Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
F S Sci ; 3(4): 340-348, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35728767

RESUMO

OBJECTIVE: To investigate the advantages of cryopreserved medulla-containing ovarian cortex grafts with those of commonly used sole cortex grafts for fertility preservation by analyzing tissue quality, neovascularization processes, and the number of vital follicles. DESIGN: Experimental setting of cryopreserved bovine ovarian cortex tissue grafts with or without medulla tissue. SETTING: Laboratory animal research at Ulm University, Ulm, Germany. ANIMALS: Bovine ovaries and fertilized chicken eggs. INTERVENTION(S): Experimental setting of bovine ovarian tissue samples grafted on the chicken chorioallantoic membrane (CAM) after cryopreservation and thawing to examine histologic tissue integrity, apoptosis and proliferation immunohistochemically, blood vessel counts and determine the presence of neutral red-stained vital follicles. MAIN OUTCOME MEASURE(S): We used hematoxylin and eosin staining to visualize tissue structures, immunohistochemistry with anti-caspase 3 to detect apoptosis, anti-Ki67 to examine proliferation, blood vessel count on the chicken CAM to investigate neovascularization processes, and neutral red staining to evaluate vital follicles. RESULT(S): We demonstrated that in all analyzed tissue samples, after cryopreservation, thawing, and grafting on the chicken CAM, there was excellent tissue integrity and quality, as shown by extremely rare apoptosis processes analyzed using immunohistochemical caspase 3 staining (sole cortex, 0.54%; thin medulla-containing cortex, 0.43%; thick medulla-containing cortex, 0.13%; and sole medulla, 2.82%). Moreover, we detected increased neovascularization in the vicinity of medulla and medulla-containing grafts (small blood vessels: cortex 8.7, thin medulla-containing cortex 9.9, thick medulla-containing cortex 9.7, and medulla 9.8; very small blood vessels: cortex 7.0, thin medulla-containing cortex 13.0, thick medulla-containing cortex 12.0, and medulla 15.0), with higher Ki67-detected proliferation (cortex, 17.58%; thin medulla-containing cortex, 20.28%; thick medulla-containing cortex, 20.56%; and medulla, 29.9%). Additionally, we identified an increased number of vital follicles in medulla-containing cortex grafts compared with the number of vital follicles in sole cortex tissue (cortex, 256.1; thin medulla-containing cortex, 338.2; thick medulla-containing cortex, 346.6; and medulla, 8.1). CONCLUSION(S): In this experimental setting, bovine medulla-containing cortex tissue had excellent tissue structure and quality after cryopreservation and thawing and increased neovascularization and an augmented vital follicle count after grafting than the commonly used sole cortex tissue. Therefore, we suggest reconsidering the current cryopreservation and grafting processes in humans for fertility preservation by favoring retain medulla tissue at the ovarian cortex.


Assuntos
Preservação da Fertilidade , Humanos , Feminino , Bovinos , Animais , Ovário/transplante , Vermelho Neutro , Folículo Ovariano/transplante , Criopreservação/veterinária , Neovascularização Patológica
2.
Arch Gynecol Obstet ; 300(5): 1435-1443, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31599349

RESUMO

PURPOSE: To evaluate whether the use of a commercially available dimethylxanthine theophylline compound (SpermMobil®) for artificial sperm activation would negatively affect clinical, obstetric and perinatal outcomes. METHODS: Artificial sperm activation (ASA) was used when sperm motility after preparation was low or absent in our clinical standard procedure practice. ICSI cycles using either testicular or ejaculated sperm with concentration smaller than 5 million/ml from August 2012 to January 2018 were retrospectively analyzed (n = 815) and divided into two groups, a control group where no ASA was needed and the SpermMobil® group with ASA. RESULTS: The fertilization rate was significantly higher in the control group, but pregnancy and implantation rates did not differ significantly. Number of embryos transferred, good quality embryos for ET and number of frozen blastocysts were similar in both groups. Clinical pregnancy loss was significantly reduced in the SpermMobil® group, which was reflected in slightly better live birth rates than in the control group. Furthermore, there were no significant differences regarding gestational age, weight, height and z score for singletons or multiples in the SpermMobil® (n = 27 and n = 10) or control (n = 144 and n = 67) groups. There were no reports of malformation, perinatal mortality or intensive therapy in the SpermMobil® group, whereas in the control group, 12 babies needed intensive care, besides one intrauterine death. CONCLUSION: The use of SpermMobil® in samples with mostly immotile sperm not only facilitates the embryologists work but also optimizes the treatment outcomes for those patients with a bad prognosis. This is the first report of obstetric and perinatal outcomes after applying a theophylline derivative in human clinical use.


