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










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1284184, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020932

RESUMEN

Much of the foundation for lifelong spermatogenesis is established prior to puberty, and disruptions during this developmental window negatively impact fertility long into adulthood. However, the factors that coordinate prepubertal germline development are incompletely understood. Here, we report that core-binding factor subunit-ß (CBFß) plays critical roles in prepubertal development and the onset of spermatogenesis. Using a mouse conditional knockout (cKO) approach, inactivation of Cbfb in the male germline resulted in rapid degeneration of the germline during the onset of spermatogenesis, impaired overall sperm production, and adult infertility. Utilizing a different Cre driver to generate another Cbfb cKO model, we determined that the function of CBFß in the male germline is likely limited to undifferentiated spermatogonia despite expression in other germ cell types. Within undifferentiated spermatogonia, CBFß regulates proliferation, survival, and overall maintenance of the undifferentiated spermatogonia population. Paradoxically, we discovered that CBFß also distally regulates meiotic progression and spermatid formation but only with Cbfb cKO within undifferentiated spermatogonia. Spatial transcriptomics revealed that CBFß modulates cell cycle checkpoint control genes associated with both proliferation and meiosis. Taken together, our findings demonstrate that core programs established within the prepubertal undifferentiated spermatogonia population are necessary for both germline maintenance and sperm production.

2.
Methods Mol Biol ; 2656: 309-324, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37249878

RESUMEN

Spermatogonial stem cells (SSCs) are the fundamental units from which continuous spermatogenesis arises. Although our knowledge regarding the basic properties of SSCs has grown, driven primarily through the advancement of techniques and technologies to study SSCs, the mechanisms controlling their fate remain largely unknown. Among the modern strategies to evaluate SSCs, lineage tracing is among the few established approaches that allow for functional assessment of stem cell capacity. As a result, lineage tracing continues to forge new discoveries underlying the basic attributes of SSCs as well as the molecular factors that govern SSC function. Traditional approaches to lineage tracing with dyes or radioactive labels suffer from progressive loss after successive cell divisions or unintentional label transfer to neighboring cells. To address these limitations, genetic approaches primarily leveraging transgenic technologies have prevailed as the preferred avenue for modern lineage tracing. This chapter will discuss current protocols for effective genetic lineage tracing and address applications of this technology, considerations when designing lineage tracing experiments, and the methods involved in utilizing lineage tracing to study SSCs and other cell populations.


Asunto(s)
Células Madre Germinales Adultas , Espermatogonias , Masculino , Humanos , Espermatogénesis/genética , Células Madre/fisiología
3.
Nat Commun ; 14(1): 2111, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069147

RESUMEN

In sexual reproduction, sperm contribute half the genomic material required for creation of offspring yet core molecular mechanisms essential for their formation are undefined. Here, the α-arrestin molecule arrestin-domain containing 5 (ARRDC5) is identified as an essential regulator of mammalian spermatogenesis. Multispecies testicular tissue transcriptome profiling indicates that expression of Arrdc5 is testis enriched, if not specific, in mice, pigs, cattle, and humans. Knockout of Arrdc5 in mice leads to male specific sterility due to production of low numbers of sperm that are immotile and malformed. Spermiogenesis, the final phase of spermatogenesis when round spermatids transform to spermatozoa, is defective in testes of Arrdc5 deficient mice. Also, epididymal sperm in Arrdc5 knockouts are unable to capacitate and fertilize oocytes. These findings establish ARRDC5 as an essential regulator of mammalian spermatogenesis. Considering the role of arrestin molecules as modulators of cellular signaling and ubiquitination, ARRDC5 is a potential male contraceptive target.


Asunto(s)
Arrestinas , Infertilidad Masculina , Testículo , Animales , Bovinos , Humanos , Masculino , Ratones , Arrestinas/genética , Arrestinas/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Ratones Noqueados , Morfogénesis , Semen/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Porcinos , Testículo/metabolismo
4.
Biol Reprod ; 106(6): 1175-1190, 2022 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-35244684

