Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Reprod Biomed Online ; 26(5): 420-30, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23523379

RESUMO

Preimplantation genetic diagnosis (PGD) for inherited disorders is presently applied for more than 300 different conditions. The most frequent PGD indication is cystic fibrosis (CF), the largest series of which is reviewed here, totalling 404 PGD cycles. This involved testing for 52 different CFTR mutations with almost half of the cases (195/404 cycles) performed for ΔF508 mutation, one-quarter (103/404 cycles) for six other frequent mutations and only a few for the remaining 45 CFTR mutations. There were 44 PGD cycles performed for 25 CF-affected homozygous or double-heterozygous CF patients (18 male and seven female partners), which involved testing simultaneously for three mutations, resulting in birth of 13 healthy CF-free children and no misdiagnosis. PGD was also performed for six couples at a combined risk of producing offspring with CF and another genetic disorder. Concomitant testing for CFTR and other mutations resulted in birth of six healthy children, free of both CF and another genetic disorder in all but one cycle. A total of 96 PGD cycles for CF were performed with simultaneous aneuploidy testing, including microarray-based 24-chromosome analysis, as a comprehensive PGD for two or more conditions in the same biopsy material.


Assuntos
Aneuploidia , Aberrações Cromossômicas , Fibrose Cística/diagnóstico , Fibrose Cística/genética , Diagnóstico Pré-Implantação/métodos , Fibrose Cística/epidemiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Erros de Diagnóstico , Feminino , Humanos , Masculino , Mutação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Risco
2.
Reprod Biomed Online ; 24(4): 443-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22386593

RESUMO

Preimplantation genetic diagnosis (PGD) has been applied for more than 200 different inherited conditions, with expanding application to common disorders with genetic predisposition. One of the recent indications for PGD has been inherited cardiac disease, for which no preclinical diagnosis and preventive management may exist and which may lead to premature or sudden death. This paper presents the first, as far as is known, cumulative experience of PGD for inherited cardiac diseases, including familial hypertrophic and dilated cardiomyopathy, cardioencephalomyopathy and Emery-Dreifuss muscular dystrophy. A total of 18 PGD cycles were performed, resulting in transfer in 15 of them, which yielded nine unaffected pregnancies and the births of seven disease- or disease predisposition-free children. The data open the prospect of PGD for inherited cardiac diseases, allowing couples carrying cardiac disease predisposing genes to reproduce without much fear of having offspring with these genes, which are at risk for premature or sudden death. Preimplantation genetic diagnosis (PGD) is currently an established clinical procedure in assisted reproduction and genetic practices. Its application has been expanding beyond traditional indications of prenatal diagnosis and currently includes common disorders with genetic predisposition, such as inherited forms of cancer. This applies also to the diseases with no current prospect of treatment, which may manifest despite presymptomatic diagnosis and follow up, when PGD may provide the only relief for the at-risk couples to reproduce. One of the recent indications for PGD has been inherited cardiac disease, for which no preclinical diagnosis and preventive management may exist and which may lead to premature or sudden death. We present here our first cumulative experience of PGD for inherited cardiac diseases, including familial hypertrophic and dilated cardiomyopathy, cardioencephalomyopathy and Emery-Dreifuss muscular dystrophy. A total of 18 PGD cycles for these disorders was performed, resulting in transfer in 15 of them, which yielded nine unaffected pregnancies and birth of seven disease- or disease predisposition-free children. The data open the prospect of PGD for inherited cardiac diseases, allowing couples carrying cardiac disease predisposing genes to reproduce without much fear of having offspring with these genes at risk for premature or sudden death.


Assuntos
Cardiopatias/diagnóstico , Cardiopatias/genética , Diagnóstico Pré-Implantação , Adulto , Análise Mutacional de DNA , Transferência Embrionária , Feminino , Fertilização in vitro/métodos , Predisposição Genética para Doença , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Cardiopatias/prevenção & controle , Humanos , Masculino , Linhagem , Gravidez , Diagnóstico Pré-Implantação/métodos , Estudos Retrospectivos , Adulto Jovem
3.
Mol Hum Reprod ; 17(5): 275-85, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21320873

RESUMO

Introduced >20 years ago, the use of polar bodies (PBs), involving sequential removal and genetic analysis of the first (PB1) and second (PB2) PB, provides the option for pre-embryonic diagnosis, when the objection to the embryo biopsy procedures makes preimplantation genetic diagnosis (PGD) non-applicable. PB-based approach has presently been utilized in PGD for genetic and chromosomal disorders, applied either separately, or together with embryo biopsy approaches, especially if there are two or more PGD indications. We present here the world's largest experience of 938 PGD cycles for single-gene disorders performed by PB testing for 146 different monogenic conditions, which resulted in the birth of 345 healthy children (eight pregnancies are still ongoing), providing strong evidence that PB-based PGD is a reliable and safe procedure, with an extremely high accuracy rate of over 99%. With application of microarray technology, PB-based approach will be utilized for increasing number of indications, involving simultaneous testing for 24 chromosomes and single-gene disorders.


Assuntos
Transtornos Cromossômicos/diagnóstico , Transtornos Cromossômicos/genética , Cromossomos Humanos/genética , Corpos Polares/metabolismo , Diagnóstico Pré-Implantação/métodos , Blastocisto/metabolismo , Blastocisto/patologia , Blastômeros/metabolismo , Blastômeros/patologia , Transtornos Cromossômicos/embriologia , Transtornos Cromossômicos/patologia , Embrião de Mamíferos , Feminino , Fertilização in vitro , Testes Genéticos/métodos , Humanos , Recém-Nascido , Masculino , Corpos Polares/patologia , Gravidez , Diagnóstico Pré-Implantação/estatística & dados numéricos , Análise Serial de Tecidos
4.
Reprod Biomed Online ; 22(4): 350-61, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21324748

RESUMO

Standard preimplantation genetic diagnosis (PGD) cannot be applied for de-novo mutations (DNM), because neither origin nor relevant haplotypes are available for testing in single cells. PGD strategies were developed for 80 families with 38 genetic disorders, determined by 33 dominant, three recessive and two X-linked DNM. All three recessive mutations were of paternal origin, while of 93 dominant mutations, 40 were paternal, 46 maternal and seven detected in affected children. The development of specific PGD strategy for each couple involved DNA analysis of the parents and affected children prior to PGD, including a mutation verification, polymorphic marker evaluation, whole and single sperm testing to establish the normal and mutant haplotypes and PGD by polar body analysis and/or embryo biopsy. Overall, 151 PGD cycles were performed for 80 families, for which a specific PGD design has been established. The application of these protocols resulted in pre-selection and transfer of 219 (1.72 per cycle) DNM-free embryos in 127 (84.1%) PGD cycles, yielding 63 (49.6%) unaffected pregnancies and birth of 59 (46.5%) healthy children, confirmed to be free of DNM. The data show feasibility of PGD for DNM, which may routinely be performed with accuracy of over 99%, using the established PGD strategy.


Assuntos
Haplótipos/genética , Mutação/genética , Diagnóstico Pré-Implantação/métodos , Técnicas de Reprodução Assistida , Humanos , Padrões de Herança/genética , Mosaicismo , Linhagem
5.
Hemoglobin ; 35(5-6): 547-55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21910603

RESUMO

Hemoglobinopathies are the most frequent indications for preimplantation genetic diagnosis (PGD), allowing couples at-risk of bearing offspring with thalassemia and sickle cell disease to reproduce without fear of having an affected child. The present experience includes PGD for sickle cell disease, α- and ß-thalassemia (α- and ß-thal). We present here the results of the world's largest experience of over 395 PGD cycles for hemoglobin (Hb) disorders, resulting in the birth of 98 healthy, hemoglobinopathy-free children, with seven pregnancies still ongoing. One-third of these cases were performed in combination with HLA typing, allowing the birth of unaffected children who were also HLA identical to the affected siblings with hemoglobinopathies in these families, with successful or pending stem cell transplantation in a dozen of them. The results show that PGD is presently a practical approach for prevention of hemoglobinopathies, gradually also becoming a useful approach to improving access to HLA-compatible stem cell transplantation for this group of diseases.


Assuntos
Hemoglobinopatias/diagnóstico , Diagnóstico Pré-Implantação , Análise Mutacional de DNA , Feminino , Haplótipos , Teste de Histocompatibilidade , Humanos , Mutação , Linhagem , Gravidez , Resultado da Gravidez , Talassemia/diagnóstico , Globinas beta/genética
6.
Int J Gynaecol Obstet ; 150(1): 34-40, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32524593

RESUMO

The 10% rate of preterm birth rate worldwide has not been proved amenable to reduction. Avoiding multiple embryo transfer in assisted reproductive technologies (ART) using in vitro fertilization is one unassailable method. Preimplantation genetic testing (PGT) to select only a single euploid embryo for transfer is one unequivocal way, maintaining 50%-60% pregnancy rates while avoiding twins. Contemporary methodology entails trophectoderm biopsy of a 5-6-day blastocyst, and cryopreservation of biopsied embryos while awaiting analysis by next generation sequencing. Embryo biopsy is safe, analytic validity for chromosomal analysis high, and global access to PGT high.


Assuntos
Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Nascimento Prematuro/prevenção & controle , Transferência Embrionária/efeitos adversos , Transferência Embrionária/métodos , Feminino , Fertilização in vitro/efeitos adversos , Fertilização in vitro/métodos , Humanos , Gravidez
7.
J Mol Diagn ; 22(2): 220-227, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31751677

RESUMO

We investigated the potential of next-generation sequencing (NGS) as an alternative method for preimplantation genetic testing of monogenic disease (PGT-M) with human leukocyte antigen (HLA) matching and for noninvasive prenatal diagnosis follow-up. The case involved parents who were carriers of the Fanconi anemia complementation group G (FANCG) 260delG mutation. After clinical PGT using conventional short tandem repeat and mutation analysis, two euploid disease-free embryos were transferred, resulting in a twin pregnancy. Using the original embryo whole genome amplification products from 10 embryos, NGS confirmed the genotypes of the eight nontransferred embryos for both mutation status and HLA combination. NGS also confirmed that the two transferred embryos, which resulted in a twin pregnancy, were euploid, Fanconi disease free, and HLA matched to their sick sibling. At 15 weeks' gestation, noninvasive prenatal diagnosis of the maternal cell-free DNA determined fetal fractions of 14% and 6.6% for twins 1 and 2, respectively. The maternal plasma FANCG 260delG mutation ratio was measured at 46.2%, consistent with the presence of a carrier fetus and a normal fetus. These findings provide proof of concept that NGS has clinical utility as a safe and effective PGT-M method for embryo genotyping as well as more complex direct HLA matching. In addition, NGS can be used to confirm the original PGT-M and HLA matching embryo results in early pregnancy without the need for invasive prenatal diagnosis.


Assuntos
Feto , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Teste Pré-Natal não Invasivo/métodos , Diagnóstico Pré-Implantação/métodos , Análise de Célula Única/métodos , Aneuploidia , Proteína do Grupo de Complementação G da Anemia de Fanconi/genética , Feminino , Marcadores Genéticos , Testes Genéticos/métodos , Técnicas de Genotipagem , Antígenos HLA/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Teste de Histocompatibilidade , Humanos , Masculino , Teste Pré-Natal não Invasivo/normas , Gravidez , Gravidez de Gêmeos , Diagnóstico Pré-Implantação/normas
8.
Fertil Steril ; 111(1): 21-23, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30611408

RESUMO

Couples at risk for autosomal recessive congenital adrenal hyperplasia often request anticipatory guidance and genetic counseling. Initially, hormones in amniotic fluid were measured to distinguish affected female fetuses from unaffected fetuses. With the molecular era, more-targeted approaches became possible. Prenatal genetic diagnosis via amniocentesis or chorionic villus sampling was used to determine the need for continuing fetal therapy (dexamethasone), allowing cessation if the fetus was unaffected. Newer methods now allow diagnosis earlier in gestation, further shortening the treatment time for unaffected female fetuses who will not develop genital ambiguity. Preimplantation genetic testing permits transfer only of an unaffected female or male fetus. Analysis of maternal cell-free DNA based on quantitative differences in the amount of allele parental DNA permits affected pregnancies to be differentiated from unaffected pregnancies.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Hiperplasia Suprarrenal Congênita/terapia , Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Diagnóstico Pré-Natal/métodos , Hiperplasia Suprarrenal Congênita/diagnóstico , Transferência Embrionária/métodos , Feminino , Aconselhamento Genético/métodos , Humanos , Masculino , Gravidez
9.
Fertil Steril ; 112(4): 622-630, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31561864

RESUMO

Disorders of genetic etiology exist in 2%-3% of live-born infants. Identifying couples with increased susceptibility for offspring with anomalies or genetic disorders is increasingly effective as a result of molecular advances. Preimplantation genetic testing (PGT) with the use of trophectoderm biopsy, 24-chromosome testing, and molecular testing have allowed wider applicability for avoiding a clinical pregnancy termination. Cell-free DNA in maternal blood is another targeted option, although invasive prenatal genetic diagnosis provides the greatest amount of genetic information. DNA-based methods to detect subtle chromosomal abnormalities are much more sensitive than traditional karyotypes and do not require cultured cells. Aneuploidy and structural chromosomal abnormalities can be readily detected with the use of small amounts of DNA, if necessary amplified, as in PGT. Novel approaches exist for detecting perturbations in single-gene disorders. Not only has the molecular basis for many monogenic disorders been elucidated, but modest costs for DNA sequencing has made testing feasible. As the number of testable genetic disorders has increased, principles underlying screening have advanced. Genetic screening for disorders of high incidence in certain ethnic groups was initiated decades ago; however, limitations exist, and reduction in live-born incidence is not infrequently small. Expanded carrier screening is now offered in panethnic fashion, extending surveillance to couples of mixed ethnicities and involving many more genetic conditions. Targeted gene panels (e.g., adult-onset cancer genes) further increase the number of genetic disorders amenable to screening, often leading to PGT.


Assuntos
Aberrações Cromossômicas , Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Aneuploidia , Variações do Número de Cópias de DNA , Feminino , Triagem de Portadores Genéticos , Humanos , Infertilidade/genética , Idade Materna , Gravidez , Translocação Genética
10.
Methods Mol Biol ; 1885: 23-43, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30506188

RESUMO

Preimplantation genetic diagnosis (PGD) can be considered the earliest form of prenatal testing. It was first used in humans over 26 years ago. At its inception, PGD could only be performed for a limited number of genetic disorders. Technological advances in molecular biology and cytogenomics have been utilized in the field of PGD to greatly expand the spectrum of genetic disorders that can now be detected in early human embryos.


Assuntos
Testes Genéticos/métodos , Diagnóstico Pré-Implantação/métodos , Aborto Habitual/genética , Aneuploidia , Biópsia , Blastocisto/citologia , Blastocisto/metabolismo , Aberrações Cromossômicas , Criopreservação , Desenvolvimento Embrionário/genética , Feminino , Estudos de Associação Genética/métodos , Marcadores Genéticos , Predisposição Genética para Doença , Humanos , Gravidez , Taxa de Gravidez , Translocação Genética
11.
Expert Rev Mol Diagn ; 17(12): 1071-1088, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29039978

RESUMO

INTRODUCTION: Preimplantation genetic testing (PGT) is now a widely applied procedure in genetic practices and ART, with more than one third of ART Centers in US already utilizing PGT technology. Its indications have also been significantly extended to include common late-onset disorders and non-genetic conditions, such as testing for HLA match. Areas covered: This is a critical review of the developments in PGT, with emphasis on their outstanding limitations and directions for the future research and practice in the area of PGT. Expert commentary: The application of the new higher resolution PGT technologies has led to the identification of genetic variations, the biological and clinical importance of which is not sufficiently understood. It is obvious that the current selection process of embryos with the highest developmental potential requires a further improvement, as significant proportion of transferred euploid embryos still fail to result in an ongoing clinical pregnancy. More research will be needed to upgrade PGT for different conditions into a single universal test in the same biopsy material. To avoid a potential damage of embryo biopsy procedures, one of the important challenges will be the development of non-invasive approaches to PGT.


Assuntos
Testes Genéticos/tendências , Diagnóstico Pré-Implantação/tendências , Técnicas de Reprodução Assistida/tendências , Aneuploidia , Biópsia , Feminino , Testes Genéticos/métodos , Variação Genética/genética , Humanos , Gravidez , Diagnóstico Pré-Implantação/métodos
12.
J Steroid Biochem Mol Biol ; 165(Pt A): 124-130, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27108943

RESUMO

Prenatal treatment of congenital adrenal hyperplasia (CAH) has long involved prenatal treatment with dexamethasone, administered to the pregnant woman to prevent genital masculinization of an affected female fetus. Although it is unnecessary to treat unaffected or affected males because their genital development would not be disturbed, there has only been incremental progress in determining fetal gender sufficiently each to avoid treating males and unaffected females. Invasive procedures were initially necessary, with first-trimester amniocentesis at 15-20 weeks and then chorionic villus sampling (CVS) at 10-12 weeks gestation. Two approaches now allow personalized treatment of affected female fetuses prior to female genital differentiation. Only preimplantation genetic diagnosis (PGD) is available prior to clinical pregnancy. Recent technological advances have further allowed both single gene diagnosis (e.g., CAH) and aneuploidy detection concomitantly, resulting in far better pregnancy rates than heretofore possible in assisted reproduction technology.


Assuntos
Hiperplasia Suprarrenal Congênita/genética , Diagnóstico Pré-Implantação/métodos , Diagnóstico Pré-Natal/métodos , Hiperplasia Suprarrenal Congênita/diagnóstico , Amniocentese/métodos , Aneuploidia , Biópsia , Blastocisto/citologia , Vilosidades Coriônicas/metabolismo , Fase de Clivagem do Zigoto , Criopreservação , Dexametasona/uso terapêutico , Feminino , Sangue Fetal/citologia , Genótipo , Antígenos HLA/metabolismo , Humanos , Masculino , Oócitos/citologia , Gravidez , Diferenciação Sexual , Transplante de Células-Tronco , Células-Tronco/citologia
13.
Ann N Y Acad Sci ; 1054: 223-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16339669

RESUMO

There has been progress in the application of stem cell transplantation for treatment of an increasing number of severe congenital and acquired bone marrow disorders, currently restricted by the availability of human leukocyte antigen (HLA)-matched related donors. Preimplantation HLA typing has recently been introduced to improve the access to stem cell therapy for inherited bone marrow failures. Preimplantation genetic diagnosis (PGD) provides an option not only for avoiding an affected pregnancy with thalassemia and other inherited disorders but also for preselection of the HLA-compatible donors for affected siblings. Multiple short tandem repeat markers throughout the HLA region are applied for this purpose, allowing 100% accuracy of HLA typing, through picking up possible recombination in the HLA region, as well as the copy number of chromosome 6, which affect accuracy of preimplantation HLA typing. Present experience of preimplantation HLA typing includes preimplantation HLA typing in 180 cycles, 122 of which were done as part of PGD for Fanconi anemia, thalassemia, Wiscott-Aldrich syndrome, hyper-immunoglobulin M syndrome, hypohidrotic ectodermal dysplasia with immune deficiency, and X-linked adrenoleukodystrophy, and 58 for the sole purpose of HLA typing for leukemias and for aplastic and Diamond-Blackfan anemia. The applied method resulted in the accurate preselection and transfer of 100% HLA-matched embryos, yielding already three dozen clinical pregnancies and the birth of two dozen HLA-matched children to the siblings requiring stem cell transplantation. Successful therapy with HLA-matched stem cells, obtained from these PGD children, has been achieved already for Diamond-Blackfan anemia hypohidrotic ectodermal dysplasia with immune deficiency and thalassemia.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Doenças Genéticas Inatas/genética , Engenharia Genética/métodos , Antígenos HLA/genética , Teste de Histocompatibilidade/métodos , Diagnóstico Pré-Implantação , Obtenção de Tecidos e Órgãos/métodos , Anemia de Diamond-Blackfan/diagnóstico , Anemia de Diamond-Blackfan/embriologia , Anemia de Diamond-Blackfan/prevenção & controle , Anemia de Diamond-Blackfan/cirurgia , Blastocisto , Bancos de Sangue , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/embriologia , Displasia Ectodérmica/prevenção & controle , Displasia Ectodérmica/cirurgia , Transferência Embrionária , Fertilização in vitro , Doenças Fetais/diagnóstico , Doenças Genéticas Inatas/embriologia , Doenças Genéticas Inatas/prevenção & controle , Doenças Genéticas Inatas/cirurgia , Engenharia Genética/ética , Humanos , Diagnóstico Pré-Implantação/ética , Irmãos , Sequências de Repetição em Tandem , Talassemia/embriologia , Talassemia/genética , Talassemia/prevenção & controle , Talassemia/cirurgia , Obtenção de Tecidos e Órgãos/ética
14.
Fertil Steril ; 103(2): 503-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25516085

RESUMO

OBJECTIVE: To study the feasibility, accuracy, and reproductive outcome of 24-chromosome aneuploidy testing (24-AT), combined with preimplantation genetic diagnosis (PGD) for single-gene disorders (SGDs) or human leukocyte antigen (HLA) typing in the same biopsy sample. DESIGN: Retrospective study. SETTING: Preimplantation genetic diagnosis center. PATIENT(S): A total of 238 PGD patients, average age 36.8 years, for whom 317 combined PGD cycles were performed, involving 105 different conditions, with or without HLA typing. INTERVENTION(S): Whole-genome amplification product, obtained in 24-AT, was used for PGD and/or HLA typing in the same blastomere or blastocyst biopsy samples. MAIN OUTCOME MEASURE(S): Proportion of the embryos suitable for transfer detected in these blastomere or blastocyst samples, and the resulting pregnancy and spontaneous abortion rates. RESULT(S): Embryos suitable for transfer were detected in 42% blastocyst and 25.1% blastomere samples, with a total of 280 unaffected, HLA-matched euploid embryos detected for transfer in 212 cycles (1.3 embryos per transfer), resulting in 145 (68.4%) unaffected pregnancies and birth of 149 healthy, HLA-matched children. This outcome is significantly different from that of our 2,064 PGD cycle series without concomitant 24-AT, including improved pregnancy (68.4% vs. 45.4%) and 3-fold spontaneous abortion reduction (5.5% vs. 15%) rates. CONCLUSION(S): The introduced combined approach is a potential universal PGD test, which in addition to achieving extremely high diagnostic accuracy, significantly improves reproductive outcomes of PGD for SGDs and HLA typing in patients of advanced reproductive age.


Assuntos
Aneuploidia , Testes Genéticos/métodos , Antígenos HLA/genética , Diagnóstico Pré-Implantação/métodos , Adulto , Feminino , Humanos , Gravidez , Estudos Retrospectivos
15.
Reprod Biomed Online ; 2(1): 17-19, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12537820

RESUMO

Long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency is a fatal autosomal recessive metabolic disorder, presenting during infancy. Preimplantation genetic diagnosis (PGD) provides an option for establishing an unaffected pregnancy, avoiding the risk for termination of pregnancy following prenatal diagnosis. The method for pre-selection of mutation-free oocytes for LCHAD deficiency was developed by testing the first and second polar body removed from oocytes by micromanipulation techniques in the framework of in-vitro fertilization. To avoid misdiagnosis, testing was done using hemi-nested polymerase chain reaction (PCR), with outer primers designed to lie outside the pseudogene, eliminating false priming. Four of 12 tested oocytes were predicted to be unaffected, based on the heterozygous first and mutant second polar body. The embryos resulting from these mutation-free oocytes were replaced, yielding a singleton clinical pregnancy and birth of a healthy child following confirmation by prenatal diagnosis.

16.
Reprod Biomed Online ; 2(2): 102-105, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12537806

RESUMO

Preimplantation genetic diagnosis (PGD) was introduced for high-risk couples to avoid establishing affected pregnancies potentially requiring termination following prenatal diagnosis. This opens the possibility for PGD for late onset disorders with genetic predisposition, including inherited cancer predisposition, because only embryos free from the predisposing gene may be transferred back to the patient, with no potential risk for pregnancy termination. PGD was performed for two couples, one with maternally and one with paternally derived p53 tumour-suppressor mutations, 902insC in exon 8 and G524A in exon 5, respectively. This involved a standard IVF protocol, allowing oocytes or embryos to be tested prior to their transfer back to uterus. Maternal mutation was tested by sequential PCR analysis of the first and second polar bodies, removed following maturation and fertilization of oocytes, while paternal mutation analysis required embryo biopsy at the cleavage stage. To avoid misdiagnosis due to allele drop out, multiplex nested PCR was applied, involving p53 mutation analysis simultaneously with the linked short tandem repeats in intron 1. Of 10 oocytes tested in two PGD cycles for 902insC mutation, four unaffected oocytes were pre-selected for transfer yielding no clinical pregnancy. Of 18 embryos analysed in two cycles for G524A mutation, seven mutation-free embryos were detected, two of which were transferred in each cycle, resulting in a singleton pregnancy and birth of a mutation-free child. This is the first PGD for inherited cancer predisposition determined by p53 tumour suppressor mutations, resulting in a clinical pregnancy and birth of a child free from inherited cancer predisposition.

17.
Fertil Steril ; 80(4): 1047-51, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14556832

RESUMO

OBJECTIVE: To use preimplantation genetic diagnosis (PGD) to achieve a Kell 1 (K1) allele-free pregnancy in couples at risk for producing a child with hemolytic disease of the newborn (HDN) caused by maternofetal incompatibility in sensitized mothers. DESIGN: DNA analysis of biopsied blastomeres from cleavage-stage embryos in IVF-ET with the goal of identifying and transferring back to patients the K1 allele-free embryos. SETTING: IVF program at the Reproductive Genetics Institute, Chicago, Illinois, and IVF Michigan, Rochester Hills, Michigan. PATIENT(S): Two at-risk couples with a history of neonatal death caused by HDN due to K1/K2 genotype in a male partner. INTERVENTION(S): Biopsy of single blastomeres and testing for paternal K1 allele in each embryo after standard IVF. MAIN OUTCOME MEASURE(S): DNA analysis of blastomeres indicating whether corresponding embryos were K1 allele-free for the purpose of transferring only embryos without the K1 allele. RESULT(S): Of 36 embryos tested in five cycles from two couples, 18 were predicted to be K1 allele-free. Of these, 9 were transferred, resulting in a K1 allele-free twin pregnancy and the birth of two healthy children. CONCLUSION(S): PGD of the K1 genotype resulted in the birth of healthy twins confirmed to be free of the K1 allele. PGD in couples with a heterozygous K1/K2 male partner provides an option for avoiding HDN in sensitized mothers.


Assuntos
Sistema do Grupo Sanguíneo de Kell/genética , Diagnóstico Pré-Implantação , Adulto , Alelos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Frequência do Gene , Ligação Genética , Genótipo , Humanos , Recém-Nascido , Íntrons/genética , Masculino , Parto , Linhagem , Gravidez , Gravidez Múltipla , Gêmeos
18.
Fertil Steril ; 82(4): 926-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15482771

RESUMO

OBJECTIVE: To use preimplantation genetic diagnosis for achieving a polycystic kidney disease (PKD)-free pregnancy for a couple in which the female partner was affected by PKD but whose PKD1 or PKD2 carrier status was not established. DESIGN: Case report. SETTING: The IVF program of Reproductive Genetics Institute, Chicago, Illinois. PATIENT(S): An at-risk couple with the female partner affected by PKD, whose PKD1 or PKD2 carrier status was not established. INTERVENTION(S): Removal of PB1 and PB2 and testing for three closely linked markers to PKD1 (Kg8, D16S664, and SM7) and four closely linked markers to PKD2 (D4S2922, D4S2458, D4S423, and D4S1557) after standard IVF. MAIN OUTCOME MEASURE(S): Deoxyribonucleic acid analysis of PB1 and PB2 indicating whether corresponding oocytes were PKD1 or PKD2 allele free, for the purpose of transferring only embryos resulting from mutation-free oocytes. RESULT(S): Of 11 oocytes tested by PB1 and PB2 DNA analysis, 7 were predicted to contain PKD1 or PKD2, with the remaining 4 free of both mutations. Three embryos resulting from these oocytes were transferred, yielding a twin pregnancy and the birth of two unaffected children. CONCLUSION(S): This is the first preimplantation genetic diagnosis for PKD, which resulted in the birth of healthy twins confirmed to be free of PKD1 and PKD2. Preimplantation genetic diagnosis based on linked marker analysis provides an alternative for avoiding the pregnancy and birth of children with PKD, even in at-risk couples without exact PKD1 or PKD2 carrier information.


Assuntos
Doenças Renais Policísticas/diagnóstico , Doenças Renais Policísticas/genética , Diagnóstico Pré-Implantação/métodos , Mapeamento Cromossômico/métodos , Implantação do Embrião , Feminino , Marcadores Genéticos , Humanos , Recém-Nascido , Masculino , Proteínas de Membrana/genética , Gravidez , Proteínas/genética , Canais de Cátion TRPP , Gêmeos/genética
19.
JAMA ; 291(17): 2079-85, 2004 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15126435

RESUMO

CONTEXT: Preimplantation genetic diagnosis (PGD) has become an option for couples for whom termination of an affected pregnancy identified by traditional prenatal diagnosis is unacceptable and is applicable to indications beyond those of prenatal diagnosis, such as HLA matching to affected siblings to provide stem cell transplantation. OBJECTIVE: To describe preimplantation HLA typing, not involving identification of a causative gene, for couples who had children with bone marrow disorders at need for HLA-matched stem cell transplantation. DESIGN, SETTING, AND PARTICIPANTS: HLA matching procedures conducted at a single site during 2002-2003 in an in vitro fertilization program for 9 couples with children affected by acute lymphoid leukemia, acute myeloid leukemia, or Diamond-Blackfan anemia requiring HLA-matched stem cell transplantation. In 13 clinical cycles, DNA in single blastomeres removed from 8-cell embryos following in vitro fertilization was analyzed for HLA genes simultaneously with analysis for short tandem repeats in the HLA region to select and transfer only those embryos that were HLA matched to affected siblings. MAIN OUTCOME MEASURES: Results of HLA matching and pregnancy outcome. RESULTS: As a result of testing a total of 199 embryos, 45 (23%) HLA-matched embryos were selected, of which 28 were transferred in 12 clinical cycles, resulting in 5 singleton pregnancies and birth of 5 HLA-matched healthy children. CONCLUSION: This is the first known experience of preimplantation HLA typing performed without PGD for a causative gene, providing couples with a realistic option of having HLA-matched offspring to serve as potential donors of stem cells for their affected siblings.


Assuntos
Doação Dirigida de Tecido , Desenvolvimento Embrionário , Teste de Histocompatibilidade , Transplante de Células-Tronco , Anemia de Diamond-Blackfan/terapia , Feminino , Fertilização in vitro , Antígenos HLA , Humanos , Lactente , Leucemia Linfoide/terapia , Leucemia Mieloide/terapia , Gravidez , Resultado da Gravidez , Irmãos
20.
JAMA Neurol ; 71(4): 484-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24493558

RESUMO

IMPORTANCE: To describe the first case of preimplantation genetic diagnosis (PGD) and in vitro fertilization (IVF) performed for the prevention of genetic prion disease in the children of a 27-year-old asymptomatic woman with a family history of Gerstmann-Sträussler-Sheinker syndrome (GSS). OBSERVATIONS: PGD and fertilization cycles resulted in detection of 6 F198S mutation-free embryos. Of these, 2 were selected for embryo transfer to the patient's uterus, yielding a clinical twin pregnancy and birth of healthy but slightly premature offspring with normal development at age 27 months. CONCLUSION AND RELEVANCE: IVF with PGD is a viable option for couples who wish to avoid passing the disease to their offspring. Neurologists should be aware of PGD to be able to better consult at-risk families on their reproductive choices.


Assuntos
Testes Genéticos/métodos , Mutação/genética , Diagnóstico Pré-Implantação/métodos , Doenças Priônicas/diagnóstico , Doenças Priônicas/genética , Príons/genética , Adulto , Doenças em Gêmeos/diagnóstico , Doenças em Gêmeos/genética , Doenças em Gêmeos/prevenção & controle , Feminino , Seguimentos , Humanos , Recém-Nascido , Gravidez , Doenças Priônicas/prevenção & controle , Proteínas Priônicas , Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA