Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 367
Filtrar
1.
Front Immunol ; 12: 790379, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899758

RESUMO

The journey of a hematopoietic stem cell (HSC) involves the passage through successive anatomical sites where HSCs are in direct contact with their surrounding microenvironment, also known as niche. These spatial and temporal cellular interactions throughout development are required for the acquisition of stem cell properties, and for maintaining the HSC pool through balancing self-renewal, quiescence and lineage commitment. Understanding the context and consequences of these interactions will be imperative for our understanding of HSC biology and will lead to the improvement of in vitro production of HSCs for clinical purposes. The aorta-gonad-mesonephros (AGM) region is in this light of particular interest since this is the cradle of HSC emergence during the embryonic development of all vertebrate species. In this review, we will focus on the developmental origin of HSCs and will discuss the novel technological approaches and recent progress made to identify the cellular composition of the HSC supportive niche and the underlying molecular events occurring in the AGM region.


Assuntos
Genômica/tendências , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Análise de Célula Única/tendências , Nicho de Células-Tronco , Animais , Aorta/embriologia , Técnicas de Cultura de Células/tendências , Linhagem da Célula , Células Cultivadas , Difusão de Inovações , Perfilação da Expressão Gênica/tendências , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Humanos , Mesonefro/embriologia , Fenótipo , Proteômica/tendências , Transdução de Sinais , Transcriptoma
2.
Int. j. morphol ; 39(5): 1253-1263, oct. 2021. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1385493

RESUMO

SUMMARY: The rabbit is considered an ideal animal model for studies that describe abnormalities in the testicles due to the similar morphogenetic mechanisms of sexual development and diseases commonly found in humans. The aim of this study was to determine the male sexual differentiation of the New Zealand rabbit (Oryctolagus cuniculus) through development. The gestational age was estimated and classified as 9, 12, 14, 16, 18, 20, 23 and 28 gestational days. The morphological and sexual determination were performed by histological analysis of the reproductive tract in the embryos and fetuses (9-28 days) as well as by immunohistochemistry- Desert hedgehog-Dhh- (testis-specific protein on Y chromosome- 16, 20, 23 days and adult rabbits). Gonads were observed from the 14th day in an undifferentiated stage and with homogeneous aspect. Sexual differentiation was observed from the 16th day with presence of cells forming gonadal cords and Dhh+ cells in the gonadal parenchyma. From the 18th gestational day testicular cords were observed, which evolved into organized seminiferous tubules. The formation of the efferent ducts and ductus deferens and epididymis was observed on the 20th and 23rd days, respectively. The differentiation of the external genitalia occurred from the 23rd days from the anogenital distance and was identified to identify the penile structures. In summary, the features of the sexual differentiation were determined by observation of the Dhh+ protein in embryos from the 16th day to adulthood, and the morphological particularities observed from the 18th gestational day, determined by differentiation of the external genitalia from the 23rd day.


RESUMEN: El conejo se considera un modelo animal ideal para estudios que describen anomalías a nivel testícular debido a que presenta mecanismos morfogenéticos similares al desa- rrollo sexual y enfermedades que se encuentran comúnmente en los seres humanos. El objetivo de este estudio fue determinar la diferenciación sexual masculina del conejo de Nueva Zelanda (Oryctolagus cuniculus) a través del desarrollo. La edad gestacional se estimó y clasificó en 9, 12, 14, 16, 18, 20, 23 y 28 días gestacionales. La determinación morfológica y sexual se realizó mediante análisis histológico del tracto reproductivo en los embriones y fetos (9 - 28 días) así como mediante inmunohistoquímica -Desert hedgehog-Dhh- (proteína testicular específica en el cromosoma Y- 16, 20, 23 días y conejos adultos). Las gónadas se observaron a partir del día 14 en un estadio indiferenciado y con aspecto homogéneo. Se observó diferenciación sexual a partir del día 16 con presencia de células formadoras de cordones gonadales y células Dhh+ en el parénquima gonadal. A partir del día 18 de gestación se observaron cordones testiculares, que evolucionaron a túbulos seminíferos organizados. La formación de los conductos eferentes, deferentes y del epidídimo se observó a los 20 y 23 días, respectivamente. La diferenciación de los genitales externos ocurrió a partir del día 23 desde la distancia anogenital y se utilizó para identificar las estructuras del pene. En conclusión, las características de la diferenciación sexual se determinaron mediante la observación de la proteína Dhh en embriones desde el día 16 hasta la edad adulta, y las particularidades morfológicas observadas a partir del día 18 de gestación, determinadas por diferenciación de los genitales externos a partir del día 23.


