Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.570
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 86: 659-684, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28301744

ABSTRACT

The endoplasmic reticulum (ER) has a broad localization throughout the cell and forms direct physical contacts with all other classes of membranous organelles, including the plasma membrane (PM). A number of protein tethers that mediate these contacts have been identified, and study of these protein tethers has revealed a multiplicity of roles in cell physiology, including regulation of intracellular Ca2+ dynamics and signaling as well as control of lipid traffic and homeostasis. In this review, we discuss the cross talk between the ER and the PM mediated by direct contacts. We review factors that tether the two membranes, their properties, and their dynamics in response to the functional state of the cell. We focus in particular on the role of ER-PM contacts in nonvesicular lipid transport between the two bilayers mediated by lipid transfer proteins.


Subject(s)
Calcium/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , ORAI1 Protein/metabolism , Biological Transport , Calcium Signaling , Carrier Proteins/genetics , Cell Membrane/ultrastructure , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Gene Expression , Homeostasis , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Synaptotagmins/genetics , Synaptotagmins/metabolism
2.
Nat Immunol ; 21(10): 1267-1279, 2020 10.
Article in English | MEDLINE | ID: mdl-32868928

ABSTRACT

Antigen stimulation (signal 1) triggers B cell proliferation and primes B cells to recruit, engage and respond to T cell help (signal 2). Failure to receive signal 2 within a defined time window results in B cell apoptosis, yet the mechanisms that enforce dependence on co-stimulation are incompletely understood. Nr4a1-3 encode a small family of orphan nuclear receptors that are rapidly induced by B cell antigen receptor stimulation. Here, we show that Nr4a1 and Nr4a3 play partially redundant roles to restrain B cell responses to antigen in the absence of co-stimulation and do so, in part, by repressing the expression of BATF and, consequently, MYC. The NR4A family also restrains B cell access to T cell help by repressing expression of the T cell chemokines CCL3 and CCL4, as well as CD86 and ICAM1. Such NR4A-mediated regulation plays a role specifically under conditions of competition for limiting T cell help.


Subject(s)
B-Lymphocytes/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Communication , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Immunity, Humoral , Immunomodulation , Lymphocyte Activation , Mice , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction
3.
Cell ; 159(1): 58-68, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25259920

ABSTRACT

Complex gene regulatory networks require transcription factors (TFs) to bind distinct DNA sequences. To understand how novel TF specificity evolves, we combined phylogenetic, biochemical, and biophysical approaches to interrogate how DNA recognition diversified in the steroid hormone receptor (SR) family. After duplication of the ancestral SR, three mutations in one copy radically weakened binding to the ancestral estrogen response element (ERE) and improved binding to a new set of DNA sequences (steroid response elements, SREs). They did so by establishing unfavorable interactions with ERE and abolishing unfavorable interactions with SRE; also required were numerous permissive substitutions, which nonspecifically improved cooperativity and affinity of DNA binding. Our findings indicate that negative determinants of binding play key roles in TFs' DNA selectivity and-with our prior work on the evolution of SR ligand specificity during the same interval-show how a specific new gene regulatory module evolved without interfering with the integrity of the ancestral module.


Subject(s)
Evolution, Molecular , Gene Regulatory Networks , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Response Elements , Animals , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Receptors, Steroid/metabolism
4.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38775023

ABSTRACT

Regenerative ability often declines as animals mature past embryonic and juvenile stages, suggesting that regeneration requires redirection of growth pathways that promote developmental growth. Intriguingly, the Drosophila larval epithelia require the hormone ecdysone (Ec) for growth but require a drop in circulating Ec levels to regenerate. Examining Ec dynamics more closely, we find that transcriptional activity of the Ec-receptor (EcR) drops in uninjured regions of wing discs, but simultaneously rises in cells around the injury-induced blastema. In parallel, blastema depletion of genes encoding Ec biosynthesis enzymes blocks EcR activity and impairs regeneration but has no effect on uninjured wings. We find that local Ec/EcR signaling is required for injury-induced pupariation delay following injury and that key regeneration regulators upd3 and Ets21c respond to Ec levels. Collectively, these data indicate that injury induces a local source of Ec within the wing blastema that sustains a transcriptional signature necessary for developmental delay and tissue repair.


Subject(s)
Drosophila Proteins , Ecdysone , Regeneration , Wings, Animal , Animals , Ecdysone/metabolism , Wings, Animal/metabolism , Wings, Animal/growth & development , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Epithelium/metabolism , Gene Expression Regulation, Developmental , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Larva/metabolism , Larva/growth & development , Signal Transduction , Drosophila , Receptors, Steroid/metabolism , Receptors, Steroid/genetics
5.
Mol Cell ; 73(3): 458-473.e7, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30581148

ABSTRACT

Cholesterol is highly enriched at the plasma membrane (PM), and lipid transfer proteins may deliver cholesterol to the PM in a nonvesicular manner. Here, through a mini-screen, we identified the oxysterol binding protein (OSBP)-related protein 2 (ORP2) as a novel mediator of selective cholesterol delivery to the PM. Interestingly, ORP2-mediated enrichment of PM cholesterol was coupled with the removal of phosphatidylinositol 4, 5-bisphosphate (PI(4,5)P2) from the PM. ORP2 overexpression or deficiency impacted the levels of PM cholesterol and PI(4,5)P2, and ORP2 efficiently transferred both cholesterol and PI(4,5)P2in vitro. We determined the structure of ORP2 in complex with PI(4,5)P2 at 2.7 Å resolution. ORP2 formed a stable tetramer in the presence of PI(4,5)P2, and tetramerization was required for ORP2 to transfer PI(4,5)P2. Our results identify a novel pathway for cholesterol delivery to the PM and establish ORP2 as a key regulator of both cholesterol and PI(4,5)P2 of the PM.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Hepatocytes/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptors, Steroid/metabolism , Biological Transport , Cell Line, Tumor , HEK293 Cells , Humans , Models, Molecular , Protein Multimerization , Protein Structure, Quaternary , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Structure-Activity Relationship
6.
Nature ; 588(7838): 503-508, 2020 12.
Article in English | MEDLINE | ID: mdl-33299178

ABSTRACT

Most proteins assemble into multisubunit complexes1. The persistence of these complexes across evolutionary time is usually explained as the result of natural selection for functional properties that depend on multimerization, such as intersubunit allostery or the capacity to do mechanical work2. In many complexes, however, multimerization does not enable any known function3. An alternative explanation is that multimers could become entrenched if substitutions accumulate that are neutral in multimers but deleterious in monomers; purifying selection would then prevent reversion to the unassembled form, even if assembly per se does not enhance biological function3-7. Here we show that a hydrophobic mutational ratchet systematically entrenches molecular complexes. By applying ancestral protein reconstruction and biochemical assays to the evolution of steroid hormone receptors, we show that an ancient hydrophobic interface, conserved for hundreds of millions of years, is entrenched because exposure of this interface to solvent reduces protein stability and causes aggregation, even though the interface makes no detectable contribution to function. Using structural bioinformatics, we show that a universal mutational propensity drives sites that are buried in multimeric interfaces to accumulate hydrophobic substitutions to levels that are not tolerated in monomers. In a database of hundreds of families of multimers, most show signatures of long-term hydrophobic entrenchment. It is therefore likely that many protein complexes persist because a simple ratchet-like mechanism entrenches them across evolutionary time, even when they are functionally gratuitous.


Subject(s)
Evolution, Molecular , Hydrophobic and Hydrophilic Interactions , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Multimerization , Binding Sites/genetics , DNA/metabolism , Humans , Ligands , Models, Molecular , Multiprotein Complexes/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Aggregates , Protein Domains , Protein Multimerization/genetics , Protein Stability , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Solvents/chemistry
7.
Dev Biol ; 508: 8-23, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38199580

ABSTRACT

Steroid hormones play various physiological roles including metabolism and reproduction. Steroid hormones in insects are ecdysteroids, and the major form in Drosophila melanogaster is ecdysone. In Drosophila males, the accessory gland is responsive to nutrient-dependent regulation of fertility/fecundity. The accessory gland is composed of two types of binucleated epithelial cells: a main cell and a secondary cell (SC). The transcription factors Defective proventriculus (Dve), Abdominal-B, and Ecdysone receptors (EcRs) are strongly expressed in adult SCs. We show that this EcR expression is regulated by parallel pathways of nutrient signaling and the Dve activity. Induction of Dve expression is also dependent on nutrient signaling, and it becomes nutrient signal-independent during a restricted period of development. Forced dve expression during the restricted period significantly increased the number of SCs. Here, we provide evidence that the level of nutrient signal-dependent Dve expression during the restricted period determines the number of SCs, and that ecdysone signaling is also crucial to optimize male fecundity through nutrient signal-dependent survival and maturation of SCs.


Subject(s)
Drosophila Proteins , Receptors, Steroid , Animals , Male , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Ecdysone/metabolism , Fertility , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Steroids/metabolism
8.
Nat Immunol ; 14(3): 230-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334790

ABSTRACT

Regulatory T cells (T(reg) cells) develop from progenitor thymocytes after the engagement of T cell antigen receptors (TCRs) with high-affinity ligands, but the underlying molecular mechanisms are still unclear. Here we show that the Nr4a nuclear receptors, which are encoded by immediate-early genes upregulated by TCR stimulation in thymocytes, have essential roles in T(reg) cell development. Mice that lacked all Nr4a factors could not produce T(reg) cells and died early owing to systemic autoimmunity. Nr4a receptors directly activated the promoter of the gene encoding the transcription factor Foxp3, and forced activation of Nr4a receptors bypassed low-strength TCR signaling to drive the T(reg) cell developmental program. Our results suggest that Nr4a receptors have key roles in determining CD4(+) T cell fates in the thymus and thus contribute to immune homeostasis.


Subject(s)
DNA-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Receptors, Antigen, T-Cell/metabolism , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/genetics , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genes, Immediate-Early , Homeostasis , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , Thymocytes/metabolism
9.
Mol Cell ; 67(6): 947-961.e5, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28890336

ABSTRACT

The Hsp90 system in the eukaryotic cytosol is characterized by a cohort of co-chaperones that bind to Hsp90 and affect its function. Although progress has been made regarding the underlying biochemical mechanisms, how co-chaperones influence Hsp90 client proteins in vivo has remained elusive. By investigating the effect of 12 Hsp90 co-chaperones on the activity of different client proteins in yeast, we find that deletion of co-chaperones can have a neutral or negative effect on client activity but can also lead to more active clients. Only a few co-chaperones are active on all clients studied. Closely related clients and even point mutants can depend on different co-chaperones. These effects are direct because differences in client-co-chaperone interactions can be reconstituted in vitro. Interestingly, some co-chaperones affect client conformation in vivo. Thus, co-chaperones adapt the Hsp90 cycle to the requirements of the client proteins, ensuring optimal activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Plasticity , HSP90 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/genetics , Genotype , HSP90 Heat-Shock Proteins/genetics , Mutation , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Phenotype , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 119(40): e2208935119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161884

ABSTRACT

Steroid hormones perform diverse biological functions in developing and adult animals. However, the mechanistic basis for their tissue specificity remains unclear. In Drosophila, the ecdysone steroid hormone is essential for coordinating developmental timing across physically separated tissues. Ecdysone directly impacts genome function through its nuclear receptor, a heterodimer of the EcR and ultraspiracle proteins. Ligand binding to EcR triggers a transcriptional cascade, including activation of a set of primary response transcription factors. The hierarchical organization of this pathway has left the direct role of EcR in mediating ecdysone responses obscured. Here, we investigate the role of EcR in controlling tissue-specific ecdysone responses, focusing on two tissues that diverge in their response to rising ecdysone titers: the larval salivary gland, which undergoes programmed destruction, and the wing imaginal disc, which initiates morphogenesis. We find that EcR functions bimodally, with both gene repressive and activating functions, even at the same developmental stage. EcR DNA binding profiles are highly tissue-specific, and transgenic reporter analyses demonstrate that EcR plays a direct role in controlling enhancer activity. Finally, despite a strong correlation between tissue-specific EcR binding and tissue-specific open chromatin, we find that EcR does not control chromatin accessibility at genomic targets. We conclude that EcR contributes extensively to tissue-specific ecdysone responses. However, control over access to its binding sites is subordinated to other transcription factors.


Subject(s)
Chromatin , Drosophila Proteins , Drosophila melanogaster , Ecdysone , Enhancer Elements, Genetic , Gene Expression Regulation , Receptors, Steroid , Animals , Chromatin/metabolism , DNA/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Ecdysone/metabolism , Ligands , Receptors, Steroid/genetics , Receptors, Steroid/metabolism
11.
Mod Pathol ; 37(5): 100474, 2024 May.
Article in English | MEDLINE | ID: mdl-38508521

ABSTRACT

Recurrent gene fusions have been observed in epithelioid and myxoid variants of uterine leiomyosarcoma. PGR::NR4A3 fusions were recently described in a subset of epithelioid leiomyosarcomas exhibiting rhabdoid morphology. In this study, we sought to expand the clinical, morphologic, immunohistochemical, and genetic features of gynecologic leiomyosarcomas harboring NR4A3 rearrangements with PGR and novel fusion partners. We identified 9 gynecologic leiomyosarcomas harboring PGR::NR4A3, CARMN::NR4A3, ACTB::NR4A3, and possible SLCO5A1::NR4A3 fusions by targeted RNA sequencing. Tumors frequently affected premenopausal women, involving the uterine corpus, uterine cervix, or pelvis. All were similarly characterized by lobules of monomorphic epithelioid and/or spindled cells arranged in sheets, cords, trabeculae, and micro- and macrocysts associated with abundant myxoid matrix and hemorrhage, creating labyrinth-like or pulmonary edema-like architecture. Myogenic differentiation with frequent estrogen receptor and progesterone receptor staining and no CD10 expression characterized all tumors. All cases showed high NR4A3 RNA expression levels and NOR1 (NR4A3) nuclear staining similar to salivary gland acinic cell carcinomas and a subset of extraskeletal myxoid chondrosarcomas harboring NR4A3 rearrangements. NOR1 (NR4A3) immunohistochemistry may serve as a useful diagnostic marker of NR4A3 fusion-positive gynecologic leiomyosarcomas.


Subject(s)
Leiomyosarcoma , Receptors, Thyroid Hormone , Humans , Female , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Middle Aged , Adult , Receptors, Thyroid Hormone/genetics , Receptors, Steroid/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , DNA-Binding Proteins/genetics , Aged , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Oncogene Proteins, Fusion/genetics , Gene Fusion
12.
PLoS Pathog ; 18(9): e1010837, 2022 09.
Article in English | MEDLINE | ID: mdl-36137163

ABSTRACT

The balance between immunity and reproduction is essential for many key physiological functions. We report that to maintain an optimal fertility, 20-hydroxyecdysone (20E) and the ecdysone receptor (EcR) downregulate the immune deficiency (IMD) pathway during the post blood meal phase (PBM) of the Aedes aegypti reproductive cycle. RNA interference-mediated depletion of EcR elicited an increased expression of the IMD pathway components, and these mosquitoes were more resistant to infection by Gram-negative bacteria. Moreover, 20E and EcR recruit Pirk-like, the mosquito ortholog of Drosophila melanogaster Pirk. CRISPR-Cas9 knockout of Pirk-like has shown that it represses the IMD pathway by interfering with IMD-mediated formation of amyloid aggregates. 20E and EcR disruption of the amyloid formation is pivotal for maintaining normal yolk protein production and fertility. Additionally, 20E and its receptor EcR directly induce Pirk-like to interfere with cRHIM-mediated formation of amyloid. Our study highlights the vital role of 20E in governing the trade-off between immunity and reproduction. Pirk-like might be a potential target for new methods to control mosquito reproduction and pathogen transmission.


Subject(s)
Aedes , Receptors, Steroid , Aedes/metabolism , Animals , Drosophila melanogaster/metabolism , Ecdysone , Ecdysterone/genetics , Egg Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Receptors, Steroid/genetics , Reproduction
13.
Blood ; 139(7): 1052-1065, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34797912

ABSTRACT

Human T-cell leukemia virus 1 (HTLV-1) causes adult T-cell leukemia (ATL), but the mechanism underlying its initiation remains elusive. In this study, ORP4L was expressed in ATL cells but not in normal T-cells. ORP4L ablation completely blocked T-cell leukemogenesis induced by the HTLV-1 oncoprotein Tax in mice, whereas engineering ORP4L expression in T-cells resulted in T-cell leukemia in mice, suggesting the oncogenic properties and prerequisite of ORP4L promote the initiation of T-cell leukemogenesis. For molecular insight, we found that loss of miR-31 caused by HTLV-1 induced ORP4L expression in T-cells. ORP4L interacts with PI3Kδ to promote PI(3,4,5)P3 generation, contributing to AKT hyperactivation; NF-κB-dependent, p53 inactivation-induced pro-oncogene expression; and T-cell leukemogenesis. Consistently, ORP4L ablation eliminates human ATL cells in patient-derived xenograft ATL models. These results reveal a plausible mechanism of T-cell deterioration by HTLV-1 that can be therapeutically targeted.


Subject(s)
Carcinogenesis/pathology , Gene Expression Regulation, Leukemic , HTLV-I Infections/complications , Human T-lymphotropic virus 1/isolation & purification , Leukemia-Lymphoma, Adult T-Cell/pathology , Receptors, Steroid/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Proliferation , Gene Products, tax , HTLV-I Infections/virology , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/metabolism , Leukemia-Lymphoma, Adult T-Cell/virology , Mice , Prognosis , Receptors, Steroid/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Mol Biol Rep ; 51(1): 657, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740636

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis (MTB) is the causative organism of tuberculosis. Cholesterol is a crucial carbon source required for the survival of MTB in host cells. Transcription factor NR1H3 along with its important target genes ABCA1 and ApoE play important role in removal of extra cholesterol from cells. Changes in the gene expression of NR1H3, ABCA1 and ApoE can affect cholesterol homeostasis and thus the survival of MTB in host cells.Therefore, the present study was designed to analyze the mRNA expression of NR1H3, ABCA1 and ApoE in pulmonary TB (PTB) patients from the population of Punjab, India. METHODS AND RESULTS: In this study, mRNA expression of the transcription factor NR1H3 and its target genes ABCA1 and ApoE was analyzed in 89 subjects, including 41 PTB patients and 48 healthy controls (HCs) by real-time quantitative PCR. It was found that the mRNA expression of both NR1H3 and ABCA1 genes was significantly lower in TB patients than in HCs (p < 0.001). Even after sex-wise stratification of the subjects, mRNA expression of NR1H3 and ABCA1 was found to be down-regulated in both male and female TB patients. No significant difference was observed in expression of ApoE (p = 0.98). CONCLUSIONS: The present study found that the mRNA expression of NR1H3 and ABCA1 is down-regulated in TB patients from Punjab state of India.


Subject(s)
ATP Binding Cassette Transporter 1 , RNA, Messenger , Tuberculosis, Pulmonary , Humans , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Female , Male , India , Adult , RNA, Messenger/genetics , RNA, Messenger/metabolism , Middle Aged , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Mycobacterium tuberculosis/genetics , Case-Control Studies , Receptors, Steroid/genetics , Receptors, Steroid/metabolism
15.
Arch Insect Biochem Physiol ; 115(4): e22110, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38605666

ABSTRACT

20-Hydroxyecdysone (20E) plays a vital role in a series of biological processes, via the nuclear receptors, EcR/USP by activating the ecdysone regulatory cascade. To clarify the role of EcR during the development of Grapholita molesta, the complementary DNA of ecdysone receptor isoform B1 (GmEcR-B1) was obtained from the transcriptome of G. molesta and verified by PCR. Alignment analysis revealed that the deduced protein sequence of GmEcR-B1 was highly homologous to EcR proteins identified in other lepidopteran species, especially the EcR-B1 isoform in Spodoptera litura. Quantitative real-time PCR showed that GmEcRs was expressed at all test developmental stages, and the expression level of GmEcRs was relatively higher during the period of the 3rd day of fifth instar larvae to 2nd of pupa than those in other stages. Moreover, the messenger RNA of GmEcRs was much more strongly expressed in the Malpighian tubule and epidermis than those in other tissues, which suggests that this gene may function in a tissue-specific manner during larval development. Silencing of GmEcRs could significantly downregulate the transcriptional level of ecdysone-inducible genes and result in increased mortality during metamorphosis and prolonged prepupal duration. Taken together, the present results indicate that GmEcRs may directly or indirectly affect the development of G. molesta.


Subject(s)
Moths , Receptors, Steroid , Animals , Moths/metabolism , Ecdysone , Fruit/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Protein Isoforms/genetics
16.
Mol Cell ; 62(5): 665-7, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259199

ABSTRACT

Pioneer factors such as FoxA target nucleosomal DNA and initiate cooperative interactions at silent genes during development, cellular reprogramming, and steroid hormone induction. Biophysical studies previously showed that the nuclear mobility of FoxA1 is slower than for many other transcription factors, whereas a new single molecule study (Swinstead et al., 2016, Cell) shows comparable chromatin residence times for FoxA1 and steroid receptors. Despite that steroid receptors engage nucleosome-remodeling complexes, the vast majority of co-bound sites with FoxA are dependent upon FoxA, not vice versa. Taken together, the distinguishing feature of pioneer factors remains nucleosomal access rather than an exceptional residence time in chromatin.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Nucleosomes/metabolism , Receptors, Steroid/metabolism , Animals , Binding Sites , Cell Nucleus/ultrastructure , Cellular Reprogramming , Chromatin Assembly and Disassembly , DNA/genetics , Gene Expression Regulation, Developmental , Gonadal Steroid Hormones/biosynthesis , Gonadal Steroid Hormones/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Nucleosomes/genetics , Protein Binding , Receptors, Steroid/genetics
17.
Gen Comp Endocrinol ; 347: 114436, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38141859

ABSTRACT

Teleost fishes have emerged as tractable models for studying the neuroendocrine regulation of social behavior via molecular genetic techniques, such as CRISPR/Cas9 gene editing. Moreover, teleosts provide an opportunity to investigate the evolution of steroid receptors and their functions, as species within this lineage possess novel steroid receptor paralogs that resulted from a teleost-specific whole genome duplication. Although teleost fishes have grown in popularity as models for behavioral neuroendocrinology, there is not a consistent nomenclature system for steroid receptors and their genes, which may impede a clear understanding of steroid receptor paralogs and their functions. Here, we used a phylogenetic approach to assess the relatedness of protein sequences encoding steroid receptor paralogs in 18 species from 12 different orders of the Infraclass Teleostei. While most similarly named sequences grouped based on the established phylogeny of the teleost lineage, our analysis revealed several inconsistencies in the nomenclature of steroid receptor paralogs, particularly for sequences encoding estrogen receptor beta (ERß). Based on our results, we propose a nomenclature system for teleosts in which Greek symbols refer to proteins and numbers refer to genes encoding different subtypes of steroid receptors within the five major groups of this nuclear receptor subfamily. Collectively, our results bridge a critical gap by providing a cohesive naming system for steroid receptors in teleost fishes, which will serve to improve communication, promote collaboration, and enhance our understanding of the evolution and function of steroid receptors across vertebrates.


Subject(s)
Evolution, Molecular , Receptors, Steroid , Animals , Phylogeny , Fishes/genetics , Vertebrates , Receptors, Steroid/genetics , Gene Duplication
18.
Gen Comp Endocrinol ; 355: 114548, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38761872

ABSTRACT

Ecdysteroid molting hormones coordinate arthropod growth and development. Binding of 20-hydroxyecdysone (20E) to ecdysteroid receptor EcR/RXR activates a cascade of nuclear receptor transcription factors that mediate tissue responses to hormone. Insect ecdysteroid responsive and Forkhead box class O (FOXO) transcription factor gene sequences were used to extract orthologs from blackback land crab (Gecarcinus lateralis) Y-organ (YO) transcriptome: Gl-Ecdysone Receptor (EcR), Gl-Broad Complex (Br-C), Gl-E74, Gl-Hormone Receptor 3 (HR3), Gl-Hormone Receptor 4 (HR4), Gl-FOXO, and Gl-Fushi tarazu factor-1 (Ftz-f1). Quantitative polymerase chain reaction quantified mRNA levels in tissues from intermolt animals and in YO of animals induced to molt by multiple limb autotomy (MLA) or eyestalk ablation (ESA). Gl-EcR, Gl-Retinoid X Receptor (RXR), Gl-Br-C, Gl-HR3, Gl-HR4, Gl-E74, Gl-E75, Gl-Ftz-f1, and Gl-FOXO were expressed in all 10 tissues, with Gl-Br-C, Gl-E74, Gl-E75, and Gl-HR4 mRNA levels in the YO lower than those in most of the other tissues. In MLA animals, molting had no effect on Gl-Br-C, Gl-E74, and Gl-Ftz-f1 mRNA levels and little effect on Gl-EcR, Gl-E75, and Gl-HR4 mRNA levels. Gl-HR3 and Gl-FOXO mRNA levels were increased during premolt stages, while Gl-RXR mRNA level was highest during intermolt and premolt stages and lowest at postmolt stage. In ESA animals, YO mRNA levels were not correlated with hemolymph ecdysteroid titers. ESA had no effect on Gl-EcR, Gl-E74, Gl-HR3, Gl-HR4, Gl-Ftz-f1, and Gl-FOXO mRNA levels, while Gl-RXR, Gl-Br-C, and Gl-E75 mRNA levels were decreased at 3 days post-ESA. These data suggest that transcriptional up-regulation of Gl-FOXO and Gl-HR3 contributes to increased YO ecdysteroidogenesis during premolt. By contrast, transcriptional regulation of ecdysteroid responsive genes and ecdysteroidogenesis were uncoupled in the YO of ESA animals.


Subject(s)
Ecdysteroids , Molting , Animals , Molting/genetics , Ecdysteroids/metabolism , Ecdysteroids/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Ecdysterone/metabolism , Brachyura/genetics , Brachyura/metabolism , Brachyura/growth & development , Endocrine Glands/metabolism
19.
Acta Biochim Biophys Sin (Shanghai) ; 56(1): 44-53, 2024 01 25.
Article in English | MEDLINE | ID: mdl-37905340

ABSTRACT

The incidence and related death of hepatocellular carcinoma (HCC) have increased over the past decades. However, the molecular mechanisms underlying HCC pathogenesis are not fully understood. Long noncoding RNA (lncRNA) RP11-495P10.1 has been proven to be closely associated with the progression of prostate cancer, but its role and specific mechanism in HCC are still unknown. Here, we identify that RP11-495P10.1 is highly expressed in HCC tissues and cells and contributes to the proliferation of HCC cells. Moreover, this study demonstrates that RP11-495P10.1 affects the proliferation of HCC by negatively regulating the expression of nuclear receptor subfamily 4 group a member 3 (NR4A3). Glycometabolism reprogramming is one of the main characteristics of tumor cells. In this study, we discover that RP11-495P10.1 regulates glycometabolism reprogramming by changing the expression of pyruvate dehydrogenase kinase 1 (PDK1) and pyruvate dehydrogenase (PDH), thus contributing to the proliferation of HCC cells. Furthermore, knockdown of RP11-495P10.1 increases enrichment of H3K27Ac in the promoter of NR4A3 by promoting the activity of PDH and the production of acetyl-CoA, which leads to the increased transcription of NR4A3. Altogether, RP11-495P10.1 promotes HCC cell proliferation by regulating the reprogramming of glucose metabolism and acetylation of the NR4A3 promoter via the PDK1/PDH axis, which provides an lncRNA-oriented therapeutic strategy for the diagnosis and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA, Long Noncoding , Receptors, Steroid , Humans , Male , Acetylation , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glucose , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism
20.
PLoS Genet ; 17(2): e1009352, 2021 02.
Article in English | MEDLINE | ID: mdl-33529191

ABSTRACT

Diapause, a programmed developmental arrest primarily induced by seasonal environmental changes, is very common in the animal kingdom, and found in vertebrates and invertebrates alike. Diapause provides an adaptive advantage to animals, as it increases the odds of surviving adverse conditions. In insects, individuals perceive photoperiodic cues and modify endocrine signaling to direct reproductive diapause traits, such as ovary arrest and increased fat accumulation. However, it remains unclear as to which endocrine factors are involved in this process and how they regulate the onset of reproductive diapause. Here, we found that the long day-mediated drop in the concentration of the steroid hormone ecdysone is essential for the preparation of photoperiodic reproductive diapause in Colaphellus bowringi, an economically important cabbage beetle. The diapause-inducing long-day condition reduced the expression of ecdysone biosynthetic genes, explaining the drop in the titer of 20-hydroxyecdysone (20E, the active form of ecdysone) in female adults. Application of exogenous 20E induced vitellogenesis and ovarian development but reduced fat accumulation in the diapause-destined females. Knocking down the ecdysone receptor (EcR) in females destined for reproduction blocked reproductive development and induced diapause traits. RNA-seq and hormone measurements indicated that 20E stimulates the production of juvenile hormone (JH), a key endocrine factor in reproductive diapause. To verify this, we depleted three ecdysone biosynthetic enzymes via RNAi, which confirmed that 20E is critical for JH biosynthesis and reproductive diapause. Importantly, impairing Met function, a component of the JH intracellular receptor, partially blocked the 20E-regulated reproductive diapause preparation, indicating that 20E regulates reproductive diapause in both JH-dependent and -independent manners. Finally, we found that 20E deficiency decreased ecdysis-triggering hormone signaling and reduced JH production, thereby inducing diapause. Together, these results suggest that 20E signaling is a pivotal regulator that coordinates reproductive plasticity in response to environmental inputs.


Subject(s)
Coleoptera/genetics , Diapause/genetics , Ecdysone/metabolism , Gene Expression Regulation, Developmental , Photoperiod , Animals , Coleoptera/metabolism , Ecdysterone/metabolism , Female , Juvenile Hormones/deficiency , Juvenile Hormones/genetics , Metamorphosis, Biological/genetics , Ovary/growth & development , Ovary/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Reproduction/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL