Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 14(3): 643-668.e1, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35690337

RESUMO

BACKGROUND & AIMS: Mesenchymal-epithelial crosstalk (MEC) in the stomach is executed by pathways such as bone morphogenetic protein (BMP) and extracellular signal-regulated kinase (ERK). Mis-regulation of MEC disrupts gastric homeostasis and causes tumorigenesis. Protein Kinase A (PKA) crosstalks with BMP and ERK signaling; however, PKA function(s) in stomach development and homeostasis remains undefined. METHODS: We generated a novel Six2-Cre+/-PKAcαRfl/wt (CA-PKA) mouse in which expression of constitutive-active PKAcαR was induced in gastric mesenchyme progenitors. Lineage tracing determined spatiotemporal activity of Six2-Cre in the stomach. For phenotyping CA-PKA mice histological, co-immunofluorescence, immunoblotting, mRNA sequencing, and bioinformatics analyses were performed. RESULTS: Lineage tracing showed that Six2-Cre activity in the stomach is restricted to the mesenchymal compartment. CA-PKA mice showed disruption of gastric homeostasis characterized by aberrant mucosal development and epithelial hyperproliferation; ultimately developing multiple features of gastric corpus preneoplasia including decreased parietal cells, mucous cell hyperplasia, spasmolytic peptide expressing metaplasia with intestinal characteristics, and dysplastic and invasive cystic glands. Furthermore, mutant corpus showed marked chronic inflammation characterized by infiltration of lymphocytes and myeloid-derived suppressor cells along with the upregulation of innate and adaptive immune system components. Striking upregulation of inflammatory mediators and STAT3 activation was observed. Mechanistically, we determined there is an activation of ERK1/2 and downregulation of BMP/SMAD signaling characterized by marked upregulation of BMP inhibitor gremlin 1. CONCLUSIONS: We report a novel role of PKA signaling in gastric MEC execution and show that PKA activation in the gastric mesenchyme drives preneoplasia by creating a proinflammatory and proproliferative microenvironment associated with the downregulation of BMP/SMAD signaling and activation of ERK1/2.


Assuntos
Mucosa Gástrica , Estômago , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Mucosa Gástrica/patologia , Mesoderma/metabolismo , Camundongos
2.
Eur J Histochem ; 63(3)2019 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-31544449

RESUMO

Nephron progenitors (NPs) and nephrogenesis have been extensively studied in mice and humans and have provided insights into the mechanisms of renal development, disease and possibility of NP-based therapies. However, molecular features of NPs and their derivatives in the canine fetal kidney (CFK) remain unknown. This study was focused to characterize the expression of potential markers of canine NPs and their derivatives by immuno-fluorescence and western blot analysis. Transcription factors (TFs) SIX1 and SIX2, well-characterized human NP markers, were expressed in NPs surrounding the ureteric bud in the CFK. Canine NPs also expressed ITGA8 and NCAM1, surface markers previously used to isolate NPs from the mouse and human fetal kidneys. TF, PAX2 was detected in the ureteric bud, NPs and their derivative structures such as renal vesicle and S-shaped body. This study highlights the similarities in dog, mouse and human renal development and characterizes markers to identify canine NPs and their derivatives. These results will facilitate the isolation of canine NPs and their functional characterization to develop NP-based therapies for canine renal diseases.


Assuntos
Células Epiteliais/metabolismo , Néfrons/metabolismo , Fatores de Transcrição/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Cães/embriologia , Células Epiteliais/citologia , Feminino , Imunofluorescência , Néfrons/citologia , Gravidez
3.
Am J Pathol ; 188(1): 84-94, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29107072

RESUMO

Ectopic cAMP signaling is pathologic in polycystic kidney disease; however, its spatiotemporal actions are unclear. We characterized the expression of phosphorylated Creb (p-Creb), a target and mediator of cAMP signaling, in developing and cystic kidney models. We also examined tubule-specific effects of cAMP analogs in cystogenesis in embryonic kidney explants. In wild-type mice, p-Creb marked nephron progenitors (NP), early epithelial NP derivatives, ureteric bud, and cortical stroma; p-Creb was present in differentiated thick ascending limb of Henle, collecting duct, and stroma; however, it disappeared in mature NP-derived proximal tubules. In Six2cre;Frs2αFl/Fl mice, a renal cystic model, ectopic p-Creb stained proximal tubule-derived cystic segments that lost the differentiation marker lotus tetragonolobus lectin. Furthermore, lotus tetragonolobus lectin-negative/p-Creb-positive cyst segments (re)-expressed Ncam1, Pax2, and Sox9 markers of immature nephron structures and dedifferentiated proximal tubules after acute kidney injury. These dedifferentiation markers were co-expressed with p-Creb in renal cysts in Itf88 knockout mice subjected to ischemia and Six2cre;Pkd1Fl/Fl mice, other renal cystogenesis models. 8-Br-cAMP addition to wild-type embryonic kidney explants induced proximal tubular cystogenesis and p-Creb expression; these effects were blocked by co-addition of protein kinase A inhibitor. Thus p-Creb/cAMP signaling is appropriate in NP and early nephron derivatives, but disappears in mature proximal tubules. Moreover, ectopic p-Creb expression/cAMP signaling marks dedifferentiated proximal tubular cystic segments. Furthermore, proximal tubules are predisposed to become cystic after cAMP stimulation.


Assuntos
Desdiferenciação Celular/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Doenças Renais Císticas/metabolismo , Túbulos Renais Proximais/metabolismo , Animais , Modelos Animais de Doenças , Doenças Renais Císticas/patologia , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Knockout , Fosforilação
4.
Pediatr Res ; 82(6): 1022-1029, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29135976

RESUMO

BackgroundFibroblast growth factor receptor 2 (Fgfr2) deletion from murine peri-Wolffian duct stroma (ST) results in aberrant ureteric bud induction, abnormal ureteral insertion into the bladder, and high rates of vesicoureteral reflux (VUR). It is unclear which receptor docking protein(s) is/are responsible for Fgfr2 actions in these tissues. We investigated whether the docking protein, fibroblast receptor substrate 2α (Frs2α), had a role in peri-Wolffian duct ST similar to Fgfr2.MethodsWe conditionally deleted Frs2α in peri-Wolffian duct ST with a Tbx18cre mouse line (Frs2αST-/-). We assessed for ureteric induction defects and alterations in downstream targets mediating defects. We performed euthanized cystograms and assessed ureter-bladder junctions by three-dimensional (3D) reconstructions.ResultsEmbryonic day (E) 11.5 Frs2αST-/- embryos had many displaced ureteric bud induction sites when compared with controls. E11.0 Frs2αST-/- embryos had decreased Bmp4 expression and signaling, which can cause abnormal ureteric bud induction. Postnatal day 1 (P1) and P30 Frs2αST-/- mice had higher VUR rates and grades vs. CONTROLS: Mutant refluxing ureters that inserted improperly into the bladder had shortened intravesicular tunnels (IVTs) when compared with controlsConclusionFrs2αST-/- embryos have aberrant ureteric induction sites, improper ureteral insertion, shortened intravesicular lengths, and VUR. Induction site defects appear secondary to reduced Bmp4 expression, similar to Fgfr2 mutants.


Assuntos
Proteínas de Membrana/genética , Ureter/embriologia , Refluxo Vesicoureteral/genética , Ductos Mesonéfricos/metabolismo , Animais , Apoptose , Proteína Morfogenética Óssea 4/genética , Proliferação de Células , Camundongos , Camundongos Knockout , Ureter/patologia
5.
Sci Rep ; 6: 36736, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27853247

RESUMO

Six2cre-mediated deletion of Frs2α (Six2creFrs2αKO), a major fibroblast growth factor receptor (Fgfr) docking protein in mouse nephron progenitors results in perinatal renal hypoplasia; however, postnatal Six2creFrs2αKO kidneys develop cysts. We sought to determine the pathogenesis of Six2creFrs2αKO cyst formation. We performed histological assays, Western blots, and quantitative PCR (qPCR). While embryonic day (E) 18.5 Six2Frs2αKO kidneys were hypoplastic and not cystic, postnatal day (P) 7 mutants had proximal tubular-derived cysts that nearly replaced the renal parenchyma by P21. Mutants had high proximal tubular proliferation rates and interstitial fibrosis, similar to known polycystic kidney disease (PKD) models. Six2creFrs2αKO kidneys also had upregulation of Wnt/ßcatenin signaling, macrophage infiltration and chemokine production (e.g. ectopic Ccl2 in non-dilated proximal tubules), and augmented hedgehog signaling, features also seen in other PKD models. We saw increased Gli1 (hedgehog readout) in postnatal Six2creFrs2αKO interstitium and ectopic sonic hedgehog (Shh) in subsets of non-dilated P7 mutant proximal tubules (likely driving the stromal Gli expression). As ectopic tubular Shh and Ccl2 expression is seen after acute kidney injury (AKI), we interrogated another bone fide AKI marker, Kim1 and noted ectopic expression in P7 non-dilated proximal tubules. These observations suggest that aberrantly activated "AKI" pathways may drive pathogenesis in PKD.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Rim/embriologia , Proteínas de Membrana/genética , Doenças Renais Policísticas/genética , Fatores de Transcrição/genética , Animais , Apoptose , Nitrogênio da Ureia Sanguínea , Proliferação de Células , Quimiocinas/metabolismo , Fibrose/metabolismo , Proteínas Hedgehog/metabolismo , Receptor Celular 1 do Vírus da Hepatite A/genética , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Proteínas de Homeodomínio/metabolismo , Inflamação , Rim/crescimento & desenvolvimento , Rim/metabolismo , Túbulos Renais/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Doenças Renais Policísticas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , beta Catenina/metabolismo
6.
Sci Rep ; 6: 28132, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27324437

RESUMO

Activation of protein kinase A (PKA) by follicle stimulating hormone (FSH) transduces the signal that drives differentiation of ovarian granulosa cells (GCs). An unresolved question is whether PKA is sufficient to initiate the complex program of GC responses to FSH. We compared signaling pathways and gene expression profiles of GCs stimulated with FSH or expressing PKA-CQR, a constitutively active mutant of PKA. Both FSH and PKA-CQR stimulated the phosphorylation of proteins known to be involved in GC differentiation including CREB, ß-catenin, AKT, p42/44 MAPK, GAB2, GSK-3ß, FOXO1, and YAP. In contrast, FSH stimulated the phosphorylation of p38 MAP kinase but PKA-CQR did not. Microarray analysis revealed that 85% of transcripts that were up-regulated by FSH were increased to a comparable extent by PKA-CQR and of the transcripts that were down-regulated by FSH, 76% were also down-regulated by PKA-CQR. Transcripts regulated similarly by FSH and PKA-CQR are involved in steroidogenesis and differentiation, while transcripts more robustly up-regulated by PKA-CQR are involved in ovulation. Thus, PKA, under the conditions of our experimental approach appears to function as a master upstream kinase that is sufficient to initiate the complex pattern of intracellular signaling pathway and gene expression profiles that accompany GC differentiation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hormônio Foliculoestimulante Humano/metabolismo , Células da Granulosa/fisiologia , Ovário/citologia , Animais , Diferenciação Celular , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Feminino , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Mutação/genética , Ratos , Ratos Endogâmicos , Transdução de Sinais , Esteroides/metabolismo , beta Catenina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Semin Cell Dev Biol ; 59: 27-34, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26805442

RESUMO

PTPN11 (also known as SHP2) is a ubiquitously expressed non-receptor tyrosine phosphatase that regulates cell survival, proliferation, differentiation, migration and adhesion. Naturally occurring mutations in the PTPN11 gene cause Noonan and LEOPARD syndromes, two genetic disorders that are characterized by a spectrum of defects including male infertility. This review summarizes four cellular and molecular mechanisms by which PTPN11 acts to support male fertility. First, PTPN11 is required for the proliferation and survival of spermatogonial stem cells (SSCs) that are essential to replenish the germ cells that will become sperm. Second, PTPN11 regulation of cellular adhesion functions in Sertoli cells is required to maintain the blood-testis barrier (BTB) that protects meiotic and post-meiotic germ cells. Third, expression of PTPN11 in Sertoli cells is essential to prevent premature differentiation and exhaustion of the SSC population and to maintain the SSC niche. Finally, in Leydig cells, PTPN11 supports mitochondrial fusion and the expression of acyl-CoA synthetase (ACSL4) needed for the production of steroids including testosterone, which is required for fertility.


Assuntos
Fertilidade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Humanos , Masculino , Modelos Biológicos , Espermatogênese , Espermatozoides/citologia , Espermatozoides/enzimologia , Testículo/enzimologia
8.
Dev Biol ; 400(1): 82-93, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25641696

RESUMO

Previous studies using transgenic Pax3cre mice have revealed roles for fibroblast growth factor receptors (Fgfrs) and Fgfr substrate 2α (Frs2α) signaling in early metanephric mesenchyme patterning and in ureteric morphogenesis. The role of Fgfr/Frs2α signaling in nephron progenitors is unknown. Thus, we generated mouse models using BAC transgenic Six2EGFPcre (Six2cre) mediated deletion of Fgfrs and/or Frs2α in nephron progenitors. Six2cre mediated deletion of Fgfr1 or Fgfr2 alone led to no obvious kidney defects. Six2creFgfr1(flox/flox)Fgfr2(flox/flox) (Fgfr1/2(NP-/-)) mice generate a discernable kidney; however, they develop nephron progenitor depletion starting at embryonic day 12.5 (E12.5) and later demonstrate severe cystic dysplasia. To determine the role of Frs2α signaling downstream of Fgfr2 in Fgfr1/2(NP-/-) mice, we generated Six2cre(,)Fgfr1(flox/flox)Fgfr2(LR/LR) (Fgfr1(NP-/-)Fgfr2(LR/LR)) mice that have point mutations in the Frs2α binding site of Fgfr2. Like Fgfr1/2(NP-/-) mice, Fgfr1(NP-/-)Fgfr2(LR/LR) develop nephron progenitor depletion, but it does not start until E14.5 and older mice have less severe cystic dysplasia than Fgfr1/2(NP-/-) To determine the role of Frs2α alone in nephron progenitors, we generated Six2creFrs2'A(flox/flox) (Frs2a(NP-/-)) mice. Frs2a(NP-/-)mice also develop nephron progenitor depletion and renal cysts, although these occurred later and were less severe than in the other Six2cre mutant mice. The nephron progenitor loss in all Six2cre mutant lines was associated with decreased Cited1 expression and increased apoptosis versus controls. FAC-sorted nephron progenitors in Six2cre Frs2'A(flox/flox) mice demonstrated evidence of increased Notch activity versus controls, which likely drives the progenitor defects. Thus, Fgfr1 and Fgfr2 have synergistic roles in maintaining nephron progenitors; furthermore, Fgfr signaling in nephron progenitors appears to be mediated predominantly by Frs2α.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/fisiologia , Néfrons/embriologia , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Citometria de Fluxo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase , Receptores Notch/metabolismo , Transativadores/metabolismo
9.
Stem Cells ; 32(3): 741-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24123360

RESUMO

SHP2 is a widely expressed protein tyrosine phosphatase required for signal transduction from multiple cell surface receptors. Gain and loss of function SHP2 mutations in humans are known to cause Noonan and LEOPARD syndromes, respectively, that are characterized by numerous pathological conditions including male infertility. Using conditional gene targeting in the mouse, we found that SHP2 is required for maintaining spermatogonial stem cells (SSCs) and the production of germ cells required for male fertility. After deleting SHP2, spermatogenesis was halted at the initial step during which transit-amplifying undifferentiated spermatogonia are produced from SSCs. In the absence of SHP2, proliferation of SSCs and undifferentiated spermatogonia was inhibited, thus germ cells cannot be replenished and SSCs cannot undergo renewal. However, germ cells beyond the undifferentiated spermatogonia stage of development at the time of SHP2 knockout were able to complete their maturation to become sperm. In cultures of SSCs and their progeny, inhibition of SHP2 activity reduced growth factor-mediated intracellular signaling that regulates SSC proliferation and cell fate. Inhibition of SHP2 also decreased the number of SSCs present in culture and caused SSCs to detach from supporting cells. Injection of mice with an SHP2 inhibitor blocked the production of germ cells from SSCs. Together, our studies show that SHP2 is essential for SSCs to maintain fertility and indicates that the pathogenesis of infertility in humans with SHP2 mutations is due to compromised SSC functions that block spermatogenesis.


Assuntos
Fertilidade , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Espermatogônias/citologia , Espermatogônias/enzimologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Envelhecimento , Animais , Adesão Celular , Contagem de Células , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Deleção de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Transdução de Sinais , Espermatogênese
10.
Biol Reprod ; 89(5): 128, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24089200

RESUMO

The four isoforms of serine/threonine phosphoprotein phosphatase 1 (PP1), derived from three genes, are among the most conserved proteins known. The Ppp1cc gene encodes two alternatively spliced variants, PP1 gamma1 (PPP1CC1) and PP1 gamma2 (PPP1CC2). Global deletion of the Ppp1cc gene, which causes loss of both isoforms, results in male infertility due to impaired spermatogenesis. This phenotype was assumed to be due to the loss of PPP1CC2, which is abundant in testis. While PPP1CC2 is predominant, other PP1 isoforms are also expressed in testis. Given the significant homology between the four PP1 isoforms, the lack of compensation by the other PP1 isoforms for loss of one, only in testis, is surprising. Here we document, for the first time, expression patterns of the PP1 isoforms in postnatal developing and adult mouse testis. The timing and sites of testis expression of PPP1CC1 and PPP1CC2 in testis are nonoverlapping. PPP1CC2 is the only one of the four PP1 isoforms not detected in sertoli cells and spermatogonia. Conversely, PPP1CC2 may be the only PP1 isoform expressed in postmeiotic germ cells. Deletion of the Ppp1cc gene in germ cells at the differentiated spermatogonia stage of development and beyond in Stra8 promoter-driven Cre transgenic mice results in oligo-terato-asthenozoospermia and male infertility, thus phenocopying global Ppp1cc null (-/-) mice. Taken together, these results confirm that spermatogenic defects observed in the global Ppp1cc knockout mice and in mice expressing low levels of PPP1CC2 in testis are due to compromised functions of PPP1CC2 in meiotic and postmeiotic germ cells.


Assuntos
Oligospermia/genética , Proteína Fosfatase 1/genética , Espermatozoides/metabolismo , Animais , Feminino , Expressão Gênica , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Meiose/genética , Camundongos , Camundongos Knockout , Oligospermia/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Fosfatase 1/metabolismo , Espermatogênese/genética , Testículo/metabolismo
11.
Biol Reprod ; 88(3): 59, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23325809

RESUMO

The blood-testis barrier (BTB) is a large junctional complex composed of tight junctions, adherens junctions, and gap junctions between adjacent Sertoli cells in the seminiferous tubules of the testis. Maintenance of the BTB as well as the controlled disruption and reformation of the barrier is essential for spermatogenesis and male fertility. Tyrosine phosphorylation of BTB proteins is known to regulate the integrity of adherens and tight junctions found at the BTB. SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) and a key regulator of growth factor-mediated tyrosine kinase signaling pathways. We found that SHP2 is localized to Sertoli-Sertoli cell junctions in rat testis. The overexpression of a constitutive active SHP2 mutant, SHP2 Q79R, up-regulated the BTB disruptor ERK1/2 via Src kinase in primary rat Sertoli cells in culture. Furthermore, focal adhesion kinase (FAK), which also supports BTB integrity, was found to interact with SHP2 and constitutive activation of SHP2 decreased FAK tyrosine phosphorylation. Expression of the SHP2 Q79R mutant in primary cultured Sertoli cells also resulted in the loss of tight junction and adherens junction integrity that corresponded with the disruption of the actin cytoskeleton and mislocalization of adherens junction and tight junction proteins N-cadherin, ß-catenin, and ZO-1 away from the plasma membrane. These results suggest that SHP2 is a key regulator of BTB integrity and Sertoli cell support of spermatogenesis and fertility.


Assuntos
Barreira Hematotesticular/metabolismo , Junções Intercelulares/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Células de Sertoli/enzimologia , Animais , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Mol Endocrinol ; 25(2): 238-52, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21177760

RESUMO

Testosterone and FSH act in synergy to produce the factors required to maximize the production of spermatozoa and male fertility. However, the molecular mechanisms by which these hormones support spermatogenesis are not well established. Recently, we identified a nonclassical mechanism of testosterone signaling in cultured rat Sertoli cells. We found that testosterone binding to the androgen receptor recruits and activates Src tyrosine kinase. Src then causes the activation of the epidermal growth factor receptor, which results in the phosphorylation and activation of the ERK MAPK and the cAMP response element-binding protein transcription factor. In this report, we find that FSH inhibits testosterone-mediated activation of ERK and the MAPK pathway in Sertoli cells via the protein kinase A-mediated inhibition of Raf kinase. In addition, FSH, as well as inhibitors of Src and ERK kinase activity, reduced germ cell attachment to Sertoli cells in culture. Using pathway-specific androgen receptor mutants we found that the nonclassical pathway is required for testosterone-mediated increases in germ cell attachment to Sertoli cells. Studies of seminiferous tubule explants determined that Src kinase, but not ERK kinase, activity is required for the release of sperm from seminiferous tubule explants. These findings suggest the nonclassical testosterone-signaling pathway acts via Src and ERK kinases to facilitate the adhesion of immature germ cells to Sertoli cells and through Src to permit the release of mature spermatozoa. In contrast, FSH acts to limit testosterone-mediated ERK kinase activity and germ cell attachment.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hormônio Foliculoestimulante/metabolismo , Células de Sertoli/fisiologia , Espermatozoides/fisiologia , Testosterona/metabolismo , Quinases raf/metabolismo , Quinases da Família src/metabolismo , Animais , Adesão Celular , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Masculino , Fosforilação , Regiões Promotoras Genéticas/genética , Ratos , Túbulos Seminíferos , Transdução de Sinais , Espermatogênese , Testosterona/genética
13.
Spermatogenesis ; 1(4): 354-365, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22332119

RESUMO

The 14-3-3 family of proteins interacts with various cellular phosphoproteins and regulates multiple cell signaling cascades. Identification of 14-3-3 interactors is important to define 14-3-3 functions in various biological pathways. The binding partners of protein 14-3-3 in testis are not known. The main goal of this study was to identify the 14-3-3 interactome in testis to determine the 14-3-3 regulated cellular processes in testis. We used transgenic mice expressing tandem affinity tagged 14-3-3ζ (TAP-14-3-3ζ) driven by the ubiquitin promoter to isolate 14-3-3 binding proteins. The 14-3-3 complexes in testis were isolated using a two-step tandem affinity purification (TAP) followed by identification with liquid chromatography/tandem mass spectrometry (LC-MS/MS). A total of 135 proteins were found to be associated with 14-3-3 in vivo in testis. Comparison of the testis 14-3-3 proteome with known 14-3-3 binding proteins showed that 71 of the proteins identified in this study are novel 14-3-3 interactors. Eight of these novel 14-3-3 interacting proteins are predominantly expressed in testis. The 14-3-3 interactors predominant in testis are: protein phosphatase1γ2 (PP1γ2), spermatogenesis associated 18 (SPATA18), phosphoglycerate kinase-2 (PGK2), testis specific gene A-2 (TSGA-2), dead box polypeptide 4 (DDX4), piwi homolog 1, protein kinase NYD-SP25 and EAN57. The fact that some of these proteins are indispensable for spermatogenesis suggests that their binding to 14-3-3 may be important for their function in germ cell division and maturation. These findings are discussed in context of the putative functions of 14-3-3 in spermatogenesis.

14.
Biol Reprod ; 79(6): 1183-91, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18753613

RESUMO

Posttranslational modification of proteins by phosphorylation is involved in regulation of sperm function. Protein phosphatase 1 gamma isoform 2 (PPP1CC_v2) and protein YWHA (also known as 14-3-3) are likely to be key molecules in pathways involving sperm protein phosphorylation. We have shown that phosphorylated PPP1CC_v2 is bound to protein YWHAZ in spermatozoa. In somatic cells, protein YWHA is known to bind a number of phosphoproteins involved in signaling and energy metabolism. Thus, in addition to PPP1CC_v2, it is likely that sperm contain other YWHA-binding proteins. A goal of the present study was to identify these sperm YWHA-binding proteins. The binding proteins were isolated by affinity chromatography with GST-YWHAZ followed by elution with a peptide, R-11, which is known to disrupt YWHA complexes. The YWHA-binding proteins in sperm can be classified as those involved in fertilization, acrosome reaction, energy metabolism, protein folding, and ubiquitin-mediated proteolysis. A subset of these putative YWHA-binding proteins contain known amino acid consensus motifs, not only for YWHA binding but also for PPP1C binding. Identification of sperm PPP1CC_v2-binding proteins by microcystin-agarose chromatography confirmed that PPP1CC_v2 and YWHA interactomes contain several common proteins. These are metabolic enzymes phosphoglycerate kinase 2, hexokinase 1, and glucose phosphate isomerase; proteins involved in sperm-egg fusion; angiotensin-converting enzyme, sperm adhesion molecule, and chaperones; heat shock 70-kDa protein 5 (glucose-regulated protein 78 kDa; and heat shock 70-kDa protein 1-like. These proteins are likely to be phosphoproteins and potential PPP1CC_v2 substrates. Our data suggest that in addition to potential regulation of a number of important sperm functions, YWHA may act as an adaptor molecule for a subset of PPP1CC_v2 substrates.