Assuntos
Animais , Masculino , Coelhos , Diferenciação Celular , Desenvolvimento Embrionário e Fetal , Gônadas/crescimento & desenvolvimento , Gônadas/embriologia , Túbulos Seminíferos , Diferenciação Sexual , Imuno-Histoquímica
3.
Genetics ; 217(2)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33724412

RESUMO

People with NR5A1 mutations experience testicular dysgenesis, ovotestes, or adrenal insufficiency, but we do not completely understand the origin of this phenotypic diversity. NR5A1 is expressed in gonadal soma precursor cells before expression of the sex-determining gene SRY. Many fish have two co-orthologs of NR5A1 that likely partitioned ancestral gene subfunctions between them. To explore ancestral roles of NR5A1, we knocked out nr5a1a and nr5a1b in zebrafish. Single-cell RNA-seq identified nr5a1a-expressing cells that co-expressed genes for steroid biosynthesis and the chemokine receptor Cxcl12a in 1-day postfertilization (dpf) embryos, as does the mammalian adrenal-gonadal (interrenal-gonadal) primordium. In 2dpf embryos, nr5a1a was expressed stronger in the interrenal-gonadal primordium than in the early hypothalamus but nr5a1b showed the reverse. Adult Leydig cells expressed both ohnologs and granulosa cells expressed nr5a1a stronger than nr5a1b. Mutants for nr5a1a lacked the interrenal, formed incompletely differentiated testes, had no Leydig cells, and grew far larger than normal fish. Mutants for nr5a1b formed a disorganized interrenal and their gonads completely disappeared. All homozygous mutant genotypes lacked secondary sex characteristics, including male breeding tubercles and female sex papillae, and had exceedingly low levels of estradiol, 11-ketotestosterone, and cortisol. RNA-seq showed that at 21dpf, some animals were developing as females and others were not, independent of nr5a1 genotype. By 35dpf, all mutant genotypes greatly under-expressed ovary-biased genes. Because adult nr5a1a mutants form gonads but lack an interrenal and conversely, adult nr5a1b mutants lack a gonad but have an interrenal, the adrenal, and gonadal functions of the ancestral nr5a1 gene partitioned between ohnologs after the teleost genome duplication, likely owing to reciprocal loss of ancestral tissue-specific regulatory elements. Identifying such elements could provide hints to otherwise unexplained cases of Differences in Sex Development.


Assuntos
Glândulas Suprarrenais/metabolismo , Proteínas de Ligação a DNA/genética , Disgenesia Gonadal/genética , Gônadas/metabolismo , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/genética , Glândulas Suprarrenais/embriologia , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Gônadas/embriologia , Masculino , Fenótipo , Processos de Determinação Sexual , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
4.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158211

RESUMO

Cadherins are a group of membrane proteins responsible for cell adhesion. They are crucial for cell sorting and recognition during the morphogenesis, but they also play many other roles such as assuring tissue integrity and resistance to stretching, mechanotransduction, cell signaling, regulation of cell proliferation, apoptosis, survival, carcinogenesis, etc. Within the cadherin superfamily, E- and N-cadherin have been especially well studied. They are involved in many aspects of sexual development and reproduction, such as germline development and gametogenesis, gonad development and functioning, and fertilization. E-cadherin is expressed in the primordial germ cells (PGCs) and also participates in PGC migration to the developing gonads where they become enclosed by the N-cadherin-expressing somatic cells. The differential expression of cadherins is also responsible for the establishment of the testis or ovary structure. In the adult testes, N-cadherin is responsible for the integrity of the seminiferous epithelium, regulation of sperm production, and the establishment of the blood-testis barrier. Sex hormones regulate the expression and turnover of N-cadherin influencing the course of spermatogenesis. In the adult ovaries, E- and N-cadherin assure the integrity of ovarian follicles and the formation of corpora lutea. Cadherins are expressed in the mature gametes and facilitate the capacitation of sperm in the female reproductive tract and gamete contact during fertilization. The germ cells and accompanying somatic cells express a series of different cadherins; however, their role in gonads and reproduction is still unknown. In this review, we show what is known and unknown about the role of cadherins in the germline and gonad development, and we suggest topics for future research.