Assuntos
Fertilização in vitro/métodos , Injeções de Esperma Intracitoplásmicas/métodos , Teofilina/efeitos adversos , Teofilina/uso terapêutico , Adulto , Feminino , Humanos , Masculino , Gravidez , Resultado da Gravidez , Estudos Retrospectivos , Teofilina/farmacologia
3.
Dev Dyn ; 244(4): 591-606, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25690936

RESUMO

BACKGROUND: Pluripotency, self-renewal, and differentiation are special features of embryonic stem (ES) cells, thereby providing valuable perspectives in regenerative medicine. Developmental processes require a fine-tuned organization, mainly regulated by the well-known JAK/STAT, PI3K/AKT, and ERK/MAPK pathways. SPREDs (Sprouty related proteins with EVH1 domain) were discovered as inhibitors of the ERK/MAPK signaling pathway, whereas nothing was known about their functions in ES cells and during early differentiation, so far. RESULTS: We generated SPRED1 and SPRED2 overexpressing and SPRED2 knockout murine ES cells to analyze the functions of SPRED proteins in ES cells and during early differentiation. Overexpression of SPREDs increases significantly the self-renewal and clonogenicity of murine ES cells, whereas lack of SPRED2 reduces proliferation and increases apoptosis. During early differentiation in embryoid bodies, SPREDs promote the pluripotent state and inhibit differentiation whereby mesodermal differentiation into cardiomyocytes is considerably delayed and inhibited. LIF- and growth factor-stimulation revealed that SPREDs inhibit ERK/MAPK activation in murine ES cells. However, no effects were detectable on LIF-induced activation of the JAK/STAT3, or PI3K/AKT signaling pathway by SPRED proteins. CONCLUSIONS: We show that SPREDs promote self-renewal and inhibit mesodermal differentiation of murine ES cells by selective suppression of the ERK/MAPK signaling pathway in pluripotent cells.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fator Inibidor de Leucemia/metabolismo , Mesoderma/metabolismo , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
4.
Stem Cells ; 33(4): 1113-29, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25524439

RESUMO

During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.


Assuntos
Proteínas de Homeodomínio/biossíntese , Proteínas com Homeodomínio LIM/antagonistas & inibidores , Proteínas com Homeodomínio LIM/biossíntese , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/biossíntese , Animais , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/metabolismo , Células HEK293 , Proteína Homeobox Nkx-2.5 , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica/fisiologia , Xenopus
5.
Dev Dyn ; 243(5): 699-711, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24288149

RESUMO

BACKGROUND: ATIPs (Angiotensin receptor type 2 [AT2] interacting proteins) are described as AT2 interacting protein variants, whereas their expression and functions during development are not known yet. RESULTS: Here, we provide a detailed expression pattern of ATIP variants during mouse development by visualizing Mtus1 promotor activity, Mtus1 RNA, and subsequent ATIP protein expression. ATIPs are strongly expressed in the developing cardiovascular system, including the vascular plexus of the yolk sac and the fetal vascular part of the placenta. Moreover, ATIP is expressed spatially distinct during eye and limb bud development, and in later stages in lung and nervous system. The three murine ATIP isoforms are expressed in a distinct manner, whereupon isoform 1 and 4 are correlated to cardiovascular, lung, and limb bud development and isoform 3 is restricted to brain and eye development. Interestingly, Mtus1 expression is not necessarily correlated to Agtr2 expression, suggesting novel but yet unknown functions for ATIP, independent of AT2 signaling. CONCLUSIONS: ATIPs seem to be mainly involved in the developmental regulation of the cardiovascular system and may act in different AT2-dependent and -independent manners. Hence, these results deliver valuable information to further elucidate the different functions of ATIPs in the processes of mammalian development.


Assuntos
Sistema Cardiovascular/embriologia , Proteínas de Transporte/biossíntese , Embrião de Mamíferos/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Supressoras de Tumor/biossíntese , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Sistema Cardiovascular/citologia , Proteínas de Transporte/genética , Embrião de Mamíferos/citologia , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Proteínas Supressoras de Tumor/genética
6.
Stem Cells Dev ; 21(8): 1225-38, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21967637

RESUMO

Nanog is a homeodomain transcription factor associated with the acquisition of pluripotency. Genome analyses of lower and higher vertebrates revealed that the existence of Nanog is restricted to gnathostomata but absent from agnatha and invertebrates. To elucidate the function of Nanog in nonmammalia, we identified the Danio rerio ortholog of Nanog and characterized its role in gain and loss of function experiments. We found Nanog to be crucial for survival of early zebrafish embryos, because depletion of Nanog led to gastrulation defects with subsequent lethality. Mouse Nanog overexpression could rescue these defects. Vice versa, zebrafish Nanog was found to promote proliferation and to inhibit differentiation of mouse embryonic stem cells in the absence of leukemia inhibitory factor. These findings indicate functional conservation of Nanog from teleost fishes to mammals. However, Nanog was lost in the genome of the anurans Xenopus laevis and Xenopus tropicalis. Phylogenetic analysis revealed that deletion probably occurred in a common anuran ancestor along with chromosomal translocations. The closest homologs of Nanog in Xenopus are the Vent proteins. We, therefore, investigated whether the Xvent genes might substitute for Nanog function in Xenopus. Although we found some similarities in phenotypes after overexpression and in the regulation of several marker genes, Xvent1/2 and Nanog cannot substitute each other. Depletion of Nanog in zebrafish cannot be rescued by ectopic expression of Xvent, and Xvent depletion in Xenopus cannot be overcome by ectopic expression of zebrafish Nanog.


Assuntos
Proteínas de Homeodomínio/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , DNA/metabolismo , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Variação Genética/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Humanos , Fator Inibidor de Leucemia/farmacologia , Camundongos , Proteína Homeobox Nanog , Ligação Proteica/efeitos dos fármacos , Especificidade da Espécie , Proteínas de Xenopus/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
Dev Dyn ; 240(12): 2634-45, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22072574

RESUMO

The T-box transcription factor Tbx5 is involved in several developmental processes including cardiogenesis. Early steps of cardiac development are characterised by the formation of two cardiogenic lineages, the first (FHF) and the second heart field (SHF) lineage, which arise from a common cardiac progenitor cell population. To further investigate the function of Tbx5 during cardiogenesis, we generated a murine embryonic stem cell line constitutively overexpressing Tbx5. Differentiation of these cells is characterised by an earlier and increased appearance of contracting cardiomyocytes that beat with a higher frequency than control cells. In semi-quantitative and quantitative RT-PCR analyses, we observed an up-regulation of cardiac marker genes such as Troponin T, endogenous Tbx5, and Nkx2.5 and a down-regulation of others like BMP4 and Hand2. Similar data were gained in Xenopus laevis arguing for a conserved function of Tbx5. Furthermore, markers of the conduction system and atrial cardiomyocytes were increased.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Átrios do Coração/embriologia , Proteínas com Domínio T/biossíntese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Linhagem Celular , Regulação para Baixo/fisiologia , Células-Tronco Embrionárias/citologia , Átrios do Coração/citologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Troponina T/genética , Troponina T/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
8.
J Biol Chem ; 286(11): 9477-88, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21199868

RESUMO

Sprouty-related proteins with EVH1 (enabled/vasodilator-stimulated phosphoprotein homology 1) domain (SPREDs) are inhibitors of MAPK signaling. To elucidate SPRED2 in vivo function, we characterized body homeostasis in SPRED2(-/-) mice. They showed a doubled daily water uptake, induced by elevated serum osmolality, originating from increased blood salt load. Accordingly, serum aldosterone was doubled, accompanied by augmented adrenal aldosterone synthase (AS) expression. Surprisingly, serum vasopressin (AVP) was unaltered, and, as evidenced by halved angiotensin II (Ang II) levels, the renin angiotensin system (RAS) was down-regulated. Adrenocorticotropic hormone (ACTH) was significantly elevated in SPRED2(-/-) mice, together with its secretagogue corticotropin-releasing hormone (CRH) and its downstream target corticosterone. ERK phosphorylation in brains was augmented, and hypothalamic CRH mRNA levels were elevated, both contributing to the increased CRH release. Our data were supported by CRH promoter reporter assays in hypothalamic mHypoE-44 cells, revealing a SPRED-dependent inhibition of Ets (ERK/E-twenty-six)-dependent transcription. Furthermore, SPRED suppressed CRH production in these cells. In conclusion, our study suggests that SPRED2 deficiency leads to an increased MAPK signaling, which results in an augmented CRH promoter activity. The subsequent CRH overproduction causes an up-regulation of downstream hypothalamic-pituitary-adrenal (HPA) hormone secretion. This constitutes a possible trigger for the observed compulsive grooming in SPRED2(-/-) mice and may, together with hyperplasia of aldosterone-producing cells, contribute to the hyperaldosteronism and homeostatic imbalances.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Repressoras/metabolismo , Animais , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Fosforilação/fisiologia , Proteínas Repressoras/genética
9.
Dev Dyn ; 239(11): 3072-85, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20882678

RESUMO

SPREDs (Sprouty-related proteins with Ena/Vasodilator-stimulated phosphoprotein homology-1 domain) are known membrane-associated modulators of receptor tyrosine kinases by inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway. Although Spred2(-/-) mice exhibit dwarfism and increase of early haematopoiesis, the precise expression and role of SPRED2 in mouse development remains unknown. Here, we demonstrate a detailed Spred2 expression pattern during mouse development using X-Gal stainings from samples of a gene-trapped Spred2 mouse line. In early stages, Spred2 was highly expressed in ectodermal and mesodermal tissues, and later on in developing neural tissue, heart, lung, intestine, urogenital tract, and limbs. Strikingly, we observed that Spred2 was mainly expressed at leading edges of further outgrowing structures and in folds of newly forming grooves. Therefore, SPRED2 is likely involved in the regulation of dynamic developmental processes. These new data provide valuable information for further studies regarding the still enigmatic physiological SPRED functions during mouse development.


Assuntos
Proteínas Repressoras/metabolismo , Animais , Ectoderma/metabolismo , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética
10.
J Biol Chem ; 285(11): 8408-21, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20064932

RESUMO

Xenopus Oct25 is a POU family subclass V (POU-V) transcription factor with a distinct domain structure. To investigate the contribution of different domains to the function of Oct25, we have performed gain of function analyses. Deletions of the N- or C-terminal regions and of the Hox domain (except its nuclear localization signal) result in mutants being indistinguishable from the wild type protein in the suppression of genes promoting germ layer formation. Deletion of the complete POU domain generates a mutant that has no effect on embryogenesis. However, disruption of the alpha-helical structures in the POU domain, even by a single amino acid mutation, causes reversal of protein function. Overexpression of such mutants leads to dorsalization of embryos and formation of secondary axial structures. The underlying mechanism is an enhanced transcription of genes coding for antagonists of the ligands for ventralizing bone morphogenetic protein and Wnt pathways. Corresponding deletion mutants of Xenopus Oct60, Oct91, or mouse Oct4 also exhibit such a dominant-negative effect. Therefore, our results reveal that the integrity of the POU domain is crucial for the function of POU-V transcription factors in the regulation of genes that promote germ layer formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fatores do Domínio POU , Proteínas de Xenopus , Xenopus/genética , Ativinas/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Deleção de Genes , Humanos , Rim/citologia , Camundongos , Mutagênese/fisiologia , Proteína Nodal/metabolismo , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fatores do Domínio POU/química , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Transcrição Gênica/fisiologia , Xenopus/embriologia , Proteínas de Xenopus/química , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , beta Catenina/metabolismo
11.
Bioessays ; 29(9): 897-907, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17691106

RESUMO

Spreds form a new protein family with an N-terminal Enabled/VASP homology 1 domain (EVH1), a central c-Kit binding domain (KBD) and a C-terminal Sprouty-related domain (SPR). They are able to inhibit the Ras-ERK signalling pathway after various mitogenic stimulations. In mice, Spred proteins are identified as regulators of bone morphogenesis, hematopoietic processes, allergen-induced airway eosinophilia and hyperresponsiveness. They inhibit cell motility and metastasis and have a high potential as tumor markers and suppressors of carcinogenesis. Moreover, in vertebrates, XtSpreds help together with XtSprouty proteins to coordinate gastrulation and mesoderm specification. Here, we give an overview of this new field and summarize the domain functions, binding partners, expression patterns and the cellular localizations, regulations and functions of Spred proteins and try to give perspectives for future scientific directions.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Proteínas Adaptadoras de Transdução de Sinal , Animais , Regulação da Expressão Gênica , Hematopoese , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Morfogênese , Neoplasias/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas ras/metabolismo
12.
J Biol Chem ; 281(48): 36477-81, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16987806

RESUMO

Sprouty-related proteins with an EVH1 domain (Spreds) belong to a new protein family harboring a conserved N-terminal EVH1 domain, which is related to the VASP (vasodilator-stimulated phosphoprotein) EVH1 domain (Enabled/VASP homology 1 domain) and a C-terminal Sprouty-related domain, typical for Sprouty proteins. Spreds were, like Sproutys, initially discovered as inhibitors of the Ras/MAPK pathway, and the SPR (Sprouty-related) domains of both protein families seem to be very important for many protein interactions and cellular processes. VASP was initially characterized as a proline-rich substrate of protein kinases A and G in human platelets and later shown to be a scaffold protein, regulating both signal transduction pathways and the actin filament system. The VASP-EVH1 domain is known to bind specifically to a FP(4) binding motif, which is, for example, present in the focal adhesion proteins vinculin and zyxin. In this review we give a structural and functional overview on these three protein families and ask whether nature plays a modular protein domain puzzle with stable exchangeable elements or if these closely related domains have various functions when pasted in a different protein context.


Assuntos
Moléculas de Adesão Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas de Membrana/fisiologia , Proteínas dos Microfilamentos/fisiologia , Fosfoproteínas/fisiologia , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Plaquetas/metabolismo , Proteínas do Citoesqueleto/química , Glicoproteínas/química , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Estrutura Terciária de Proteína , Transdução de Sinais , Vinculina/química , Zixina
13.
Gene Expr Patterns ; 6(3): 247-55, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16378760

RESUMO

Spreds (Sprouty-related proteins with an Ena/Vasodilator-stimulated phosphoprotein homology-1 domain) are a new protein family inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway. Different RNA and protein studies already revealed an almost ubiquitous Spred-2 expression pattern. But until now, only few data were available on the in situ Spred-2 promoter activity. Here, we show a detailed in situ analysis of a mouse strain with a trapped Spred-2 gene, bringing a beta-galactosidase and neomycin fusion gene (beta-geo) under the control of the endogenous Spred-2 promoter. This allowed us to monitor Spred-2 promoter activity in practically every organ and their corresponding sub-compartments. X-Gal staining of newborn and adult mice revealed a nearly congruent Spred-2 promoter activity pattern. Our detailed data provide information for further studies of the still enigmatic physiological functions of Spred-2 in various organs by identifying the tissues with strong Spred-2 promoter activity.


Assuntos
Marcação de Genes , Vetores Genéticos , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Animais , Animais Recém-Nascidos , Linhagem Celular , Cruzamentos Genéticos , DNA Complementar/genética , Éxons , Feminino , Galactosídeos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Imuno-Histoquímica , Indóis/metabolismo , Íntrons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso/metabolismo , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Células-Tronco/citologia
14.
J Biol Chem ; 280(31): 28572-80, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15946934

RESUMO

The impact of the fibroblast growth factor receptor 3 (FGFR3)-mediated signaling pathway on bone growth has been demonstrated by various genetic approaches. Overexpression of fibroblast growth factors (FGFs), several gain-of-function mutations in the FGFR3, and constitutive activation of mitogen-activated protein kinase (MAPK) kinase (MEK1) in chondrocytes have been shown to cause dwarfism in mice by activation of the MAPK signaling pathway. To investigate the previously reported inhibitory role of Spred in the FGFR3/MAPK pathway, we generated mice with a trapped Spred-2 gene. Here we show that lack of functional Spred-2 protein in mice caused a dwarf phenotype, similar to achondroplasia, the most common form of human dwarfism. Spred-2(-/-) mice showed reduced growth and body weight, they had a shorter tibia length, and showed narrower growth plates as compared with wild-type mice. We detected promoter activity and protein expression of Spred-2 in chondrocytes, suggesting an important function of Spred-2 in chondrocytes and bone development. Stimulation of chondrocytes with different FGF concentrations showed earlier and augmented ERK phosphorylation in Spred-2(-/-) chondrocytes in comparison to Spred-2(+/+) chondrocytes. Our observations suggest a model in which loss of Spred-2 inhibits bone growth by inhibiting chondrocyte differentiation through up-regulation of the MAPK signaling pathway.


Assuntos
Nanismo/genética , Proteínas Repressoras/genética , Animais , Desenvolvimento Ósseo/genética , Nanismo/patologia , Feminino , Deleção de Genes , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Histochem Cell Biol ; 122(6): 527-38, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15580519

RESUMO

Spred-1 and Spred-2 (Sprouty-related protein with an EVH1 domain) are recently described members of the EVH1 (Ena/VASP-homology domain 1) family. Both Spred-1 and Spred-2 are membrane-associated substrates of receptor tyrosine kinases and they act as negative regulators of the Ras pathway upon growth factor stimulation. Since the Spred family members seem to exert overlapping molecular functions, the isotype-specific function of each member remains enigmatic. To date, no comprehensive expression profiling of Spred proteins has been shown. Therefore, we compared mRNA and protein expression patterns of Spred-1 and Spred-2 systematically in mouse organs. Furthermore, we focused on the tissue-specific expression of Spred-2 in adult human tissues, the subcellular localization, and the potential role of Spred-2 in the organism. Our studies show that expression patterns of Spred-1 and Spred-2 differ markedly among various tissues and cell types. In mouse, Spred-1 and Spred-2 were found to be expressed predominantly in brain, whereas Spred-2 was found to be more widely expressed in various adult tissues than Spred-1. In humans, Spred-2 was found to be strongly expressed in glandular epithelia and, at the subcellular level, its immunoreactivity was associated with secretory vesicles. Using confocal microscopy we found Spred-2 to be strongly colocalized with Rab11 and, to a lesser extent, with Rab5a GTPase, an observation that was not made for Spred-1. We conclude that the two members of the recently discovered Spred protein family, Spred-1 and Spred-2, show a highly specific expression pattern in various tissues reflecting a specific physiological role for the individual Spred isoforms in these tissues. Furthermore, it becomes most likely that Spred-2 is involved in the regulation of secretory pathways.


Assuntos
Proteínas Repressoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Formação de Anticorpos , Especificidade de Anticorpos , Encéfalo/metabolismo , Decídua/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fígado/metabolismo , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Microscopia Confocal , Miocárdio/metabolismo , Gravidez , Proteínas Repressoras/biossíntese , Proteínas Repressoras/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo , Testículo/ultraestrutura , Proteínas rab de Ligação ao GTP/biossíntese
16.
J Biol Chem ; 279(27): 28220-6, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15078889

RESUMO

Calcium and Ca(2+)-dependent signals play a crucial role in sperm motility and mammalian fertilization, but the molecules and mechanisms underlying these Ca(2+)-dependent pathways are incompletely understood. Here we show that homozygous male mice with a targeted gene deletion of isoform 4 of the plasma membrane calcium/calmodulin-dependent calcium ATPase (PMCA), which is highly enriched in the sperm tail, are infertile due to severely impaired sperm motility. Furthermore, the PMCA inhibitor 5-(and-6)-carboxyeosin diacetate succinimidyl ester reduced sperm motility in wild-type animals, thus mimicking the effects of PMCA4 deficiency on sperm motility and supporting the hypothesis of a pivotal role of the PMCA4 on the regulation of sperm function and intracellular Ca(2+) levels.


Assuntos
ATPases Transportadoras de Cálcio/biossíntese , Fertilidade , Motilidade dos Espermatozoides , Processamento Alternativo , Animais , Northern Blotting , Southern Blotting , Western Blotting , Cálcio/metabolismo , Proteínas de Transporte de Cátions , Clonagem Molecular , DNA Complementar/metabolismo , Fertilização in vitro , Fluoresceínas/farmacologia , Corantes Fluorescentes/farmacologia , Genótipo , Humanos , Masculino , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , ATPases Transportadoras de Cálcio da Membrana Plasmática , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Succinimidas/farmacologia , Testículo/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...