Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Genes (Basel) ; 12(11)2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34828434

RESUMO

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.


Assuntos
Hidropisia Fetal/genética , Fenótipo , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , Adulto , Feminino , Humanos , Hidropisia Fetal/patologia , Porfiria Eritropoética/patologia , Gravidez
3.
Nat Commun ; 10(1): 1136, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850590

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Cromossomos Humanos Par 10 , Quebras de DNA de Cadeia Dupla , Desoxirribonuclease I/genética , Edição de Genes/métodos , Terapia Genética/métodos , Uroporfirinogênio III Sintetase/genética , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Deleção Cromossômica , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/genética , DNA/metabolismo , Desoxirribonuclease I/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Genoma Humano , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células K562 , Modelos Biológicos , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Porfiria Eritropoética/terapia , Cultura Primária de Células , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Reparo de DNA por Recombinação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Uroporfirinogênio III Sintetase/metabolismo
4.
Sci Transl Med ; 10(459)2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30232228

RESUMO

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.


Assuntos
Ciclopirox/uso terapêutico , Reposicionamento de Medicamentos , Porfiria Eritropoética/tratamento farmacológico , Sítio Alostérico , Animais , Fenômenos Biofísicos , Linhagem Celular , Ciclopirox/farmacocinética , Modelos Animais de Doenças , Homeostase , Camundongos , Fenótipo , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/patologia , Uroporfirinogênio III Sintetase/antagonistas & inibidores , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo
5.
Br J Dermatol ; 179(2): 486-490, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28865079

RESUMO

Congenital erythropoietic porphyria is a rare autosomal recessive disease caused by a deficiency of uroporphyrinogen III synthase, owing to mutations in UROS in chromosome 10. Occasionally, patients show a mild, late-onset disease, without germline UROS mutations, associated with haematological malignancies. We report a 65-year-old patient with photosensitivity, overexcretion of porphyrins and thrombocytopenia. Bone marrow analysis gave a diagnosis of myelodysplastic syndrome (MDS) with the presence of a derivative chromosome 3, possibly due to an inversion including 3q21 and 3q26 break points. After allogeneic stem-cell transplantation, complete remission of MDS and uroporphyria was achieved. To our knowledge, this is the first reported case of acquired erythropoietic uroporphyria associated with MDS, with chromosome 3 alterations.


Assuntos
Cromossomos Humanos Par 3/genética , Transtornos de Início Tardio/diagnóstico , Síndromes Mielodisplásicas/diagnóstico , Porfiria Eritropoética/diagnóstico , Idoso , Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/uso terapêutico , Transfusão de Sangue , Medula Óssea/patologia , Transplante de Medula Óssea , Inversão Cromossômica , Humanos , Transtornos de Início Tardio/etiologia , Transtornos de Início Tardio/patologia , Transtornos de Início Tardio/terapia , Masculino , Síndromes Mielodisplásicas/complicações , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/terapia , Porfiria Eritropoética/etiologia , Porfiria Eritropoética/patologia , Porfiria Eritropoética/terapia , Porfirinas/sangue , Porfirinas/urina , Pele/patologia , Resultado do Tratamento
6.
Proc Natl Acad Sci U S A ; 114(38): E8045-E8052, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28874591

RESUMO

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.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Endopeptidase Clp , Mutação de Sentido Incorreto , Porfiria Eritropoética , Protoporfirinas/biossíntese , 5-Aminolevulinato Sintetase/genética , Adolescente , Substituição de Aminoácidos , Endopeptidase Clp/genética , Endopeptidase Clp/metabolismo , Estabilidade Enzimática/genética , Feminino , Humanos , Masculino , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Protoporfirinas/genética
7.
Hum Mol Genet ; 26(8): 1565-1576, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28334762

RESUMO

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.


Assuntos
Mutação de Sentido Incorreto/genética , Porfiria Eritropoética/genética , Relação Estrutura-Atividade , Uroporfirinogênio III Sintetase/genética , Biologia Computacional , Homeostase , Humanos , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma/química , Inibidores de Proteassoma/uso terapêutico , Dobramento de Proteína , Uroporfirinogênio III Sintetase/química
8.
Dermatol Online J ; 23(2)2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-28329491

RESUMO

Patients with the rare genodermatosis congenitalerythropoietic porphyria (CEP, Gunther disease)develop erosions and scarring on sun-exposedsites caused by phototoxin mediated damage.Compromised skin barrier function places patientsat higher risk of infection and long term sequelaeinclude scarring. We report a long term follow up ofa 60 year old patient born with CEP and provide anextensive literature review of CEP including recentupdates on potential management options. Multiplepatient interviews and collection of biochemistry datawere conducted for the case discussion. All Australianpathology laboratories in each state performingporphyria testing were surveyed in mid 2015 to verifyexistence of other cases of CEP in Australia with onlyone case of true congenital porphyria identifiedand one adult onset case. Congenital erythropoieticporphyria is a rare condition with no cure currentlyavailable. It is important to diagnose patients earlyto prevent and minimize complications such asscarring and secondary infection, provide longterm skin checks, and advise patients about lifestylemodification.


Assuntos
Cicatriz/patologia , Porfiria Eritropoética/fisiopatologia , Infecções Cutâneas Estafilocócicas/fisiopatologia , Cicatriz/etiologia , Erros de Diagnóstico , Seguimentos , Humanos , Estudos Longitudinais , Masculino , Staphylococcus aureus Resistente à Meticilina , Pessoa de Meia-Idade , Porfiria Cutânea Tardia/diagnóstico , Porfiria Eritropoética/complicações , Porfiria Eritropoética/diagnóstico , Porfiria Eritropoética/patologia , Infecções Cutâneas Estafilocócicas/etiologia , Infecções Cutâneas Estafilocócicas/patologia
9.
Br J Haematol ; 173(3): 365-79, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26969896

RESUMO

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.


Assuntos
Porfiria Eritropoética/patologia , Fator de Transcrição GATA1/genética , Heme/biossíntese , Humanos , Mutação , Fenótipo , Porfiria Eritropoética/etiologia , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfirinas/biossíntese , Porfirinas/metabolismo , Uroporfirinogênio III Sintetase
10.
Expert Rev Mol Med ; 18: e3, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26953528

RESUMO

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.


Assuntos
Endoderma/metabolismo , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Neoplasias/genética , Animais , Sistema Cardiovascular/crescimento & desenvolvimento , Sistema Cardiovascular/metabolismo , Diferenciação Celular , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endoderma/citologia , Endoderma/crescimento & desenvolvimento , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fatores de Transcrição GATA/metabolismo , Sistema Hematopoético/crescimento & desenvolvimento , Sistema Hematopoético/metabolismo , Humanos , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Mutação , Neoplasias/metabolismo , Neoplasias/patologia , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Transdução de Sinais
11.
Med. clín (Ed. impr.) ; 145(10): 433-435, nov. 2015. tab
Artigo em Espanhol | IBECS | ID: ibc-145253

RESUMO

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)


Assuntos
Feminino , Humanos , Anorexia Nervosa/metabolismo , Anorexia Nervosa/patologia , Anemia/sangue , Anemia/metabolismo , Porfiria Eritropoética/patologia , Transtornos de Alimentação na Infância/diagnóstico , Hematologia/métodos , Anorexia Nervosa/complicações , Anorexia Nervosa/diagnóstico , Anemia/complicações , Anemia/patologia , Porfiria Eritropoética/genética , Transtornos de Alimentação na Infância/complicações , Hematologia/normas
12.
Blood ; 126(2): 257-61, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25972160

RESUMO

Congenital erythropoietic porphyria (CEP) is an autosomal recessive disorder of heme synthesis characterized by reduced activity of uroporphyrinogen III synthase and the accumulation of nonphysiologic isomer I porphyrin metabolites, resulting in ineffective erythropoiesis and devastating skin photosensitivity. Management of the disease primarily consists of supportive measures. Increased activity of 5-aminolevulinate synthase 2 (ALAS2) has been shown to adversely modify the disease phenotype. Herein, we present a patient with CEP who demonstrated a remarkable improvement in disease manifestations in the setting of iron deficiency. Hypothesizing that iron restriction improved her symptoms by decreasing ALAS2 activity and subsequent porphyrin production, we treated the patient with off-label use of deferasirox to maintain iron deficiency, with successful results. We confirmed the physiology of her response with marrow culture studies.


Assuntos
Benzoatos/uso terapêutico , Eritropoese/efeitos dos fármacos , Quelantes de Ferro/uso terapêutico , Deficiências de Ferro , Porfiria Eritropoética/tratamento farmacológico , Tolerância a Radiação/efeitos dos fármacos , Triazóis/uso terapêutico , Adulto , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células Cultivadas , Deferasirox , Evolução Fatal , Feminino , Humanos , Lactente , Luz/efeitos adversos , Uso Off-Label , Porfiria Eritropoética/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Irmãos
14.
BMJ Case Rep ; 20132013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23704443

RESUMO

On review of past 10 years medical records, we could find four typical cases of porphyria with rare ocular manifestations. Cases 1, 2 and 4 have presented with features suggestive of acute scleritis. Based on clinical, biochemical and dermatological evaluation, all these three cases were diagnosed to have congenital erythropoietic porphyria. Case 1 was initially managed with scleral patch graft which on subsequent melt was managed with double layered amniotic membrane grafting along with conjunctival advancement and lateral paramedian tarsorrhaphy in both the eyes. Cases 2 and 4 were managed conservatively with artificial tear drops and general protective measures. Case 3 was presented with multiple failed grafts due to repeated ulceration and infection. Owing to multiple failed grafts, Boston keratoprosthesis was done and the patient is doing well with stable kertaoprosthesis at the last follow-up visit.


Assuntos
Infecções Oculares/diagnóstico , Porfiria Cutânea Tardia/diagnóstico , Porfiria Cutânea Tardia/cirurgia , Porfiria Eritropoética/diagnóstico , Porfiria Eritropoética/terapia , Esclera/patologia , Esclerite/diagnóstico , Adolescente , Adulto , Âmnio/transplante , Infecções Oculares/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Porfiria Cutânea Tardia/complicações , Porfiria Cutânea Tardia/patologia , Porfiria Eritropoética/complicações , Porfiria Eritropoética/patologia , Próteses e Implantes , Esclerite/etiologia
15.
Am J Hematol ; 88(5): 343-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23417980

RESUMO

Lentiviral modification combined with ex vivo erythroid differentiation was used to stably inhibit RhAG expression, a critical component of the Rh(rhesus) membrane complex defective in the Rh(null) syndrome. The cultured red cells generated recapitulate the major alterations of native Rh(null) cells regarding antigen expression, membrane deformability, and gas transport function, providing the proof of principle for their use as model of Rh(null) syndrome and to investigate Rh complex biogenesis in human primary erythroid cells. Using this model, we were able to reveal for the first time that RhAG extinction alone is sufficient to explain ICAM-4 and CD47 loss observed on native Rh(null) RBCs. Together with the effects of RhAG forced expression in Rh(null) progenitors, this strongly strengthens the hypothesis that RhAG is critical to Rh complex formation. The strategy is also promising for diagnosis purpose in order to overcome the supply from rare blood donors and is applicable to other erythroid defects and rare phenotypes, providing models to dissect membrane biogenesis of multicomplex proteins in erythroid cells, with potential clinical applications in transfusion medicine.


Assuntos
Proteínas Sanguíneas/metabolismo , Antígeno CD47/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Eritroides/metabolismo , Doenças Genéticas Inatas/metabolismo , Glicoproteínas de Membrana/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Anemia Hemolítica Congênita/metabolismo , Anemia Hemolítica Congênita/patologia , Anemia Hipoplástica Congênita/metabolismo , Anemia Hipoplástica Congênita/patologia , Proteínas Sanguíneas/antagonistas & inibidores , Proteínas Sanguíneas/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Células Eritroides/patologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Feminino , Sangue Fetal , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Doenças Genéticas Inatas/sangue , Doenças Genéticas Inatas/patologia , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Gravidez , Interferência de RNA , RNA Interferente Pequeno , Reticulócitos/metabolismo , Reticulócitos/patologia , Sistema do Grupo Sanguíneo Rh-Hr/sangue
16.
Actas Dermosifiliogr ; 104(3): 212-9, 2013 Apr.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-22766189

RESUMO

In recent years, important advances have been made in our understanding of the genetics of porphyrias, particularly with respect to erythropoietic protoporphyria (EPP) and congenital erythropoietic porphyria (CEP), 2 forms of erythropoietic porphyria no longer considered to be monogenic. The identification of mutations in genes not previously associated with these disorders as causative factors or modulators of severity has helped to explain the presence of genotypic and phenotypic differences between patients carrying the same mutations. These advances have also led to the identification of causative genetic defects in patients who, based on molecular studies, had no mutations in the uroporphyrinogen III synthase gene UROS (in CEP) or in the ferrochelatase gene FECH (in EPP). Better understanding and characterization of the genetics of porphyrias will allow us to determine genotypic and phenotypic correlations and improve the molecular classification of these diseases, which will have both practical and prognostic implications.


Assuntos
Porfiria Eritropoética , Humanos , Fenótipo , Porfiria Eritropoética/genética , Porfiria Eritropoética/patologia
17.
Gastroenterology ; 141(4): 1509-19, 1519.e1-3, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21762662

RESUMO

BACKGROUND & AIMS: Chronic, progressive hepatobiliary disease is the most severe complication of erythropoietic protoporphyria (EPP) and can require liver transplantation, although the mechanisms that lead to liver failure are unknown. We characterized protoporphyrin-IX (PPIX)-linked hepatobiliary disease in BALB/c and C57BL/6 (Fechm1Pas) mice with mutations in ferrochelatase as models for EPP. METHODS: Fechm1Pas and wild-type (control) mice were studied at 12-14 weeks of age. PPIX was quantified; its distribution in the liver, serum levels of lipoprotein-X, liver histology, contents of bile salt and cholesterol phospholipids, and expression of genes were compared in mice of the BALB/c and C57BL/6 backgrounds. The in vitro binding affinity of PPIX for bile components was determined. RESULTS: Compared with mice of the C57BL/6 background, BALB/c Fechm1Pas mice had a more severe pattern of cholestasis, fibrosis with portoportal bridging, bile acid regurgitation, sclerosing cholangitis, and hepatolithiasis. In C57BL/6 Fechm1Pas mice, PPIX was sequestrated mainly in the cytosol of hepatocytes and Kupffer cells, whereas, in BALB/c Fechm1Pas mice, PPIX was localized within enlarged bile canaliculi. Livers of C57BL/6 Fechm1Pas mice were protected through a combination of lower efflux of PPIX and reduced synthesis and export of bile acid. CONCLUSIONS: PPIX binds to bile components and disrupts the physiologic equilibrium of phospholipids, bile acids, and cholesterol in bile. This process might be involved in pathogenesis of sclerosing cholangitis from EPP; a better understanding might improve diagnosis and development of reagents to treat or prevent liver failure in patients with EPP.


Assuntos
Colangite Esclerosante/prevenção & controle , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Porfiria Eritropoética/metabolismo , Protoporfirinas/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Colangite Esclerosante/genética , Colangite Esclerosante/metabolismo , Colangite Esclerosante/patologia , Colesterol/metabolismo , Modelos Animais de Doenças , Ferroquelatase/genética , Ferroquelatase/metabolismo , Regulação da Expressão Gênica , Genótipo , Hepatócitos/patologia , Células de Kupffer/patologia , Lipoproteína-X/sangue , Cirrose Hepática/metabolismo , Cirrose Hepática/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Fosfolipídeos/metabolismo , Mutação Puntual , Porfiria Eritropoética/complicações , Porfiria Eritropoética/genética , Porfiria Eritropoética/patologia , Índice de Gravidade de Doença
18.
Blood ; 118(6): 1443-51, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21653323

RESUMO

Mutations in the uroporphyrinogen III synthase (UROS) gene cause congenital erythropoietic porphyria (CEP), an autosomal-recessive inborn error of erythroid heme biosynthesis. Clinical features of CEP include dermatologic and hematologic abnormalities of variable severity. The discovery of a new type of erythroid porphyria, X-linked dominant protoporphyria (XLDPP), which results from increased activity of 5-aminolevulinate synthase 2 (ALAS2), the rate-controlling enzyme of erythroid heme synthesis, led us to hypothesize that the CEP phenotype may be modulated by sequence variations in the ALAS2 gene. We genotyped ALAS2 in 4 unrelated CEP patients exhibiting the same C73R/P248Q UROS genotype. The most severe of the CEP patients, a young girl, proved to be heterozygous for a novel ALAS2 mutation: c.1757 A > T in exon 11. This mutation is predicted to affect the highly conserved and penultimate C-terminal amino acid of ALAS2 (Y586). The rate of 5-aminolevulinate release from Y586F was significantly increased over that of wild-type ALAS2. The contribution of the ALAS2 gain-of-function mutation to the CEP phenotype underscores the importance of modifier genes underlying CEP. We propose that ALAS2 gene mutations should be considered not only as causative of X-linked sideroblastic anemia (XLSA) and XLDPP but may also modulate gene function in other erythropoietic disorders.


Assuntos
5-Aminolevulinato Sintetase/genética , Mutação de Sentido Incorreto , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Sequência de Aminoácidos , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Sequência de Bases , Pré-Escolar , Eletroforese em Gel de Poliacrilamida , Saúde da Família , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Genótipo , Humanos , Lactente , Cinética , Masculino , Dados de Sequência Molecular , Linhagem , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/metabolismo , Homologia de Sequência de Aminoácidos , Índice de Gravidade de Doença , Espectrofotometria , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/metabolismo
20.
Blood ; 115(5): 1062-9, 2010 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19965637

RESUMO

Splicing mutations account for approximately 10% of lesions causing genetic diseases, but few branchpoint sequence (BPS) lesions have been reported. In 3 families with autosomal recessive congenital erythropoietic porphyria (CEP) resulting from uroporphyrinogen III synthase (URO-synthase) deficiency, sequencing the promoter, all 10 exons and the intron/exon boundaries did not detect a mutation. Northern analyses of lymphoblast mRNAs from 2 patients and reverse-transcribed polymerase chain reaction (RT-PCR) of lymphoblast mRNAs from all 3 patients revealed multiple longer transcripts involving intron 9 and low levels of wild-type message. Sequencing intron 9 RT-PCR products and genomic DNA in each case revealed homozygosity for a novel BPS mutation (c.661-31T-->G) and alternatively spliced transcripts containing 81, 246, 358, and 523 nucleotides from intron 9. RT-PCR revealed aberrant transcripts in both wild-type and CEP lymphoblasts, whereas BPS mutation reduced the wild-type transcript and enzyme activity in CEP lymphoblasts to approximately 10% and 15% of normal, respectively. Although the +81-nucleotide alternative transcript was in-frame, it only contributed approximately 0.2% of the lymphoblast URO-synthase activity. Thus, the BPS mutation markedly reduced the wild-type transcript and enzyme activity, thereby causing the disease. This is the first BPS mutation in the last intron, presumably accounting for the observed 100% intron retention without exon skipping.


Assuntos
Processamento Alternativo/genética , Mutação , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , Adolescente , Adulto , Northern Blotting , Análise Mutacional de DNA , Éxons/genética , Saúde da Família , Feminino , Humanos , Íntrons/genética , Linfócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...