Assuntos
Proteínas 14-3-3/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Bovinos , Cromatografia de Afinidade , Imunoprecipitação , Isoenzimas/biossíntese , Isoenzimas/genética , Masculino , Espectrometria de Massas , Microcistinas/metabolismo , Fosfoglicerato Quinase/biossíntese , Fosfoglicerato Quinase/genética , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Ligação Proteica , Proteômica , Transdução de Sinais/genética , Espermatozoides/metabolismo
15.
Biol Reprod ; 79(2): 337-47, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18463355

RESUMO

Proteins in the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein family (YWHA; also known as 14-3-3) are involved in the regulation of many intracellular processes. We have examined the interaction of YWHA with peptidylarginine deiminase type VI (PADI6), an abundant protein in mammalian oocytes, eggs, and early embryos. Peptidylarginine deiminases catalyze the posttranslational modification of peptidylarginine to citrulline. PADI6 is associated with oocyte cytoplasmic sheets, and PADI6-deficient mice are infertile because of disruption of development beyond the two-cell stage. We found that PADI6 undergoes a dramatic developmental change in phosphorylation during oocyte maturation. This change in phosphorylation is linked to an interaction of PADI6 with YWHA in the mature egg. Recombinant glutathione S-transferase YWHA pull-down experiments and transgenic tandem affinity purification with liquid chromatography-mass spectrometry demonstrate a binding interaction between YWHA and PADI6 in mature eggs. YWHA proteins modulate or complement intracellular events involving phosphorylation-dependent switching or protein modification. These results indicate that phosphorylation and/or YWHA binding may serve as a means of intracellular PADI6 regulation.


Assuntos
Proteínas 14-3-3/metabolismo , Hidrolases/metabolismo , Oócitos/metabolismo , Oogênese/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Dados de Sequência Molecular , Oócitos/química , Oócitos/crescimento & desenvolvimento , Fosforilação , Ligação Proteica , Proteína-Arginina Desiminase do Tipo 6 , Desiminases de Arginina em Proteínas , Triptofano Hidroxilase/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
16.
Asian J Androl ; 9(4): 445-52, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17589781

RESUMO

The serine/threonine phosphatase (PP1) isoform PP1 gamma 2, predominantly expressed in the testis, is a key enzyme in spermatozoa. High PP1 gamma 2 catalytic activity holds motility in check in immature spermatozoa. Inhibition of PP1 gamma 2 causes motility initiation in immature spermatozoa and motility stimulation and changes in flagellar beat parameters in mature spermatozoa. The PP1 gamma 2 isoform is present in all mammalian spermatozoa studied: mouse, rat, hamster, bovine, non-human primate and man. We have now identified at least four of its regulatory proteins that regulate distinct pools of PP1 gamma 2 within spermatozoa. Our studies provide new insights into biochemical mechanisms underlying development and regulation of sperm motility. We hypothesize that changes in sperm PP1 gamma 2 activity as a result of phosphorylation and reversible binding of the regulatory proteins to the catalytic subunit are critical in the development and regulation of motility and the ability of sperm to fertilize eggs. Targeted disruption of the Ppp1cc gene, which encodes the PP1 gamma 1 or PP1 gamma 2 isoforms, causes male infertility in mice as a result of impaired spermiogenesis. Our observations suggest that, in addition to motility, the protein phosphatase PP1 gamma 2 might play an isoform-specific function in the development of specialized flagellar structures of mammalian spermatozoa.


Assuntos
Epididimo/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/enzimologia , Animais , Bovinos , Cricetinae , Epididimo/fisiologia , Homeostase , Humanos , Masculino , Camundongos , Fosfoproteínas Fosfatases/genética , Espermatozoides/fisiologia , Testículo/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...