Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Hum Reprod ; 26(7): 469-484, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32402059

RESUMO

Idiopathic azoospermia (IA) refers to azoospermia without a clear aetiology. Due to the unclear aetiology and pathological mechanism of IA, there is no effective treatment for IA. The development of assisted reproductive and microsperm extraction technologies has brought hope to patients with IA with fertility problems. However, there are still many patients with IA whose testes lack healthy sperm, causing infertility. Therefore, it is key to identify how testicular spermatogenic failure can be reversed to promote spermatogenesis in patients with IA to resolve fertility problems; these goals are a great challenge in reproductive medicine. The underlying genetic factors seem to be important pathological factors of IA. Understanding the role of genetic factors in the pathological mechanism of spermatogenic failure in patients with IA is of great value for future studies and treatments and is also an important reference for the reproductive health of males and their offspring. A method combining sequencing technology and bioinformatics analysis is an important means to understand the genetic pathological mechanisms. We used bioinformatics analysis to study the public human IA dataset. We found that the pathogenic mechanism of IA may be related to abnormal ciliary structure and function and disrupted RNA metabolism in spermatogenic cells. Disrupted m6A regulation of spermatogenesis may be an important pathological mechanism of IA and warrants attention. Finally, we screened for key genes and potential therapeutic drugs to determine future research directions.


Assuntos
Azoospermia/metabolismo , Cromossomos Humanos Y/fisiologia , Infertilidade Masculina/metabolismo , Infertilidade Masculina/fisiopatologia , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/fisiopatologia , Cromossomos Humanos Y/metabolismo , Biologia Computacional , Humanos , Masculino , Locos de Características Quantitativas/genética , Espermatogênese/genética , Espermatozoides/fisiologia
2.
PLoS Genet ; 15(9): e1008369, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31525193

RESUMO

The Y chromosome harbors nine multi-copy ampliconic gene families expressed exclusively in testis. The gene copies within each family are >99% identical to each other, which poses a major challenge in evaluating their copy number. Recent studies demonstrated high variation in Y ampliconic gene copy number among humans. However, how this variation affects expression levels in human testis remains understudied. Here we developed a novel computational tool Ampliconic Copy Number Estimator (AmpliCoNE) that utilizes read sequencing depth information to estimate Y ampliconic gene copy number per family. We applied this tool to whole-genome sequencing data of 149 men with matched testis expression data whose samples are part of the Genotype-Tissue Expression (GTEx) project. We found that the Y ampliconic gene families with low copy number in humans were deleted or pseudogenized in non-human great apes, suggesting relaxation of functional constraints. Among the Y ampliconic gene families, higher copy number leads to higher expression. Within the Y ampliconic gene families, copy number does not influence gene expression, rather a high tolerance for variation in gene expression was observed in testis of presumably healthy men. No differences in gene expression levels were found among major Y haplogroups. Age positively correlated with expression levels of the HSFY and PRY gene families in the African subhaplogroup E1b, but not in the European subhaplogroups R1b and I1. We also found that expression of five Y ampliconic gene families is coordinated with that of their non-Y (i.e. X or autosomal) homologs. Indeed, five ampliconic gene families had consistently lower expression levels when compared to their non-Y homologs suggesting dosage regulation, while the HSFY family had higher expression levels than its X homolog and thus lacked dosage regulation.


Assuntos
Cromossomos Humanos Y/genética , Genes Ligados ao Cromossomo Y/genética , Análise de Sequência de DNA/métodos , Animais , Cromossomos Humanos Y/fisiologia , Variações do Número de Cópias de DNA/genética , Bases de Dados Genéticas , Mecanismo Genético de Compensação de Dose/genética , Mecanismo Genético de Compensação de Dose/fisiologia , Epigênese Genética/genética , Dosagem de Genes/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Genes Ligados ao Cromossomo Y/fisiologia , Fatores de Transcrição de Choque Térmico/genética , Fatores de Transcrição de Choque Térmico/metabolismo , Humanos , Masculino , Família Multigênica/genética , Testículo/metabolismo
3.
Clin Interv Aging ; 14: 1227-1241, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413553

