Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters








Publication year range
1.
Nat Struct Mol Biol ; 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388830

ABSTRACT

The RAS-MAPK pathway regulates cell proliferation, differentiation and survival, and its dysregulation is associated with cancer development. The pathway minimally comprises the small GTPase RAS and the kinases RAF, MEK and ERK. Activation of RAF by RAS is notoriously intricate and remains only partially understood. There are three RAF isoforms in mammals (ARAF, BRAF and CRAF) and two related pseudokinases (KSR1 and KSR2). RAS-mediated activation of RAF depends on an allosteric mechanism driven by the dimerization of its kinase domain. Recent work on human RAFs showed that MEK binding to KSR1 promotes KSR1-BRAF heterodimerization, which leads to the phosphorylation of free MEK molecules by BRAF. Similar findings were made with the single Drosophila RAF homolog. Here we show that the fly scaffold proteins CNK and HYP stabilize the KSR-MEK interaction, which in turn enhances RAF-KSR heterodimerization and RAF activation. The cryogenic electron microscopy structure of the minimal KSR-MEK-CNK-HYP complex reveals a ring-like arrangement of the CNK-HYP complex allowing CNK to simultaneously engage KSR and MEK, thus stabilizing the binary interaction. Together, these results illuminate how CNK contributes to RAF activation by stimulating the allosteric function of KSR and highlight the diversity of mechanisms impacting RAF dimerization as well as the regulatory potential of the KSR-MEK interaction.

2.
Nat Commun ; 14(1): 3560, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37322019

ABSTRACT

Cell motility is a critical feature of invasive tumour cells that is governed by complex signal transduction events. Particularly, the underlying mechanisms that bridge extracellular stimuli to the molecular machinery driving motility remain partially understood. Here, we show that the scaffold protein CNK2 promotes cancer cell migration by coupling the pro-metastatic receptor tyrosine kinase AXL to downstream activation of ARF6 GTPase. Mechanistically, AXL signalling induces PI3K-dependent recruitment of CNK2 to the plasma membrane. In turn, CNK2 stimulates ARF6 by associating with cytohesin ARF GEFs and with a novel adaptor protein called SAMD12. ARF6-GTP then controls motile forces by coordinating the respective activation and inhibition of RAC1 and RHOA GTPases. Significantly, genetic ablation of CNK2 or SAMD12 reduces metastasis in a mouse xenograft model. Together, this work identifies CNK2 and its partner SAMD12 as key components of a novel pro-motility pathway in cancer cells, which could be targeted in metastasis.


Subject(s)
ADP-Ribosylation Factors , Neoplasms , Humans , Mice , Animals , ADP-Ribosylation Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , ADP-Ribosylation Factor 6 , Signal Transduction/physiology , Cell Movement/physiology , Neoplasms/genetics , rac1 GTP-Binding Protein/metabolism
3.
Mol Reprod Dev ; 86(12): 1901-1908, 2019 12.
Article in English | MEDLINE | ID: mdl-31713287

ABSTRACT

Follicle growth and granulosa cell health are dependent on the secretion of estradiol from granulosa cells. Estradiol is synthesized from androgen precursor by cytochrome P450 aromatase (CYP19A1), and in cattle CYP19A1 messenger RNA has a short half-life but a long (3.5 kb) 3'-untranslated region (3'UTR), suggesting that posttranscriptional regulation may be important for control of enzyme activity. We tested this hypothesis by inserting the CYP19A1 3'UTR and fragments thereof into a reporter vector between the end of the luciferase coding region and the polyadenylation signal. The full-length aromatase 3'UTR suppressed luciferase activity to 10% of control levels, and smaller fragments showed that this inhibitory activity lies between +926 and +1134 of the 3'UTR. Protein-RNA cross-linking experiments revealed that these 3'UTR fragments formed an RNA-protein complex of approximately 70 kDa that was present in granulosa cells but not in corpus luteum, lung, liver, kidney, pancreas, or bladder extracts. The RNA-binding activity was specific to the 3'UTR, as shown by competition experiments with unlabeled RNA, and was present only in 3'UTR constructs that inhibited luciferase activity. These data suggest that posttranscriptional regulation is an important component of the control of CYP19A1 expression and involves protein binding to a specific sequence in the 3'UTR.


Subject(s)
3' Untranslated Regions , Aromatase/biosynthesis , Granulosa Cells/metabolism , Multiprotein Complexes/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Animals , Cattle , Female , Granulosa Cells/cytology
4.
Nature ; 554(7693): 549-553, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29433126

ABSTRACT

RAF family kinases have prominent roles in cancer. Their activation is dependent on dimerization of their kinase domains, which has emerged as a hindrance for drug development. In mammals, RAF family kinases include three catalytically competent enzymes (ARAF, BRAF and CRAF) and two pseudokinases (KSR1 and KSR2) that have been described as scaffolds owing to their apparent ability to bridge RAF isoforms and their substrate, mitogen-activated protein kinase kinase (MEK). Kinase suppressor of Ras (KSR) pseudokinases were also shown to dimerize with kinase-competent RAFs to stimulate catalysis allosterically. Although GTP-bound RAS can modulate the dimerization of RAF isoforms by engaging their RAS-binding domains, KSR1 and KSR2 lack an RAS-binding domain and therefore the regulatory principles underlying their dimerization with other RAF family members remain unknown. Here we show that the selective heterodimerization of BRAF with KSR1 is specified by direct contacts between the amino-terminal regulatory regions of each protein, comprising in part a novel domain called BRS in BRAF and the coiled-coil-sterile α motif (CC-SAM) domain in KSR1. We also discovered that MEK binding to the kinase domain of KSR1 asymmetrically drives BRAF-KSR1 heterodimerization, resulting in the concomitant stimulation of BRAF catalytic activity towards free MEK molecules. These findings demonstrate that KSR-MEK complexes allosterically activate BRAF through the action of N-terminal regulatory region and kinase domain contacts and challenge the accepted role of KSR as a scaffold for MEK recruitment to RAF.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Allosteric Regulation , Crystallography, X-Ray , Enzyme Activation , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Domains , Protein Multimerization , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
5.
Nat Commun ; 8(1): 1211, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29084939

ABSTRACT

First-generation RAF inhibitors paradoxically induce ERK signaling in normal and tumor cells exhibiting RAS activity. Compound-induced RAF dimerization through stabilization of the RAF ON/active state by inhibitors has emerged as a critical contributing factor. RAF inhibitors also enhance RAS-RAF association. Although this event is thought to play a key role in priming RAF activation, the underlying mechanism is not known. Here we report that RAF inhibitors induce the disruption of intramolecular interactions between the kinase domain and its N-terminal regulatory region independently of RAS activity. This provides a molecular basis to explain the induction of RAS-RAF association by RAF inhibitors, as well as the co-operativity observed between RAS activity and RAF kinase inhibitors in driving RAF activation. Profiling of second-generation RAF inhibitors confirmed their improved mode of action, but also revealed liabilities that allowed us to discern two properties of an ideal RAF inhibitor: high-binding affinity to all RAF paralogs and maintenance of the OFF/autoinhibited state of the enzyme.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Allosteric Regulation/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Models, Biological , Mutation/genetics , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/chemistry , Protein Multimerization , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics
6.
PLoS Biol ; 14(8): e1002539, 2016 08.
Article in English | MEDLINE | ID: mdl-27552662

ABSTRACT

RAS-induced MAPK signaling is a central driver of the cell proliferation apparatus. Disruption of this pathway is widely observed in cancer and other pathologies. Consequently, considerable effort has been devoted to understanding the mechanistic aspects of RAS-MAPK signal transmission and regulation. While much information has been garnered on the steps leading up to the activation and inactivation of core pathway components, comparatively little is known on the mechanisms controlling their expression and turnover. We recently identified several factors that dictate Drosophila MAPK levels. Here, we describe the function of one of these, the deubiquitinase (DUB) USP47. We found that USP47 acts post-translationally to counteract a proteasome-mediated event that reduces MAPK half-life and thereby dampens signaling output. Using an RNAi-based genetic interaction screening strategy, we identified UBC6, POE/UBR4, and UFD4, respectively, as E2 and E3 enzymes that oppose USP47 activity. Further characterization of POE-associated factors uncovered KCMF1 as another key component modulating MAPK levels. Together, these results identify a novel protein degradation module that governs MAPK levels. Given the role of UBR4 as an N-recognin ubiquitin ligase, our findings suggest that RAS-MAPK signaling in Drosophila is controlled by the N-end rule pathway and that USP47 counteracts its activity.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Animals, Genetically Modified , Calmodulin-Binding Proteins/genetics , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Immunoblotting , MAP Kinase Signaling System/genetics , Models, Biological , Mutation , Protein Stability , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Wings, Animal/metabolism
7.
Genetics ; 197(4): 1237-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899161

ABSTRACT

The Ras/MAPK-signaling pathway plays pivotal roles during development of metazoans by controlling cell proliferation and cell differentiation elicited, in several instances, by receptor tyrosine kinases (RTKs). While the internal mechanism of RTK-driven Ras/MAPK signaling is well understood, far less is known regarding its interplay with other co-required signaling events involved in developmental decisions. In a genetic screen designed to identify new regulators of RTK/Ras/MAPK signaling during Drosophila eye development, we identified the small GTPase Rap1, PDZ-GEF, and Canoe as components contributing to Ras/MAPK-mediated R7 cell differentiation. Rap1 signaling has recently been found to participate in assembling cadherin-based adherens junctions in various fly epithelial tissues. Here, we show that Rap1 activity is required for the integrity of the apical domains of developing photoreceptor cells and that reduced Rap1 signaling hampers the apical accumulation of the Sevenless RTK in presumptive R7 cells. It thus appears that, in addition to its role in cell-cell adhesion, Rap1 signaling controls the partitioning of the epithelial cell membrane, which in turn influences signaling events that rely on apico-basal cell polarity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/genetics , Eye Proteins/metabolism , Eye/embryology , Receptor Protein-Tyrosine Kinases/metabolism , rap1 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Polarity/genetics , Chromosome Mapping , Drosophila/embryology , Drosophila Proteins/genetics , Epithelial Cells/metabolism , Eye Proteins/genetics , Gene Expression Regulation, Developmental , MAP Kinase Signaling System , Organogenesis/genetics , Photoreceptor Cells, Invertebrate/metabolism , Receptor Protein-Tyrosine Kinases/genetics , rap1 GTP-Binding Proteins/genetics
8.
PLoS Biol ; 12(3): e1001809, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24643257

ABSTRACT

The small GTPase RAS is among the most prevalent oncogenes. The evolutionarily conserved RAF-MEK-MAPK module that lies downstream of RAS is one of the main conduits through which RAS transmits proliferative signals in normal and cancer cells. Genetic and biochemical studies conducted over the last two decades uncovered a small set of factors regulating RAS/MAPK signaling. Interestingly, most of these were found to control RAF activation, thus suggesting a central regulatory role for this event. Whether additional factors are required at this level or further downstream remains an open question. To obtain a comprehensive view of the elements functionally linked to the RAS/MAPK cascade, we used a quantitative assay in Drosophila S2 cells to conduct a genome-wide RNAi screen for factors impacting RAS-mediated MAPK activation. The screen led to the identification of 101 validated hits, including most of the previously known factors associated to this pathway. Epistasis experiments were then carried out on individual candidates to determine their position relative to core pathway components. While this revealed several new factors acting at different steps along the pathway--including a new protein complex modulating RAF activation--we found that most hits unexpectedly work downstream of MEK and specifically influence MAPK expression. These hits mainly consist of constitutive splicing factors and thereby suggest that splicing plays a specific role in establishing MAPK levels. We further characterized two representative members of this group and surprisingly found that they act by regulating mapk alternative splicing. This study provides an unprecedented assessment of the factors modulating RAS/MAPK signaling in Drosophila. In addition, it suggests that pathway output does not solely rely on classical signaling events, such as those controlling RAF activation, but also on the regulation of MAPK levels. Finally, it indicates that core splicing components can also specifically impact alternative splicing.


Subject(s)
Alternative Splicing , Drosophila Proteins/genetics , Drosophila/metabolism , MAP Kinase Signaling System , ras Proteins/metabolism , Animals , Cell Line , Cluster Analysis , Drosophila/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Epistasis, Genetic , Gene Expression Regulation , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/physiology , RNA Interference
9.
Nature ; 461(7263): 542-5, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19727074

ABSTRACT

The ERK (extracellular signal-regulated kinase) pathway is an evolutionarily conserved signal transduction module that controls cellular growth, differentiation and survival. Activation of receptor tyrosine kinases (RTKs) by the binding of growth factors initiates GTP loading of RAS, which triggers the initial steps in the activation of the ERK pathway by modulating RAF family kinase function. Once activated, RAF participates in a sequential cascade of phosphorylation events that activate MEK, and in turn ERK. Unbridled signalling through the ERK pathway caused by activating mutations in RTKs, RAS or RAF has been linked to several human cancers. Of note, one member of the RAF family, BRAF, is the most frequently mutated oncogene in the kinase superfamily. Not surprisingly, there has been a colossal effort to understand the underlying regulation of this family of kinases. In particular, the process by which the RAF kinase domain becomes activated towards its substrate MEK remains of topical interest. Here, using Drosophila Schneider S2 cells, we demonstrate that RAF catalytic function is regulated in response to a specific mode of dimerization of its kinase domain, which we term the side-to-side dimer. Moreover, we find that the RAF-related pseudo-kinase KSR (kinase suppressor of Ras) also participates in forming side-to-side heterodimers with RAF and can thereby trigger RAF activation. This mechanism provides an elegant explanation for the longstanding conundrum about RAF catalytic activation, and also provides an explanation for the capacity of KSR, despite lacking catalytic function, to directly mediate RAF activation. We also show that RAF side-to-side dimer formation is essential for aberrant signalling by oncogenic BRAF mutants, and identify an oncogenic mutation that acts specifically by promoting side-to-side dimerization. Together, our data identify the side-to-side dimer interface of RAF as a potential therapeutic target for intervention in BRAF-dependent tumorigenesis.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Protein Multimerization , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Animals , Binding Sites , Biocatalysis , Cell Line , Drosophila Proteins/genetics , Enzyme Activation , Humans , Models, Molecular , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Structure-Activity Relationship
10.
Genetics ; 181(2): 567-79, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19064708

ABSTRACT

The Jun N-terminal kinase and p38 pathways, also known as stress-activated protein kinase (SAPK) pathways, are signaling conduits reiteratively used throughout the development and adult life of metazoans where they play central roles in the control of apoptosis, immune function, and environmental stress responses. We recently identified a Drosophila Ser/Thr phosphatase of the PP2C family, named Alphabet (Alph), which acts as a negative regulator of the Ras/ERK pathway. Here we show that Alph also plays an inhibitory role with respect to Drosophila SAPK signaling during development as well as under stress conditions such as oxidative or genotoxic stresses. Epistasis experiments suggest that Alph acts at a step upstream of the MAPKKs Hep and Lic. Consistent with this interpretation, biochemical experiments identify the upstream MAPKKKs Slpr, Tak1, and Wnd as putative substrates. Together with previous findings, this work identifies Alph as a general attenuator of MAPK signaling in Drosophila.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Cell Line , DNA Primers/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Epistasis, Genetic , Eye Abnormalities/genetics , Female , Genes, Insect , Genes, Lethal , Genotype , MAP Kinase Signaling System , Male , Microscopy, Electron, Scanning , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Oxidative Stress , Phenotype , Phosphoprotein Phosphatases/deficiency , Phosphoprotein Phosphatases/genetics , Protein Kinases/genetics , RNA Interference , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 105(8): 2836-41, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287031

ABSTRACT

RAF kinase functions in the mitogen-activated protein kinase (MAPK) pathway to transmit growth signals to the downstream kinases MEK and ERK. Activation of RAF catalytic activity is facilitated by a regulatory complex comprising the proteins CNK (Connector enhancer of KSR), HYP (Hyphen), and KSR (Kinase Suppressor of Ras). The sterile alpha-motif (SAM) domain found in both CNK and HYP plays an essential role in complex formation. Here, we have determined the x-ray crystal structure of the SAM domain of CNK in complex with the SAM domain of HYP. The structure reveals a single-junction SAM domain dimer of 1:1 stoichiometry in which the binding mode is a variation of polymeric SAM domain interactions. Through in vitro and in vivo mutational analyses, we show that the specific mode of dimerization revealed by the crystal structure is essential for RAF signaling and facilitates the recruitment of KSR to form the CNK/HYP/KSR regulatory complex. We present two docking-site models to account for how SAM domain dimerization might influence the formation of a higher-order CNK/HYP/KSR complex.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Drosophila Proteins/genetics , Models, Molecular , Protein Kinases/metabolism , Protein Structure, Tertiary , Signal Transduction/genetics , raf Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Crystallization , Dimerization , Drosophila Proteins/metabolism , Escherichia coli , Humans
12.
J Endocrinol ; 190(1): 107-15, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16837615

ABSTRACT

In the present study, we determined the potential for post-transcriptional regulation of cytochrome P450 aromatase (Cyp19), cytochrome P450 side-chain cleavage (Cyp11a) and 17beta-hydroxysteroid dehydrogenase I (Hsd17b1) mRNA. Bovine granulosa cells were cultured in non-luteinizing conditions that permit long-term oestradiol secretion. Half-lives of mRNA were measured by Northern and/or reverse transcriptase (RT)-PCR after inhibition of gene transcription. In FSH-stimulated cells, the Cyp11a and Hsd17b1 mRNAs had half-lives greater than 12 h. The half-life of Cyp19 mRNA was significantly shorter at 3 h. The addition of the translation inhibitor cycloheximide to FSH-stimulated cells significantly increased Cyp19 mRNA half-life to approximately 12 h. Stimulation of cells with insulin resulted in Cyp19 mRNA half-life that was double (P<0.05) that in FSH-stimulated cells. We conclude that bovine Cyp19 mRNA is very labile under physiological conditions, and that Cyp19 expression is under hormonal control at a post-transcriptional level.


Subject(s)
Aromatase/genetics , Gene Expression Regulation, Enzymologic , Granulosa Cells/metabolism , RNA, Messenger/metabolism , Animals , Blotting, Northern/methods , Cattle , Cells, Cultured , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Female , Flavonoids/pharmacology , Follicle Stimulating Hormone/pharmacology , Half-Life , Insulin/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical
13.
Genes Dev ; 20(7): 807-19, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16600912

ABSTRACT

RAF is a critical effector of the small GTPase RAS in normal and malignant cells. Despite intense scrutiny, the mechanism regulating RAF activation remains partially understood. Here, we show that the scaffold KSR (kinase suppressor of RAS), a RAF homolog known to assemble RAF/MEK/ERK complexes, induces RAF activation in Drosophila by a mechanism mediated by its kinase-like domain, but which is independent of its scaffolding property or putative kinase activity. Interestingly, we found that KSR is recruited to RAF prior to signal activation by the RAF-binding protein CNK (connector enhancer of KSR) in association with a novel SAM (sterile alpha motif) domain-containing protein, named Hyphen (HYP). Moreover, our data suggest that the interaction of KSR to CNK/HYP stimulates the RAS-dependent RAF-activating property of KSR. Together, these findings identify a novel protein complex that controls RAF activation and suggest that KSR does not only act as a scaffold for the MAPK (mitogen-activated protein kinase) module, but may also function as a RAF activator. By analogy to catalytically impaired, but conformationally active B-RAF oncogenic mutants, we discuss the possibility that KSR represents a natural allosteric inducer of RAF catalytic function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , raf Kinases/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Catalytic Domain , Cell Line , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Genes, Insect , MAP Kinase Signaling System , Models, Biological , Mutation , Protein Structure, Tertiary , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , raf Kinases/chemistry , raf Kinases/genetics , ras Proteins/genetics
14.
Biol Reprod ; 71(3): 887-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15128599

ABSTRACT

Remodeling of the extracellular matrix (ECM) occurs during antral follicle growth, and the plasminogen activators (PA) have been implicated in this process in rodents. In the present study, we measured the expression and secretion of PA and the PA inhibitor protease nexin-1 (SerpinE2) in antral and basal bovine granulosa cells from small (<6 mm), medium (6-8 mm), and large follicles (>8 mm) during 6 days of culture in serum-free medium. Casein zymography revealed that the cells secreted predominantly tissue-type PA (tPA) with urokinase (uPA) being associated mainly with cell lysates, and Western blot demonstrated that the cells secreted SerpinE2. Overall, secreted tPA activity was higher in cultures of cells from small follicles compared with large follicles, and secreted SerpinE2 levels were higher in cultures of cells from large follicles. In cultures of cells from small follicles, secreted tPA levels increased with time of culture for antral but not basal cells, and SerpinE2 levels increased with time for basal but not antral cells. In cultures of granulosa cells from large follicles, tPA activity increased significantly with time of culture, whereas SerpinE2 levels decreased. Cell-associated uPA activity decreased with time in cells from medium and large follicles. Reverse-transcription polymerase chain reaction and Northern blot analysis showed that SerpinE2 secretion was regulated largely at the transcriptional level, whereas tPA secretion was not. The data suggest stage-dependent regulation of granulosa cell PA and SerpinE2 production, consistent with a role in ECM remodeling during follicle growth.


Subject(s)
Carrier Proteins/metabolism , Granulosa Cells/enzymology , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Amyloid beta-Protein Precursor , Animals , Carrier Proteins/genetics , Cattle , Cells, Cultured , Estradiol/metabolism , Extracellular Matrix/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/metabolism , In Vitro Techniques , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/physiology , Phenotype , Protease Nexins , Receptors, Cell Surface/genetics , Serpin E2
SELECTION OF CITATIONS
SEARCH DETAIL