RESUMEN

Spermatogenic regeneration is key for male fertility and relies on activities of an undifferentiated spermatogonial population. Here, a high-throughput approach with primary cultures of mouse spermatogonia was devised to rapidly predict alterations in functional capacity. Combining the platform with a large-scale RNAi screen of transcription factors, we generated a repository of new information from which pathway analysis was able to predict candidate molecular networks regulating regenerative functions. Extending from this database, the SRCAP-CREBBP/EP300 (Snf2-related CREBBP activator protein-CREB binding protein/E1A binding protein P300) complex was found to mediate differential levels of histone acetylation between stem cell and progenitor spermatogonia to influence expression of key self-renewal genes including the previously undescribed testis-specific transcription factor ZSCAN2 (zinc finger and SCAN domain containing 2). Single cell RNA sequencing analysis revealed that ZSCAN2 deficiency alters key cellular processes in undifferentiated spermatogonia such as translation, chromatin modification, and ubiquitination. In Zscan2 knockout mice, while spermatogenesis was moderately impacted during steady state, regeneration after cytotoxic insult was significantly impaired. Altogether, these findings have validated the utility of our high-throughput screening approach and have generated a transcription factor database that can be utilized for uncovering novel mechanisms governing spermatogonial functions.


Asunto(s)
Espermatogénesis , Espermatogonias , Animales , Diferenciación Celular , Masculino , Ratones , Espermatogénesis/fisiología , Células Madre , Testículo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Adv Sci (Weinh) ; 8(22): e2102157, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34647690

RESUMEN

Obesity during pregnancy leads to adverse health outcomes in offspring. However, the initial effects of maternal obesity (MO) on embryonic organogenesis have yet to be thoroughly examined. Using unbiased single-cell transcriptomic analyses (scRNA-seq), the effects of MO on the myogenic process is investigated in embryonic day 9.5 (E9.5) mouse embryos. The results suggest that MO induces systematic hypoxia, which is correlated with enhanced BMP signaling and impairs skeletal muscle differentiation within the dermomyotome (DM). The Notch-signaling effectors, HES1 and HEY1, which also act down-stream of BMP signaling, suppress myogenic differentiation through transcriptionally repressing the important myogenic regulator MEF2C. Moreover, the major hypoxia effector, HIF1A, enhances expression of HES1 and HEY1 and blocks myogenic differentiation in vitro. In summary, this data demonstrate that MO induces hypoxia and impairs myogenic differentiation by up-regulating BMP signaling within the DM, which may account for the disruptions of skeletal muscle development and function in progeny.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Desarrollo de Músculos , Obesidad Materna/embriología , Obesidad Materna/metabolismo , Complicaciones del Embarazo/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo , Transducción de Señal
6.
Biol Reprod ; 105(6): 1591-1602, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34494084

RESUMEN

Sertoli cells are a critical component of the testis environment for their role in maintaining seminiferous tubule structure, establishing the blood-testis barrier, and nourishing maturing germ cells in a specialized niche. This study sought to uncover how Sertoli cells are regulated in the testis environment via germ cell crosstalk in the mouse. We found two major clusters of Sertoli cells as defined by their transcriptomes in Stages VII-VIII of the seminiferous epithelium and a cluster for all other stages. Additionally, we examined transcriptomes of germ cell-deficient testes and found that these existed in a state independent of either of the germ cell-sufficient clusters. Altogether, we highlight two main transcriptional states of Sertoli cells in an unperturbed testis environment, and a germ cell-deficient environment does not allow normal Sertoli cell transcriptome cycling and results in a state unique from either of those seen in Sertoli cells from a germ cell-sufficient environment.


Asunto(s)
Células de Sertoli/citología , Transducción de Señal , Espermatozoides/fisiología , Animales , Masculino , Ratones
7.
Development ; 148(9)2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33929507

RESUMEN

The stem cell-containing undifferentiated spermatogonial population in mammals, which ensures continual sperm production, arises during development from prospermatogonial precursors. Although a period of quiescence is known to occur in prospermatogonia prior to postnatal spermatogonial transition, the importance of this has not been defined. Here, using mouse models with conditional knockout of the master cell cycle regulator Rb1 to disrupt normal timing of the quiescence period, we found that failure to initiate mitotic arrest during fetal development leads to prospermatogonial apoptosis and germline ablation. Outcomes of single-cell RNA-sequencing analysis indicate that oxidative phosphorylation activity and inhibition of meiotic initiation are disrupted in prospermatogonia that fail to enter quiescence on a normal timeline. Taken together, these findings suggest that key layers of programming are laid down during the quiescent period in prospermatogonia to ensure proper fate specification and fitness in postnatal life.


Asunto(s)
División Celular/fisiología , Espermatogonias/citología , Espermatogonias/crecimiento & desarrollo , Células Madre/citología , Animales , Apoptosis , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Proteínas de Unión a Retinoblastoma/genética , Análisis de Secuencia de ARN , Espermatogénesis/fisiología , Espermatogonias/metabolismo , Espermatozoides , Transcriptoma
8.
Andrology ; 8(4): 852-861, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32356598

RESUMEN

BACKGROUND: The germline serves as a conduit for transmission of genetic and epigenetic information from one generation to the next. In males, spermatozoa are the final carriers of inheritance and their continual production is supported by a foundational population of spermatogonial stem cells (SSCs) that forms from prospermatogonial precursors during the early stages of neonatal development. In mammals, the timing for which SSCs are specified and the underlying mechanisms guiding this process remain to be completely understood. OBJECTIVES: To propose an evolving concept for how the foundational SSC population is established. MATERIALS AND METHODS: This review summarizes recent and historical findings from peer-reviewed publications made primarily with mouse models while incorporating limited studies from humans and livestock. RESULTS AND CONCLUSION: Establishment of the SSC population appears to follow a biphasic pattern involving a period of fate programming followed by an establishment phase that culminates in formation of the SSC population. This model for establishment of the foundational SSC population from precursors is anticipated to extend across mammalian species and include humans and livestock, albeit on different timescales.


Asunto(s)
Células Madre Germinales Adultas , Espermatogénesis/fisiología , Espermatogonias , Animales , Bovinos , Humanos , Masculino , Ratones
9.
Am J Physiol Endocrinol Metab ; 318(4): E554-E563, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32069073

RESUMEN

Insulin pulsatility is important to hepatic response in regulating blood glucose. Growing evidence suggests that insulin-secreting pancreatic ß-cells can adapt to chronic disruptions of pulsatility to rescue this physiologically important behavior. We determined the time scale for adaptation and examined potential ion channels underlying it. We induced the adaptation both by chronic application of the ATP-sensitive K+ [K(ATP)] channel blocker tolbutamide and by application of the depolarizing agent potassium chloride (KCl). Acute application of tolbutamide without pretreatment results in elevated Ca2+ as measured by fura-2AM and the loss of endogenous pulsatility. We show that after chronic exposure to tolbutamide (12-24 h), Ca2+ oscillations occur with subsequent acute tolbutamide application. The same experiment was conducted with potassium chloride (KCl) to directly depolarize the ß-cells. Once again, following chronic exposure to the cell stimulator, the islets produced Ca2+ oscillations when subsequently exposed to tolbutamide. These experiments suggest that it is the chronic stimulation, and not tolbutamide desensitization, that is responsible for the adaptation that rescues oscillatory ß-cell activity. This compensatory response also causes islet glucose sensitivity to shift rightward following chronic tolbutamide treatment. Mathematical modeling shows that a small increase in the number of K(ATP) channels in the membrane is one adaptation mechanism that is compatible with the data. To examine other compensatory mechanisms, pharmacological studies provide support that Kir2.1 and TEA-sensitive channels play some role. Overall, this investigation demonstrates ß-cell adaptability to overstimulation, which is likely an important mechanism for maintaining glucose homeostasis in the face of chronic stimulation.


Asunto(s)
Adaptación Fisiológica , Señalización del Calcio , Islotes Pancreáticos/metabolismo , Canales de Potasio/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Hiperinsulinismo Congénito/metabolismo , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Canales KATP/metabolismo , Masculino , Ratones , Modelos Teóricos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/metabolismo , Cloruro de Potasio , Estimulación Química , Tolbutamida/farmacología
10.
Nat Commun ; 10(1): 2787, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31243281

RESUMEN

Continuity, robustness, and regeneration of cell lineages relies on stem cell pools that are established during development. For the mammalian spermatogenic lineage, a foundational spermatogonial stem cell (SSC) pool arises from prospermatogonial precursors during neonatal life via mechanisms that remain undefined. Here, we mapped the kinetics of this process in vivo using a multi-transgenic reporter mouse model, in silico with single-cell RNA sequencing, and functionally with transplantation analyses to define the SSC trajectory from prospermatogonia. Outcomes revealed that a heterogeneous prospermatogonial population undergoes dynamic changes during late fetal and neonatal development. Differential transcriptome profiles predicted divergent developmental trajectories from fetal prospermatogonia to descendant postnatal spermatogonia. Furthermore, transplantation analyses demonstrated that a defined subset of fetal prospermatogonia is fated to function as SSCs. Collectively, these findings suggest that SSC fate is preprogrammed within a subset of fetal prospermatogonia prior to building of the foundational pool during early neonatal development.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Linaje de la Célula , Testículo/embriología , Animales , Diferenciación Celular , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes Reporteros , Masculino , Ratones , Ratones Transgénicos , ARN/genética , Espermatogénesis/fisiología , Espermatogonias/fisiología
11.
Cell Rep ; 25(6): 1650-1667.e8, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30404016

RESUMEN

Spermatogenesis is a complex and dynamic cellular differentiation process critical to male reproduction and sustained by spermatogonial stem cells (SSCs). Although patterns of gene expression have been described for aggregates of certain spermatogenic cell types, the full continuum of gene expression patterns underlying ongoing spermatogenesis in steady state was previously unclear. Here, we catalog single-cell transcriptomes for >62,000 individual spermatogenic cells from immature (postnatal day 6) and adult male mice and adult men. This allowed us to resolve SSC and progenitor spermatogonia, elucidate the full range of gene expression changes during male meiosis and spermiogenesis, and derive unique gene expression signatures for multiple mouse and human spermatogenic cell types and/or subtypes. These transcriptome datasets provide an information-rich resource for studies of SSCs, male meiosis, testicular cancer, male infertility, or contraceptive development, as well as a gene expression roadmap to be emulated in efforts to achieve spermatogenesis in vitro.


Asunto(s)
Mamíferos/genética , Análisis de la Célula Individual , Espermátides/citología , Espermatogénesis/genética , Espermatogonias/citología , Transcriptoma/genética , Adulto , Envejecimiento/genética , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Haploidia , Humanos , Masculino , Meiosis , Ratones Endogámicos C57BL , Transducción de Señal , Espermátides/metabolismo , Espermatogonias/metabolismo , Células Madre/citología , Células Madre/metabolismo , Testículo/citología
12.
Endocrinology ; 159(11): 3747-3760, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239634

RESUMEN

An early sign of islet failure in type 2 diabetes (T2D) is the loss of normal patterns of pulsatile insulin release. Disruptions in pulsatility are associated with a left shift in glucose sensing that can cause excessive insulin release in low glucose (relative hyperinsulinemia, a hallmark of early T2D) and ß-cell exhaustion, leading to inadequate insulin release during hyperglycemia. Our hypothesis was that reducing excessive glucokinase activity in diabetic islets would improve their function. Isolated mouse islets were exposed to glucose and varying concentrations of the glucokinase inhibitor d-mannoheptulose (MH) to examine changes in intracellular calcium ([Ca2+]i) and insulin secretion. Acutely exposing islets from control CD-1 mice to MH in high glucose (20 mM) dose dependently reduced the size of [Ca2+]i oscillations detected by fura-2 acetoxymethyl. Glucokinase activation in low glucose (3 mM) had the opposite effect. We then treated islets from male and female db/db mice (age, 4 to 8 weeks) and heterozygous controls overnight with 0 to 10 mM MH to determine that 1 mM MH produced optimal oscillations. We then used 1 mM MH overnight to measure [Ca2+]i and insulin simultaneously in db/db islets. MH restored oscillations and increased insulin secretion. Insulin secretion rates correlated with MH-induced increases in amplitude of [Ca2+]i oscillations (R2 = 0.57, P < 0.01, n = 10) but not with mean [Ca2+]i levels in islets (R2 = 0.05, not significant). Our findings show that correcting glucose sensing can restore proper pulsatility to diabetic islets and improved pulsatility correlates with enhanced insulin secretion.


Asunto(s)
Calcio/metabolismo , Glucoquinasa/antagonistas & inhibidores , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Manoheptulosa/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Glucoquinasa/metabolismo , Glucosa , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos
13.
Endocrinology ; 158(7): 2043-2051, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28460125

RESUMEN

Protein kinase A (PKA) has recently been shown to mimic the actions of follicle-stimulating hormone (FSH) by activating signaling pathways that promote granulosa cell (GC) differentiation, such as phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK). We sought to elucidate the mechanism by which PKA, a Ser/Thr kinase, intersected the PI3K/AKT and MAPK/ERK pathways that are canonically activated by receptor tyrosine kinases (RTKs). Our results show that for both of these pathways, the RTK is active in the absence of FSH yet signaling down the pathways to commence transcriptional responses requires FSH-stimulated PKA activation. For both pathways, PKA initiates signaling by regulating the activity of a protein phosphatase (PP). For the PI3K/AKT pathway, PKA activates the Ser/Thr PP1 complexed with the insulinlike growth factor 1 receptor (IGF-1R) and insulin receptor substrate 1 (IRS1) to dephosphorylate Ser residues on IRS1, authorizing phosphorylation of IRS1 by the IGF-1R to activate PI3K. Treatment of GCs with FSH and exogenous IGF-1 initiates synergistic IRS1 Tyr phosphorylation and resulting gene activation. The mechanism by which PKA activates PI3K is conserved in preovulatory GCs, MCF7 breast cancer cells, and FRTL thyroid cells. For the MAPK/ERK pathway, PKA promotes inactivation of the MAPK phosphatase (MKP) dual specificity phosphatase (DUSP) MKP3/DUSP6 to permit MEK-phosphorylated ERK to accumulate downstream of the epidermal growth factor receptor. Thus, for the two central signaling pathways that regulate gene expression in GCs, FSH via PKA intersects canonical RTK-regulated signaling by modulating the activity of PPs.


Asunto(s)
Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células de la Granulosa/fisiología , Proteínas Tirosina Quinasas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Células MCF-7 , Fosforilación , Ratas , Transducción de Señal/genética
14.
J Biol Chem ; 291(53): 27160-27169, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27856640

RESUMEN

G protein-coupled receptors (GPCRs) activate PI3K/v-AKT thymoma viral oncoprotein (AKT) to regulate many cellular functions that promote cell survival, proliferation, and growth. However, the mechanism by which GPCRs activate PI3K/AKT remains poorly understood. We used ovarian preantral granulosa cells (GCs) to elucidate the mechanism by which the GPCR agonist FSH via PKA activates the PI3K/AKT cascade. Insulin-like growth factor 1 (IGF1) is secreted in an autocrine/paracrine manner by GCs and activates the IGF1 receptor (IGF1R) but, in the absence of FSH, fails to stimulate YXXM phosphorylation of IRS1 (insulin receptor substrate 1) required for PI3K/AKT activation. We show that PKA directly phosphorylates the protein phosphatase 1 (PP1) regulatory subunit myosin phosphatase targeting subunit 1 (MYPT1) to activate PP1 associated with the IGF1R-IRS1 complex. Activated PP1 is sufficient to dephosphorylate at least four IRS1 Ser residues, Ser318, Ser346, Ser612, and Ser789, and promotes IRS1 YXXM phosphorylation by the IGF1R to activate the PI3K/AKT cascade. Additional experiments indicate that this mechanism also occurs in breast cancer, thyroid, and preovulatory granulosa cells, suggesting that the PKA-dependent dephosphorylation of IRS1 Ser/Thr residues is a conserved mechanism by which GPCRs signal to activate the PI3K/AKT pathway downstream of the IGF1R.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células Cultivadas , Femenino , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
15.
J Biol Chem ; 291(37): 19701-12, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27422819

RESUMEN

Within the ovarian follicle, granulosa cells (GCs) surround and support immature oocytes. FSH promotes the differentiation and proliferation of GCs and is essential for fertility. We recently reported that ERK activation is necessary for FSH to induce key genes that define the preovulatory GC. This research focused on the phosphoregulation by FSH of ERK within GCs. FSH-stimulated ERK phosphorylation on Thr(202)/Tyr(204) was PKA-dependent, but MEK(Ser(217)/Ser(221)) phosphorylation was not regulated; rather, MEK was already active. However, treatment of GCs with the EGF receptor inhibitor AG1478, a dominant-negative RAS, an Src homology 2 domain-containing Tyr phosphatase inhibitor (NSC 87877), or the MEK inhibitor PD98059 blocked FSH-dependent ERK(Thr(202)/Tyr(204)) phosphorylation, demonstrating the requirement for upstream pathway components. We hypothesized that FSH via PKA enhances ERK phosphorylation by inhibiting the activity of a protein phosphatase that constitutively dephosphorylates ERK in the absence of FSH, allowing MEK-phosphorylated ERK to accumulate in the presence of FSH because of inactivation of the phosphatase. GCs treated with different phosphatase inhibitors permitted elimination of both Ser/Thr and Tyr phosphatases and implicated dual specificity phosphatases (DUSPs) in the dephosphorylation of ERK. Treatment with MAP kinase phosphatase (MKP3, DUSP6) inhibitors increased ERK(Thr(202)/Tyr(204)) phosphorylation in the absence of FSH to levels comparable with ERK phosphorylated in the presence of FSH. ERK co-immunoprecipitated with Myc-FLAG-tagged MKP3(DUSP6). GCs treated with MKP3(DUSP6) inhibitors blocked and PKA inhibitors enhanced dephosphorylation of recombinant ERK2-GST in an in vitro phosphatase assay. Together, these results suggest that FSH-stimulated ERK activation in GCs requires the PKA-dependent inactivation of MKP3(DUSP6).


Asunto(s)
Fosfatasa 6 de Especificidad Dual/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/enzimología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfatasa 6 de Especificidad Dual/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Femenino , Flavonoides/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley
16.
Sci Rep ; 6: 28132, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27324437

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante Humana/metabolismo , Células de la Granulosa/fisiología , Ovario/citología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mutación/genética , Ratas , Ratas Endogámicas , Transducción de Señal , Esteroides/metabolismo , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Mol Cell Endocrinol ; 434: 116-26, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27328024

RESUMEN

FSH promotes maturation of ovarian follicles. One pathway activated by FSH in granulosa cells (GCs) is phosphatidylinositol-3 kinase/AKT. The AKT target FOXO1 is reported to function primarily as a repressor of FSH genes, including Ccnd2 and Inha. Based on its broad functions in other tissues, we hypothesized that FOXO1 may regulate many more GC genes. We transduced GCs with empty adenovirus or constitutively active FOXO1 followed by treatment with FSH for 24 h, and conducted RNA deep sequencing. Results show that FSH regulates 3772 genes ≥2.0-fold; 60% of these genes are activated or repressed by FOXO1. Pathway Studio Analysis revealed enrichment of genes repressed by FOXO1 in metabolism, signaling, transport, development, and activated by FOXO1 in signaling, cytoskeletal functions, and apoptosis. Gene regulation was verified by q-PCR (eight genes) and ChIP analysis (two genes). We conclude that FOXO1 regulates the majority of FSH target genes in GCs.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Redes Reguladoras de Genes/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Animales , Células Cultivadas , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/citología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Análisis de Secuencia de ARN/métodos
18.
J Biol Chem ; 291(9): 4547-60, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26702053

RESUMEN

The ubiquitous phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates many cellular functions. However, the mechanism by which G protein-coupled receptors (GPCRs) signal to activate PI3K is poorly understood. We have used ovarian granulosa cells as a model to investigate this pathway, based on evidence that the GPCR agonist follicle-stimulating hormone (FSH) promotes the protein kinase A (PKA)-dependent phosphorylation of insulin receptor substrate 1 (IRS1) on tyrosine residues that activate PI3K. We report that in the absence of FSH, granulosa cells secrete a subthreshold concentration of insulin-like growth factor-1 (IGF-1) that primes the IGF-1 receptor (IGF-1R) but fails to promote tyrosine phosphorylation of IRS1. FSH via PKA acts to sensitize IRS1 to the tyrosine kinase activity of the IGF-1R by activating protein phosphatase 1 (PP1) to promote dephosphorylation of inhibitory Ser/Thr residues on IRS1, including Ser(789). Knockdown of PP1ß blocks the ability of FSH to activate PI3K in the presence of endogenous IGF-1. Activation of PI3K thus requires both PKA-mediated relief of IRS1 inhibition and IGF-1R-dependent tyrosine phosphorylation of IRS1. Treatment with FSH and increasing concentrations of exogenous IGF-1 triggers synergistic IRS1 tyrosine phosphorylation at PI3K-activating residues that persists downstream through protein kinase B (AKT) and FOXO1 (forkhead box protein O1) to drive synergistic expression of genes that underlies follicle maturation. Based on the ability of GPCR agonists to synergize with IGFs to enhance gene expression in other cell types, PP1 activation to relieve IRS1 inhibition may be a more general mechanism by which GPCRs act with the IGF-1R to activate PI3K/AKT.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína Fosfatasa 1/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Activación Enzimática , Femenino , Células de la Granulosa/citología , Humanos , Proteínas Sustrato del Receptor de Insulina/agonistas , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/genética , Mutación , Fosfatidilinositol 3-Quinasa/química , Fosforilación , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/genética , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Ratas Sprague-Dawley , Receptor IGF Tipo 1/agonistas , Receptor IGF Tipo 1/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Tirosina/metabolismo
19.
Mol Endocrinol ; 27(8): 1295-310, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23754802

RESUMEN

Ovarian follicles lacking FSH or FSH receptors fail to progress to a preovulatory stage, resulting in infertility. One hallmark of the preovulatory follicle is the presence of luteinizing hormone/choriogonadotropin receptors (LHCGR) on granulosa cells (GCs). However, the mechanisms by which FSH induces Lhcgr gene expression are poorly understood. Our results show that protein kinase A (PKA) and phosphoinositide 3-kinase (PI3K)/AKT pathways are required for FSH to activate both the murine Lhcgr-luciferase reporter and expression of Lhcgr mRNA in rat GCs. Based on results showing that an adenovirus (Ad) expressing a steroidogenic factor 1 (SF1) mutant that cannot bind ß-catenin abolished FSH-induced Lhcgr mRNA, we evaluated the role of ß-catenin in the regulation of Lhcgr gene expression. FSH promoted the PKA-dependent, PI3K-independent phosphorylation of ß-catenin on Ser552 and Ser665. FSH activated the ß-catenin/T-cell factor (TCF) artificial promoter-reporter TOPFlash via a PKA-dependent, PI3K-independent pathway, and dominant-negative (DN) TCF abolished FSH-activated Lhcgr-luciferase reporter and induction of Lhcgr mRNA. Microarray analysis of GCs treated with Ad-DN-TCF and FSH identified the Lhcgr as the most down-regulated gene. Chromatin immunoprecipitation results placed ß-catenin phosphorylated on Ser552 and Ser675 and SF1 on the Lhcgr promoter in FSH-treated GCs; TCF3 was constitutively associated with the Lhcgr promoter. Transduction with an Ad-phospho-ß-catenin mutant (Ser552/665/Asp) enhanced Lhcgr mRNA expression in FSH-treated cells greater than 3-fold. Finally, we identified a recognized PI3K/AKT target, forkhead box O1, as a negative regulator of Lhcgr mRNA expression. These results provide new understanding of the complex regulation of Lhcgr gene expression in GCs.


Asunto(s)
Células de la Granulosa/metabolismo , Folículo Ovárico/metabolismo , Receptores de Gonadotropina/metabolismo , Receptores de HL/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Femenino , Hormona Folículo Estimulante/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores de Gonadotropina/biosíntesis , Receptores de Gonadotropina/genética , Receptores de HL/biosíntesis , Receptores de HL/genética , Transducción de Señal , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Transfección , beta Catenina/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(44): E2979-88, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23045700

RESUMEN

Controlled maturation of ovarian follicles is necessary for fertility. Follicles are restrained at an immature stage until stimulated by FSH secreted by pituitary gonadotropes. FSH acts on granulosa cells within the immature follicle to inhibit apoptosis, promote proliferation, stimulate production of steroid and protein hormones, and induce ligand receptors and signaling intermediates. The phosphoinositide 3-kinase (PI3K)/AKT (protein kinase B) pathway is a pivotal signaling corridor necessary for transducing the FSH signal. We report that protein kinase A (PKA) mediates the actions of FSH by signaling through multiple targets to activate PI3K/AKT. PKA uses a route that promotes phosphorylation of insulin receptor substrate-1 (IRS-1) on Tyr(989), a canonical binding site for the 85-kDa regulatory subunit of PI3K that allosterically activates the catalytic subunit. PI3K activation leads to activation of AKT through phosphorylation of AKT on Thr(308) and Ser(473). The adaptor growth factor receptor bound protein 2-associated binding protein 2 (GAB2) is present in a preformed complex with PI3K heterodimer and IRS-1, it is an A-kinase anchoring protein that binds the type I regulatory subunit of PKA, and it is phosphorylated by PKA on Ser(159). Overexpression of GAB2 enhances FSH-stimulated AKT phosphorylation. GAB2, thus, seems to coordinate signals from the FSH-stimulated rise in cAMP that leads to activation of PI3K/AKT. The ability of PKA to commandeer IRS-1 and GAB2, adaptors that normally integrate receptor/nonreceptor tyrosine kinase signaling into PI3K/AKT, reveals a previously unrecognized route for PKA to activate a pathway that promotes proliferation, inhibits apoptosis, enhances translation, and initiates differentiation of granulosa cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Folículo Estimulante/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Regulación Alostérica , Animales , Dominio Catalítico , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Activación Enzimática , Femenino , Fosforilación , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...