Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Proc Natl Acad Sci U S A ; 117(9): 4902-4909, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32075916

ABSTRACT

Disagreements about the phenotype of estrogen receptor ß (ERß) knockout mouse, created by removing the DNA-binding domain of the ERß gene or interruption of the gene with a neocassette (Oliver Smithies ERß knockout mice [ERßOS-/-]), prompted us to create an ERß knockout mouse by deleting the ERß gene with the use of CRISPR/Cas9 technology. We confirmed that the ERß gene was eliminated from the mouse genome and that no ERß mRNA or protein was detectable in tissues of this mouse. Overall the phenotype of the ventral prostate (VP) and mammary gland (MG) in ERßcrispr-/- mice was similar to, but more severe than, that in the ERßOS-/-mice. In the VP of 6-mo-old ERßcrispr-/- mice there was epithelial hyperplasia, fibroplasia, inflammation, stromal overgrowth, and intraductal cancer-like lesions. This was accompanied by an increase in Ki67 and P63 and loss in DACH1 and PURα, two androgen receptor (AR) repressors. In the MG there was overexpression of estrogen receptor α and progesterone receptor, loss of collagen, increase in proliferation and expression of metalloproteases, and invasive epithelium. Surprisingly, by 18 mo of age, the number of hyperplastic foci was reduced, the ducts of the VP and MG became atrophic, and, in the VP, there was massive immune infiltration and massive desquamation of the luminal epithelial cells. These changes were coincident with reduced levels of androgens in males and estrogens in females. We conclude that ERß is a tumor suppressor gene in the VP and MG where its loss increases the activity AR and ERα, respectively.


Subject(s)
Estrogen Receptor beta/genetics , Mammary Glands, Animal/metabolism , Phenotype , Prostate/metabolism , Receptors, Androgen/metabolism , Sequence Deletion , Androgens/metabolism , Animals , CRISPR-Cas Systems , Chemokines/metabolism , DNA-Binding Proteins/metabolism , Epithelium/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Eye Proteins , Female , Hyperplasia/pathology , Inflammation , Ki-67 Antigen/metabolism , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Prostate/pathology , RNA, Messenger/metabolism , Signal Transduction , Stromal Cells , Trans-Activators , Transcription Factors/metabolism , Transcriptome , Tumor Suppressor Proteins/metabolism
2.
Int J Mol Sci ; 24(15)2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37569842

ABSTRACT

Liver X receptors (LXRα and LXRß) are oxysterol-activated nuclear receptors that play key roles in cholesterol homeostasis, the central nervous system, and the immune system. We have previously reported that LXRαß-deficient mice are more susceptible to dextran sodium sulfate (DSS)-induced colitis than their WT littermates, and that an LXR agonist protects against colitis in mice mainly via the regulation of the immune system in the gut. We now report that both LXRα and LXRß are expressed in the colonic epithelium and that in aging LXRαß-/- mice there is a reduction in the intensity of goblet cells, mucin (MUC2), TFF3, and estrogen receptor ß (ERß) levels. The cytoplasmic compartment of the surface epithelial cells was markedly reduced and there was a massive invasion of macrophages in the lamina propria. The expression and localization of ß-catenin, α-catenin, and E-cadherin were not changed, but the shrinkage of the cytoplasm led to an appearance of an increase in staining. In the colonic epithelium there was a reduction in the expression of plectin, a hemidesmosome protein whose loss in mice leads to spontaneous colitis, ELOVL1, a fatty acid elongase protein coding gene whose overexpression is found in colorectal cancer, and non-neuronal choline acetyltransferase (ChAT) involved in the regulation of epithelial cell adhesion. We conclude that in aging LXRαß-/- mice, the phenotype in the colon is due to loss of ERß expression.


Subject(s)
Colitis , Estrogen Receptor beta , Mice , Animals , Estrogen Receptor beta/metabolism , Mice, Knockout , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Intestinal Mucosa/metabolism , Dextran Sulfate/toxicity , Mice, Inbred C57BL
3.
Mol Psychiatry ; 26(11): 6380-6393, 2021 11.
Article in English | MEDLINE | ID: mdl-33963286

ABSTRACT

Astrocytes are integral components of synaptic transmission, and their dysfunction leads to neuropsychiatric disorders such as anxiety and depression. Liver X receptor ß (LXRß) is expressed in astrocytes, and LXRß global knockout mice shows impaired synaptic formation. In order to define the role of LXRß in astrocytes, we used a conditional Cre-loxP system to specifically remove LXRß from astrocytes. We found that this deletion caused anxiety-like but not depressive-like behaviors in adult male mice. This behavioral phenotype could be completely reproduced by selective deletion of LXRß in astrocytes in the medial prefrontal cortex (mPFC). Pyramidal neurons in layer V of mPFC are involved in mood behaviors. We found that there was an increased spontaneous excitatory synaptic transmission in layer V pyramidal neurons of the mPFC of these mice. This was concurrent with increased dendritic complexity, despite normal appearance and number of dendritic spines. In addition, gene ontology analysis of RNA sequencing revealed that deletion of astrocytic LXRß led to the enrichment of the process of synaptic transmission in mPFC. Finally, we also confirmed that renormalized excitatory synaptic transmission in layer V pyramidal neurons alleviated the anxiety in mice with astrocytic LXRß deletion in mPFC. Together, our findings reveal that astrocytic LXRß in mPFC is critical in the regulation of synaptic transmission, and this provides a potential new target for treatment of anxiety-like behavior.


Subject(s)
Astrocytes , Prefrontal Cortex , Animals , Anxiety/genetics , Astrocytes/physiology , Liver X Receptors/genetics , Male , Mice , Mice, Knockout , Synaptic Transmission/physiology
4.
Neuroendocrinology ; 111(1-2): 27-44, 2021.
Article in English | MEDLINE | ID: mdl-31991411

ABSTRACT

BACKGROUND: Male estrogen receptor beta (ERß) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERß loss have not been investigated. OBJECTIVE: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERß loss on OL differentiation were confirmed in vitro. CONCLUSION: ERß is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.


Subject(s)
Estrogen Receptor beta/genetics , Locomotion/genetics , Motor Cortex/physiopathology , Myelin Sheath/physiology , Psychomotor Performance , Synaptic Transmission , Animals , Gene Expression , Male , Mice , Mice, Knockout , Motor Cortex/metabolism , Oligodendroglia/pathology
5.
Biochem Biophys Res Commun ; 529(2): 231-237, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703416

ABSTRACT

Estrogen receptor beta (ERß), encoded by the Esr2 gene, is one of two nuclear receptors that mediate the functions of the steroid hormone estradiol. The binding of estradiol to the receptor results in enhanced transcription of many genes that have estrogen response elements in promoter or enhancer regions. Several genetically modified mouse lines with mutations or deletions of exons in the Esr2 gene have been developed and results from analysis of these are not completely consistent, especially regarding ERß's role in fertility. To address these controversies, we have used the CRISPR/Cas9 genome editing system to make a deletion of the entire Esr2 gene in the mouse genome and determined the effect of this mutation on fertility. We show that female Esr2 deleted mice, Esr2ΔE1-10, are subfertile at young age, with fewer litters and smaller litter size, and that they become infertile/have severely reduced fertility at around six months of age, while the male Esr2ΔE1-10 mice are fertile. Ovaries from Esr2ΔE1-10 mice are smaller than those from wild-type littermates and the morphology of the ovary displays very few corpora lutea, indicating a defect in ovulation. We also show that the estradiol levels are reduced at diestrus, the phase in the estrous cycle when levels are expected to start to increase before ovulation. Our results verify that ERß has an important function in female reproduction, likely as a regulator of serum estradiol levels, and that its loss does not affect male reproductive function.


Subject(s)
Estrogen Receptor beta/genetics , Fertility , Animals , CRISPR-Cas Systems , Exons , Female , Gene Deletion , Male , Mice , Ovary/physiology , Ovulation
6.
Proc Natl Acad Sci U S A ; 114(48): E10428-E10437, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133394

ABSTRACT

The ability to propagate mature cells and tissue from pluripotent stem cells offers enormous promise for treating many diseases, including neurodegenerative diseases. Before such cells can be used successfully in neurodegenerative diseases without causing unwanted cell growth and migration, genes regulating growth and migration of neural stem cells need to be well characterized. Estrogen receptor beta (ERß) is essential for migration of neurons and glial cells in the developing mouse brain. To examine whether ERß influences differentiation of mouse embryonic stem cells (mESC) into neural lineages, we compared control and ERß knockout (BERKO) mESCs at defined stages of neural development and examined the effects of an ERß-selective ligand (LY3201) with a combination of global and targeted gene-expression profiling and the expression of key pluripotency markers. We found that ERß was induced in embryoid bodies (EBs) and neural precursor cells (NPCs) during development. Proliferation was higher in BERKO NPCs and was inhibited by LY3201. Neurogenesis was reduced in BERKO ES cells, and oligodendrogliogenesis was enhanced. BERKO EBs expressed higher levels of key ectodermal and neural progenitor markers and lower levels of markers for mesoderm and endoderm lineages. ERß-regulated factors are involved in cell adhesion, axon guidance, and signaling of Notch and GABA receptor pathways, as well as factors important for the differentiation of neuronal precursors into dopaminergic neurons (Engrailed 1) and for the oligodendrocyte fate acquisition (Olig2). Our data suggest that ERß is an important component for differentiation into midbrain neurons as well as for preventing precocious oligodendrogliogenesis.


Subject(s)
Cell Differentiation/physiology , Estrogen Receptor beta/physiology , Mesencephalon/physiology , Mouse Embryonic Stem Cells/physiology , Neural Stem Cells/physiology , Regeneration/physiology , Animals , Benzopyrans/pharmacology , Biomarkers/metabolism , Cell Culture Techniques , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dopaminergic Neurons/physiology , Estrogen Receptor beta/agonists , Female , Gene Expression Profiling , Homeodomain Proteins/metabolism , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Neurogenesis/physiology , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/physiology , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 114(19): E3816-E3822, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439009

ABSTRACT

As estrogen receptor ß-/- (ERß-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERß-/- mice and their WT littermates. We also treated young and old WT mice with an ERß-selective agonist and evaluated protein expression. The most significant findings were that ERß down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERß agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERß-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor ß signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERß in opposing AR signaling, proliferation, and inflammation suggests that ERß-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.


Subject(s)
Aging/metabolism , Down-Regulation , Estrogen Receptor beta/metabolism , Prostate/metabolism , Receptors, Androgen/biosynthesis , Signal Transduction , Aging/genetics , Aging/pathology , Androgens/metabolism , Animals , Chemokine CXCL16/biosynthesis , Chemokine CXCL16/genetics , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Clusterin/biosynthesis , Clusterin/genetics , Estrogen Receptor beta/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Keratins/biosynthesis , Keratins/genetics , Male , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/biosynthesis , Nedd4 Ubiquitin Protein Ligases/genetics , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Prostate/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Androgen/genetics , Smad7 Protein/biosynthesis , Smad7 Protein/genetics
8.
Mol Pharmacol ; 94(4): 1220-1231, 2018 10.
Article in English | MEDLINE | ID: mdl-30115672

ABSTRACT

Recent meta-analyses found an association between prenatal exposure to the antidepressant fluoxetine (FLX) and an increased risk of autism in children. This developmental disorder has been related to dysfunctions in the brains' rewards circuitry, which, in turn, has been linked to dysfunctions in dopaminergic (DA) signaling. The present study investigated if FLX affects processes involved in dopaminergic neuronal differentiation. Mouse neuronal precursors were differentiated into midbrain dopaminergic precursor cells (mDPCs) and concomitantly exposed to clinically relevant doses of FLX. Subsequently, dopaminergic precursors were evaluated for expression of differentiation and stemness markers using quantitative polymerase chain reaction. FLX treatment led to increases in early regional specification markers orthodenticle homeobox 2 (Otx2) and homeobox engrailed-1 and -2 (En1 and En2). On the other hand, two transcription factors essential for midbrain dopaminergic (mDA) neurogenesis, LIM homeobox transcription factor 1 α (Lmx1a) and paired-like homeodomain transcription factor 3 (Pitx3) were downregulated by FLX treatment. The stemness marker nestin (Nes) was increased, whereas the neuronal differentiation marker ß3-tubulin (Tubb3) decreased. Additionally, we observed that FLX modulates the expression of several genes associated with autism spectrum disorder and downregulates the estrogen receptors (ERs) α and ß Further investigations using ERß knockout (BERKO) mDPCs showed that FLX had no or even opposite effects on several of the genes analyzed. These findings suggest that FLX affects differentiation of the dopaminergic system by increasing production of dopaminergic precursors, yet decreasing their maturation, partly via interference with the estrogen system.


Subject(s)
Cell Differentiation/drug effects , Dopaminergic Neurons/drug effects , Fluoxetine/pharmacology , Mesencephalon/drug effects , Animals , Autism Spectrum Disorder/metabolism , Cells, Cultured , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Down-Regulation/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/metabolism , Mesencephalon/metabolism , Mice , Neurogenesis/drug effects , Otx Transcription Factors/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Tubulin/metabolism
9.
Reproduction ; 151(3): 253-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700939

ABSTRACT

Estrogen receptor beta (ERß/ESR2) has a central role in mouse ovaries, as ERß knockout (BERKO) mice are subfertile due to an increase in fibrosis around the maturing follicle and a decrease in blood supply. This has a consequence that these follicles rarely rupture to release the mature oocyte. Matrix metalloproteinases (MMPs) are modulators of the extracellular matrix, and the expression of one specific MMP, MMP-19, is normally increased in granulosa cells during their maturation until ovulation. In this study, we demonstrate that MMP-19 levels are downregulated in BERKO mouse ovaries. Using human MCF-7 cells that overexpress ERß, we could identify MMP-19 to be a transcriptional target of ligand-bound activated ERß acting on a specificity protein-1 binding site. These data provide a molecular explanation for the observed follicle rupture defect that contributes to the subfertility of female BERKO mice.


Subject(s)
Estrogen Receptor beta/metabolism , Matrix Metalloproteinases, Secreted/metabolism , Ovary/metabolism , Animals , Chorionic Gonadotropin , Down-Regulation , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Matrix Metalloproteinases, Secreted/genetics , Mice, Inbred C57BL , Mice, Knockout , Ovulation , Sp1 Transcription Factor/metabolism
10.
Biochem Biophys Res Commun ; 466(4): 650-5, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26393907

ABSTRACT

Estrogen receptor alpha (ERα) is an important regulator of the estrous cycle and mice with global ERα deletion, as well as some conditional knockout mouse lines, have an interruption in the estrous cycle. In this study we observed that conditional ERα knockout mice where the Cre gene is regulated by the rat insulin promoter (RIP), RIP-Cre/ERα(KO) mice, have a 3.7-fold increase in serum 17ß-estradiol levels, blocked estrous cycle, and develop a fluid-filled uterus (hydrometra). Using a proteomics approach, we identified three proteins, lactoferrin, complement C3 and chitinase 3-like protein 1 (CHI3L1), as highly expressed proteins in hydrometra fluid. The mRNA levels of the corresponding genes were more than 50-fold higher in RIP-Cre/ERα(KO) uterus compared to controls. High expression of CHI3L1 in the uterine fluid was not reflected as elevated levels in the serum. The high expression of lactoferrin, complement C3 and CHI3L1 in the uterine fluid, in association with elevated estrogen levels, prompted us to address if the expression of these genes is related to reproduction. However, gonadotropin treatment of mice reduced the uterine expression of these genes in a model of in vitro fertilization. Our findings identify lactoferrin, complement C3 and CHI3L1 as highly expressed proteins in hydrometra fluid in association with chronically elevated serum estradiol levels.


Subject(s)
Glycoproteins/metabolism , Serpins/metabolism , Uterus/metabolism , Animals , Chitinase-3-Like Protein 1 , Complement C3/genetics , Complement C3/metabolism , Estradiol/blood , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrous Cycle/blood , Estrous Cycle/genetics , Estrous Cycle/metabolism , Female , Gene Expression , Glycoproteins/blood , Glycoproteins/genetics , Lactoferrin/genetics , Lactoferrin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Serpins/genetics , Uterine Diseases/genetics , Uterine Diseases/metabolism
11.
Sci Adv ; 10(4): eadj1354, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38266095

ABSTRACT

The brain-specific enzyme CYP46A1 controls cholesterol turnover by converting cholesterol into 24S-hydroxycholesterol (24OH). Dysregulation of brain cholesterol turnover and reduced CYP46A1 levels are observed in Alzheimer's disease (AD). In this study, we report that CYP46A1 overexpression in aged female mice leads to enhanced estrogen signaling in the hippocampus and improved cognitive functions. In contrast, age-matched CYP46A1 overexpressing males show anxiety-like behavior, worsened memory, and elevated levels of 5α-dihydrotestosterone in the hippocampus. We report that, in neurons, 24OH contributes to these divergent effects by activating sex hormone signaling, including estrogen receptors. CYP46A1 overexpression in female mice protects from memory impairments induced by ovariectomy while having no effects in gonadectomized males. Last, we measured cerebrospinal fluid levels of 24OH in a clinical cohort of patients with AD and found that 24OH negatively correlates with neurodegeneration markers only in women. We suggest that CYP46A1 activation is a valuable pharmacological target for enhancing estrogen signaling in women at risk of developing neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Memory Disorders , Male , Female , Humans , Animals , Mice , Aged , Cholesterol 24-Hydroxylase , Memory Disorders/etiology , Cholesterol , Cognition , Alzheimer Disease/genetics , Estrogens
12.
Endocrinology ; 164(6)2023 04 17.
Article in English | MEDLINE | ID: mdl-37075218

ABSTRACT

Granulosa cell tumors (GCTs) are rare ovarian tumors comprising an adult and a juvenile subtype. They have a generally good prognosis, but the survival rate drastically declines in patients with late-stage or recurring tumors. Due to the rarity of GCTs, the tumor type is largely understudied and lacks a specific treatment strategy. Estrogen receptor beta (ERß/ESR2) has been found to be highly expressed in GCTs, which could be of therapeutic importance since it can be targeted with small molecules. However, its role in GCTs is not known. In this review, we summarize the current knowledge about the action of ERß in the ovary and discuss its prospective role in GCTs.


Subject(s)
Granulosa Cell Tumor , Ovarian Neoplasms , Female , Humans , Estrogen Receptor beta/genetics , Granulosa Cell Tumor/metabolism , Neoplasm Recurrence, Local , Ovarian Neoplasms/metabolism
13.
Cell Death Dis ; 14(2): 129, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792589

ABSTRACT

Lipid and cholesterol metabolism play a crucial role in tumor cell behavior and in shaping the tumor microenvironment. In particular, enzymatic and non-enzymatic cholesterol metabolism, and derived metabolites control dendritic cell (DC) functions, ultimately impacting tumor antigen presentation within and outside the tumor mass, dampening tumor immunity and immunotherapeutic attempts. The mechanisms accounting for such events remain largely to be defined. Here we perturbed (oxy)sterol metabolism genetically and pharmacologically and analyzed the tumor lipidome landscape in relation to the tumor-infiltrating immune cells. We report that perturbing the lipidome of tumor microenvironment by the expression of sulfotransferase 2B1b crucial in cholesterol and oxysterol sulfate synthesis, favored intratumoral representation of monocyte-derived antigen-presenting cells, including monocyte-DCs. We also found that treating mice with a newly developed antagonist of the oxysterol receptors Liver X Receptors (LXRs), promoted intratumoral monocyte-DC differentiation, delayed tumor growth and synergized with anti-PD-1 immunotherapy and adoptive T cell therapy. Of note, looking at LXR/cholesterol gene signature in melanoma patients treated with anti-PD-1-based immunotherapy predicted diverse clinical outcomes. Indeed, patients whose tumors were poorly infiltrated by monocytes/macrophages expressing LXR target genes showed improved survival over the course of therapy. Thus, our data support a role for (oxy)sterol metabolism in shaping monocyte-to-DC differentiation, and in tumor antigen presentation critical for responsiveness to immunotherapy. The identification of a new LXR antagonist opens new treatment avenues for cancer patients.


Subject(s)
Melanoma , Monocytes , Mice , Animals , Monocytes/metabolism , Cell Differentiation , Cholesterol/metabolism , Antigen Presentation , Dendritic Cells/metabolism , Tumor Microenvironment
14.
Mol Cell Proteomics ; 9(7): 1411-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20348541

ABSTRACT

The estrogen receptors (ERs) are ligand-dependent transcription factors that activate transcription by binding to estrogen response elements. Estrogen-mediated effects are tissue- and cell type-specific, determined by the cofactor recruitment to the ERs among other factors. To understand these differences in estrogen action, it is important to identify the various compositions of the ER complexes (ER receptosomes). In this report, we describe a fast and efficient method for the isolation of the ERalpha receptosome for proteomics analysis. Using immobilized estrogen response element on a Sepharose column in combination with two-dimensional electrophoresis and MALDI-TOF MS, significant amounts of proteins could be isolated and identified. Differences in ERalpha complex composition with the ER ligands 17beta-estradiol, 4-hydroxytamoxifen, and ICI-182,780 could also be observed. Thus, this approach provides an easy and relevant way of identifying ERalpha cofactor and transcription factor recruitment under different conditions.


Subject(s)
Endosomes/metabolism , Estrogen Receptor alpha/metabolism , Macromolecular Substances/metabolism , Proteome/analysis , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Cell Line , Electrophoresis, Gel, Two-Dimensional , Endosomes/chemistry , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Humans , Macromolecular Substances/chemistry , Molecular Sequence Data , Response Elements
15.
Antioxidants (Basel) ; 11(2)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35204179

ABSTRACT

Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.

16.
Neural Regen Res ; 17(9): 1861-1866, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35142659

ABSTRACT

Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.

17.
Essays Biochem ; 65(6): 913-925, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34623401

ABSTRACT

The female sex hormone estrogen has been ascribed potent neuroprotective properties. It signals by binding and activating estrogen receptors that, depending on receptor subtype and upstream or downstream effectors, can mediate gene transcription and rapid non-genomic actions. In this way, estrogen receptors in the brain participate in modulating neural differentiation, proliferation, neuroinflammation, cholesterol metabolism, synaptic plasticity, and behavior. Circulating sex hormones decrease in the course of aging, more rapidly at menopause in women, and slower in men. This review will discuss what this drop entails in terms of modulating neuroprotection and resilience in the aging brain downstream of spatiotemporal estrogen receptor alpha (ERα) and beta (ERß) signaling, as well as in terms of the sex differences observed in Alzheimer's disease (AD) and Parkinson's disease (PD). In addition, controversies related to ER expression in the brain will be discussed. Understanding the spatiotemporal signaling of sex hormones in the brain can lead to more personalized prevention strategies or therapies combating neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Receptors, Estrogen , Aging , Alzheimer Disease/metabolism , Brain/metabolism , Female , Humans , Male , Receptors, Estrogen/metabolism
18.
Vitam Horm ; 116: 313-326, 2021.
Article in English | MEDLINE | ID: mdl-33752823

ABSTRACT

The female sex hormone estradiol (E2, 17ß-estradiol) has important functions in the developing brain. In addition to regulating sexual differentiation of the brain, E2 participates in the development of brain areas involved in functions unrelated to reproduction, such as cognition. E2 signals mainly thorough two estrogen receptors; estrogen receptor alpha (ERα) and beta (ERß). While ERα has distinct functions for sexual imprinting of the developing brain, ERß is considered to participate in the development of brain areas related to cognitive function. In this chapter we will focus on ERß's role during neural development. We will discuss the contributions of sex chromosomal and sex hormonal effects in this process and place it in relation to recent data on ERß obtained from stem cell models. Finally, we will discuss the lessons learned from mouse and stem cell models in understanding ERß's role in neural development and how new stem cell models, by addressing the human relevance, may help to advance our progress in this field.


Subject(s)
Estrogen Receptor alpha , Estrogen Receptor beta , Animals , Estradiol , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Mice , Receptors, Estrogen
19.
Stem Cells Transl Med ; 10(7): 987-995, 2021 07.
Article in English | MEDLINE | ID: mdl-33634954

ABSTRACT

Hematopoietic stem cell- (HSC) and induced pluripotent stem (iPS) cell-derived natural killer (NK) cells containing engineered functions, such as chimeric antigen receptors (CAR), offer great promise for the treatment of seemingly incurable oncological malignancies. Today, some of the main challenges of CAR cell-based therapeutics are the long manufacturing time and safety of the cell sources used. Additional challenges include avoiding graft vs host disease (GVHD) and cytokine release syndrome (CRS). Here, we show compelling evidence for the use of NK cell therapeutics as a reliable off-the-shelf option, as they address key issues. Furthermore, we highlight how iPS cells and directed differentiation toward HSC and NK cells address industrial scalability and safety.


Subject(s)
Hematopoietic Stem Cells , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells , Killer Cells, Natural , Receptors, Chimeric Antigen , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/cytology
20.
Heliyon ; 7(3): e06568, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33855238

ABSTRACT

[This corrects the article DOI: 10.1016/j.heliyon.2020.e05535.].

SELECTION OF CITATIONS
SEARCH DETAIL