Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Blood Adv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38748870

ABSTRACT

The regulation of RBC homeostasis by erythropoietin (EPO) is critical for O2 transport and maintaining the adequate number of RBCs in vertebrates. Therefore, dysregulation in EPO synthesis results in disease conditions like polycythemia in the case of excessive EPO production and anemia, which occurs when EPO is inadequately produced. EPO plays a crucial role in treating anemic patients; however, its overproduction can increase blood viscosity, potentially leading to fatal heart failure. Consequently, the identification of druggable transcription factors (TFs) and their associated ligands capable of regulating EPO offers a promising therapeutic approach to address EPO-related disorders. This study unveils a novel regulatory mechanism involving two pivotal nuclear receptors (NRs), Rev-erbα and RORα, in the control of EPO gene expression. Rev-erbα acts as a cell-intrinsic negative regulator, playing a vital role in maintaining erythropoiesis at the correct level. It accomplishes this by directly binding to newly identified response elements within the human and mouse EPO gene promoter, thereby repressing EPO production. These findings are further supported by the discovery that a Rev-erbα agonist (SR9011) effectively suppresses hypoxia-induced EPO expression in mice. In contrast, RORα functions as a positive regulator of EPO gene expression, also binding to the same response elements in the promoter to induce EPO production. Finally, the results of this study revealed that the two NRs, Rev-erbα, and RORα, influence EPO synthesis in a negative and positive manner, suggesting that the modulating activity of these two NRs could provide a method to target disorders linked with EPO dysregulation.

2.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38609183

ABSTRACT

Elevated ER stress has been linked to the pathogenesis of several disease conditions including neurodegeneration. In this study, we have holistically determined the differential expression of all the nuclear receptors (NRs) in the presence of classical ER stress inducers. Activation of Nr1h4 and Thrb by their cognate ligands (GW4064 and T3) ameliorates the tunicamycin (TM)-induced expression of ER stress genes. A combination of both ligands is effective in mitigating cell death induced by TM. Further exploration of their protective effects in the Parkinson's disease (PD) model shows that they reduce MPP+-induced dissipation of mitochondrial membrane potential and ROS generation in an in vitro PD model in neuronal cells. Furthermore, the generation of an experimental murine PD model reveals that simultaneous treatment of GW4064 and T3 protects mice from ER stress, dopaminergic cell death, and functional deficits in the MPTP mouse model of PD. Thus, activation of Nr1h4 and Thrb by their respective ligands plays an indispensable role in ER stress amelioration and mounts protective effects in the MPTP mouse model of PD.


Subject(s)
Parkinson Disease , Animals , Mice , Cell Death , Disease Models, Animal , Dopamine , Dopaminergic Neurons , Thyroid Hormone Receptors beta
3.
Cell Mol Life Sci ; 79(6): 308, 2022 May 21.
Article in English | MEDLINE | ID: mdl-35596832

ABSTRACT

Nuclear receptors are a unique family of transcription factors that play cardinal roles in physiology and plethora of human diseases. The adopted orphan nuclear receptor Nr1d1 is a constitutive transcriptional repressor known to modulate several biological processes. In this study, we found that Nr1d1 plays a decisive role in T helper (Th)-cell polarization and transcriptionally impedes the formation of Th2 cells by directly binding to the promoter region of GATA binding protein 3 (GATA3) gene. Nr1d1 interacts with its cellular companion, the nuclear receptor corepressor and histone deacetylase 3 to form a stable repression complex on the GATA3 promoter. The presence of Nr1d1 also imparts protection against associated inflammatory responses in murine model of asthma and its ligand SR9011 eased disease severity by suppressing Th2 responses. Moreover, Chip-seq profiling uncovered Nr1d1 interactions with other gene subsets that impedes Th2-linked pathways and regulates metabolism, immunity and brain functions, therefore, providing empirical evidence regarding the genetic link between asthma and other comorbid conditions. Thus, Nr1d1 emerges as a molecular switch that could be targeted to subdue asthma.


Subject(s)
Asthma , Th2 Cells , Animals , Cell Differentiation/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression , Humans , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Th1 Cells
4.
Autophagy ; 17(9): 2273-2289, 2021 09.
Article in English | MEDLINE | ID: mdl-32917126

ABSTRACT

Macrophage derived foam cells in atherosclerotic plaques are the major factor responsible for the pathogenesis of atherosclerosis (AS). During advanced AS, macrophage-specific macroautophagy/autophagy is dysfunctional. 1, 25-dihydroxy vitamin D3 (VitD3) and its receptor VDR (vitamin D receptor) are reported to inhibit foam cell formation and induce autophagy; however, the role of VitD3-VDR-induced autophagy and foam cell formation in AS has not been explored. Here we find that VitD3 significantly recovered oxidized low-density lipoprotein-impaired autophagy, as well as increased autophagy-mediated lipid breakdown in mouse bone marrow-derived macrophages and human monocyte-derived macrophages, thus inhibiting the conversion of macrophages into foam cells. Importantly, VitD3 functions through its receptor VDR to upregulate autophagy and attenuate the accumulation of lipids in macrophages. Moreover, this study is the first occasion to report the interesting link between VitD3 signaling and PTPN6/SHP-1 (protein tyrosine phosphatase non-receptor type 6) in macrophages. VitD3-induced autophagy was abrogated in the presence of the PTPN6/Ptpn6 shRNA or inhibitor. VDR along with RXRA (retinoid X receptor alpha), and NCOA1 (nuclear receptor coactivator 1), are recruited to a specific response element located on the gene promoter and induce PTPN6 expression. PTPN6 contributes to VitD3-mediated autophagy by regulating autophagy-related genes via activation of MAPK1 (mitogen-activated protein kinase 1) and CEBPB (CCAAT enhancer binding protein beta). Furthermore, expression of PTPN6 is also crucial for VitD3-mediated inhibition of macrophage foam cell formation through autophagy. Thus, VitD3-VDR-PTPN6 axis-regulated autophagy attenuates foam cell formation in macrophages.


Subject(s)
Autophagy , Cholecalciferol , Foam Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Receptors, Calcitriol , Animals , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Calcitriol/metabolism
5.
Cell Cycle ; 18(24): 3589-3602, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31739702

ABSTRACT

Hepatocellular carcinoma appears as an extremely angiogenic solid tumor marked by apoptosis evasion, dysregulated cell cycle and low sensitivity to chemotherapy. TGF-ß, a multifunctional cytokine, plays a pleiotropic role in the tumor microenvironment and has implications in cancer drug resistance. The current study provides novel evidence that TGF-ß signaling contributes to drug resistance in liver cancer cells by inducing the expression of xenobiotic nuclear receptor PXR. We observed that PXR increases the expression of drug efflux transporters; therefore, accounting for exacerbated drug resistance. Additionally, anti-apoptotic nature of PXR contributes to TGF-ß mediated chemoresistance as seen by procaspase-3 and Mcl-1 cellular levels. TGF-ß binding to the TGF-ß receptor triggers a complex downstream signaling cascade through a non-canonical SMAD-independent ERK pathway that leads to increased PXR expression. Activated ERK activates ETS1 transcription factor which is a critical regulator of endogenous PXR expression in hepatic cells. Loss of function of ETS1 abrogates the TGF-ß induced PXR expression. Together these findings indicate that PXR modulates TGF-ß induced resistance to chemotherapy in liver cancer cells. This underscores the need for combinatorial approaches with focus on PXR antagonism to improve drug effectiveness in hepatocellular carcinoma.Abbreviations: HCC: Hepatocellular Carcinoma; FDA: Food and Drug Administration; TGF-ß: Transforming growth factor-ß; PXR: Pregnane X receptor; CAR: Constitutive androstane receptor; P-gp/ABCB1: P-glycoproteins/ATP-binding cassette transporter subfamily B member 1; MRP1/ABCC1 and MRP2/ABCC2: Multidrug-resistance associated proteins; BCRP/ABCG2: Breast cancer resistant protein; DMEs: Drug-metabolizing enzymes; CFDA: 5,6-carboxyfluorescein diacetate; ETS1: Transcription factor E26 transformation specific sequence 1.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Pregnane X Receptor/genetics , Transforming Growth Factor beta/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Inactivation, Metabolic/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Proto-Oncogene Protein c-ets-1/genetics , Pyrazoles/pharmacology , Quinolines/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors
6.
Autophagy ; 15(7): 1280-1295, 2019 07.
Article in English | MEDLINE | ID: mdl-30669929

ABSTRACT

Macroautophagy/autophagy is a complex self-degradative mechanism responsible for clearance of non functional organelles and proteins. A range of factors influences the autophagic process, and disruptions in autophagy-related mechanisms lead to disease states, and further exacerbation of disease. Despite in-depth research into autophagy and its role in pathophysiological processes, the resources available to use it for therapeutic purposes are currently lacking. Herein we report the Autophagy Small Molecule Database (AutophagySMDB; http://www.autophagysmdb.org/ ) of small molecules and their cognate protein targets that modulate autophagy. Presently, AutophagySMDB enlists ~10,000 small molecules which regulate 71 target proteins. All entries are comprised of information such as EC50 (half maximal effective concentration), IC50 (half maximal inhibitory concentration), Kd (dissociation constant) and Ki (inhibition constant), IUPAC name, canonical SMILE, structure, molecular weight, QSAR (quantitative structure activity relationship) properties such as hydrogen donor and acceptor count, aromatic rings and XlogP. AutophagySMDB is an exhaustive, cross-platform, manually curated database, where either the cognate targets for small molecule or small molecules for a target can be searched. This database is provided with different search options including text search, advanced search and structure search. Various computational tools such as tree tool, cataloging tools, and clustering tools have also been implemented for advanced analysis. Data and the tools provided in this database helps to identify common or unique scaffolds for designing novel drugs or to improve the existing ones for autophagy small molecule therapeutics. The approach to multitarget drug discovery by identifying common scaffolds has been illustrated with experimental validation. Abbreviations: AMPK: AMP-activated protein kinase; ATG: autophagy related; AutophagySMDB: autophagy small molecule database; BCL2: BCL2, apoptosis regulator; BECN1: beclin 1; CAPN: calpain; MTOR: mechanistic target of rapamycin kinase; PPARG: peroxisome proliferator activated receptor gamma; SMILES: simplified molecular input line entry system; SQSTM1: sequestosome 1; STAT3: signal transducer and activator of transcription.


Subject(s)
Autophagy-Related Proteins/drug effects , Autophagy/drug effects , Databases, Pharmaceutical , Small Molecule Libraries/chemistry , Autophagy/genetics , Autophagy-Related Proteins/antagonists & inhibitors , Cataloging , Humans , Inhibitory Concentration 50 , Search Engine , Small Molecule Libraries/pharmacology , Software
7.
Sci Rep ; 8(1): 2296, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396519

ABSTRACT

Mycobacterium tuberculosis instigates interactions with host factors to promote its survival within the host inimical conditions. Among such factors, nuclear receptors (NRs) seem to be promising candidates owing to their role in bacterial pathogenesis. However, only few members of NR superfamily have been implicated in M. tuberculosis infection and there is a dearth of comprehensive knowledge about expression or function of the entire superfamily. In this study, we performed detailed expression analysis and identified key NRs getting differentially regulated in murine macrophages and dendritic cells (DC) upon infection with H37Rv. The murine macrophages and DCs infected with H37Rv entailed overlapping changes in the expression of certain NRs which reflect upon the possibility that both cells might utilize similar transcriptional programs upon M. tuberculosis infection. We identified Nr4a3 and Rora, which have not been implicated in M. tuberculosis pathogenesis, undergo similar changes in expression in macrophages and DCs upon H37Rv infection. Interestingly, a similar pattern in their expression was also observed in infected human monocyte derived macrophages and the findings corroborated well with PBMCs obtained from TB patients. This all-inclusive analysis provides the basis for a precise approach in identifying NRs that can be targeted therapeutically in intracellular bacterial infections.


Subject(s)
Dendritic Cells/microbiology , Macrophages/microbiology , Mycobacterium tuberculosis/growth & development , Receptors, Cytoplasmic and Nuclear/analysis , Animals , Cells, Cultured , Gene Expression Profiling , Humans , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/genetics
8.
J Biol Chem ; 293(10): 3747-3757, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29358328

ABSTRACT

Mycobacterium tuberculosis is the causative agent of tuberculosis (TB). It acquires phenotypic drug resistance inside macrophages, and this resistance mainly arises from host-induced stress. However, whether cellular drug-efflux mechanisms in macrophages contribute to nonresponsiveness of M. tuberculosis to anti-TB drugs is unclear. Here, we report that xenobiotic nuclear receptors mediate TB drug nonresponsiveness by modulating drug-efflux transporters in macrophages. This was evident from expression analysis of drug-efflux transporters in macrophages isolated from TB patients. Among patients harboring rifampicin-susceptible M. tuberculosis, we observed increased intracellular survival of M. tuberculosis upon rifampicin treatment of macrophages isolated from patients not responding to anti-TB drugs compared with macrophages from patients who did respond. Of note, M. tuberculosis infection and rifampicin exposure synergistically modulated macrophage drug-efflux transporters in vitro We also found that the xenobiotic nuclear receptor pregnane X receptor (PXR) modulates macrophage drug-efflux transporter expression and activity, which compromised the anti-TB efficacy of rifampicin. We further validated this finding in a TB mouse model in which use of the PXR antagonist ketoconazole rescued rifampicin anti-TB activity. We conclude that PXR activation in macrophages compromises the efficacy of the anti-TB drug rifampicin. Alternative therapeutic strategies, such as use of the rifampicin derivatives rifapentine and rifabutin, which do not activate PXR, or of a PXR antagonist, may be effective for tackling drug nonresponsiveness of M. tuberculosis that arises from drug-efflux systems of the host.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Drug Resistance, Bacterial , Host-Pathogen Interactions/drug effects , Macrophages/metabolism , Mycobacterium tuberculosis/drug effects , Pregnane X Receptor/metabolism , Rifampin/pharmacology , ATP-Binding Cassette Transporters/agonists , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antibiotics, Antitubercular/therapeutic use , Cells, Cultured , Drug Resistance, Bacterial/drug effects , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Genes, Reporter/drug effects , Humans , Ketoconazole/pharmacology , Lung/drug effects , Lung/metabolism , Lung/microbiology , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Male , Mice, Inbred C57BL , Microbial Viability/drug effects , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Pregnane X Receptor/agonists , Pregnane X Receptor/antagonists & inhibitors , Pregnane X Receptor/genetics , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rifampin/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology
9.
J Immunol ; 197(1): 244-55, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27233963

ABSTRACT

Mycobacterium tuberculosis can evade host defense processes, thereby ensuring its survival and pathogenesis. In this study, we investigated the role of nuclear receptor, pregnane X receptor (PXR), in M. tuberculosis infection in human monocyte-derived macrophages. In this study, we demonstrate that PXR augments M. tuberculosis survival inside the host macrophages by promoting the foamy macrophage formation and abrogating phagolysosomal fusion, inflammation, and apoptosis. Additionally, M. tuberculosis cell wall lipids, particularly mycolic acids, crosstalk with human PXR (hPXR) by interacting with its promiscuous ligand binding domain. To confirm our in vitro findings and to avoid the reported species barrier in PXR function, we adopted an in vivo mouse model expressing hPXR, wherein expression of hPXR in mice promotes M. tuberculosis survival. Therefore, pharmacological intervention and designing antagonists to hPXR may prove to be a promising adjunct therapy for tuberculosis.


Subject(s)
Macrophages/immunology , Mycobacterium tuberculosis/immunology , Receptors, Steroid/metabolism , Tuberculosis/immunology , Xenobiotics/metabolism , Animals , Apoptosis , Cell Line , Cell Survival , Humans , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Phagosomes , Pregnane X Receptor , Receptors, Steroid/genetics , Transgenes/genetics
10.
Autophagy ; 11(11): 1987-1997, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26390081

ABSTRACT

NR1D1 (nuclear receptor subfamily 1, group D, member 1), an adopted orphan nuclear receptor, is widely known to orchestrate the expression of genes involved in various biological processes such as adipogenesis, skeletal muscle differentiation, and lipid and glucose metabolism. Emerging evidence suggests that various members of the nuclear receptor superfamily perform a decisive role in the modulation of autophagy. Recently, NR1D1 has been implicated in augmenting the antimycobacterial properties of macrophages and providing protection against Mycobacterium tuberculosis infection by downregulating the expression of the IL10 gene in human macrophages. This antiinfective property of NR1D1 suggests the need for an improved understanding of its role in other host-associated antimycobacterial pathways. The results presented here demonstrate that in human macrophages either ectopic expression of NR1D1 or treatment with its agonist, GSK4112, enhanced the number of acidic vacuoles as well as the level of MAP1LC3-II, a signature molecule for determination of autophagy progression, in a concentration- and time-dependent manner. Conversely, a decrease in NR1D1 in knockdown cells resulted in the reduced expression of lysosomal-associated membrane protein 1, LAMP1, commensurate with a decrease in the level of transcription factor EB, TFEB. This is indicative of that NR1D1 may have a regulatory role in lysosome biogenesis. NR1D1 being a repressor, its positive regulation on LAMP1 and TFEB is suggestive of an indirect byzantine mechanism of action. Its role in the modulation of autophagy and lysosome biogenesis together with its ability to repress IL10 gene expression supports the theory that NR1D1 has a pivotal antimycobacterial function in human macrophages.

SELECTION OF CITATIONS
SEARCH DETAIL
...