Assuntos
Caderinas/fisiologia , Fertilidade/fisiologia , Gônadas/crescimento & desenvolvimento , Reprodução/fisiologia , Animais , Feminino , Fertilidade/genética , Gametogênese/genética , Células Germinativas/fisiologia , Gônadas/embriologia , Humanos , Masculino , Morfogênese/genética , Reprodução/genética
5.
Int J Mol Sci ; 21(18)2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-32927658

RESUMO

The gonads are unique among the body's organs in having a developmental choice: testis or ovary formation. Gonadal sex differentiation involves common progenitor cells that form either Sertoli and Leydig cells in the testis or granulosa and thecal cells in the ovary. Single-cell analysis is now shedding new light on how these cell lineages are specified and how they interact with the germline. Such studies are also providing new information on gonadal maturation, ageing and the somatic-germ cell niche. Furthermore, they have the potential to improve our understanding and diagnosis of Disorders/Differences of Sex Development (DSDs). DSDs occur when chromosomal, gonadal or anatomical sex are atypical. Despite major advances in recent years, most cases of DSD still cannot be explained at the molecular level. This presents a major pediatric concern. The emergence of single-cell genomics and transcriptomics now presents a novel avenue for DSD analysis, for both diagnosis and for understanding the molecular genetic etiology. Such -omics datasets have the potential to enhance our understanding of the cellular origins and pathogenesis of DSDs, as well as infertility and gonadal diseases such as cancer.


Assuntos
Transtornos do Desenvolvimento Sexual/etiologia , Gônadas/embriologia , Desenvolvimento Sexual , Análise de Célula Única , Transtornos do Desenvolvimento Sexual/diagnóstico , Genômica , Humanos , Análise de Sequência de RNA
6.
Sci Rep ; 9(1): 13462, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530896

RESUMO

SRY is the master regulator of male sex determination in eutherian mammals. In mice, Sry expression is transcriptionally and epigenetically controlled in a developmental stage-specific manner. The Sry promoter undergoes demethylation in embryonic gonadal somatic cells at the sex-determining period. However, its molecular mechanism and in vivo significance remain unclear. Here, we report that the Sry promoter is actively demethylated during gonadal development, and TET2 plays a fundamental role in Sry demethylation. Tet2-deficient mice showed absence of 5-hydroxymethylcytosine in the Sry promoter. Furthermore, Tet2 deficiency diminished Sry expression, indicating that TET2-mediated DNA demethylation regulates Sry expression positively. We previously showed that the deficiency of the H3K9 demethylase Jmjd1a compromises Sry expression and induces male-to-female sex reversal. Tet2 deficiency enhanced the sex reversal phenotype of Jmjd1a-deficient mice. Thus, TET2-mediated active DNA demethylation and JMJD1A-mediated H3K9 demethylation contribute synergistically to sex determination.


Assuntos
Desmetilação do DNA , Proteínas de Ligação a DNA/metabolismo , Gônadas , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Proteína da Região Y Determinante do Sexo/genética , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Dioxigenases , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Processos de Determinação Sexual , Fator Esteroidogênico 1/metabolismo
7.
Sex Dev ; 13(4): 195-204, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32008010

RESUMO

MAPKs affect gonadal differentiation in mice and humans, but whether this applies to all mammals is as yet unknown. Thus, we investigated MAPK expression during gonadal differentiation and after treatment with oestrogen in a distantly related mammal, the marsupial tammar wallaby, using our model of oestrogen-induced gonadal sex reversal. High-throughput RNA-sequencing was carried out on gonads collected from developing tammar 2 days before birth to 8 days after birth to characterise MAPK and key sexual differentiation markers. Day 25 foetal testes were cultured for 120 h in control medium or medium supplemented with exogenous oestrogen and processed for RNA-seq to identify changes in gene expression in response to oestrogen. MAPK pathway genes in the tammar were highly conserved at the sequence and amino acid level with those of mice and humans. Marsupial MAP3K1 and MAP3K4 clustered together in a separate branch from eutherian mammals. There was a marked decrease in the expression of male-determining genes SOX9 and AMH and increase in the female marker FOXL2 in oestrogen-treated male gonads. Only MAP3K1 expression increased in male gonads in response to oestrogen while other MAPK genes remained unaffected. This study suggests that MAP3K1 can be influenced by exogenous oestrogens during gonadal differentiation in this marsupial.


Assuntos
Perfilação da Expressão Gênica , Gônadas/embriologia , Gônadas/enzimologia , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 4/genética , Macropodidae/embriologia , Macropodidae/genética , Animais , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Marcadores Genéticos , Gônadas/efeitos dos fármacos , MAP Quinase Quinase Quinase 1/metabolismo , MAP Quinase Quinase Quinase 4/metabolismo , Masculino , Filogenia , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
8.
Adv Anat Embryol Cell Biol ; 230: 1-70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30543033

RESUMO

The observation of two precursor groups of the early stem cells (Groups I and II) leads to the realization that a first amount of fetal stem cells (Group I) migrate from the AMG (Aortal-Mesonephric-Gonadal)-region into the aorta and its branching vessels. A second group (Group II) gains quite a new significance during human development. This group presents a specific developmental step which is found only in the human. This continuation of the early development along a different way indicates a general alteration of the stem cell biology. This changed process in the stem cell scene dominates the further development of the human stem cells. It remains unclear where this phylogenetic step first appears. By far not all advanced mammals show this second group of stem cells and their axonal migration. Essentially only primates seem to be involved in this special development.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/ultraestrutura , Gônadas/citologia , Gônadas/embriologia , Células APUD/citologia , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiologia , Córtex Suprarrenal/ultraestrutura , Medula Suprarrenal/citologia , Medula Suprarrenal/embriologia , Medula Suprarrenal/fisiologia , Aorta/citologia , Aorta/embriologia , Aorta/ultraestrutura , Sistema Nervoso Autônomo/citologia , Sistema Nervoso Autônomo/embriologia , Sistema Nervoso Autônomo/fisiologia , Orientação de Axônios/fisiologia , Movimento Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Gônadas/fisiologia , Gônadas/ultraestrutura , Desenvolvimento Humano/fisiologia , Humanos , Microscopia Eletrônica , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/fisiologia , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/ultraestrutura , Paragânglios Cromafins/citologia , Paragânglios Cromafins/fisiologia , Paragânglios Cromafins/ultraestrutura , Teratoma/embriologia , Teratoma/fisiopatologia
9.
Thromb Haemost ; 118(8): 1370-1381, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29991091

RESUMO

The development of new strategies based on cell therapy approaches to correct haemophilia A (HA) requires further insights into new cell populations capable of producing coagulation factor VIII (FVIII) and presenting stable engraftment potential. The major producers of FVIII in the adult are liver sinusoidal endothelial cells (LSECs) and in a lesser degree bone marrow-derived cells, both of which have been shown to ameliorate the bleeding phenotype in adult HA mice after transplantation. We have previously shown that cells from the foetal liver (FL) and the aorta-gonads-mesonephros (AGM) haematopoietic locations possess higher LSEC engraftment potential in newborn mice compared with adult-derived LSECs, constituting likely therapeutic targets for the treatment of HA in neonates. However, less is known about the production of FVIII in embryonic locations. Quantitative polymerase chain reaction and Western blot analysis were performed to assess the relative level of FVIII production in different embryonic tissues and at various developmental stages, identifying the FL and AGM region from day 12 (E12) as prominent sources of FVIII. Furthermore, FL-derived VE-cad+CD45-Lyve1+/- endothelial/endothelial progenitor cells, presenting vascular engraftment potential, produced high levels of F8 ribonucleic acid compared with CD45+ blood progenitors or Dlk1+ hepatoblasts. In addition, we show that the E11 AGM explant cultures expanded cells with LSEC repopulation activity, instrumental to further understand signals for in vitro generation of LSECs. Taking into account the capacity for FVIII expression, culture expansion and newborn engraftment potential, these results support the use of cells with foetal characteristics for correction of FVIII deficiency in young individuals.


Assuntos
Aorta/metabolismo , Células Progenitoras Endoteliais/metabolismo , Fator VIII/metabolismo , Gônadas/metabolismo , Hemofilia A/metabolismo , Fígado/metabolismo , Mesonefro/metabolismo , Animais , Aorta/embriologia , Aorta/transplante , Diferenciação Celular , Células Progenitoras Endoteliais/transplante , Fator VIII/genética , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Gônadas/embriologia , Gônadas/transplante , Hemofilia A/genética , Hemofilia A/cirurgia , Fígado/embriologia , Mesonefro/embriologia , Mesonefro/transplante , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transplante de Células-Tronco/métodos , Técnicas de Cultura de Tecidos
10.
Elife ; 72018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29905527

RESUMO

The origin of Langerhans cells (LCs), which are skin epidermis-resident macrophages, remains unclear. Current lineage tracing of LCs largely relies on the promoter-Cre-LoxP system, which often gives rise to contradictory conclusions with different promoters. Thus, reinvestigation with an improved tracing method is necessary. Here, using a laser-mediated temporal-spatial resolved cell labeling method, we demonstrated that most adult LCs originated from the ventral wall of the dorsal aorta (VDA), an equivalent to the mouse aorta, gonads, and mesonephros (AGM), where both hematopoietic stem cells (HSCs) and non-HSC progenitors are generated. Further fine-fate mapping analysis revealed that the appearance of LCs in adult zebrafish was correlated with the development of HSCs, but not T cell progenitors. Finally, we showed that the appearance of tissue-resident macrophages in the brain, liver, heart, and gut of adult zebrafish was also correlated with HSCs. Thus, the results of our study challenged the EMP-origin theory for LCs.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Células de Langerhans/fisiologia , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/embriologia , Aorta/crescimento & desenvolvimento , Gônadas/citologia , Gônadas/embriologia , Gônadas/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células de Langerhans/citologia , Macrófagos/metabolismo , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/crescimento & desenvolvimento , Camundongos , Microscopia Confocal , Peixe-Zebra
11.
Mol Hum Reprod ; 24(5): 233-243, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29528446

RESUMO

STUDY QUESTION: Which set of antibodies can be used to identify migratory and early post-migratory human primordial germ cells (hPGCs)? STUDY FINDING: We validated the specificity of 33 antibodies for 31 markers, including POU5F1, NANOG, PRDM1 and TFAP2C as specific markers of hPGCs at 4.5 weeks of development of Carnegie stage (CS12-13), whereas KIT and SOX17 also marked the intra-aortic hematopoietic stem cell cluster in the aorta-gonad-mesonephros (AGM). WHAT IS KNOWN ALREADY: The dynamics of gene expression during germ cell development in mice is well characterized and this knowledge has proved crucial to allow the development of protocols for the in vitro derivation of functional gametes. Although there is a great interest in generating human gametes in vitro, it is still unclear which markers are expressed during the early stages of hPGC development and many studies use markers described in mouse to benchmark differentiation of human PGC-like cells (hPGCLCs). Early post-implantation development differs significantly between mice and humans, and so some germ cells markers, including SOX2, SOX17, IFITM3 and ITGA6 may not identify mPGCs and hPGCs equally well. STUDY DESIGN, SIZE, DURATION: This immunofluorescence study investigated the expression of putative hPGC markers in the caudal part of a single human embryo at 4.5 weeks of development. PARTICIPANTS/MATERIALS, SETTING, METHODS: We have investigated by immunofluorescence the expression of a set of 33 antibodies for 31 markers, including pluripotency, germ cell, adhesion, migration, surface, mesenchymal and epigenetic markers on paraffin sections of the caudal part, including the AGM region, of a single human embryo (CS12-13). The human material used was anonymously donated with informed consent from elective abortions without medical indication. MAIN RESULTS AND THE ROLE OF CHANCE: We observed germ cell specific expression of NANOG, TFAP2C and PRDM1 in POU5F1+ hPGCs in the AGM. The epigenetic markers H3K27me3 and 5mC were sufficient to distinguish hPGCs from the surrounding somatic cells. Some mPGC-markers were not detected in hPGCs, but marked other tissues; whereas other markers, such as ALPL, SOX17, KIT, TUBB3, ITGA6 marked both POU5F1+ hPGCs and other cells in the AGM. We used a combination of multiple markers, immunostaining different cellular compartments when feasible, to decrease the chance of misidentifying hPGCs. LARGE SCALE DATA: Non-applicable. LIMITATIONS REASONS FOR CAUTION: Material to study early human development is unique and very rare thus restricting the sample size. We have used a combination of antibodies limited by the number of paraffin sections available. WIDER IMPLICATIONS OF THE FINDINGS: Most of our knowledge on early gametogenesis has been obtained from model organisms such as mice and is extrapolated to humans. However, since there is a dedicated effort to produce human artificial gametes in vitro, it is of great importance to determine the expression and specificity of human-specific germ cell markers. We provide a systematic analysis of the expression of 31 different markers in paraffin sections of a CS12-13 embryo. Our results will help to set up a toolbox of markers to evaluate protocols to induce hPGCLCs in vitro. STUDY FUNDING AND COMPETING INTEREST(S): M.G.F. was funded by Fundação para a Ciência e Tecnologia (FCT) [SFRH/BD/78689/2011] and S.M.C.S.L. was funded by the Interuniversity Attraction Poles (IAP, P7/07) and the European Research Council Consolidator (ERC-CoG-725722-OVOGROWTH). The authors declare no conflict of interest.


Assuntos
Aorta/citologia , Gametogênese/fisiologia , Células Germinativas/citologia , Gônadas/citologia , Mesonefro/citologia , Aorta/embriologia , Aorta/metabolismo , Biomarcadores/metabolismo , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células Germinativas/metabolismo , Gônadas/embriologia , Gônadas/metabolismo , Humanos , Mesonefro/embriologia , Mesonefro/metabolismo
12.
Development ; 145(2)2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29358215

RESUMO

Hematopoietic stem cells (HSCs) develop in discrete anatomical niches, migrating during embryogenesis from the aorta-gonad-mesonephros (AGM) region to the fetal liver, and finally to the bone marrow, where most HSCs reside throughout adult life. These niches provide supportive microenvironments that specify, expand and maintain HSCs. Understanding the constituents and molecular regulation of HSC niches is of considerable importance as it could shed new light on the mechanistic principles of HSC emergence and maintenance, and provide novel strategies for regenerative medicine. However, controversy exists concerning the cellular complexity of the bone marrow niche, and our understanding of the different HSC niches during development remains limited. In this Review, we summarize and discuss what is known about the heterogeneity of the HSC niches at distinct stages of their ontogeny, from the embryo to the adult bone marrow, drawing predominantly on data from mouse studies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Aorta/embriologia , Linhagem da Célula , Feminino , Gônadas/embriologia , Neoplasias Hematológicas/patologia , Sistema Hematopoético/embriologia , Humanos , Masculino , Mesonefro/embriologia , Camundongos , Placenta/citologia , Placenta/fisiologia , Gravidez , Células Estromais/citologia , Células Estromais/fisiologia , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia
13.
Sex Dev ; 12(1-3): 106-122, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29131109

RESUMO

Malignant gonadal germ cell tumors, referred to as germ cell cancers (GCC), occur with increased frequency in individuals who have specific types of differences (disorders) of sex development (DSD). Recent population-based studies have identified new environmental and genetic risk factors that have led to a 'genvironment' hypothesis, which may potentially be helpful in risk assessment in DSD-related GCC. In DSD, the malignancy risk is highly heterogeneous, but recent studies allow now to discriminate between high- and low-risk conditions. Gonadal biopsy is in some cases the best procedure of choice to assess the risk, and with the availability of immunohistochemical biomarkers [OCT3/4 (POU5F1), TSPY, SOX9, FOXL2 and KITLG (SCF)], a reliable classification of GCC and its precursors can be made. The opportunities in the field of virtual diagnostic pathology will be presented, having possibilities for rare diseases in general and DSD specifically. It is expected that the International DSD Registry will stimulate international collaborations, facilitating better diagnostic procedures as well as research.


Assuntos
Transtornos do Desenvolvimento Sexual/patologia , Transtornos do Desenvolvimento Sexual/terapia , Gônadas/patologia , Gônadas/embriologia , Humanos , Guias de Prática Clínica como Assunto , Fatores de Risco , Diferenciação Sexual , Telemedicina
14.
Sci Rep ; 7(1): 15961, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162857

RESUMO

The precise timing and sequence of changes in expression of key genes and proteins during human sex-differentiation and onset of steroidogenesis was evaluated by whole-genome expression in 67 first trimester human embryonic and fetal ovaries and testis and confirmed by qPCR and immunohistochemistry (IHC). SRY/SOX9 expression initiated in testis around day 40 pc, followed by initiation of AMH and steroidogenic genes required for androgen production at day 53 pc. In ovaries, gene expression of RSPO1, LIN28, FOXL2, WNT2B, and ETV5, were significantly higher than in testis, whereas GLI1 was significantly higher in testis than ovaries. Gene expression was confirmed by IHC for GAGE, SOX9, AMH, CYP17A1, LIN28, WNT2B, ETV5 and GLI1. Gene expression was not associated with the maternal smoking habits. Collectively, a precise temporal determination of changes in expression of key genes involved in human sex-differentiation is defined, with identification of new genes of potential importance.


Assuntos
Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Diferenciação Sexual/genética , Adolescente , Adulto , Contagem de Células , Feminino , Marcadores Genéticos , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Fumar/efeitos adversos , Coloração e Rotulagem , Esteroides/biossíntese , Fatores de Tempo , Adulto Jovem
15.
J Exp Med ; 214(12): 3731-3751, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29093060

RESUMO

In the developing embryo, hematopoietic stem cells (HSCs) emerge from the aorta-gonad-mesonephros (AGM) region, but the molecular regulation of this process is poorly understood. Recently, the progression from E9.5 to E10.5 and polarity along the dorso-ventral axis have been identified as clear demarcations of the supportive HSC niche. To identify novel secreted regulators of HSC maturation, we performed RNA sequencing over these spatiotemporal transitions in the AGM region and supportive OP9 cell line. Screening several proteins through an ex vivo reaggregate culture system, we identify BMPER as a novel positive regulator of HSC development. We demonstrate that BMPER is associated with BMP signaling inhibition, but is transcriptionally induced by BMP4, suggesting that BMPER contributes to the precise control of BMP activity within the AGM region, enabling the maturation of HSCs within a BMP-negative environment. These findings and the availability of our transcriptional data through an accessible interface should provide insight into the maintenance and potential derivation of HSCs in culture.


Assuntos
Aorta/metabolismo , Diferenciação Celular , Gônadas/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/metabolismo , Animais , Aorta/embriologia , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular/genética , Análise por Conglomerados , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Mesoderma/metabolismo , Mesonefro/embriologia , Camundongos Endogâmicos C57BL , Transdução de Sinais , Proteínas Smad/metabolismo , Nicho de Células-Tronco/genética , Fatores de Tempo
16.
J Biol Chem ; 292(49): 20281-20291, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29042436

RESUMO

Wilms tumor protein 1 (WT1) has been implicated in the control of several genes in sexual development, but its function in gonad formation is still unclear. Here, we report that WT1 stimulates expression of Kdr, the gene encoding VEGF receptor 2, in murine embryonic gonads. We found that WT1 and KDR are co-expressed in Sertoli cells of the testes and somatic cells of embryonic ovaries. Vivo-morpholino-mediated WT1 knockdown decreased Kdr transcripts in cultured embryonic gonads at multiple developmental stages. Furthermore, WT1 bound to the Kdr promoter in the chromatin of embryonic testes and ovaries. Forced expression of the WT1(-KTS) isoform, which functions as a transcription factor, increased KDR mRNA levels, whereas the WT1(+KTS) isoform, which acts presumably on the post-transcriptional level, did not. ChIP indicated that WT1(-KTS), but not WT1(+KTS), binds to the KDR promoter. Treatment with the KDR tyrosine kinase inhibitor SU1498 or the KDR ligand VEGFA revealed that KDR signaling represses the testis-promoting gene Sox9 in embryonic XX gonads. WT1 knockdown abrogated the stimulatory effect of SU1498-mediated KDR inhibition on Sox9 expression. Exposure to 1% O2 to mimic the low-oxygen conditions in the embryo increased Vegfa expression but did not affect Sox9 mRNA levels in gonadal explants. However, incubation in 1% O2 in the presence of SU1498 significantly reduced Sox9 transcripts in cultured testes and increased Sox9 levels in ovaries. These findings demonstrate that both the local oxygen environment and WT1, which enhances KDR expression, contribute to sex-specific Sox9 expression in developing murine gonads.


Assuntos
Gônadas/metabolismo , Hipóxia/fisiopatologia , Proteínas Repressoras/genética , Fatores de Transcrição SOX9/genética , Transcrição Gênica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Masculino , Camundongos , Ovário/embriologia , Ovário/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Testículo/embriologia , Testículo/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas WT1
17.
Yale J Biol Med ; 90(3): 449-461, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28955183

RESUMO

The role of steroids in human medicine is well recognized, but the major contributions made by the large domestic animals as a source of material in the discovery, isolation, and determination of the structure of the steroid hormones is less well appreciated. After a brief reminder of the early efforts to obtain a reliable source of steroids for clinical use, the narrative here is to outline one example where success was ultimately achieved for estrogen replacement therapy. Whereas knowledge of the high concentrations of estrogens in urine of pregnant women and mares dates from the late 1920s, it was not until the 1940s that the latter was shown to be a practical source. Initially, the placenta was held to be responsible, but the involvement of the fetus in each case was eventually established. The remarkable enlargement of the human fetal adrenal glands and the fetal gonads in the horse, with characteristic features of steroid secreting tissues, suggested their participation. Ultimately, it was 16-hydroxylation by the fetal liver that resulted in estriol being the major estrogen type, by far, in late human pregnancy. In the mare, the pattern of estrogen production reflected that of the growth and later regression of the fetal gonads. The characteristic production ring-B, unsaturated estrogens in the mare is derived from an alternative pathway involving retention of the additional double bond in the biosynthesis of equilin.


Assuntos
Placenta/embriologia , Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/metabolismo , Animais , Estrogênios/metabolismo , Estrona/metabolismo , Feminino , Gônadas/embriologia , Gônadas/metabolismo , Cavalos , Humanos , Placenta/metabolismo , Gravidez , Esteroides/metabolismo
18.
J Exp Med ; 214(11): 3347-3360, 2017 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-28931624

RESUMO

T lymphocytes are key cellular components of the adaptive immune system and play a central role in cell-mediated immunity in vertebrates. Despite their heterogeneities, it is believed that all different types of T lymphocytes are generated exclusively via the differentiation of hematopoietic stem cells (HSCs). Using temporal-spatial resolved fate-mapping analysis and time-lapse imaging, here we show that the ventral endothelium in the zebrafish aorta-gonad-mesonephros and posterior blood island, the hematopoietic tissues previously known to generate HSCs and erythromyeloid progenitors, respectively, gives rise to a transient wave of T lymphopoiesis independent of HSCs. This HSC-independent T lymphopoiesis occurs early and generates predominantly CD4 Tαß cells in the larval but not juvenile and adult stages, whereas HSC-dependent T lymphopoiesis emerges late and produces various subtypes of T lymphocytes continuously from the larval stage to adulthood. Our study unveils the existence, origin, and ontogeny of HSC-independent T lymphopoiesis in vivo and reveals the complexity of the endothelial-hematopoietic transition of the aorta.


Assuntos
Aorta/citologia , Embrião não Mamífero/citologia , Endotélio Vascular/citologia , Células-Tronco Hematopoéticas/citologia , Linfopoese , Linfócitos T/citologia , Animais , Animais Geneticamente Modificados , Aorta/embriologia , Aorta/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/citologia , Gônadas/embriologia , Gônadas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hibridização In Situ , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/metabolismo , Microscopia Confocal , Linfócitos T/metabolismo , Imagem com Lapso de Tempo/métodos , Peixe-Zebra
19.
PLoS One ; 12(8): e0183049, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28817611

RESUMO

Glycoproteins such as growth factor receptors and extracellular matrix have well-known functions in development and cancer progression, however, the glycans at sites of modification are often heterogeneous molecular populations which makes their functional characterization challenging. Here we provide evidence for a specific, discrete, well-defined glycan modification and regulation of a stage-specific cell migration in Caenorhabditis elegans. We show that a chain-terminating, putative null mutation in the gene encoding a predicted ß1,4-N-acetylgalactosaminyltransferase, named ngat-1, causes a maternally rescued temperature sensitive (ts) defect in the second phase of the three phase migration pattern of the posterior, but not the anterior, hermaphrodite Distal Tip Cell (DTC). An amino-terminal partial deletion of ngat-1 causes a similar but lower penetrance ts phenotype. The existence of multiple ts alleles with distinctly different molecular DNA lesions, neither of which is likely to encode a ts protein, indicates that NGAT-1 normally prevents innate temperature sensitivity for phase 2 DTC pathfinding. Temperature shift analyses indicate that the ts period for the ngat-1 mutant defect ends by the beginning of post-embryonic development-nearly 3 full larval stages prior to the defective phase 2 migration affected by ngat-1 mutations. NGAT-1 homologs generate glycan-terminal GalNAc-ß1-4GlcNAc, referred to as LacdiNAc modifications, on glycoproteins and glycolipids. We also found that the absence of the GnT1/Mgat1 activity [UDP-N-acetyl-D-glucosamine:α-3-D-mannoside ß-1,2-N-acetylglucosaminyltransferase 1 (encoded by C. elegans gly-12, gly-13, and gly-14 and homologous to vertebrate GnT1/Mgat1)], causes a similar spectrum of DTC phenotypes as ngat-1 mutations-primarily affecting posterior DTC phase 2 migration and preventing manifestation of the same innate ts period as ngat-1. GnT1/Mgat1 is a medial Golgi enzyme known to modify mannose residues and initiate N-glycan branching, an essential step in the biosynthesis of hybrid, paucimannose and complex-type N-glycans. Quadruple mutant animals bearing putative null mutations in ngat-1 and the three GnT genes (gly-12, gly-13, gly-14) were not enhanced for DTC migration defects, suggesting NGAT-1 and GnT1 act in the same pathway. These findings suggest that GnTI generates an N-glycan substrate for NGAT-1 modification, which is required at restrictive temperature (25°C) to prevent, stabilize, reverse or compensate a perinatal thermo-labile process (or structure) causing late larval stage DTC phase 2 migration errors.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Movimento Celular , Gônadas/citologia , N-Acetilgalactosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Neurônios/metabolismo , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Gônadas/embriologia , Gônadas/metabolismo , Mutação , N-Acetilgalactosaminiltransferases/metabolismo , N-Acetilglucosaminiltransferases/genética , Neurônios/citologia
20.
Endocrinology ; 158(9): 3030-3041, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575219

RESUMO

Aromatase (encoded by the cyp19a1a and cyp19a1b genes) plays a central role in sex differentiation in fish, but its precise roles during sex differentiation are still largely unknown. Here, we systematically generated cyp19a1a and cyp19a1b mutant lines as well as a cyp19a1a;cyp19a1b double mutant line in zebrafish using transcription activatorlike effector nucleases. Our results showed that cyp19a1a mutants and cyp19a1a;cyp19a1b double mutants, but not cyp19a1b mutants, had impaired sex differentiation, and all cyp19a1a mutants and cyp19a1a;cyp19a1b double mutants were males. During sex differentiation, the ovary-like gonads were not observed and the male sex differentiation program was delayed in the cyp19a1a-null fish, and these phenotypes could be partially rescued by 17ß-estradiol treatment. Gene expression analysis indicated that male and female sex differentiation-related genes were significantly decreased in the cyp19a1a mutant. Collectively, our results revealed dual functions of the cyp19a1a gene during sex differentiation: cyp19a1a is not only indispensable for female sex differentiation but also required for male sex differentiation.


Assuntos
Aromatase/fisiologia , Diferenciação Sexual/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Aromatase/genética , Embrião não Mamífero , Estradiol/farmacologia , Feminino , Fertilidade/efeitos dos fármacos , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Transferência de Genes , Gônadas/efeitos dos fármacos , Gônadas/embriologia , Gônadas/metabolismo , Masculino , Mutação , Diferenciação Sexual/efeitos dos fármacos , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA