Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 15(5): 361, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796462

ABSTRACT

Disease models of neurodegeneration with brain iron accumulation (NBIA) offer the possibility to explore the relationship between iron dyshomeostasis and neurodegeneration. We analyzed hiPS-derived astrocytes from PANK2-associated neurodegeneration (PKAN), an NBIA disease characterized by progressive neurodegeneration and high iron accumulation in the globus pallidus. Previous data indicated that PKAN astrocytes exhibit alterations in iron metabolism, general impairment of constitutive endosomal trafficking, mitochondrial dysfunction and acquired neurotoxic features. Here, we performed a more in-depth analysis of the interactions between endocytic vesicles and mitochondria via superresolution microscopy experiments. A significantly lower number of transferrin-enriched vesicles were in contact with mitochondria in PKAN cells than in control cells, confirming the impaired intracellular fate of cargo endosomes. The investigation of cytosolic and mitochondrial iron parameters indicated that mitochondrial iron availability was substantially lower in PKAN cells compared to that in the controls. In addition, PKAN astrocytes exhibited defects in tubulin acetylation/phosphorylation, which might be responsible for unregulated vesicular dynamics and inappropriate iron delivery to mitochondria. Thus, the impairment of iron incorporation into these organelles seems to be the cause of cell iron delocalization, resulting in cytosolic iron overload and mitochondrial iron deficiency, triggering mitochondrial dysfunction. Overall, the data elucidate the mechanism of iron accumulation in CoA deficiency, highlighting the importance of mitochondrial iron deficiency in the pathogenesis of disease.


Subject(s)
Astrocytes , Cytosol , Iron Overload , Iron , Mitochondria , Astrocytes/metabolism , Astrocytes/pathology , Humans , Mitochondria/metabolism , Cytosol/metabolism , Iron/metabolism , Iron Overload/metabolism , Iron Overload/pathology , Tubulin/metabolism , Phosphorylation , Iron Deficiencies , Acetylation
2.
Plants (Basel) ; 12(1)2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36616135

ABSTRACT

Pollen tubes are tip-growing cells that create safe routes to convey sperm cells to the embryo sac for double fertilization. Recent studies have purified and biochemically characterized detergent-insoluble membranes from tobacco pollen tubes. These microdomains, called lipid rafts, are rich in sterols and sphingolipids and are involved in cell polarization in organisms evolutionarily distant, such as fungi and mammals. The presence of actin in tobacco pollen tube detergent-insoluble membranes and the preferential distribution of these domains on the apical plasma membrane encouraged us to formulate the intriguing hypothesis that sterols and sphingolipids could be a "trait d'union" between actin dynamics and polarized secretion at the tip. To unravel the role of sterols and sphingolipids in tobacco pollen tube growth, we used squalestatin and myriocin, inhibitors of sterol and sphingolipid biosynthesis, respectively, to determine whether lipid modifications affect actin fringe morphology and dynamics, leading to changes in clear zone organization and cell wall deposition, thus suggesting a role played by these lipids in successful fertilization.

3.
Kidney Int ; 98(4): 883-896, 2020 10.
Article in English | MEDLINE | ID: mdl-32919786

ABSTRACT

Loss-of-function mutations in the OCRL gene, which encodes the phosphatidylinositol [PI] 4,5-bisphosphate [PI(4,5)P2] 5-phosphatase OCRL, cause defective endocytosis and proximal tubule dysfunction in Lowe syndrome and Dent disease 2. The defect is due to increased levels of PI(4,5)P2 and aberrant actin polymerization, blocking endosomal trafficking. PI 3-phosphate [PI(3)P] has been recently identified as a coactivator with PI(4,5)P2 in the actin pathway. Here, we tested the hypothesis that phosphoinositide 3-kinase (PI3K) inhibitors may rescue the endocytic defect imparted by OCRL loss, by rebalancing phosphoinositide signals to the actin machinery. The broad-range PI3K inhibitor copanlisib and class IA p110α PI3K inhibitor alpelisib reduced aberrant actin polymerization in OCRL-deficient human kidney cells in vitro. Levels of PI 3,4,5-trisphosphate, PI(4,5)P2 and PI(3)P were all reduced with alpelisib treatment, and siRNA knockdown of the PI3K catalytic subunit p110α phenocopied the actin phenotype. In a humanized OcrlY/- mouse model, alpelisib reduced endosomal actin staining while restoring stress fiber architecture and levels of megalin at the plasma membrane of proximal tubule cells, reflected by improved endocytic uptake of low molecular weight proteins in vivo. Thus, our findings support the link between phosphoinositide lipids, actin polymerization and endocytic trafficking in the proximal tubule and represent a proof-of-concept for repurposing alpelisib in Lowe syndrome/Dent disease 2.


Subject(s)
Dent Disease , Oculocerebrorenal Syndrome , Actins , Humans , Mice , Oculocerebrorenal Syndrome/genetics , Phosphatidylinositol 3-Kinases , Phosphatidylinositol Phosphates , Phosphoinositide-3 Kinase Inhibitors , Phosphoric Monoester Hydrolases/genetics , Thiazoles
4.
Commun Biol ; 3(1): 27, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31941999

ABSTRACT

The structural plasticity of G-protein coupled receptors (GPCRs) enables the long-range transmission of conformational changes induced by specific orthosteric site ligands and other pleiotropic factors. Here, we demonstrate that the ligand binding cavity in the sphingosine 1-phosphate receptor S1PR1, a class A GPCR, is in allosteric communication with both the ß-arrestin-binding C-terminal tail, and a receptor surface involved in oligomerization. We show that S1PR1 oligomers are required for full response to different agonists and ligand-specific association with arrestins, dictating the downstream signalling kinetics. We reveal that the active form of the immunomodulatory drug fingolimod, FTY720-P, selectively harnesses both these intramolecular networks to efficiently recruit ß-arrestins in a stable interaction with the receptor, promoting deep S1PR1 internalization and simultaneously abrogating ERK1/2 phosphorylation. Our results define a molecular basis for the efficacy of fingolimod for people with multiple sclerosis, and attest that GPCR signalling can be further fine-tuned by the oligomeric state.


Subject(s)
Allosteric Regulation , Models, Molecular , Protein Conformation , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Cell Line , Cell Membrane/metabolism , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacology , Humans , Kinetics , Phosphorylation , Proprotein Convertases/chemistry , Proprotein Convertases/metabolism , Protein Binding , Protein Multimerization , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Signal Transduction , Structure-Activity Relationship , beta-Arrestins/chemistry , beta-Arrestins/metabolism
6.
Mol Cell Biol ; 38(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30201806

ABSTRACT

The translational capability of ribosomes deprived of specific nonfundamental ribosomal proteins may be altered. Physiological mechanisms are scanty, and it is unclear whether free ribosomal proteins can cross talk with the signaling machinery. RACK1 (receptor for activated C kinase 1) is a highly conserved scaffold protein, located on the 40S subunit near the mRNA exit channel. RACK1 is involved in a variety of intracellular contexts, both on and off the ribosomes, acting as a receptor for proteins in signaling, such as the protein kinase C (PKC) family. Here we show that the binding of RACK1 to ribosomes is essential for full translation of capped mRNAs and efficient recruitment of eukaryotic initiation factor 4E (eIF4E). In vitro, when RACK1 is partially depleted, supplementing the ribosome machinery with wild-type RACK1 restores the translational capability, whereas the addition of a RACK1 mutant that is unable to bind ribosomes does not. Outside the ribosome, RACK1 has a reduced half-life. By accumulating in living cells, free RACK1 exerts an inhibitory phenotype, impairing cell cycle progression and repressing global translation. Here we present RACK1 binding to ribosomes as a crucial way to regulate translation, possibly through interaction with known partners on or off the ribosome that are involved in signaling.

7.
Platelets ; 28(6): 585-594, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27897101

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and fatal intracranial cancer in humans and exhibits intense and aberrant angiogenesis that sustains its malignancy and involves several angiogenic signals. Among them, vascular endothelial growth factor (VEGF) plays a key role and is overexpressed in GBM. Different cells appear to act as triggers of the aberrant angiogenesis, and, among them, platelets act as key participants. In order to provide further insights into the platelet features and angiogenic role in GBM, this study investigated the effects of platelet releasate on GBM-derived endothelial cells (GECs) and the levels of VEGF and endostatin, as pro- and anti-angiogenic components of platelet releasate from GBM patients. We demonstrate for the first time that: 1) platelet releasate exerts powerful pro-angiogenic effect on GECs, suggesting it might exert a role in the aberrant angiogenesis of GBM; 2) ADP and thrombin stimulation leads to significantly higher level of VEGF, but not of endostatin, in the releasate of platelets from GBM patients than those from healthy subjects; and 3) the intraplatelet concentrations of VEGF were significantly elevated in GBM patients as compared to controls. Moreover, we found a direct correlation between platelet-released VEGF and overall survival in our patient cohort. Although preliminary, these findings prompt further investigations to clarify the biologic relevance of platelet VEGF in GBM and prospective studies for screening GBM patients for anti-VEGF therapy and/or to optimize this treatment.


Subject(s)
Blood Platelets/metabolism , Brain Neoplasms/metabolism , Endothelial Cells/metabolism , Glioblastoma/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Blood Platelets/pathology , Brain Neoplasms/pathology , Endothelial Cells/pathology , Female , Glioblastoma/pathology , Humans , Male , Middle Aged , Neovascularization, Pathologic/pathology , Tumor Cells, Cultured
8.
Nat Commun ; 7: 11903, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27301576

ABSTRACT

Stem cell identity depends on the integration of extrinsic and intrinsic signals, which directly influence the maintenance of their epigenetic state. Although Myc transcription factors play a major role in stem cell self-renewal and pluripotency, their integration with signalling pathways and epigenetic regulators remains poorly defined. We addressed this point by profiling the gene expression and epigenetic pattern in ESCs whose growth depends on conditional Myc activity. Here we show that Myc potentiates the Wnt/ß-catenin signalling pathway, which cooperates with the transcriptional regulatory network in sustaining ESC self-renewal. Myc activation results in the transcriptional repression of Wnt antagonists through the direct recruitment of PRC2 on these targets. The consequent potentiation of the autocrine Wnt/ß-catenin signalling induces the transcriptional activation of the endogenous Myc family members, which in turn activates a Myc-driven self-reinforcing circuit. Thus, our data unravel a Myc-dependent self-propagating epigenetic memory in the maintenance of ESC self-renewal capacity.


Subject(s)
Gene Regulatory Networks/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Self Renewal/drug effects , Epigenesis, Genetic/drug effects , Feedback, Physiological/drug effects , Gene Regulatory Networks/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Polycomb-Group Proteins/metabolism , Transcription, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
9.
Stem Cells Dev ; 25(14): 1095-107, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27217090

ABSTRACT

Central nervous system (CNS) inflammation is primarily driven by microglial cells which secrete proinflammatory cytokines and undergo proliferation upon activation, as it occurs in neurodegenerative diseases. Uncontrolled or prolonged CNS inflammation is potentially harmful and can result in cellular damage. Recently, many studies have focused on human adipose tissue as an attractive source of cytokines with immunosuppressive properties that potentially modulate inflammation. Our study aimed to evaluate if different methods of human tissue collection could affect adipose mesenchymal stem cell (ADSC)-derived cytokine secretion and investigate the effects of ADSC secretome in modulating microglia activation and the possible implication of sphingosine-1-phosphate (S1P) in these effects. Our results demonstrate that the conditioned medium (CM) of ADSCs isolated by two different processing methods (lipoaspirate and Lipogems) significantly inhibited the lipopolysaccharide (LPS)-induced effects on microglia activation, including microglial expression of CD68, cytokine secretion, proliferation, and migration. Pulse studies with radiolabeled sphingosine demonstrated that LPS treatment of resting microglia induced a significant increase of both cellular and extracellular S1P. Moreover, and of relevance, FTY720, a functional antagonist of S1P receptor, inhibited the multiple LPS-induced proinflammatory effects on microglia, and S1P suppressed the anti-inflammatory effect of ADSC-CM. This suggests that LPS-mediated microglial activation is countered by ADSC-CM through the modulation of sphingosine kinase/S1P signalling.


Subject(s)
Adipose Tissue/cytology , Inflammation/pathology , Lysophospholipids/metabolism , Mesenchymal Stem Cells/metabolism , Microglia/pathology , Proteome/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Adult , Cell Proliferation/drug effects , Cell Separation , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Down-Regulation/drug effects , Female , Fingolimod Hydrochloride/pharmacology , Humans , Inflammation/metabolism , Lipopolysaccharides , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Microglia/metabolism , Middle Aged , Phenotype , Signal Transduction/drug effects , Sphingosine/metabolism
10.
Oncotarget ; 6(19): 17514-31, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26020805

ABSTRACT

The vacuolar H+ ATPase (V-ATPase) is a proton pump responsible for acidification of cellular microenvironments, an activity exploited by tumors to survive, proliferate and resist to therapy. Despite few observations, the role of V-ATPase in human tumorigenesis remains unclear.We investigated the expression of ATP6V0C, ATP6V0A2, encoding two subunits belonging to the V-ATPase V0 sector and ATP6V1C, ATP6V1G1, ATPT6V1G2, ATP6V1G3, which are part of the V1 sector, in series of adult gliomas and in cancer stem cell-enriched neurospheres isolated from glioblastoma (GBM) patients. ATP6V1G1 expression resulted significantly upregulated in tissues of patients with GBM and correlated with shorter patients' overall survival independent of clinical variables.ATP6V1G1 knockdown in GBM neurospheres hampered sphere-forming ability, induced cell death, and decreased matrix invasion, a phenotype not observed in GBM monolayer cultures. Treating GBM organotypic cultures or neurospheres with the selective V-ATPase inhibitor bafilomycin A1 reproduced the effects of ATP6V1G1 siRNA and strongly suppressed expression of the stem cell markers Nestin, CD133 and transcription factors SALL2 and POU3F2 in neurospheres.These data point to ATP6V1G1 as a novel marker of poor prognosis in GBM patients and identify V-ATPase inhibition as an innovative therapeutic strategy for GBM.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Vacuolar Proton-Translocating ATPases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/enzymology , Brain Neoplasms/mortality , Cell Movement , Cell Survival , Female , Fluorescent Antibody Technique , Glioblastoma/enzymology , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Transfection , Young Adult
11.
J Biol Chem ; 287(2): 1242-51, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22110132

ABSTRACT

Correct function of spermatogonia is critical for the maintenance of spermatogenesis throughout life, but the cellular pathways regulating undifferentiated spermatogonia proliferation, differentiation, and survival are only partially known. We show here that long glucocorticoid-induced leucine zipper (L-GILZ) is highly expressed in spermatogonia and primary spermatocytes and controls spermatogenesis. Gilz deficiency in knock-out (gilz KO) mice leads to a complete loss of germ cell lineage within first cycles of spermatogenesis, resulting in male sterility. Spermatogenesis failure is intrinsic to germ cells and is associated with increased proliferation and aberrant differentiation of undifferentiated spermatogonia and with hyperactivity of Ras signaling pathway as indicated by an increase of ERK and Akt phosphorylation. Spermatogonia differentiation does not proceed beyond the prophase of the first meiotic division due to massive apoptosis associated with accumulation of unrepaired chromosomal damage. These results identify L-GILZ as a novel important factor for undifferentiated spermatogonia function and spermatogenesis.


Subject(s)
Cell Differentiation/physiology , Signal Transduction/physiology , Spermatogenesis/physiology , Spermatogonia/metabolism , Transcription Factors/metabolism , ras Proteins/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Meiosis/physiology , Mice , Mice, Knockout , Phosphorylation/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt , Spermatogonia/cytology , Transcription Factors/genetics , ras Proteins/genetics
12.
Gene ; 458(1-2): 37-44, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20347019

ABSTRACT

Estrogen receptors (ER), members of the nuclear steroid receptor superfamily, act to activate transcription through ligand-dependent recruitment of coregulators and chromatin modifications. A series of synthetic A-ring reduced 19-nortestosterone-derived progestins has the capacity to selectively bind ERalpha for activated transcription, and to recruit coregulatory factors. In this study, we have analyzed the ability of synthetic 19-nortestosterone derivatives to visibly alter the configuration of ER-target gene chromatin using a novel mammalian promoter transcriptional biosensor (PRL-array) stably transfected into the genome of HeLa cells (PRL-HeLa cells). Results from synthetic steroid-treated cells expressing functional GFP-ERalpha or YFP-ERbeta chimeras were compared to those obtained with estradiol (E(2)) and the antiestrogen tamoxifen. In the presence of synthetic ligands or E(2) a concentration-dependent increase in area of the biosensor array was observed in GFP-ERalpha-expressing PRL-HeLa cells. No significant differences were found between the effects obtained with natural and synthetic steroids. Similarly, E(2) or synthetic steroids-treated PRL-HeLa cells also resulted in similar colocalization of SRC-1- and RNAPII-immunofluorescence at the array. YFP-ERbeta-expressing PRL-HeLa cells treated with E(2) showed increases in array area that were similar to ERalpha; however, treatment of YFP-ERbeta-expressing cells with synthetic ligands was indistinguishable from vehicle controls. These data indicate that A-ring reduced 19-nortestosterone derivatives have an estrogen-like effect on chromatin, including recruitment of transcription factors through selective interactions with ERalpha.


Subject(s)
Biosensing Techniques , Chromatin/metabolism , Receptors, Estrogen/metabolism , Chromatin/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , HeLa Cells , Humans , Ligands , Nandrolone/pharmacology , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Receptors, Estrogen/genetics , Transcription, Genetic
13.
Circ Res ; 106(3): 559-72, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20007915

ABSTRACT

RATIONALE: The Notch signaling pathway is important for cell-cell communication that controls tissue formation and homeostasis during embryonic and adult life, but the precise cell targets of Notch signaling in the mammalian heart remain poorly defined. OBJECTIVE: To investigate the functional role of Notch signaling in the cardiomyocyte compartment of the embryonic and adult heart. METHODS AND RESULTS: Here, we report that either conditional overexpression of Notch1 intracellular domain (NICD1) or selective silencing of Notch signaling in the embryonic cardiomyocyte compartment results in developmental defects and perinatal lethality. In contrast, augmentation of endogenous Notch reactivation after myocardial infarction in the adult, either by inducing cardiomyocyte-specific Notch1 transgene expression or by intramyocardial delivery of a Notch1 pseudoligand, increases survival rate, improves cardiac functional performance, and minimizes fibrosis, promoting antiapoptotic and angiogenic mechanisms. CONCLUSIONS: These results reveal a strict requirement for cell-autonomous modulation of Notch signaling during heart morphogenesis, and illustrate how the same signaling pathway that promotes congenital heart defects when perturbed in the embryo can be therapeutically redeployed for the treatment of adult myocardial damage.


Subject(s)
Myocytes, Cardiac/physiology , Receptor, Notch1/physiology , Age Factors , Animals , Cell Differentiation , Collateral Circulation/physiology , Fetal Heart/cytology , Gene Expression Regulation , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/cytology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Protein Structure, Tertiary , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Regeneration , Signal Transduction/drug effects , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology
14.
Breast Cancer Res Treat ; 119(1): 71-85, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19205871

ABSTRACT

Estrogens play a crucial role in breast tumor growth, which is the rationale for the use of antiestrogens, such as tamoxifen, in women with estrogen receptor (ER)-alpha-positive breast cancer. However, hormone resistance is a major clinical problem. Altered growth factor signaling to the ERalpha pathway has been shown to be associated with the development of clinical resistance. We previously have identified a mutation that replaces arginine for lysine at residue 303 (K303R) of ERalpha, which confers hypersensitive growth in low levels of estrogen. To determine if the K303R mutation could participate in the evolution of hormone resistance, we generated MCF-7 breast cancer cells stably transfected with either wild-type (WT) or K303R ERalpha. We found that the mutation confers decreased sensitivity to tamoxifen in the presence of the growth factor heregulin, using anchorage-independent growth assays. K303R ERalpha-expressing cells were hypersensitive to growth factor signals. Our data suggest that phosphorylation of serine 305 within the hinge domain of ERalpha might play a key role in increasing ligand-independent activity of the mutant receptor. We hypothesize that the mutation adapts the receptor for enhanced bidirectional cross-talk with the HER2 growth factor receptor pathway, which then impacts on responsiveness to tamoxifen.


Subject(s)
Drug Resistance, Neoplasm , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Serine/chemistry , Tamoxifen/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , HeLa Cells , Humans , Lysine/chemistry , Neuregulin-1/metabolism , Phosphorylation , Signal Transduction , Time Factors
15.
J Neurosci ; 29(32): 10131-43, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19675247

ABSTRACT

Understanding the modulation of the neural circuitry of fear is clearly one of the most important aims in neurobiology. Protein phosphorylation in response to external stimuli is considered a major mechanism underlying dynamic changes in neural circuitry. TrkB (Ntrk2) neurotrophin receptor tyrosine kinase potently modulates synaptic plasticity and activates signal transduction pathways mainly through two phosphorylation sites [Y515/Shc site; Y816/PLCgamma (phospholipase Cgamma) site]. To identify the molecular pathways required for fear learning and amygdalar synaptic plasticity downstream of TrkB, we used highly defined genetic mouse models carrying single point mutations at one of these two sites (Y515F or Y816F) to examine the physiological relevance of pathways activated through these sites for pavlovian fear conditioning (FC), as well as for synaptic plasticity as measured by field recordings obtained from neurons of different amygdala nuclei. We show that a Y816F point mutation impairs acquisition of FC, amygdalar synaptic plasticity, and CaMKII signaling at synapses. In contrast, a Y515F point mutation affects consolidation but not acquisition of FC to tone, and also alters AKT signaling. Thus, TrkB receptors modulate specific phases of fear learning and amygdalar synaptic plasticity through two main phosphorylation docking sites.


Subject(s)
Amygdala/physiology , Fear , Learning/physiology , Membrane Glycoproteins/metabolism , Neuronal Plasticity/physiology , Protein-Tyrosine Kinases/metabolism , Synapses/physiology , Animals , Binding Sites/genetics , Binding Sites/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Classical/physiology , Hippocampus/physiology , In Vitro Techniques , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Glycoproteins/genetics , Memory/physiology , Mice , Mice, Mutant Strains , Phosphorylation/physiology , Point Mutation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Synaptic Transmission/physiology
16.
Stem Cells ; 27(12): 3043-52, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19711452

ABSTRACT

Spermatogenesis is maintained by a pool of spermatogonial stem cells (SSCs). Analyses of the molecular profile of SSCs have revealed the existence of subsets, indicating that the stem cell population is more heterogeneous than previously believed. However, SSC subsets are poorly characterized. In rodents, the first steps in spermatogenesis have been extensively investigated, both under physiological conditions and during the regenerative phase that follows germ cell damage. In the widely accepted model, the SSCs are type Asingle (As) spermatogonia. Here, we tested the hypothesis that As spermatogonia are phenotypically heterogeneous by analyzing glial cell line-derived neurotrophic factor (GDNF) family receptor alpha1 (GFRA1) expression in whole-mounted seminiferous tubules, via cytofluorimetric analysis and in vivo colonogenic assays. GFRA1 is a coreceptor for GDNF, a Sertoli cell-derived factor essential for SSC self-renewal and proliferation. Morphometric analysis demonstrated that 10% of As spermatogonia did not express GFRA1 but were colonogenic, as shown by germ cell transplantation assay. In contrast, cells selected for GFRA1 expression were not colonogenic in vivo. In human testes, GFRA1 was also heterogeneously expressed in Adark and in Apale spermatogonia, the earliest spermatogonia. In vivo 5-bromo-2'-deoxyuridine administration showed that both GFRA1(+) and GFRA1(-) As spermatogonia were engaged in the cell cycle, a finding supported by the lack of long-term label-retaining As spermatogonia. GFRA1 expression was asymmetric in 5% of paired cells, suggesting that As subsets may be generated by asymmetric cell division. Our data support the hypothesis of the existence of SSC subsets and reveal a previously unrecognized heterogeneity in the expression profile of As spermatogonia in vivo.


Subject(s)
Cell Shape , Spermatogonia/cytology , Stem Cells/cytology , Aging , Animals , Cell Separation , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Spermatogonia/metabolism , Stem Cells/metabolism
17.
Genesis ; 46(8): 424-30, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18693277

ABSTRACT

An increasing number of genes have been implicated in skeletal muscle fiber diversity. To study the contribution of diverse genetic elements to the regulation of fiber-type composition, we generated a transgenic mouse in which CRE recombinase expression is driven by muscle-specific regulatory sequences of the myosin light chain 1/3 locus (MLC). Using ROSA26 conditional reporter mice, we detected expression of the MLC-Cre transgene starting from embryonic day 12.5 (E12.5). By E15, recombination was detected in all muscle-derived structures. Immunohistochemical analysis revealed CRE activity was restricted to fast-twitch (type II) and excluded from slow-twitch (type I) fibers of skeletal muscle. The MLC-Cre transgenic mouse can be used in conjunction with conditional alleles to study both developmental patterning and maintenance of fast fiber-type phenotypes.


Subject(s)
Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/metabolism , Myosin Light Chains/genetics , Regulatory Sequences, Nucleic Acid , Animals , Genetic Vectors , Integrases/genetics , Mice , Mice, Transgenic , Muscle, Skeletal/embryology , Myosin Light Chains/metabolism
18.
PLoS One ; 3(5): e2286, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18509470

ABSTRACT

Estrogen receptor-alpha (ER) transcription function is regulated in a ligand-dependent (e.g., estradiol, E2) or ligand-independent (e.g., growth factors) manner. Our laboratory seeks to understand these two modes of action. Using a cell line that contains a visible prolactin enhancer/promoter array (PRL-HeLa) regulated by ER, we analyzed ER response to E2 and EGF by quantifying image-based results. Data show differential recruitment of GFP-ER to the array, with the AF1 domain playing a vital role in EGF-mediated responsiveness. Temporal analyses of large-scale chromatin dynamics, and accumulation of array-localized reporter mRNA over 24 hours showed that the EGF response consists of a single pulse of reporter mRNA accumulation concomitant with transient increase in array decondensation. Estradiol induced a novel cyclical pattern of mRNA accumulation with a sustained increase in array decondensation. Collectively, our work shows that there is a stimuli-specific pattern of large-scale chromatin modification and transcript levels by ER.


Subject(s)
Chromatin/metabolism , Epidermal Growth Factor/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , RNA, Messenger/genetics , Transcription, Genetic , HeLa Cells , Humans , In Situ Hybridization, Fluorescence
19.
Breast ; 16(6): 597-605, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17604627

ABSTRACT

HER2, frequently associated with low p27 expression in breast tumors, when activated has been found to upmodulate p53 in tumor cells. The aim of this work was to investigate the role of p53 in the connection between HER2 and p27. Fifty-two breast tumor specimens, characterized for p53 mutations, were analyzed immunohistochemically (IHC) for HER2, p53 and p27 expression. p27, inversely associated with HER2, was found in 29% of tumors with IHC-negative mutated p53 versus 93% of tumors with accumulation of p53 protein and 59% with wild-type p53 (p=0.001), indicating a direct association between p53 and p27 expression. HER2-overexpressing cell lines carrying wild-type or null p53 protein, and treated with heregulin beta1 (HRG), were analyzed for expression and subcellular localization of p53 and p27. In HER2-overexpressing cells stimulated with HRG, p27 protein expression increased in parallel with p53 with no corresponding increase in p27 transcript. No p27 increase was observed in p53-null cells. Transfection with wild-type p53 restored p27 upmodulation in HRG-stimulated cells, indicating a crucial role of p53 in determining p27 upmodulation following HER2 activation. Together, our data demonstrate the crucial role of p53 in determining p27 upmodulation following HER2 activation. This could have implications in the response to Transtuzumab therapy.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, erbB-2/physiology , Proliferating Cell Nuclear Antigen/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Aged, 80 and over , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Middle Aged , Signal Transduction , Up-Regulation
20.
Mol Cell Biol ; 27(17): 6163-76, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606625

ABSTRACT

Signal transduction cascades involving Rho-associated kinases (ROCK), the serine/threonine kinases downstream effectors of Rho, have been implicated in the regulation of diverse cellular functions including cytoskeletal organization, cell size control, modulation of gene expression, differentiation, and transformation. Here we show that ROCK2, the predominant ROCK isoform in skeletal muscle, is progressively up-regulated during mouse myoblast differentiation and is highly expressed in the dermomyotome and muscle precursor cells of mouse embryos. We identify a novel and evolutionarily conserved ROCK2 splicing variant, ROCK2m, that is preferentially expressed in skeletal muscle and strongly up-regulated during in vivo and in vitro differentiation processes. The specific knockdown of ROCK2 or ROCK2m expression in C2C12 myogenic cells caused a significant and selective impairment of the expression of desmin and of the myogenic regulatory factors Mrf4 and MyoD. We demonstrate that in myogenic cells, ROCK2 and ROCK2m are positive regulators of the p42 and p44 mitogen-activated protein kinase-p90 ribosomal S6 kinase-eucaryotic elongation factor 2 intracellular signaling pathways and, thereby, positively regulate the hypertrophic effect elicited by insulin-like growth factor 1 and insulin, linking the multifactorial functions of ROCK to an important control of the myogenic maturation.


Subject(s)
Isoenzymes/metabolism , Muscle Development/physiology , Signal Transduction/physiology , rho-Associated Kinases/metabolism , Alternative Splicing , Animals , Cell Differentiation/physiology , Cells, Cultured , Desmin/genetics , Desmin/metabolism , Enzyme Activation , Humans , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Isoenzymes/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , rho-Associated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL