Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 43(12): 631-649, 2023.
Article in English | MEDLINE | ID: mdl-38014992

ABSTRACT

PTP1B plays a key role in developing different types of cancer. However, the molecular mechanism underlying this effect is unclear. To identify molecular targets of PTP1B that mediate its role in tumorigenesis, we undertook a SILAC-based phosphoproteomic approach, which allowed us to identify Cdk3 as a novel PTP1B substrate. Substrate trapping experiments and docking studies revealed stable interactions between the PTP1B catalytic domain and Cdk3. In addition, we observed that PTP1B dephosphorylates Cdk3 at tyrosine residue 15 in vitro and interacts with it in human glioblastoma cells. Next, we found that pharmacological inhibition of PTP1B or its depletion with siRNA leads to cell cycle arrest with diminished activity of Cdk3, hypophosphorylation of Rb, and the downregulation of E2F target genes Cdk1, Cyclin A, and Cyclin E1. Finally, we observed that the expression of a constitutively active Cdk3 mutant bypasses the requirement of PTP1B for cell cycle progression and expression of E2F target genes. These data delineate a novel signaling pathway from PTP1B to Cdk3 required for efficient cell cycle progression in an Rb-E2F dependent manner in human GB cells.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Cell Division , Signal Transduction , Cell Cycle Checkpoints , Cell Cycle/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
2.
Int J Mol Sci ; 24(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36768437

ABSTRACT

In 2013, recognizing that Colorectal Cancer (CRC) is the second leading cause of death by cancer worldwide and that it was a neglected disease increasing rapidly in Mexico, the community of researchers at the Biomedicine Research Unit of the Facultad de Estudios Superiores Iztacala from the Universidad Nacional Autónoma de México (UNAM) established an intramural consortium that involves a multidisciplinary group of researchers, technicians, and postgraduate students to contribute to the understanding of this pathology in Mexico. This article is about the work developed by the Mexican Colorectal Cancer Research Consortium (MEX-CCRC): how the Consortium was created, its members, and its short- and long-term goals. Moreover, it is a narrative of the accomplishments of this project. Finally, we reflect on possible strategies against CRC in Mexico and contrast all the data presented with another international strategy to prevent and treat CRC. We believe that the Consortium's characteristics must be maintained to initiate a national strategy, and the reported data could be useful to establish future collaborations with other countries in Latin America and the world.


Subject(s)
Colorectal Neoplasms , Students , Humans , Mexico , Interdisciplinary Studies , Therapies, Investigational , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/therapy
3.
Biosci Rep ; 41(11)2021 11 26.
Article in English | MEDLINE | ID: mdl-34726241

ABSTRACT

Protein tyrosine phosphorylation is one of the major post-translational modifications in eukaryotic cells and represents a critical regulatory mechanism of a wide variety of signaling pathways. Aberrant protein tyrosine phosphorylation has been linked to various diseases, including metabolic disorders and cancer. Few years ago, protein tyrosine phosphatases (PTPs) were considered as tumor suppressors, able to block the signals emanating from receptor tyrosine kinases. However, recent evidence demonstrates that misregulation of PTPs activity plays a critical role in cancer development and progression. Here, we will focus on PTP1B, an enzyme that has been linked to the development of type 2 diabetes and obesity through the regulation of insulin and leptin signaling, and with a promoting role in the development of different types of cancer through the activation of several pro-survival signaling pathways. In this review, we discuss the molecular aspects that support the crucial role of PTP1B in different cellular processes underlying diabetes, obesity and cancer progression, and its visualization as a promising therapeutic target.


Subject(s)
Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Metabolic Diseases/drug therapy , Neoplasms/drug therapy , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Signal Transduction/drug effects
4.
Cells ; 10(4)2021 04 04.
Article in English | MEDLINE | ID: mdl-33916643

ABSTRACT

Lysophosphatidic acid (LPA) induces a wide range of cellular processes and its signaling is increased in several cancers including glioblastoma (GBM), a high-grade astrocytoma, which is the most common malignant brain tumor. LPA1 receptor is expressed in GBM cells and its signaling pathways activate protein kinases C (PKCs). A downstream target of PKC, involved in GBM progression, is the intracellular progesterone receptor (PR), which can be phosphorylated by this enzyme, increasing its transcriptional activity. Interestingly, in GBM cells, PKCα isotype translocates to the nucleus after LPA stimulation, resulting in an increase in PR phosphorylation. In this study, we determined that LPA1 receptor activation induces protein-protein interaction between PKCα and PR in human GBM cells; this interaction increased PR phosphorylation in serine400. Moreover, LPA treatment augmented VEGF transcription, a known PR target. This effect was blocked by the PR selective modulator RU486; also, the activation of LPA1/PR signaling promoted migration of GBM cells. Interestingly, using TCGA data base, we found that mRNA expression of LPAR1 increases according to tumor malignancy and correlates with a lower survival in grade III astrocytomas. These results suggest that LPA1/PR pathway regulates GBM progression.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Progesterone/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Lysophospholipids/pharmacology , Phosphoric Diester Hydrolases/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Front Cell Dev Biol ; 9: 759259, 2021.
Article in English | MEDLINE | ID: mdl-35111748

ABSTRACT

p21-Activated kinase-1 (Pak1) is frequently overexpressed and/or amplified in human breast cancer and is necessary for transformation of mammary epithelial cells. Here, we show that Pak1 interacts with and phosphorylates the Calcium/Calmodulin-dependent Protein Kinase II (CaMKII), and that pharmacological inhibition or depletion of Pak1 leads to diminished activity of CaMKII. We found a strong correlation between Pak1 and CaMKII expression in human breast cancer samples, and combined inhibition of Pak1 and CaMKII with small-molecule inhibitors was synergistic and induced apoptosis more potently in Her2 positive and triple negative breast cancer (TNBC) cells. Co-adminstration of Pak and CaMKII small-molecule inhibitors resulted in a dramatic reduction of proliferation and an increase in apoptosis in a 3D cell culture setting, as well as an impairment in migration and invasion of TNBC cells. Finally, mice bearing xenografts of TNBC cells showed a significant delay in tumor growth when treated with small-molecule inhibitors of Pak and CaMKII. These data delineate a signaling pathway from Pak1 to CaMKII that is required for efficient proliferation, migration and invasion of mammary epithelial cells, and suggest new therapeutic strategies in breast cancer.

6.
Front Endocrinol (Lausanne) ; 11: 568375, 2020.
Article in English | MEDLINE | ID: mdl-33117284

ABSTRACT

The estrogen receptor alpha (ERα) is a ligand-activated transcription factor whose activity is modulated by its interaction with multiple protein complexes. In this work, we have identified the protein interferon alpha inducible protein 27 (IFI27/ISG12) as a novel ERα-associated protein. IFI27/ISG12 transcription is regulated by interferon and estradiol and its overexpression is associated to reduced overall survival in ER+ breast cancer patients but its function in mammary gland tissue remains elusive. In this study we showed that overexpression of IFI27/ISG12 in breast cancer cells attenuates ERα transactivation activity and the expression of ERα-dependent genes. Our results demonstrated that IFI27/ISG12 overexpression in MCF-7 cells reduced their proliferation rate in 2-D and 3-D cell culture assays and impaired their ability to migrate in a wound-healing assay. We show that IFI27/ISG12 downregulation of ERα transactivation activity is mediated by its ability to facilitate the interaction between ERα and CRM1/XPO1 that mediates the nuclear export of large macromolecules to the cytoplasm. IFI27/ISG12 overexpression was shown to impair the estradiol-dependent proliferation and tamoxifen-induced apoptosis in breast cancer cells. Our results suggest that IFI27/ISG12 may be an important factor in regulating ERα activity in breast cancer cells by modifying its nuclear versus cytoplasmic protein levels. We propose that IFI27/ISG12 may be a potential target of future strategies to control the growth and proliferation of ERα-positive breast cancer tumors.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation/physiology , Estrogen Receptor alpha/biosynthesis , Karyopherins/biosynthesis , Membrane Proteins/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcriptional Activation/physiology , Breast Neoplasms/genetics , Databases, Genetic , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Humans , Karyopherins/genetics , MCF-7 Cells , Membrane Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Tamoxifen/pharmacology , Transcriptional Activation/drug effects , Exportin 1 Protein
7.
Int J Cancer ; 145(11): 3126-3139, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31407335

ABSTRACT

Inflammation is currently considered a hallmark of cancer and plays a decisive role in different stages of tumorigenesis, including initiation, promotion, progression, metastasis and resistance to antitumor therapies. Colorectal cancer is a disease widely associated with local chronic inflammation. Additionally, extrinsic factors such as infection may beneficially or detrimentally alter cancer progression. Several reports have noted the ability of various parasitic infections to modulate cancer development, favoring tumor progression in many cases and inhibiting tumorigenesis in others. The aim of our study was to determine the effects of excreted/secreted products of the helminth Taenia crassiceps (TcES) as a treatment in a murine model of colitis-associated colon cancer (CAC). Here, we found that after inducing CAC, treatment with TcES was able to reduce inflammatory cytokines such as IL-1ß, TNF-α, IL-33 and IL-17 and significantly attenuate colon tumorigenesis. This effect was associated with the inhibition of signal transducer and activator of transcription 3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) phosphorylation. Furthermore, we determined that TcES interfered with LPS-induced NF-κB p65 activation in human colonic epithelial cell lines in a Raf-1 proto-oncogene-dependent manner. Moreover, in three-dimensional cultures, TcES promoted reorganization of the actin cytoskeleton, altering cell morphology and forming colonospheres, features associated with a low grade of aggressiveness. Our study demonstrates a remarkable effect of helminth-derived molecules on suppressing ongoing colorectal cancer by downregulating proinflammatory and protumorigenic signaling pathways.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Azoxymethane/adverse effects , Colitis/drug therapy , Colonic Neoplasms/drug therapy , Helminth Proteins/administration & dosage , Taenia/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Colitis/chemically induced , Colitis/complications , Colonic Neoplasms/etiology , Disease Models, Animal , Female , Helminth Proteins/pharmacology , Humans , Interleukin-1beta/metabolism , Interleukin-33/metabolism , Mice , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Mas , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
DNA Repair (Amst) ; 65: 42-46, 2018 05.
Article in English | MEDLINE | ID: mdl-29597073

ABSTRACT

p21-activated kinase 1 (PAK1) is a serine/threonine kinase activated by the small GTPases Rac1 and Cdc42. It is located in the chromosome 11q13 and is amplified and/or overexpressed in several human cancer types including 25-30% of breast tumors. This enzyme plays a pivotal role in the control of a number of fundamental cellular processes by phosphorylating its downstream substrates. In addition to its role in the cytoplasm, it is well documented that PAK1 also plays crucial roles in the nucleus participating in mitotic events and gene expression through its association and/or phosphorylation of several transcription factors, transcriptional co-regulators and cell cycle-related proteins, including Aurora kinase A (AURKA), polo-like kinase 1 (PLK1), the forkhead transcription factor (FKHR), estrogen receptor α (ERα), and Snail. More recently, PAK signaling has emerged as a component of the DNA damage response (DDR) as PAK1 activity influences the cellular sensitivity to ionizing radiation and promotes the expression of several genes involved in the Fanconi Anemia/BRCA pathway. This review will focus on the nuclear functions of PAK1 and its role in the regulation of DNA damage repair.


Subject(s)
DNA Repair , p21-Activated Kinases/metabolism , Animals , DNA/metabolism , Humans , Signal Transduction
10.
Oncotarget ; 7(47): 76590-76603, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27740936

ABSTRACT

Cells that are deficient in homologous recombination, such as those that have mutations in any of the Fanconi Anemia (FA)/BRCA genes, are hypersensitive to inhibition of poly(ADP-ribose) polymerase (PARP). However, FA/BRCA-deficient tumors represent a small fraction of breast cancers, which might restrict the therapeutic utility of PARP inhibitor monotherapy. The gene encoding the serine-threonine protein kinase p21-activated kinase 1 (PAK1) is amplified and/or overexpressed in several human cancer types including 25-30% of breast tumors. This enzyme controls many cellular processes by phosphorylating both cytoplasmic and nuclear substrates. Here, we show that depletion or pharmacological inhibition of PAK1 down-regulated the expression of genes involved in the FA/BRCA pathway and compromised the ability of cells to repair DNA by Homologous Recombination (HR), promoting apoptosis and reducing colony formation. Combined inhibition of PAK1 and PARP in PAK1 overexpressing breast cancer cells had a synergistic effect, enhancing apoptosis, suppressing colony formation, and delaying tumor growth in a xenograft setting. Because reduced PAK1 activity impaired FA/BRCA function, inhibition of this kinase in PAK1 amplified and/or overexpressing breast cancer cells represents a plausible strategy for expanding the utility of PARP inhibitors to FA/BRCA-proficient cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Fanconi Anemia Complementation Group Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chromosomes, Human, Pair 11/genetics , DNA Damage/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Fanconi Anemia Complementation Group Proteins/deficiency , Female , Gene Amplification , Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination , Humans , Mice , Xenograft Model Antitumor Assays , p21-Activated Kinases/genetics
11.
Curr Biol ; 26(12): 1556-1563, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27238285

ABSTRACT

The protein kinases Mst1 and Mst2 have tumor suppressor activity, but their mode of regulation is not well established. Mst1 and Mst2 are broadly expressed and may have certain overlapping functions in mammals, as deletions of both Mst1 and Mst2 together are required for tumorigenesis in mouse models [1-3]. These kinases act via a three-component signaling cascade comprising Mst1 and Mst2, the protein kinases Lats1 and Lats2, and the transcriptional coactivators Yap and Taz [4-6]. Mst1 and Mst2 contain C-terminal SARAH domains that mediate their homodimerization as well as heterodimerization with other SARAH domain-containing proteins, which may regulate Mst1/Mst2 activity. Here we show that, in addition to forming homodimers, Mst1 and Mst2 heterodimerize in cells, this interaction is mediated by their SARAH domains and is favored over homodimers, and these heterodimers have much-reduced protein kinase activity compared to Mst1 or Mst2 homodimers. Mst1/Mst2 heterodimerization is strongly promoted by oncogenic H-ras, and this effect requires activation of the Erk pathway. Cells lacking Mst1, in which Mst1/Mst2 heterodimers are not possible, are resistant to H-ras-mediated transformation and maintain active hippo pathway signaling compared to wild-type cells or cells lacking both Mst1 and Mst2. Our results suggest that H-ras, via an Erk-dependent mechanism, downregulates Mst1/Mst2 activity by inducing the formation of inactive Mst1/Mst2 heterodimers.


Subject(s)
Genes, ras , Protein Multimerization , Protein Serine-Threonine Kinases/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Serine-Threonine Kinase 3
12.
Mol Cell Biol ; 35(23): 3990-4005, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391956

ABSTRACT

p21-activated kinases (Paks) have been shown to regulate cytoskeleton rearrangements, cell proliferation, attachment, and migration in a variety of cellular contexts, including endothelial cells. However, the role of endothelial Pak in embryo development has not been reported, and currently, there is no consensus on the endothelial function of individual Pak isoforms, in particular p21-activated kinase 2 (Pak2), the main Pak isoform expressed in endothelial cells. In this work, we employ genetic and molecular studies that show that Pak2, but not Pak1, is a critical mediator of development and maintenance of endothelial cell function. Endothelial depletion of Pak2 leads to early embryo lethality due to flawed blood vessel formation in the embryo body and yolk sac. In adult endothelial cells, Pak2 depletion leads to severe apoptosis and acute angiogenesis defects, and in adult mice, endothelial Pak2 deletion leads to increased vascular permeability. Furthermore, ubiquitous Pak2 deletion is lethal in adult mice. We show that many of these defects are mediated through a newly unveiled Pak2/Bmk1 pathway. Our results demonstrate that endothelial Pak2 is essential during embryogenesis and also for adult blood vessel maintenance, and they also pinpoint the Bmk1/Erk5 pathway as a critical mediator of endothelial Pak2 signaling.


Subject(s)
Endothelium/embryology , Endothelium/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Signal Transduction , p21-Activated Kinases/metabolism , Animals , Capillary Permeability , Cardiovascular Abnormalities/embryology , Cardiovascular Abnormalities/genetics , Cardiovascular Abnormalities/metabolism , Cardiovascular System/embryology , Cardiovascular System/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Embryo Loss , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Endothelium/cytology , Female , Gene Deletion , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells , Male , Mice, Inbred C57BL , RNA Interference , p21-Activated Kinases/genetics
13.
Cancer Res ; 73(12): 3671-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23576562

ABSTRACT

p21-Activated kinase-1 (Pak1) is frequently upregulated in human breast cancer and is required for transformation of mammary epithelial cells by ErbB2. Here, we show that loss of Pak1, but not the closely related Pak2, leads to diminished expression of ß-catenin and its target genes. In MMTV-ErbB2 transgenic mice, loss of Pak1 prolonged survival, and mammary tissues of such mice showed loss of ß-catenin. Expression of a ß-catenin mutant bearing a phospho-mimetic mutation at Ser 675, a specific Pak1 phosphorylation site, restored transformation to ErbB2-positive, Pak1-deficient mammary epithelial cells. Mice bearing xenografts of ErbB2-positive breast cancer cells showed tumor regression when treated with small-molecule inhibitors of Pak or ß-catenin, and combined inhibition by both agents was synergistic. These data delineate a signaling pathway from ErbB2 to Pak to ß-catenin that is required for efficient transformation of mammary epithelial cells, and suggest new therapeutic strategies in ErbB2-positive breast cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Receptor, ErbB-2/metabolism , beta Catenin/metabolism , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Humans , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, SCID , Mice, Transgenic , Pyrazoles/pharmacology , Pyrroles/pharmacology , RNA Interference , Receptor, ErbB-2/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics , p21-Activated Kinases/genetics
14.
Cancer Res ; 69(11): 4582-8, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435911

ABSTRACT

Protein tyrosine phosphatase (PTP) 1B plays a major role in inhibiting signaling from the insulin and leptin receptors. Recently, PTP1B was found to have an unexpected positive role in ErbB2 signaling in a mouse model of breast cancer, but the mechanism underlying this effect has been unclear. Using human breast epithelial cells grown in a three-dimensional matrix, we found that PTP1B, but not the closely related enzyme T-cell PTP, is required for ErbB2 transformation in vitro. Activation of ErbB2, but not ErbB1, increases PTP1B expression, and increased expression of PTP1B activates Src and induces a Src-dependent transformed phenotype. These findings identify a molecular mechanism by which PTP1B links an important oncogenic receptor tyrosine kinase to signaling pathways that promote aberrant cell division and survival in human breast epithelial cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Genes, erbB-2/physiology , Mammary Glands, Human/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Cell Division/genetics , Cell Division/physiology , Cell Survival/genetics , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proto-Oncogene Proteins pp60(c-src)/physiology , Signal Transduction/genetics , Transfection
15.
Brain Res Bull ; 69(3): 276-81, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16564422

ABSTRACT

In this work we have determined the role of the 26S proteasome in the regulation of the content of progesterone receptors (PR-A and PR-B), estrogen receptors (ER-alpha and ER-beta), the coactivator SRC-1 and the corepressor SMRT in the rat brain during the estrous cycle. The 26S proteasome inhibitor MG132 was injected once into the lateral ventricle on proestrous day; and 24h later, on estrous day we evaluated the content of PR and ER isoforms, SRC-1 and SMRT in the hypothalamus, the preoptic area and the hippocampus by Western blot. A significant increase in the content of both PR isoforms, ER-beta and SRC-1 was observed after the administration of MG132 in the three studied cerebral regions. SMRT content was increased in the hypothalamus and the preoptic area and a significant increase in ER-alpha content was only observed in the preoptic area. These results suggest that essential proteins that participate in progesterone and estrogen actions in the brain should be regulated by the 26S proteasome in a tissue-specific manner in physiological conditions.


Subject(s)
Brain/drug effects , DNA-Binding Proteins/metabolism , Estrous Cycle/drug effects , Gene Expression Regulation/drug effects , Progesterone/metabolism , Proteasome Endopeptidase Complex/pharmacology , Receptors, Estrogen/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western/methods , Brain/metabolism , Estrous Cycle/physiology , Female , Histone Acetyltransferases , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivator 1 , Rats , Rats, Wistar
16.
J Steroid Biochem Mol Biol ; 85(1): 25-31, 2003 May.
Article in English | MEDLINE | ID: mdl-12798354

ABSTRACT

In this work, we determined the expression pattern and the hormonal regulation of progesterone receptor (PR) isoforms in the rat lung of ovariectomized female rats after estradiol (E2) and progesterone (P4) treatments. We also evaluated the content of estrogen receptor beta (ER-beta) which is the ER isoform expressed in the lung. RNA and proteins were extracted and processed for reverse transcription (RT) coupled to polymerase chain reaction (PCR) and Western blot, respectively. The expression of both PR isoforms in the lung at mRNA and at protein levels was up-regulated by E2 while P4 down-regulated it at mRNA level. P4 did not modify PR isoforms protein content unlike its effect in the uterus where both PR isoforms were down-regulated by their ligand at mRNA and protein levels. PR-A was the predominant isoform, both in the lung and in the uterus. In the lung, ER-beta was down-regulated by E2 while P4 did not significantly modify the effect of E2. These results suggest that both PR isoforms should be expressed in the rat lung, and that their expression should be differentially regulated at mRNA and at protein levels by P4. We also suggest that the up-regulation of PR isoforms by E2 in the lung is mediated by ER-beta.


Subject(s)
Estradiol/pharmacology , Lung/metabolism , Progesterone/pharmacology , Receptors, Progesterone/biosynthesis , Animals , Blotting, Western , Estrogen Receptor beta , Female , Protein Isoforms , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Uterus/metabolism
17.
Neuroendocrinology ; 76(5): 267-71, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12457037

ABSTRACT

The aim of this study was to investigate the participation of the 26S proteasome in the regulation of progesterone receptor (PR) concentrations in the rat brain in vivo. Ovariectomized adult female rats were treated with estradiol (10 microg/100 g s.c.), estradiol + progesterone (400 microg/100 g), and vehicle (corn oil/10% ethanol) in the presence or absence of the proteasome inhibitor Z-Ile-Glu (OBu(1))-Ala-Leu-H (PSI, 300 microg/100 g). Proteins were extracted from the preoptic area, the hippocampus, and the frontal cortex, and processed for Western blot. Estradiol-induced PR expression in the preoptic area and the hippocampus, whereas progesterone did not modify the effect of estradiol. Neither estradiol nor progesterone modified PR content in the frontal cortex. PSI treatment increased PR content in the preoptic area and the hippocampus. This increase was significant in both regions after 24 h of the treatment with progesterone + PSI in the animals primed with estradiol. In this case, the content of both PR isoforms (PR-A and PR-B) was increased in a similar manner by PSI in the preoptic area (90 and 97%) and in the hippocampus (49 and 50%). PSI did not affect PR content in the frontal cortex. Our results suggest that the 26S proteasome could participate in the turnover of PR in the preoptic area and the hippocampus of the rat in vivo.


Subject(s)
Brain/metabolism , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Receptors, Progesterone/metabolism , Animals , Blotting, Western , Brain/drug effects , Estradiol/metabolism , Female , Frontal Lobe/metabolism , Hippocampus/metabolism , Ovariectomy , Preoptic Area/metabolism , Progesterone/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...