exposed bullous skin lesions, hemolytic anemia, red/brown urine, and teeth staining. CASE PRESENTATION: We present a unique case of a 10-year-old Asian boy with CEP who presented with recurrent epistaxis, an unusual presentation for this condition. Based on clinical presentation and laboratory findings, including elevated urine uroporphyrin and coproporphyrin I and III levels, microcytic anemia, a higher red cell distribution width (RDW), and a lower platelet count, a thorough assessment and detailed workup resulted in a diagnosis of CEP. The patient underwent a successful splenectomy and recovered without any complications. CONCLUSION: This case report aims to raise awareness among healthcare professionals about the uncommon and atypical presentation of CEP and its management options.


Subject(s)
Anemia, Hemolytic , Porphyria, Erythropoietic , Male , Humans , Child , Porphyria, Erythropoietic/complications , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Epistaxis/complications , Mutation
2.
Einstein (Sao Paulo) ; 21: eRC0256, 2023.
Article in English | MEDLINE | ID: mdl-37255061

ABSTRACT

A male infant presented with progressive jaundice immediately after birth. Fecal acholia and choluria associated with extensive bullous skin lesions in his trunk, abdomen, and upper and lower limbs developed during phototherapy. Several diagnostic hypotheses were presented, including neonatal porphyria, hemochromatosis, Alagille syndrome, and neonatal lupus. A 24-hour urine sample for the dosage of urinary porphyrins was collected, showing high results (1823.6µg in 100mL). At 50 days of life, fluorescence spectroscopy using a Wood's lamp revealed simultaneous bright red fluorescence of urine-stained diapers and sample blood. A definitive diagnosis of congenital erythropoietic porphyria was made following identification of a mutation of the uroporphyrinogen synthetases III gene on genetic testing. The patient was subsequently maintained in a low light environment since then, resulting in improvement of the lesions. Congenital erythropoietic porphyria is a disease of the group of porphyrias that presents shortly after birth with blistering occurring in regions exposed to the sun or other ultraviolet light. Atrophic scars, mutilated fingers, and bright red fluorescence of the urine and teeth may also be observed. There is no specific treatment, and prophylaxis comprising a total avoidance of sunlight is generally recommended. A high degree of suspicion is required for diagnosis. An early diagnosis can lead to less damage. Here, we present the case of a newborn with congenital erythropoietic porphyria diagnosed after presenting with bullous lesions secondary to phototherapy.


Subject(s)
Lupus Erythematosus, Systemic , Porphyria, Erythropoietic , Infant , Infant, Newborn , Humans , Male , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/therapy , Blister/complications , Phototherapy , Lupus Erythematosus, Systemic/complications , Mutation
3.
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
5.
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
6.
Sci Rep ; 11(1): 9601, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953217

ABSTRACT

Congenital erythropoietic porphyria (CEP) is a rare genetic disorder leading to accumulation of uro/coproporphyrin-I in tissues due to inhibition of uroporphyrinogen-III synthase. Clinical manifestations of CEP include bone fragility, severe photosensitivity and photomutilation. Currently there is no specific treatment for CEP, except bone marrow transplantation, and there is an unmet need for treating this orphan disease. Fluorescent porphyrins cause protein aggregation, which led us to hypothesize that uroporphyrin-I accumulation leads to protein aggregation and CEP-related bone phenotype. We developed a zebrafish model that phenocopies features of CEP. As in human patients, uroporphyrin-I accumulated in the bones of zebrafish, leading to impaired bone development. Furthermore, in an osteoblast-like cell line, uroporphyrin-I decreased mineralization, aggregated bone matrix proteins, activated endoplasmic reticulum stress and disrupted autophagy. Using high-throughput drug screening, we identified acitretin, a second-generation retinoid, and showed that it reduced uroporphyrin-I accumulation and its deleterious effects on bones. Our findings provide a new CEP experimental model and a potential repurposed therapeutic.


Subject(s)
Acitretin/therapeutic use , Bone Development/drug effects , Bone and Bones/drug effects , Porphyria, Erythropoietic/drug therapy , Uroporphyrins/metabolism , Acitretin/pharmacology , Animals , Bone and Bones/metabolism , Cell Line , Disease Models, Animal , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Uroporphyrins/genetics , Zebrafish
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.
Ann Dermatol Venereol ; 146(2): 143-159, 2019 Feb.
Article in French | MEDLINE | ID: mdl-30709634

ABSTRACT

The porphyrias are a group of metabolic disorders resulting from an innate abnormality in haem biosynthesis, and the clinical settings of which vary according to the genetic enzyme abnormality in question. These are genetic disorders with autosomal dominant or recessive inheritance of varying penetrance, and whose clinical expression differs according to the preferential location of haem precursors. Different classifications have been proposed according to genetic inheritance, the enzyme anomaly at issue, and clinical expression. The clinical classification distinguishes between acute porphyria (acute intermittent porphyria, porphyria variegata, hereditary coproporphyria), bullous cutaneous porphyrias (porphyria cutanea tarda, porphyria variegata and hereditary coproporphyria), painful photosensitive acute cutaneous porphyrias (erythropoietic protoporphyria and X-linked dominant protoporphyria), and rare recessive porphyrias (congenital erythropoietic porphyria, Doss porphyria, hepatoerythropoietic porphyria and harderoporphyria). Treatment depends on the clinical expression of the disorder.


Subject(s)
Porphyrias , Skin Diseases, Metabolic , Biopsy , Coproporphyria, Hereditary/diagnosis , Coproporphyria, Hereditary/genetics , Coproporphyria, Hereditary/therapy , Diagnosis, Differential , Heme/biosynthesis , Humans , Photosensitivity Disorders/complications , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/therapy , Porphyria Cutanea Tarda/diagnosis , Porphyria Cutanea Tarda/genetics , Porphyria Cutanea Tarda/therapy , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/therapy , Porphyrias/classification , Porphyrias/diagnosis , Porphyrias/genetics , Porphyrias/therapy , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/genetics , Protoporphyria, Erythropoietic/therapy , Skin/pathology , Skin Diseases, Metabolic/classification , Skin Diseases, Metabolic/diagnosis , Skin Diseases, Metabolic/genetics , Skin Diseases, Metabolic/therapy
12.
Mol Genet Metab ; 128(3): 288-297, 2019 11.
Article in English | MEDLINE | ID: mdl-30685241

ABSTRACT

Congenital erythropoietic porphyria (CEP) is a rare autosomal recessive disorder characterized by photosensitivity and by hematologic abnormalities in affected individuals. CEP is caused by mutations in the uroporphyrinogen synthase (UROS) gene. In three reported cases, CEP has been associated with a specific X-linked GATA1 mutation. Disease-causing mutations in either gene result in absent or markedly reduced UROS enzymatic activity. This in turn leads to the accumulation of the non-physiologic and photoreactive porphyrinogens, uroporphyrinogen I and coproporphyrinogen I, which damage erythrocytes and elicit a phototoxic reaction upon light exposure. The clinical spectrum of CEP depends on the level of residual UROS activity, which is determined by the underlying pathogenic loss-of-function UROS mutations. Disease severity ranges from non-immune hydrops fetalis in utero to late-onset disease with only mild cutaneous involvement. The clinical characteristics of CEP include exquisite photosensitivity to visible light resulting in bullous vesicular lesions which, when infected lead to progressive photomutilation of sun-exposed areas such as the face and hands. In addition, patients have erythrodontia (brownish discoloration of teeth) and can develop corneal scarring. Chronic transfusion-dependent hemolytic anemia is common and leads to bone marrow hyperplasia, which further increases porphyrin production. Management of CEP consists of strict avoidance of exposure to visible light with sun-protective clothing, sunglasses, and car and home window filters. Adequate care of ruptured vesicles and use of topical antibiotics is indicated to prevent superinfections and osteolysis. In patients with symptomatic hemolytic anemia, frequent erythrocyte cell transfusions may be necessary to suppress hematopoiesis and decrease marrow production of the phototoxic porphyrins. In severe transfection-dependent cases, bone marrow or hematopoietic stem cell transplantation has been performed, which is curative. Therapeutic approaches including gene therapy, proteasome inhibition, and pharmacologic chaperones are under investigation.


Subject(s)
Biosynthetic Pathways , Genetic Diseases, Inborn , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/physiopathology , Animals , GATA1 Transcription Factor/genetics , Genetic Therapy , Heme/metabolism , Humans , Mice , Mutation , Porphyria, Erythropoietic/complications , Porphyria, Erythropoietic/therapy
13.
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
14.
Proc Natl Acad Sci U S A ; 114(38): E8045-E8052, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874591

ABSTRACT

Loss-of-function mutations in genes for heme biosynthetic enzymes can give rise to congenital porphyrias, eight forms of which have been described. The genetic penetrance of the porphyrias is clinically variable, underscoring the role of additional causative, contributing, and modifier genes. We previously discovered that the mitochondrial AAA+ unfoldase ClpX promotes heme biosynthesis by activation of δ-aminolevulinate synthase (ALAS), which catalyzes the first step of heme synthesis. CLPX has also been reported to mediate heme-induced turnover of ALAS. Here we report a dominant mutation in the ATPase active site of human CLPX, p.Gly298Asp, that results in pathological accumulation of the heme biosynthesis intermediate protoporphyrin IX (PPIX). Amassing of PPIX in erythroid cells promotes erythropoietic protoporphyria (EPP) in the affected family. The mutation in CLPX inactivates its ATPase activity, resulting in coassembly of mutant and WT protomers to form an enzyme with reduced activity. The presence of low-activity CLPX increases the posttranslational stability of ALAS, causing increased ALAS protein and ALA levels, leading to abnormal accumulation of PPIX. Our results thus identify an additional molecular mechanism underlying the development of EPP and further our understanding of the multiple mechanisms by which CLPX controls heme metabolism.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Endopeptidase Clp , Mutation, Missense , Porphyria, Erythropoietic , Protoporphyrins/biosynthesis , 5-Aminolevulinate Synthetase/genetics , Adolescent , Amino Acid Substitution , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Enzyme Stability/genetics , Female , Humans , Male , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Porphyria, Erythropoietic/pathology , Protoporphyrins/genetics
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
17.
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
18.
Expert Rev Mol Med ; 18: e3, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26953528

ABSTRACT

The GATA family of transcription factors consists of six proteins (GATA1-6) which are involved in a variety of physiological and pathological processes. GATA1/2/3 are required for differentiation of mesoderm and ectoderm-derived tissues, including the haematopoietic and central nervous system. GATA4/5/6 are implicated in development and differentiation of endoderm- and mesoderm-derived tissues such as induction of differentiation of embryonic stem cells, cardiovascular embryogenesis and guidance of epithelial cell differentiation in the adult.


Subject(s)
Endoderm/metabolism , GATA Transcription Factors/genetics , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Neoplasms/genetics , Animals , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Cell Differentiation , Central Nervous System/growth & development , Central Nervous System/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/growth & development , Epithelial Cells/cytology , Epithelial Cells/metabolism , GATA Transcription Factors/metabolism , Hematopoietic System/growth & development , Hematopoietic System/metabolism , Humans , Mesoderm/cytology , Mesoderm/growth & development , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Porphyria, Erythropoietic/pathology , Signal Transduction
19.
Med. clín (Ed. impr.) ; 145(10): 433-435, nov. 2015. tab
Article in Spanish | IBECS | ID: ibc-145253

ABSTRACT

Fundamento y objetivo: La causa o causas de la anemia que acompaña a la anorexia nerviosa (AN) no ha sido establecida, pero no parece relacionarse con deficiencias nutricionales ni cambios medulares. El objetivo de este trabajo fue evaluar la producción de eritropoyetina (EPO) en respuesta a la anemia en un pequeño grupo de pacientes con AN y anemia. Pacientes y métodos: Los niveles de EPO en muestras de suero de 41 mujeres con AN (11 con anemia y 30 sin alteraciones en los parámetros de la serie eritroide) se compararon con la respuesta observada en un grupo de pacientes de peso normal con anemia. Resultados: Las concentraciones de EPO en pacientes con AN anémicas fueron mayores que en las no anémicas: 20,63 mU/ml (4,04 a 28,46) frente a 8,7 mU/ml (3,9 a 20,93), p = 0,0088, pero el aumento de EPO fue menor de lo esperado (27,85 mU/ml [17,7 a 118,9]), p = 0,014. La correlación entre el IMC y la diferencia entre la EPO y la EPO esperada es inversa. Conclusiones: Una producción inadecuada de EPO puede explicar en parte la anemia en la AN. Son necesarios más estudios para investigar la causa de esta respuesta (AU)


Background and objective: The cause of the anemia in anorexia nervosa (AN) has not been fully ascertained. Ferritin, folate and cobalamin values are usually within normal ranges. Anemia does not have a relationship with bone marrow changes and erythropoietin (EPO) levels have not been investigated. The objective of this study was to evaluate the EPO response in a small group of AN patients. Patients and methods: EPO levels were measured in serum samples of 41 female AN patients (11 with anemia, and 30 with normal blood cell count). The adequacy of EPO response was assessed by comparing the increase observed in a group of normal weight patients with anemia. Results: EPO concentrations in anemic AN patients were higher than in non-anemic: 20.63 mU/mL (4.04-28.46) vs 8.7 mU/mL (3.9-20.93), P = .0088, but the increase in EPO was lower than expected (27.85 mU/mL [17.7-118.9]), P = .014. BMI and the difference between actual and expected EPO were inversely correlated. Conclusions: Inadequate EPO response may partly explain anemia in AN, but further studies are necessary (AU)


Subject(s)
Female , Humans , Anorexia Nervosa/metabolism , Anorexia Nervosa/pathology , Anemia/blood , Anemia/metabolism , Porphyria, Erythropoietic/pathology , Feeding and Eating Disorders of Childhood/diagnosis , Hematology/methods , Anorexia Nervosa/complications , Anorexia Nervosa/diagnosis , Anemia/complications , Anemia/pathology , Porphyria, Erythropoietic/genetics , Feeding and Eating Disorders of Childhood/complications , Hematology/standards
20.
World J Pediatr Congenit Heart Surg ; 6(4): 646-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26467880

ABSTRACT

An 18-year-old boy with congenital erythropoietic porphyria and calcific constrictive pericarditis underwent total pericardiectomy. The cause of pericardial calcification could be deposition of porphyrins in the pericardium. Surgical importance of this rare condition is highlighted.


Subject(s)
Pericardiectomy/methods , Pericarditis, Constrictive/etiology , Porphyria, Erythropoietic/complications , Adolescent , Echocardiography , Humans , Male , Pericarditis, Constrictive/genetics , Pericarditis, Constrictive/surgery , Pericardium/metabolism , Porphyria, Erythropoietic/diagnosis , Porphyria, Erythropoietic/genetics , Porphyrins/metabolism , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...