Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Development ; 148(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-33795229

RESUMO

During development of the mouse urogenital complex, the gonads undergo changes in three-dimensional structure, body position and spatial relationship with the mesonephric ducts, kidneys and adrenals. The complexity of genital ridge development obscures potential connections between morphogenesis and gonadal sex determination. To characterize the morphogenic processes implicated in regulating gonad shape and fate, we used whole-embryo tissue clearing and light sheet microscopy to assemble a time course of gonad development in native form and context. Analysis revealed that gonad morphology is determined through anterior-to-posterior patterns as well as increased rates of growth, rotation and separation in the central domain that may contribute to regionalization of the gonad. We report a close alignment of gonad and mesonephric duct movements as well as delayed duct development in a gonad dysgenesis mutant, which together support a mechanical dependency linking gonad and mesonephric duct morphogenesis.


Assuntos
Gônadas/fisiologia , Morfogênese/fisiologia , Ductos Mesonéfricos/fisiologia , Animais , Embrião de Mamíferos/fisiologia , Feminino , Idade Gestacional , Rim/fisiologia , Masculino , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Diferenciação Sexual/fisiologia
2.
Stem Cell Rev Rep ; 12(5): 530-542, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27324145

RESUMO

The aorta-gonad-mesonephros (AGM) region contains intra-aortic clusters (IACs) thought to have acquired hematopoietic stem cell (HSC) potential in vertebrate embryos. To assess extrinsic regulation of IACs in the AGM region, we employed mouse embryos harboring a Sall1-GFP reporter gene, which allows identification of mesonephros cells based on GFP expression. Analysis of AGM region tissue sections confirmed mesonephros GFP expression. Mesonephric cells sorted at E10.5 expressed mRNA encoding Csf1, a hematopoietic cytokine, and corresponding protein, based on real-time PCR and immunocytochemistry, respectively. Further analysis indicated that some IACs express the CSF1 receptor, CSF1R. Expression of Cebpa and Irf8 mRNAs was higher in CSF1R-positive IACs, whereas that of Cebpε and Gfi1 mRNAs was lower relative to CSF1R-negative IACs, suggesting that CSF1/CSF1R signaling functions in IAC myeloid differentiation by modulating expression of these transcription factors. Colony formation assays using CSF1R-positive IACs revealed increased numbers of myeloid colonies in the presence of CSF1. Analysis using an intra-cellular signaling array indicated the greatest fold increase of Cleaved Caspase-3 in AGM cells in the presence of CSF1. Immunohistochemistry revealed that Cleaved Caspase-3 is primarily expressed in IACs in the AGM region, and incubation of IACs with CSF1 up-regulated Cleaved Caspase-3. Overall, our findings suggest that CSF1 secreted from mesonephros accelerates IAC myeloid differentiation in the AGM region, possibly via Caspase-3 cleavage.


Assuntos
Aorta/metabolismo , Aorta/fisiologia , Diferenciação Celular/fisiologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Mesonefro/metabolismo , Células Mieloides/fisiologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Caspase 3/metabolismo , Fatores Reguladores de Interferon/metabolismo , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , RNA Mensageiro/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
3.
Semin Nephrol ; 34(4): 437-44, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25217272

RESUMO

Renal tubule epithelial cells can regenerate in response to acute injury. Although this process remains poorly understood, it appears to involve the reactivation of pathways that are operative during embryonic kidney formation. A better understanding of renal regeneration may lead to the development of new therapies that can attenuate acute kidney injury or expedite recovery. The zebrafish is being used as a model to understand renal regeneration. In this review, we summarize the current knowledge on zebrafish kidney formation, describe methods for inducing acute injury, and focus on the unique capacity of the zebrafish adult kidney to undergo de novo nephron formation in response to damage.


Assuntos
Injúria Renal Aguda , Rim/fisiologia , Mesonefro/fisiologia , Regeneração/fisiologia , Animais , Rim/citologia , Túbulos Renais/citologia , Túbulos Renais/fisiologia , Mesonefro/citologia , Néfrons/fisiologia , Podócitos/fisiologia , Peixe-Zebra
4.
Curr Pediatr Rev ; 10(2): 107-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25088264

RESUMO

Renal development begins in-utero and continues throughout childhood. Almost one-third of all developmental anomalies include structural or functional abnormalities of the urinary tract. There are three main phases of in-utero renal development: Pronephros, Mesonephros and Metanephros. Within three weeks of gestation, paired pronephri appear. A series of tubules called nephrotomes fuse with the pronephric duct. The pronephros elongates and induces the nearby mesoderm, forming the mesonephric (Woffian) duct. The metanephros is the precursor of the mature kidney that originates from the ureteric bud and the metanephric mesoderm (blastema) by 5 weeks of gestation. The interaction between these two components is a reciprocal process, resulting in the formation of a mature kidney. The ureteric bud forms the major and minor calyces, and the collecting tubules while the metanephrogenic blastema develops into the renal tubules and glomeruli. In humans, all of the nephrons are formed by 32 to 36 weeks of gestation. Simultaneously, the lower urinary tract develops from the vesico urethral canal, ureteric bud and mesonephric duct. In utero, ureters deliver urine from the kidney to the bladder, thereby creating amniotic fluid. Transcription factors, extracellular matrix glycoproteins, signaling molecules and receptors are the key players in normal renal development. Many medications (e.g., aminoglycosides, cyclooxygenase inhibitors, substances that affect the renin-angiotensin aldosterone system) also impact renal development by altering the expression of growth factors, matrix regulators or receptors. Thus, tight regulation and coordinated processes are crucial for normal renal development.


Assuntos
Túbulos Renais Coletores/embriologia , Mesonefro/embriologia , Pronefro/embriologia , Sistema Renina-Angiotensina/fisiologia , Sistema Urinário/embriologia , Sistema Urogenital/embriologia , Diferenciação Celular , Matriz Extracelular , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Túbulos Renais Coletores/anatomia & histologia , Túbulos Renais Coletores/fisiologia , Mesonefro/anatomia & histologia , Mesonefro/fisiologia , Pronefro/anatomia & histologia , Pronefro/fisiologia , Transdução de Sinais , Sistema Urinário/anatomia & histologia , Sistema Urogenital/anatomia & histologia , Sistema Urogenital/fisiologia
5.
PLoS One ; 8(6): e68050, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840809

RESUMO

Development of the testis begins with the expression of the SRY gene in pre-Sertoli cells. Soon after, testis cords containing Sertoli and germ cells are formed and fetal Leydig cells subsequently develop in the interstitial space. Studies using knockout mice have indicated that multiple genes encoding growth factors and transcription factors are implicated in fetal Leydig cell differentiation. Previously, we demonstrated that the Arx gene is implicated in this process. However, how ARX regulates Leydig cell differentiation remained unknown. In this study, we examined Arx KO testes and revealed that fetal Leydig cell numbers largely decrease throughout the fetal life. Since our study shows that fetal Leydig cells rarely proliferate, this decrease in the KO testes is thought to be due to defects of fetal Leydig progenitor cells. In sexually indifferent fetal gonads of wild type, ARX was expressed in the coelomic epithelial cells and cells underneath the epithelium as well as cells at the gonad-mesonephros border, both of which have been described to contain progenitors of fetal Leydig cells. After testis differentiation, ARX was expressed in a large population of the interstitial cells but not in fetal Leydig cells, raising the possibility that ARX-positive cells contain fetal Leydig progenitor cells. When examining marker gene expression, we observed cells as if they were differentiating into fetal Leydig cells from the progenitor cells. Based on these results, we propose that ARX acts as a positive factor for differentiation of fetal Leydig cells through functioning at the progenitor stage.


Assuntos
Diferenciação Celular/genética , Genes Homeobox/genética , Proteínas de Homeodomínio/genética , Células Intersticiais do Testículo/fisiologia , Diferenciação Sexual/genética , Células-Tronco/patologia , Fatores de Transcrição/genética , Animais , Proliferação de Células , Células Epiteliais/fisiologia , Epitélio/fisiologia , Feto/fisiologia , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/fisiologia , Gônadas/crescimento & desenvolvimento , Gônadas/fisiologia , Masculino , Mesonefro/crescimento & desenvolvimento , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Testículo/crescimento & desenvolvimento , Testículo/fisiologia
6.
J Cell Physiol ; 228(3): 627-39, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22886539

RESUMO

Meiotic initiation of germ cells at 13.5 dpc (days post-coitus) indicates female sex determination in mice. Recent studies reveal that mesonephroi-derived retinoic acid (RA) is the key signal for induction of meiosis. However, whether the mesonephroi is dispensable for meiosis is unclear and the role of the ovary in this meiotic process remains to be clarified. This study provides data that RA derived from fetal ovaries is sufficient to induce germ cell meiosis in a fetal ovary culture system. When fetal ovaries were collected from 11.5 to 13.5 dpc fetuses, isolated and cultured in vitro, germ cells enter meiosis in the absence of mesonephroi. To exclude RA sourcing from mesonephroi, 11.5 dpc urogenital ridges (UGRs; mesonephroi and ovary complexes) were treated with diethylaminobenzaldehyde (DEAB) to block retinaldehyde dehydrogenase (RALDH) activity in the mesonephros and the ovary. Meiosis occurred when DEAB was withdrawn and the mesonephros was removed 2 days later. Furthermore, RALDH1, rather than RALDH2, serves as the major RA synthetase in UGRs from 12.5 to 15.5 dpc. DEAB treatment to the ovary alone was able to block germ cell meiotic entry. We also found that exogenously supplied RA dose-dependently reduced germ cell numbers in ovaries by accelerating the entry into meiosis. These results suggest that ovary-derived RA is responsible for meiosis initiation.


Assuntos
Meiose/fisiologia , Ovário/embriologia , Ovário/fisiologia , Tretinoína/fisiologia , Família Aldeído Desidrogenase 1 , Animais , Benzaldeídos/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Masculino , Mesonefro/efeitos dos fármacos , Mesonefro/embriologia , Mesonefro/fisiologia , Camundongos , Ovário/efeitos dos fármacos , Óvulo/efeitos dos fármacos , Óvulo/crescimento & desenvolvimento , Óvulo/fisiologia , Gravidez , Retinal Desidrogenase/antagonistas & inibidores , Retinal Desidrogenase/metabolismo , Processos de Determinação Sexual/fisiologia , Técnicas de Cultura de Tecidos , Tretinoína/administração & dosagem
7.
Dev Dyn ; 240(6): 1600-12, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21491542

RESUMO

The metanephros is the functional organ in adult amniotes while the mesonephros degenerates. However, parallel tubulogenetic events are thought to exist between mesonephros and metanephros. Mesonephric tubules are retained in males and differentiate into efferent ducts of the male reproductive tract. By examining the murine mesonephric expression of markers of distinct stages and regions of metanephric nephrons during tubule formation and patterning, we provide further evidence to support this common morphogenetic mechanism. Renal vesicle, early proximal and distal tubule, loop of Henle, and renal corpuscle genes were expressed by mesonephric tubules. Vip, Slc6a20b, and Slc18a1 were male-specific. In contrast, mining of the GUDMAP database identified candidate late mesonephros-specific genes, 10 of which were restricted to the male. Among the male-specific genes are candidates for regulating ion/fluid balance within the efferent ducts, thereby regulating sperm maturation and genes marking tubule-associated neurons potentially critical for normal male reproductive tract function.


Assuntos
Diferenciação Celular/genética , Genes Controladores do Desenvolvimento , Mesonefro/embriologia , Néfrons/embriologia , Néfrons/metabolismo , Animais , Padronização Corporal/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Masculino , Mesonefro/citologia , Mesonefro/metabolismo , Mesonefro/fisiologia , Camundongos , Modelos Biológicos , Organogênese/genética , Organogênese/fisiologia , Distribuição Tecidual
8.
Haematologica ; 95(6): 875-83, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20007140

RESUMO

BACKGROUND: The hemangioblast is a bi-potential precursor cell with the capacity to differentiate into hematopoietic and vascular cells. In mouse E7.0-7.5 embryos, the hemangioblast can be identified by a clonal blast colony-forming cell (BL-CFC) assay or single cell OP9 co-culture. However, the ontogeny of the hemangioblast in mid-gestation embryos is poorly defined. DESIGN AND METHODS: The BL-CFC assay and the OP9 system were combined to illustrate the hemangioblast with lymphomyeloid and vascular potential in the mouse aorta-gonad-mesonephros region. The colony-forming assay, reverse transcriptase polymerase chain reaction analysis, immunostaining and flow cytometry were used to identify the hematopoietic potential, and Matrigel- or OP9-based methods were employed to evaluate endothelial progenitor activity. RESULTS: Functionally, the aorta-gonad-mesonephros-derived BL-CFC produced erythroid/myeloid progenitors, CD19(+) B lymphocytes, and CD3(+)TCRbeta(+) T lymphocytes. Meanwhile, the BL-CFC-derived adherent cells generated CD31(+) tube-like structures on OP9 stromal cells, validating the endothelial progenitor potential. The aorta-gonad-mesonephros-derived hemangioblast was greatly enriched in CD31(+), endomucin(+) and CD105(+) subpopulations, which collectively pinpoints the endothelial layer as the main location. Interestingly, the BL-CFC was not detected in yolk sac, placenta, fetal liver or embryonic circulation. Screening of candidate cytokines revealed that interleukin-3 was remarkable in expanding the BL-CFC in a dose-dependent manner through the JAK2/STAT5 and MAPK/ERK pathways. Neutralizing interleukin-3 in the aorta-gonad-mesonephros region resulted in reduced numbers of BL-CFC, indicating the physiological requirement for this cytokine. Both hematopoietic and endothelial differentiation potential were significantly increased in interleukin-3-treated BL-CFC, suggesting a persistent positive influence. Intriguingly, interleukin-3 markedly amplified primitive erythroid and macrophage precursors in E7.5 embryos. Quantitative polymerase chain reaction analysis demonstrated declined Flk-1 and elevated Scl and von Willebrand factor transcription upon interleukin-3 stimulation, indicating accelerated hemangiopoiesis. CONCLUSIONS: The hemangioblast with lymphomyeloid potential is one of the precursors of definitive hematopoiesis in the mouse aorta-gonad-mesonephros region. Interleukin-3 has a regulatory role with regards to both the number and capacity of the dual-potential hemangioblast.


Assuntos
Aorta/fisiologia , Gônadas/fisiologia , Hemangioblastos/fisiologia , Interleucina-3/fisiologia , Mesonefro/fisiologia , Animais , Aorta/citologia , Aorta/embriologia , Células Cultivadas , Técnicas de Cocultura , Gônadas/citologia , Gônadas/embriologia , Hemangioblastos/citologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mesonefro/citologia , Mesonefro/embriologia , Camundongos , Células Estromais/citologia , Células Estromais/fisiologia
9.
Dev Growth Differ ; 51(2): 135-43, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19207184

RESUMO

Although renal regeneration is limited to repair of the proximal tubule in mammals, some bony fish are capable of renal regeneration through nephron neogenesis in the event of renal injury. We previously reported that nephron development in the medaka mesonephros is characterized by four histologically distinct stages, generally referred to as condensed mesenchyme, nephrogenic body, relatively small nephron, and the mature nephron. Developing nephrons are positive for wt1 expression during the first three of these stages. In the present study, we examined the regenerative response to renal injury, artificially induced by the administration of sublethal amounts of gentamicin in adult medaka. Similar to previous reports in other animals, the renal tubular epithelium and the glomerulus of the medaka kidney exhibited severe damage after exposure to this agent. However, kidneys showed substantial recovery after gentamicin administration, and a significant number of developing nephrons appeared 14 days after gentamicin administration (P < 0.01). Similarly, the expression of wt1 in developing nephrons also indicated the early stages of nephrogenesis. These findings show that medaka has the ability to regenerate kidney through nephron neogenesis during adulthood and that wt1 is a suitable marker for detecting nephrogenesis.


Assuntos
Mesonefro/fisiologia , Néfrons/fisiologia , Regeneração/fisiologia , Células-Tronco Adultas/fisiologia , Animais , Biomarcadores , Divisão Celular , Gentamicinas/toxicidade , Masculino , Mesonefro/química , Mesonefro/efeitos dos fármacos , Néfrons/química , Oryzias , Especificidade da Espécie , Proteínas WT1/análise
10.
Br J Haematol ; 139(2): 321-30, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17897310

RESUMO

The transforming growth factor-beta-related factor bone morphogenetic protein 4 (BMP4) is expressed in the human embryonic aorta-gonad-mesonephros (AGM) coincident with the emergence of haematopoietic cells and influences postnatal mammalian haematopoietic stem cells in vitro. To investigate the role of BMP4 in mammalian embryonic haematopoiesis, cells were isolated from murine AGM and two populations of CD34(+) cells with different levels of c-Kit expression and multipotency were identified. CD34(+)/c-Kit(high) cells express CD45 and are haematopoietic-restricted progenitors. In contrast, CD34(+)/c-Kit(low) cells are Flk1+/CD45(neg) and generate adherent colonies in ex vivo culture that resemble haemangioblast colonies identified in other systems. The addition of BMP4 to AGM cells resulted in expansion of the CD34(+)/c-Kit(low) cell pool within 48 h, via a combination of down modulation of the c-Kit receptor in CD34(+)/c-Kit(high) cells and proliferation. In long-term culture, BMP4 increased the growth/survival of CD34(+)/c-Kit(high) haematopoietic progenitors, effects that were blocked by BMP inhibitors. CD34(+)/c-Kit(high) progenitors cultured with BMP4 also generated adherent colonies typical of c-Kit(low) cells. These results suggest that BMP4 regulates c-Kit expression and differentiation potential in CD34(+) AGM cells and supports a role for BMP signalling in the maintenance of multipotency during embryonic haematopoiesis, providing an insight into stem cell homeostasis within the mammalian haematopoietic niche.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Antígenos CD34/análise , Aorta/embriologia , Proteína Morfogenética Óssea 4 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas de Transporte/farmacologia , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Gônadas/embriologia , Mesonefro/fisiologia , Camundongos , Células-Tronco Multipotentes/imunologia , Técnicas de Cultura de Tecidos
11.
Endocrinology ; 148(8): 3704-10, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17495005

RESUMO

Testicular Leydig cells produce testosterone and provide the hormonal environment required for male virilization and spermatogenesis. In utero, fetal Leydig cells (FLCs) are necessary for the development of the Wolffian duct and male external genitalia. Steroidogenic factor 1 (Sf1) is a transcriptional regulator of hormone biosynthesis genes, thus serving a central role in the Leydig cell. Desert hedgehog (Dhh), a Sertoli cell product, specifies the FLC lineage in the primordial gonad through a paracrine signaling mechanism. Postnatally, FLCs are replaced in the testis by morphologically distinct adult Leydig cells (ALCs). To study a putative interaction between Sf1 and Dhh, we crossed Sf1 heterozygous mutant mice with Dhh homozygous null mice to test the function of these two genes in vivo. All of the compound Sf1(+/-); Dhh(-/-) mutants failed to masculinize and were externally female. However, embryonic gonads contained anastomotic testis cords with Sertoli cells and germ cells, indicating that sex reversal was not attributable to a fate switch of the early gonad. Instead, external feminization was attributable to the absence of differentiated FLCs in XY compound mutant mice. ALCs also failed to develop, suggesting either a dependence of ALCs on the prenatal establishment of Leydig cell precursors or that Sf1 and Dhh are both required for ALC maturation. In summary, this study provides genetic evidence that combinatorial expression of the paracrine factor Dhh and nuclear transcription factor Sf1 is required for Leydig cell development.


Assuntos
Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Intersticiais do Testículo/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Testículo/embriologia , Testículo/fisiologia , Fatores de Transcrição/metabolismo , Animais , Hormônio Antimülleriano , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Transtornos do Desenvolvimento Sexual , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/citologia , Células Germinativas/fisiologia , Glicoproteínas/metabolismo , Proteínas Hedgehog/genética , Proteínas de Homeodomínio/genética , Células Intersticiais do Testículo/citologia , Masculino , Mesonefro/embriologia , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Mitose , Gravidez , Receptores Citoplasmáticos e Nucleares/genética , Diferenciação Sexual/fisiologia , Fator Esteroidogênico 1 , Hormônios Testiculares/metabolismo , Testículo/citologia , Fatores de Transcrição/genética
12.
J Vasc Res ; 43(6): 581-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17028443

RESUMO

BACKGROUND/AIMS: Renal senescence during normal aging is associated with specific vascular alterations and tissue degeneration. Although the degenerative program executed during embryonic kidney development is known to include vascular alterations, studies yet have to examine whether it involves replicative senescence. In this study, we assessed the potential of the quail mesonephros, a transitory embryonic kidney, as a model of human renal senescence. METHODS: Quail embryos with developing or degenerating mesonephros were studied on day 6 or day 11 of incubation, respectively. Senescence-associated beta-galactosidase activity, a marker of replicative senescence, was examined on whole mounts and sections. Senescent vascular characterization was performed by the scanning electron-microscopic analysis of vascular corrosion casts. RESULTS: Senescence-associated beta-galactosidase activity was found only in old mesonephros. Moreover, at 11 days of incubation glomerular capillaries showed discontinuities and were thinner and more tortuous than those observed at 6 days, characteristics also reported for the aging human kidney. CONCLUSION: The degenerating quail mesonephros is a potential model of renal senescence, showing biochemical and morphological characteristics of the aging human kidney.


Assuntos
Envelhecimento/fisiologia , Coturnix , Mesonefro/fisiologia , Modelos Animais , Animais , Biomarcadores , Embrião de Galinha , Mesonefro/anormalidades , Mesonefro/ultraestrutura , Microscopia Eletrônica de Varredura , Circulação Renal , beta-Galactosidase/metabolismo
13.
Endocrinology ; 147(8): 3777-88, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16675530

RESUMO

Fibroblast growth factor 9 (FGF9) signal has a role in organogenesis of the mammalian testis by controlling migration of mesonephric cells to the XY gonad, but neither it nor the FGF receptors is expressed sex-specifically. Of the Sprouty genes encoding antagonists of receptor tyrosine kinases including FGFr, mSprouty2 expression was confined to the developing testis and mesonephros. Gain of SPROUTY2 function in the male genital ridge and mesonephros malformed the vas deferens and epididymis, and diminished the number of seminiferous tubules and interstitium associating with reduced mesonephric cell migration and Fgf9 expression in embryonic testis, whereas exogenous FGF9 signaling recovered mesonephric cell migration inhibited by SPROUTY2. These phenotypes associated also with the decreased expression of Sox9, Desert hedgehog, Hsd3beta, Platelet/endothelial cell adhesion molecule, and alpha-smooth muscle actin, which are markers of the Sertoli, Leydig, endothelial, and peritubular myoid cells of the developing testis. Based on these data, we propose that the Sprouty proteins are involved normally in mediating the sexually dimorphic signaling of FGF9 and controlling cell migration from the mesonephros during testis development.


Assuntos
Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas/metabolismo , Testículo/embriologia , Testículo/fisiologia , 3-Hidroxiesteroide Desidrogenases/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Movimento Celular/fisiologia , Células Endoteliais/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fator 9 de Crescimento de Fibroblastos/metabolismo , Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Homeodomínio LIM , Laminina/genética , Células Intersticiais do Testículo/fisiologia , Masculino , Proteínas de Membrana , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Miócitos de Músculo Liso/fisiologia , Técnicas de Cultura de Órgãos , Fator de Transcrição PAX2/genética , Gravidez , Regiões Promotoras Genéticas/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Diferenciação Sexual/fisiologia , Testículo/citologia , Fatores de Transcrição , Proteínas Wnt/genética , Proteína Wnt4 , Ductos Mesonéfricos/citologia , Ductos Mesonéfricos/embriologia , Ductos Mesonéfricos/fisiologia
14.
Genes Dev ; 19(19): 2331-42, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16166372

RESUMO

Identifying the molecular pathways regulating hematopoietic stem cell (HSC) specification, self-renewal, and expansion remains a fundamental goal of both basic and clinical biology. Here, we analyzed the effects of Notch signaling on HSC number during zebrafish development and adulthood, defining a critical pathway for stem cell specification. The Notch signaling mutant mind bomb displays normal embryonic hematopoiesis but fails to specify adult HSCs. Surprisingly, transient Notch activation during embryogenesis via an inducible transgenic system led to a Runx1-dependent expansion of HSCs in the aorta-gonad-mesonephros (AGM) region. In irradiated adults, Notch activity induced runx1 gene expression and increased multilineage hematopoietic precursor cells approximately threefold in the marrow. This increase was followed by the accelerated recovery of all the mature blood cell lineages. These data define the Notch-Runx pathway as critical for the developmental specification of HSC fate and the subsequent homeostasis of HSC number, thus providing a mechanism for amplifying stem cells in vivo.


Assuntos
Linhagem da Célula/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Aorta/citologia , Aorta/fisiologia , Células Sanguíneas/citologia , Células Sanguíneas/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula/efeitos da radiação , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Desenvolvimento Embrionário/fisiologia , Desenvolvimento Embrionário/efeitos da radiação , Raios gama , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Gônadas/citologia , Gônadas/fisiologia , Hematopoese/efeitos da radiação , Células-Tronco Hematopoéticas/citologia , Homeostase/fisiologia , Homeostase/efeitos da radiação , Mesonefro/citologia , Mesonefro/fisiologia , Mutação , Irradiação Corporal Total/métodos , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
15.
J Anat ; 206(2): 165-74, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15730481

RESUMO

beta-Catenin is a structural component of adherens junctions, a regulator of the Wnt signalling pathway and a transcriptional co-activator with a key role in vascular patterning. The avian mesonephros is a transitory embryonic kidney that is used in the study of vascular development and degeneration. Here we examine beta-catenin expression in this model during vascular development and degeneration. Quail embryos with developing or degenerating mesonephros were studied, on day 6 (30HH) or day 11 of incubation (40HH), respectively. QH1 whole mounts of developing mesonephros revealed numerous angioblast-like cells situated in the paramesonephric duct that seem to invade the mesonephros. Although these cells did not express beta-catenin, the surrounding periductal mesenchymal cells translocated high levels of beta-catenin into the nucleus. In contrast, degenerating mesonephros were devoid of angioblast-like cells and beta-catenin was lower than in the developing mesonephros. beta-Catenin was significantly reduced in the glomerular capillary tuffs, indicating that it was particularly down-regulated in the vascular system. No sex-related differences in beta-catenin expression were observed in degenerating mesonephros. Furthermore, two special populations of glomerular and peritubular endothelial cells were observed in degenerating mesonephros: one translocating beta-catenin into the nucleus and the other in apoptosis that did not translocate it. In conclusion, our results indicate that the paramesonephric duct is a potential new vasculogenetic pathway, and suggest that beta-catenin plays a role in the fate of mesonephric endothelial cells.


Assuntos
Proteínas do Citoesqueleto/análise , Mesonefro/fisiologia , Codorniz/embriologia , Transativadores/análise , Animais , Apoptose , Transporte Biológico , Vasos Sanguíneos , Western Blotting/métodos , Núcleo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Feminino , Imuno-Histoquímica/métodos , Masculino , Mesonefro/irrigação sanguínea , Microscopia Eletrônica de Varredura , Transativadores/metabolismo , beta Catenina
16.
J Exp Med ; 200(7): 871-82, 2004 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-15466621

RESUMO

GATA-2 is an essential transcription factor in the hematopoietic system that is expressed in hematopoietic stem cells (HSCs) and progenitors. Complete deficiency of GATA-2 in the mouse leads to severe anemia and embryonic lethality. The role of GATA-2 and dosage effects of this transcription factor in HSC development within the embryo and adult are largely unexplored. Here we examined the effects of GATA-2 gene dosage on the generation and expansion of HSCs in several hematopoietic sites throughout mouse development. We show that a haploid dose of GATA-2 severely reduces production and expansion of HSCs specifically in the aorta-gonad-mesonephros region (which autonomously generates the first HSCs), whereas quantitative reduction of HSCs is minimal or unchanged in yolk sac, fetal liver, and adult bone marrow. However, HSCs in all these ontogenically distinct anatomical sites are qualitatively defective in serial or competitive transplantation assays. Also, cytotoxic drug-induced regeneration studies show a clear GATA-2 dose-related proliferation defect in adult bone marrow. Thus, GATA-2 plays at least two functionally distinct roles during ontogeny of HSCs: the production and expansion of HSCs in the aorta-gonad-mesonephros and the proliferation of HSCs in the adult bone marrow.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Dosagem de Genes , Células-Tronco Hematopoéticas/fisiologia , Fatores de Transcrição/fisiologia , Animais , Aorta/fisiologia , Medula Óssea/fisiologia , Radioisótopos de Césio , Proteínas de Ligação a DNA/genética , Feminino , Citometria de Fluxo , Fator de Transcrição GATA2 , Gônadas/fisiologia , Imuno-Histoquímica , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Reação em Cadeia da Polimerase/métodos , Baço/anatomia & histologia , Fatores de Tempo , Fatores de Transcrição/genética
17.
Blood ; 103(11): 4084-92, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-14962910

RESUMO

Apoptosis is an essential process in embryonic tissue remodeling and adult tissue homeostasis. Within the adult hematopoietic system, it allows for tight regulation of hematopoietic cell subsets. Previously, it was shown that B-cell leukemia 2 (Bcl-2) overexpression in the adult increases the viability and activity of hematopoietic cells under normal and/or stressful conditions. However, a role for apoptosis in the embryonic hematopoietic system has not yet been established. Since the first hematopoietic stem cells (HSCs) are generated within the aortagonad-mesonephros (AGM; an actively remodeling tissue) region beginning at embryonic day 10.5, we examined this tissue for expression of apoptosis-related genes and ongoing apoptosis. Here, we show expression of several proapoptotic and antiapoptotic genes in the AGM. We also generated transgenic mice overexpressing Bcl-2 under the control of the transcriptional regulatory elements of the HSC marker stem cell antigen-1 (Sca-1), to test for the role of cell survival in the regulation of AGM HSCs. We provide evidence for increased numbers and viability of Sca-1(+) cells in the AGM and subdissected midgestation aortas, the site where HSCs are localized. Most important, our in vivo transplantation data show that Bcl-2 overexpression increases AGM and fetal liver HSC activity, strongly suggesting that apoptosis plays a role in HSC development.


Assuntos
Apoptose/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fatores Etários , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Aorta/embriologia , Aorta/fisiologia , Sobrevivência Celular/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Gônadas/embriologia , Gônadas/fisiologia , Fígado/embriologia , Fígado/fisiologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesonefro/embriologia , Mesonefro/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Gravidez , Proteínas Proto-Oncogênicas c-kit/metabolismo
18.
Biol Reprod ; 68(6): 2033-7, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12606390

RESUMO

The first morphological event after initiation of male sex determination is seminiferous cord formation in the embryonic testis. Cord formation requires migration of pre-peritubular myoid cells from the adjacent mesonephros. The embryonic Sertoli cells are the first testicular cells to differentiate and have been shown to express neurotropin-3 (NT3), which can act on high-affinity trkC receptors expressed on migrating mesonephros cells. NT3 expression is elevated in the embryonic testis during the time of seminiferous cord formation. A trkC receptor tyrophostin inhibitor, AG879, was found to inhibit seminiferous cord formation and mesonephros cell migration. Beads containing NT3 were found to directly promote mesonephros cell migration into the gonad. Beads containing other growth factors such as epidermal growth factor (EGF) did not influence cell migration. At male sex determination the SRY gene promotes testis development and the expression of downstream sex differentiation genes such as SOX-9. Inhibition of NT3 actions caused a reduction in the expression of SOX-9. Combined observations suggest that when male sex determination is initiated, the developing Sertoli cells express NT3 as a chemotactic agent for migrating mesonephros cells, which are essential to promote embryonic testis cord formation and influence downstream male sex differentiation.


Assuntos
Quimiotaxia/fisiologia , Neurotrofina 3/fisiologia , Processos de Determinação Sexual , Testículo/embriologia , Animais , Cromatografia em Agarose , Feminino , Genes sry/genética , Proteínas de Grupo de Alta Mobilidade/genética , Imuno-Histoquímica , Masculino , Mesonefro/citologia , Mesonefro/fisiologia , Técnicas de Cultura de Órgãos , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptor trkC/fisiologia , Fatores de Transcrição SOX9 , Túbulos Seminíferos/embriologia , Células de Sertoli/fisiologia , Testículo/citologia , Fatores de Transcrição/genética
19.
Curr Opin Immunol ; 14(2): 186-91, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11869891

RESUMO

During embryogenesis there is a sequential, temporal appearance of increasingly more-complex hematopoietic cells beginning with unipotential progenitors, proceeding to multipotential (myeloid, erythroid and lymphoid) progenitors and culminating with adult-repopulating hematopoietic stem cells. Current research has established an important role for the aorta-gonads-mesonephros region of the mouse embryo in the generation of multipotential progenitors and hematopoietic stem cells. Comparisons of normal and hematopoietic-cell-mutant mouse embryos have revealed several genes pivotal in hematopoietic stem cell generation/function. Other genes have been implicated in the critical generation of lymphoid lineage potential. Thus, an understanding of the cellular and molecular interactions within the midgestation aorta-gonads-mesonephros region offers insight into the mechanisms of hematopoietic lineage specification during ontogeny and perhaps will lead to a more complete knowledge of the adult hematopoietic system.


Assuntos
Sistema Hematopoético/embriologia , Tecido Linfoide/embriologia , Mesonefro/embriologia , Proteínas Proto-Oncogênicas , Animais , Aorta/citologia , Aorta/embriologia , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/fisiologia , Gônadas/citologia , Gônadas/embriologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Sistema Hematopoético/citologia , Sistema Hematopoético/fisiologia , Fator de Transcrição Ikaros , Tecido Linfoide/citologia , Tecido Linfoide/fisiologia , Mesonefro/citologia , Mesonefro/fisiologia , Camundongos , Morfogênese , Mutação , Fatores de Transcrição/fisiologia , Saco Vitelino/embriologia , Saco Vitelino/fisiologia
20.
Kidney Int ; 61(4): 1231-42, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11918729

RESUMO

BACKGROUND: In rats, offspring born to mothers supplied low protein diets during pregnancy have fewer glomeruli than normal. We hypothesized that such nephron deficits are associated with altered cell turnover in the metanephros, the embryonic precursor of the adult kidney. METHODS: Wistar rats were supplied with one of three isocaloric diets from day 0 of pregnancy: control (18% protein) or low protein (9% or 6%) diets. All had a normal chow after birth. Groups were compared by multilevel statistical modeling. RESULTS: At two weeks postnatally, when nephrogenesis has finished, controls had 16.8 x 103 +/- 0.7 x 10(3) (mean +/- SEM) glomeruli/kidney, whereas offspring exposed to 9% diet had 5.1 x 10(3) +/- 1.2 x 10(3) fewer and those exposed to 6% diet had 6.9 x 10(3) +/- 1.7 x 10(3) fewer glomeruli/kidney (P < 0.001, both diets). At embryonic day 13 (E13), when the metanephros has just formed, control metanephroi contained 2.35 x 10(4) +/- 0.15 x 10(4) cells, with no significant differences in low protein groups. At E15, when mesenchyme begins forming primitive nephrons but glomeruli are still absent, controls had 2.00 x 10(6) +/- 0.13 x 10(6) cells. E15 embryos exposed to 9% protein had 1.09 x 10(6) +/- 0.36 x 10(6) fewer cells/metanephros than controls, while those exposed to 6% diet had 1.45 x 10(6) +/- 0.37 x 10(6) fewer (P < 0.01, both diets). Apoptotic cells were detected by molecular (in-situ end-labeling) and morphological (propidium iodide staining) techniques. In all diets, apoptosis was noted in condensing mesenchyme (nephron precursors) and loose mesenchyme (interstitial precursors). Control E13 metanephroi had 63 +/- 7 apoptotic cells/mm2, whereas those exposed to 9% diet had an increase of 77 +/- 26 cells/mm2 (P < 0.01) and those exposed to 6% diet had an increase of 55 +/- 26 cells/mm2 (P < 0.05). By E15, apoptosis was similar in all groups but metanephric mitosis was significantly increased in the 6% protein diet group. No change was found in the level of apoptosis in E13 mesonephroi. CONCLUSIONS: Maternal low protein diets reduce final numbers of glomeruli in association with enhanced deletion of mesenchymal cells at the start of kidney development. Whether aberrant nephrogenesis is a direct effect from deletion of nephron precursors, or an indirect effect from loss of supportive interstitial precursors, requires further investigation.


Assuntos
Apoptose/fisiologia , Dieta com Restrição de Proteínas , Rim/embriologia , Mesoderma/fisiologia , Prenhez/fisiologia , Animais , Contagem de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário e Fetal , Feminino , Glomérulos Renais/anatomia & histologia , Tamanho da Ninhada de Vivíparos , Mesonefro/citologia , Mesonefro/fisiologia , Mitose , Placentação , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...