RESUMO

PURPOSE: Abdominal aortic aneurysm (AAA) demonstrates many features of autoimmune diseases. Y chromosome, sex-determining region of the Y chromosome (SRY) gene, androgen receptor (AR) gene, and androgen appear as potential candidates for influence of the male immune function. This study investigated Y chromosome numbers, SRY gene, AR gene, and androgen levels in male AAAs. We also investigated the correlation between Y chromosome loss (LOY) ratio, SRY expression, androgen levels, and age. PATIENTS AND METHODS: We investigated LOY by fluorescence in situ hybridization (FISH) in 37 AAAs and compared with 12 patients with abdominal aortic atherosclerotic occlusive disease (AOD) and 91 healthy controls (HC). We investigated SRY and AR expression at mRNA level by real-time PCR in peripheral T lymphocytes in AAA compared with AOD and HC, and AR protein levels by immunohistochemistry (IHC) in AAA. LOY, SRY expression, androgen levels, and age were examined for correlations using the Spearman's rank correlation coefficient. RESULTS: LOY ratio in peripheral T lymphocytes was significantly higher in the AAA group compared with the HC (9.11% vs 5.56%, P<0.001) and AOD groups (9.11% vs 6.42%, P=0.029). The SRY mRNA expression in peripheral T lymphocytes was 4.7-fold lower expressed in the AAA group than in the HC group (P<0.001). Free plasma testosterone levels were lower in the AAA group compared with the HC group (P=0.036), whereas sex hormone-binding globulin levels were higher (P=0.020). LOY ratio and expression of SRY mRNA level increased with age in the AAA group (R=0.402 and, R=0.366, respectively). A significant correlation between AR mRNA level (R=0.692) and aortic diameter was detected. Simultaneously, in AAA tissue, the rate of LOY increased with age (R=0.547) and also positively associated with LOY in peripheral blood T lymphocytes (R=0.661). CONCLUSION: This study identified a prominent Y chromosome loss in male AAAs, which is correlated to age, lower level of SRY expression and free testosterone, providing a new clue for the mechanisms of AAA.


Assuntos
Androgênios/sangue , Aneurisma da Aorta Abdominal/epidemiologia , Dissecção Aórtica/epidemiologia , Cromossomos Humanos Y/fisiologia , Receptores Androgênicos/genética , Proteína da Região Y Determinante do Sexo/genética , Fatores Etários , Idoso , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , RNA Mensageiro , Aberrações dos Cromossomos Sexuais
4.
J Proteome Res ; 16(12): 4259-4272, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28914051

RESUMO

One of the main goals of Chromosome-Centric Human Proteome Project is to identify protein evidence for missing proteins (MPs). Here, we present a case study of the role of Y chromosome genes in organ development and how to overcome the challenges facing MPs identification by employing human pluripotent stem cell differentiation into cells of different organs yielding unprecedented biological insight into adult silenced proteins. Y chromosome is a male-specific sex chromosome which escapes meiotic recombination. From an evolutionary perspective, Y chromosome has preserved 3% of ancestral genes compared to 98% preservation of the X chromosome based on Ohno's law. Male specific region of Y chromosome (MSY) contains genes that contribute to central dogma and govern the expression of various targets throughout the genome. One of the most well-known functions of MSY genes is to decide the male-specific characteristics including sex, testis formation, and spermatogenesis, which are majorly formed by ampliconic gene families. Beyond its role in sex-specific gonad development, MSY genes in coexpression with their X counterparts, as single copy and broadly expressed genes, inhibit haplolethality and play a key role in embryogenesis. The role of X-Y related gene mutations in the development of hereditary syndromes suggests an essential contribution of sex chromosome genes to development. MSY genes, solely and independent of their X counterparts and/or in association with sex hormones, have a considerable impact on organ development. In this Review, we present major recent findings on the contribution of MSY genes to gonad formation, spermatogenesis, and the brain, heart, and kidney development and discuss how Y chromosome proteome project may exploit developmental biology to find missing proteins.


Assuntos
Cromossomos Humanos Y/fisiologia , Biologia do Desenvolvimento , Organogênese/genética , Humanos , Proteoma/genética
6.
Ann N Y Acad Sci ; 1317: 1-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24739020

RESUMO

Sexual dimorphism is a well-documented phenomenon that is observed at all levels of the animal kingdom. Historically, sex hormones (testosterone and estrogen) have been implicated as key players in a wide array of pathologies displaying sexual dimorphism in their etiology and progression. While these hormones clearly contribute to sexually dimorphic diseases, other factors may be involved in this phenomenon as well. In particular, the stress hormone cortisol exerts differential effects in both males and females. The underlying molecular basis for the sexually dimorphic actions of glucocorticoids is unknown but clearly important to understand, since synthetic glucocorticoids are the most widely prescribed medication for the treatment of chronic inflammatory diseases and hematological cancers in humans.


Assuntos
Glucocorticoides/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Caracteres Sexuais , Animais , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Feminino , Humanos , Masculino , Receptores de Glucocorticoides/metabolismo
7.
Beijing Da Xue Xue Bao Yi Xue Ban ; 46(2): 211-4, 2014 Apr 18.
Artigo em Chinês | MEDLINE | ID: mdl-24743808

RESUMO

OBJECTIVE: To investigate the effects of big and bit Y chromosome configurations on male fertility and to evaluate the relevant clinical significance. METHODS: The relevant cases were divided into A and B groups. Group A included male infertile cases. Group B included cases whose wives had adverse pregnancy history or the abnormal amniotic fluid punctures. The cytogenetics of the patients were examined by culturing peripheral-blood lymphocytes and G-banding technology, and karyotyping analysis techniques were used to study the big and bit Y chromosomes in the two different groups. RESULTS: Among 2 139 cases, 98 cases were found with abnormal karyotype of big and bit Y chromosomes. There was no significant difference in the abnormal rate of the length variation of the Y chromosomal karyotypes between the male infertility group and the adverse pregnancy outcome group. In the male infertile group (group A), there was no significant difference in the abnormal rate between the big Y chromosome and the bit Y chromosome. In the group with adverse pregnancy outcomes (group B), the abnormal rate of the big Y chromosome karyotyping was significantly higher than that of the bit Y chromosome karyotyping. The main clinical effects of groups A and B were azoospermia, oligozoospermia, poor spermia, abortion, embryonic diapause and fetal anomalies, etc . CONCLUSION: The big and bit Y chromosomal abnormality results in not only the male infertility directly, but also an important and continuous reason of adverse pregnancy outcomes, of which the detailed mechanism needs to be further investigated.


Assuntos
Azoospermia , Cromossomos Humanos Y/fisiologia , Infertilidade Masculina , Oligospermia , Aborto Espontâneo , Feminino , Humanos , Cariotipagem , Masculino , Gravidez , Resultado da Gravidez , Espermatozoides
8.
Atherosclerosis ; 233(1): 160-4, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24529138

RESUMO

OBJECTIVE: Amongst middle-aged men, haplogroup I is associated with ≈ 50% higher risk of coronary artery disease than other paternal lineages of Y chromosome. We hypothesised that carriers of haplogroup I had higher levels of aggression and estrogens and/or lower levels of androgens early in life and thus might be more prone to cardiovascular disease than men with other lineages of Y chromosome. METHODS: We reconstructed phylogenetic tree of the Y chromosome in >1000 young apparently healthy white men from the general population. Each Y chromosome was classified into one of 13 most common European lineages. Androgens (DHEA-S, androstenedione, total testosterone) and their metabolites (total estradiol, estrone) were measured by radioimmunoassays. Information on five dimensions of aggression (total, physical, verbal, anger and hostility) was collected using Buss and Perry questionnaire. RESULTS: Approximately 17% men inherited haplogroup I from their fathers. Carriers of haplogroup I showed lower scores of verbal aggression than men with other haplogroups (ß = -0.72, SE = 0.29, P = 0.012) and when further compared to carriers of most common R1a lineage and other haplogroups (ß = -1.03, SE = 0.34, P = 0.003). However, these associations did not survive a correction for multiple testing. Sex steroids did not show even nominal level of association with haplogroup I. CONCLUSION: Our data show no overall association between haplogroup I and sex-related phenotypes in young white men. These results also suggest that the previously identified association between haplogroup I and coronary artery disease is not likely mediated by unfavourable profile of sex steroids or heightened aggression early in life.


Assuntos
Cromossomos Humanos Y/genética , Cromossomos Humanos Y/fisiologia , Doença da Artéria Coronariana/genética , Haplótipos , Agressão , Hormônios Esteroides Gonadais/genética , Humanos , Masculino , Filogenia , População Branca/genética , Adulto Jovem
9.
PLoS One ; 7(2): e31485, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22355370

RESUMO

In mammalian male gametogenesis the sex chromosomes are distinctive in both gene activity and epigenetic strategy. At first meiotic prophase the heteromorphic X and Y chromosomes are placed in a separate chromatin domain called the XY body. In this process, X,Y chromatin becomes highly phosphorylated at S139 of H2AX leading to the repression of gonosomal genes, a process known as meiotic sex chromosome inactivation (MSCI), which has been studied best in mice. Post-meiotically this repression is largely maintained. Disturbance of MSCI in mice leads to harmful X,Y gene expression, eventuating in spermatocyte death and sperm heterogeneity. Sperm heterogeneity is a characteristic of the human male. For this reason we were interested in the efficiency of MSCI in human primary spermatocytes. We investigated MSCI in pachytene spermatocytes of seven probands: four infertile men and three fertile controls, using direct and indirect in situ methods. A considerable degree of variation in the degree of MSCI was detected, both between and within probands. Moreover, in post-meiotic stages this variation was observed as well, indicating survival of spermatocytes with incompletely inactivated sex chromosomes. Furthermore, we investigated the presence of H3K9me3 posttranslational modifications on the X and Y chromatin. Contrary to constitutive centromeric heterochromatin, this heterochromatin marker did not specifically accumulate on the XY body, with the exception of the heterochromatic part of the Y chromosome. This may reflect the lower degree of MSCI in man compared to mouse. These results point at relaxation of MSCI, which can be explained by genetic changes in sex chromosome composition during evolution and candidates as a mechanism behind human sperm heterogeneity.


Assuntos
Instabilidade Cromossômica , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Meiose/fisiologia , Cromatina Sexual/genética , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Estudos de Casos e Controles , Histonas/metabolismo , Humanos , Hibridização in Situ Fluorescente , Masculino , Espermatócitos/citologia , Testículo/citologia , Testículo/metabolismo
10.
Br J Sports Med ; 46(8): 614-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21540190

RESUMO

Based on DNA analysis of a historical case, the authors describe how a female athlete can be unknowingly confronted with the consequences of a disorder of sex development resulting in hyperandrogenism emerging early in her sports career. In such a situation, it is harmful and confusing to question sex and gender. Exposure to either a low or high level of endogenous testosterone from puberty is a decisive factor with respect to sexual dimorphism of physical performance. Yet, measurement of testosterone is not the means by which questions of an athlete's eligibility to compete with either women or men are resolved. The authors discuss that it might be justifiable to use the circulating testosterone level as an endocrinological parameter, to try to arrive at an objective criterion in evaluating what separates women and men in sports competitions, which could prevent the initiation of complicated, lengthy and damaging sex and gender verification procedures.


Assuntos
Desempenho Atlético/fisiologia , Transtornos do Desenvolvimento Sexual/diagnóstico , Análise para Determinação do Sexo/métodos , Desempenho Atlético/história , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Transtornos do Desenvolvimento Sexual/sangue , Transtornos do Desenvolvimento Sexual/genética , Transtornos do Desenvolvimento Sexual/história , Feminino , Identidade de Gênero , História do Século XX , Humanos , Masculino , Mosaicismo , Países Baixos , Caracteres Sexuais , Análise para Determinação do Sexo/história , Desenvolvimento Sexual/fisiologia , Testosterona/sangue
11.
Sex Transm Dis ; 37(1): 1-4, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20118672

RESUMO

Self-reported sexual behaviors are subject to bias. We previously developed a polymerase chain reaction for the detection of Y-chromosome sequences in vaginal fluid as a potential biomarker of recent sexual activity. In this study, we found menses results in lower Y-chromosome concentrations but with similar decay patterns as non-menstrual samples.


Assuntos
Cromossomos Humanos Y/fisiologia , Coito , Menstruação/fisiologia , Vagina/fisiologia , Adulto , Biomarcadores , Estudos de Casos e Controles , Feminino , Humanos , Reação em Cadeia da Polimerase , Sensibilidade e Especificidade , Infecções Sexualmente Transmissíveis/prevenção & controle , Esfregaço Vaginal
12.
Fertil Steril ; 94(1): 120-5, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19324347

RESUMO

OBJECTIVE: To investigate whether Y chromosomal genetic material has a role in the development of Müllerian aplasia in Finland. We have studied the TSPY1 gene and 38 additional male-specific fragments covering areas of both the long and short arms of the Y chromosome in Finnish patients with Müllerian aplasia. DESIGN: A retrospective study. SETTING: University hospital and genetic laboratory. PATIENT(S): A sample set of 110 Finnish patients with well-diagnosed Müllerian aplasia and 20 healthy relatives (13 mothers, 4 fathers, and 3 sisters from different families) were included in the study. One hundred healthy female controls with a background of at least one normal pregnancy with delivery were used as controls. INTERVENTION(S): Blood samples for DNA extraction. MAIN OUTCOME MEASURE(S): Detection of Y chromosomal fragments by polymerase chain reaction in female patients with Müllerian aplasia. RESULT(S): None of the female patients showed presence of the earlier reported TSPY1 gene or 38 additional Y chromosomal markers. CONCLUSION(S): Our results indicate that the studied Y-specific fragments, namely TSPY1 and 38 Y chromosomal markers, are not responsible for the syndrome in these Finnish patients with Müllerian aplasia.


Assuntos
Proteínas de Ciclo Celular/genética , Cromossomos Humanos Y/fisiologia , Ductos Paramesonéfricos/anormalidades , Feminino , Finlândia , Marcadores Genéticos/genética , Humanos , Masculino , Ductos Paramesonéfricos/fisiologia , Estudos Retrospectivos , Síndrome
13.
Gynecol Obstet Fertil ; 37(11-12): 901-7, 2009.
Artigo em Francês | MEDLINE | ID: mdl-19819744

RESUMO

Human Y chromosome evolution has progressively been directed towards a role in sex determination and reproduction. Cytogenetically visible structural abnormalities have determined long arm chromosomal regions which define the AZF factor that contains genes implicated in the spermatogenic process. By using molecular tools, the AZF factor has been subdivided into three loci, AZFa, b and c, the deletion of which leads to specific spermatogenesis impairments due to the loss of particular genes. Most AZF genes are specifically expressed in testis but their functions are far to be known precisely. Partial deletions of AZF regions have been described. Some of them have allowed to define more precise genotype-phenotype relationships whereas others are considered as variants in relation to Y chromosome polymorphism.


Assuntos
Cromossomos Humanos Y/fisiologia , Espermatogênese/fisiologia , Centrômero/genética , Mapeamento Cromossômico , Evolução Molecular Direcionada , Deleção de Genes , Heterocromatina/genética , Humanos , Masculino
14.
Gynecol Obstet Fertil ; 37(11-12): 895-900, 2009.
Artigo em Francês | MEDLINE | ID: mdl-19819743

RESUMO

Sex chromosome behaviour fundamentally differs between male and female meiosis. In oocyte, X chromosomes synapse giving a XX bivalent which is not recognizable in their morphology and behaviour from autosomal bivalents. In human male, X and Y chromosomes differ from one another in their morphology and their genetic content, leading to a limited pairing and preventing genetic recombination, excepted in homologous region PAR1. During pachytene stage of the first meiotic prophase, X and Y chromosomes undergo a progressive condensation and form a transcriptionally silenced peripheral XY body. The condensation of the XY bivalent during pachytene stage led us to describe four pachytene substages and to localize the pachytene checkpoint between substages 2 and 3. We also defined the pachytene index (PI=P1+P2/P1+P2+P3+P4) which is always less than 0.50 in normal meiosis. XY body undergoes decondensation at diplotene stage, but transcriptional inactivation of the two sex chromosomes or Meiotic Sex Chromosome Inactivation (MSCI) persists through to the end of spermatogenesis. Sex chromosome inactivation involves several proteins, some of them were now identified. Two isoforms of the HP1 protein, HP1beta and HP1gamma, are involved in the facultative heterochromatinization of the XY body, but the initiation of this process involves the phosphorylation of the protein H2AX by the kinase ATR whose recruitment depends on BRCA1. Extensive researches on the inactivation of the sex chromosomes during male meiosis will allow to a better understanding of some male infertilities.


Assuntos
Meiose/fisiologia , Cromossomos Sexuais/fisiologia , Homólogo 5 da Proteína Cromobox , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Feminino , Humanos , Masculino , Ovário/fisiologia , Recombinação Genética , Espermatócitos/fisiologia
16.
Arch Androl ; 53(1): 25-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17364461

RESUMO

The objective of our study was to evaluate the accuracy of the combination of hypoosmotic swelling (HOS) and eosin Y (Ey) exclusion tests to predict the ICSI cycles' outcome and its correlations with other sperm parameters. The functional and structural integrity of sperm membrane was evaluated with the combined HOS/Ey test in 95 ICSI cycles and the results were correlated with other sperm parameters, including concentration, motility, strict morphology, and total motile sperm count. The combined HOS/Ey test was evaluated for the prediction of the ICSI cycles' outcome parameters including fertilization, cleavage, and pregnancy rates. The HOS/Ey test presented significant relationships with concentration, motility, and strict morphology (p < 0,0001) but it couldn't predict the fertilization, cleavage, and pregnancy outcomes of ICSI cycles. The combined HOS/Ey test has strong correlations with motility and strict morphology parameters of sperm samples but is not sufficiently sensitive to estimate the outcome of ICSI cycles.


Assuntos
Membrana Celular/ultraestrutura , Cromossomos Humanos Y/fisiologia , Injeções de Esperma Intracitoplásmicas/métodos , Espermatozoides/fisiologia , Gonadotropina Coriônica/sangue , Endometriose/fisiopatologia , Feminino , Fertilização , Humanos , Masculino , Oócitos/citologia , Oócitos/fisiologia , Doenças Ovarianas/fisiopatologia , Seleção de Pacientes , Valor Preditivo dos Testes , Espermatozoides/citologia , Espermatozoides/ultraestrutura , Resultado do Tratamento
17.
Medicina (Kaunas) ; 42(11): 871-6, 2006.
Artigo em Lituano | MEDLINE | ID: mdl-17172786

RESUMO

Y chromosome differs from other human chromosomes. It is found in cells of the male persons only. Different human phenotypes are associated with non-recombinant region of Y chromosome. This chromosome is of great significance in the human sex determination and the development of male gametes (spermatogenesis). Testes develop in embryos which have the Y chromosome, and ovaries develop in embryos which have no Y chromosome. SRY (sex-determining region of the Y chromosome) gene determines the male phenotype. Translocation of this gene determines sex reversal. The aim of this article was to review the structure and biological functions of the human Y chromosome.


Assuntos
Cromossomos Humanos Y , Cromossomos Humanos Y/genética , Cromossomos Humanos Y/fisiologia , Feminino , Genes sry , Humanos , Cariotipagem , Masculino , Metáfase , Fenótipo , Processos de Determinação Sexual , Diferenciação Sexual/genética , Espermatogênese/fisiologia , Translocação Genética
19.
Rheumatology (Oxford) ; 43(8): 965-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15199216

RESUMO

OBJECTIVE: Fetal microchimerism has been hypothesized as a potential pathogenic mechanism for systemic sclerosis (SSc). This hypothesis was based on the clinical similarities between SSc and graft-vs-host disease and the identification of microchimeric cells in affected SSc tissues. The aim of this study was to compare the quantity of microchimeric cells in clinically affected and non-affected skin of female patients with SSc. METHODS: Fluorescence in situ hybridization (FISH) and real-time PCR were employed in paired skin biopsies obtained from clinically affected and unaffected areas from five female SSc patients with diffuse cutaneous SSc (dcSSC) and 10 healthy women. All women in the study had delivered a male fetus. RESULTS: FISH analysis revealed the presence of male fetal cells in 1/5 SSc patients (20.0%) compared with 0/10 healthy women (P = 0.0037), whereas quantification by real-time PCR revealed that all SSc samples were positive for male DNA compared with none of the controls. In the five patients with dcSSc, there were similar numbers of microchimeric cells in both affected and unaffected skin (P = 0.4) CONCLUSION: The presence of higher numbers of microchimeric cells in clinically unaffected SSc skin, before any clinically detectable evidence of sclerotic changes, suggests that an influx of microchimeric cells may precede the development of tissue fibrosis. This provides additional support to the hypothesis that fetal microchimerism may play a role in the pathogenesis of SSc.


Assuntos
Quimera , Escleroderma Sistêmico/etiologia , Pele/patologia , Idoso , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Feminino , Humanos , Hibridização in Situ Fluorescente/métodos , Masculino , Troca Materno-Fetal/fisiologia , Pessoa de Meia-Idade , Gravidez , Escleroderma Sistêmico/patologia
20.
Trends Endocrinol Metab ; 15(1): 6-11, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14693420

RESUMO

In mammals and birds, the sex of the gonads is determined by genes on the sex chromosomes. For example, the mammalian Y-linked gene Sry causes testis differentiation. The testes then secrete testosterone, which acts on the brain (often after conversion to estradiol) to cause masculine patterns of development. If this were the only reason for sex differences in neural development, then XX and XY brain cells would have to be deemed otherwise equivalent. This equivalence is doubtful because of recent experimental results demonstrating that some XX and XY tissues, including the brain, are sexually dimorphic even when they develop in a similar endocrine environment. Although X and Y genes probably influence brain phenotype in a sex-specific manner, much more information is needed to identify the magnitude and character of these effects.


Assuntos
Encéfalo/metabolismo , Caracteres Sexuais , Cromossomo X/fisiologia , Cromossomo Y/fisiologia , Animais , Cromossomos Humanos X/fisiologia , Cromossomos Humanos Y/fisiologia , Genes sry/genética , Genes sry/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA