Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
1.
J Cell Biol ; 223(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38656405

ABSTRACT

Cells exposed to proteotoxic stress invoke adaptive responses aimed at restoring proteostasis. Our previous studies have established a firm role for the transcription factor Nuclear factor-erythroid derived-2-related factor-1 (Nrf1) in responding to proteotoxic stress elicited by inhibition of cellular proteasome. Following proteasome inhibition, Nrf1 mediates new proteasome synthesis, thus enabling the cells to mitigate the proteotoxic stress. Here, we report that under similar circumstances, multiple components of the autophagy-lysosomal pathway (ALP) were transcriptionally upregulated in an Nrf1-dependent fashion, thus providing the cells with an additional route to cope with proteasome insufficiency. In response to proteasome inhibitors, Nrf1-deficient cells displayed profound defects in invoking autophagy and clearance of aggresomes. This phenomenon was also recapitulated in NGLY1 knockout cells, where Nrf1 is known to be non-functional. Conversely, overexpression of Nrf1 induced ALP genes and endowed the cells with an increased capacity to clear aggresomes. Overall, our results significantly expand the role of Nrf1 in shaping the cellular response to proteotoxic stress.


Subject(s)
Autophagy , NF-E2-Related Factor 1 , Proteotoxic Stress , Animals , Humans , Mice , Autophagy/genetics , Lysosomes/metabolism , NF-E2-Related Factor 1/metabolism , NF-E2-Related Factor 1/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Proteostasis , Stress, Physiological
2.
Cancer Med ; 13(7): e7021, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38562019

ABSTRACT

OBJECTIVE: Non-small-cell lung cancer (NSCLC) is a deadly form of cancer that exhibits extensive intercellular communication which contributed to chemoradiotherapy resistance. Recent evidence suggests that arrange of key proteins are involved in lung cancer progression, including gap junction proteins (GJPs). METHODS AND RESULTS: In this study, we examined the expression patterns of GJPs in NSCLC, uncovering that both gap junction protein, beta 2 (GJB2) and gap junction protein, beta 2 (GJB3) are increased in LUAD and LUSC. We observed a correlation between the upregulation of GJB2, GJB3 in clinical samples and a worse prognosis in patients with NSCLC. By examining the mechanics, we additionally discovered that nuclear factor erythroid-2-related factor 1 (NFE2L1) had the capability to enhance the expression of connexin26 and connexin 31 in the NSCLC cell line A549. In addition, the use of metformin was discovered to cause significant downregulation of gap junction protein, betas (GJBs) by limiting the presence of NFE2L1 in the cytoplasm. CONCLUSION: This emphasizes the potential of targeting GJBs as a viable treatment approach for NSCLC patients receiving metformin.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Metformin , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Connexins/genetics , Connexins/metabolism , Connexins/therapeutic use , Gap Junctions/metabolism , NF-E2-Related Factor 1/metabolism
3.
Sci Rep ; 13(1): 19900, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37963997

ABSTRACT

The NFE2L1 transcription factor (aka Nrf1) is a basic leucine zipper protein that performs a critical role in the cellular stress response pathway. Here, we characterized a novel variant of NFE2L1 referred to as NFE2L1-616. The transcript encoding NFE2L1-616 is derived from an intronic promoter, and it has a distinct first exon than other reported full-length NFE2L1 isoforms. The NFE2L1-616 protein constitutively localizes in the nucleus as it lacks the N-terminal amino acid residues that targets other full-length NFE2L1 isoforms to the endoplasmic reticulum. The expression level of NFE2L1-616 is lower than other NFE2L1 isoforms. It is widely expressed across different cell lines and tissues that were examined. NFE2L1-616 showed strong transcriptional activity driving luciferase reporter expression from a promoter containing antioxidant response element. Together, the results suggest that NFE2L1-616 variant can function as a positive regulator in the transcriptional regulation of NFE2L1 responsive genes.


Subject(s)
Antioxidant Response Elements , Gene Expression Regulation , Antioxidant Response Elements/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Cell Line , NF-E2-Related Factor 1/metabolism
4.
PLoS One ; 18(11): e0294508, 2023.
Article in English | MEDLINE | ID: mdl-38011090

ABSTRACT

The essential role of protein degradation by ubiquitin-proteasome system is exerted primarily for maintaining cellular protein homeostasis. The transcriptional activation of proteasomal genes by mTORC1 signaling depends on Nrf1, but whether this process is directly via SREBP1 remains elusive. In this study, our experiment evidence revealed that Nrf1 is not a direct target of SREBP1, although both are involved in the rapamycin-responsive regulatory networks. Closely scrutinizing two distinct transcriptomic datasets unraveled no significant changes in transcriptional expression of Nrf1 and almost all proteasomal subunits in either siSREBP2-silencing cells or SREBP1-∕-MEFs, when compared to equivalent controls. However, distinct upstream signaling to Nrf1 dislocation by p97 and its processing by DDI1/2, along with downstream proteasomal expression, may be monitored by mTOR signaling, to various certain extents, depending on distinct experimental settings in different types of cells. Our further evidence has been obtained from DDI1-∕-(DDI2insC) cells, demonstrating that putative effects of mTOR on the rapamycin-responsive signaling to Nrf1 and proteasomes may also be executed partially through a DDI1/2-independent mechanism, albeit the detailed regulatory events remain to be determined.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , NF-E2-Related Factor 1 , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , NF-E2-Related Factor 1/metabolism
5.
J Bioenerg Biomembr ; 55(6): 467-478, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37848756

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a common head and neck malignancy with increasing mortality and high recurrence. In this work, we aim to explore the functional role of NFE2 like bZIP transcription factor 1 (NFE2L1) in OSCC progression. Based on databases analysis, we found that NFE2L1 was overexpressed in OSCC tumor tissues, and elevated NFE2L1 level induced poor prognosis of OSCC patients. Our results showed that NFE2L1 is upregulated in OSCC cells and overexpression of NFE2L1 promotes cell proliferation, and reduces the sensitivity of OSCC cells to erastin-induced ferroptosis. NFE2L1 upregulation decreased the levels of Fe2+, lipid reactive oxygen species and content of malondialdehyde, and increased the level of the key negative regulator of ferroptosis, GPX4 and SLC7A11. In NFE2L1 suppressed cells, these trends were reversed. Further results of dual luciferase reporter and chromatin immunoprecipitation assays confirmed that NFE2L1 could bind to the promoter of Holliday junction recognition protein (HJURP) to increase the transcriptional activity of HJURP, thus upregulating its expression. Inhibition of HJURP attenuated the proliferation and ferroptosis inhibition in NFE2L1 upregulated cells. In vivo tumorigenicity assay further proved that NFE2L1 promotes OSCC tumor growth. In summary, NFE2L1 restrains ferroptosis by transcriptionally regulating HJURP and participates in the progress of OSCC. Thus, NFE2L1 plays a key role in OSCC development and may be a promising therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Ferroptosis , Head and Neck Neoplasms , MicroRNAs , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Mouth Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , MicroRNAs/metabolism , Gene Expression Regulation, Neoplastic , Cell Movement , NF-E2-Related Factor 1/metabolism
6.
Sci Rep ; 13(1): 15843, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37739987

ABSTRACT

Proteasomes are multi-subunit complexes that specialize in protein degradation. Cancer cells exhibit a heightened dependence on proteasome activity, presumably to support their enhanced proliferation and other cancer-related characteristics. Here, a systematic analysis of TCGA breast cancer datasets revealed that proteasome subunit transcript levels are elevated in all intrinsic subtypes (luminal, HER2-enriched, and basal-like/triple-negative) when compared to normal breast tissue. Although these observations suggest a pan-breast cancer utility for proteasome inhibitors, our further experiments with breast cancer cell lines and patient-derived xenografts (PDX) pointed to triple-negative breast cancer (TNBC) as the most sensitive subtype to proteasome inhibition. Finally, using TNBC cells, we extended our studies to in vivo xenograft experiments. Our previous work has firmly established a cytoprotective role for the transcription factor NRF1 via its ability to upregulate proteasome genes in response to proteasome inhibition. In further support of this notion, we show here that NRF1 depletion significantly reduced tumor burden in an MDA-MB-231 TNBC xenograft mouse model treated with carfilzomib. Taken together, our results point to TNBC as a particularly vulnerable breast cancer subtype to proteasome inhibition and provide a proof-of-principle for targeting NRF1 as a viable means to increase the efficacy of proteasome inhibitors in TNBC tumors.


Subject(s)
NF-E2-Related Factor 1 , Proteasome Endopeptidase Complex , Triple Negative Breast Neoplasms , Animals , Humans , Mice , Cytoplasm , Disease Models, Animal , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Proteolysis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , NF-E2-Related Factor 1/metabolism
7.
Stem Cell Rev Rep ; 19(6): 1994-2012, 2023 08.
Article in English | MEDLINE | ID: mdl-37243831

ABSTRACT

Premature ovarian failure (POF) is an important cause of female infertility and seriously impacts the physical and psychological health of patients. Mesenchymal stromal cells-derived exosomes (MSCs-Exos) have an essential role in the treatment of reproductive disorders, particularly POF. However, the biological function and therapeutic mechanism of MSCs exosomal circRNAs in POF remain to be determined. Here, with bioinformatics analysis and functional assays, circLRRC8A was found to be downregulated in senescent granulosa cells (GCs) and acted as a crucial factor in MSCs-Exos for oxidative damage protection and anti-senescence of GCs in vitro and in vivo. Mechanistic investigations revealed that circLRRC8A served as an endogenous miR-125a-3p sponge to downregulate NFE2L1 expression. Moreover, eukaryotic initiation factor 4A3 (EIF4A3), acting as a pre-mRNA splicing factor, promoted circLRRC8A cyclization and expression by directly binding to the LRRC8A mRNA transcript. Notably, EIF4A3 silencing reduced circLRRC8A expression and attenuated the therapeutic effect of MSCs-Exos on oxidatively damaged GCs. This study demonstrates a new therapeutic pathway for cellular senescence protection against oxidative damage by delivering circLRRC8A-enriched exosomes through the circLRRC8A/miR-125a-3p/NFE2L1 axis and paves the way for the establishment of a cell-free therapeutic approach for POF. CircLRRC8A may be a promising circulating biomarker for diagnosis and prognosis and an exceptional candidate for further therapeutic exploration.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Primary Ovarian Insufficiency , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , Primary Ovarian Insufficiency/metabolism , Granulosa Cells/metabolism , NF-E2-Related Factor 1/metabolism , DEAD-box RNA Helicases/metabolism , Membrane Proteins/metabolism
8.
Cell Rep ; 40(13): 111428, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36170818

ABSTRACT

Proteasome machinery is a major proteostasis control system in human cells, actively compensated upon its inhibition. To understand this compensation, we compared global protein landscapes upon the proteasome inhibition with carfilzomib, in normal fibroblasts, cells of multiple myeloma, and cancers of lung, colon, and pancreas. Molecular chaperones, autophagy, and endocytosis-related proteins are the most prominent vulnerabilities in combination with carfilzomib, while targeting of the HSP70 family chaperones HSPA1A/B most specifically sensitizes cancer cells to the proteasome inhibition. This suggests a central role of HSP70 in the suppression of the proteasome downregulation, allowing to identify pathways impinging on HSP70 upon the proteasome inhibition. HSPA1A/B indeed controls proteasome-inhibition-induced autophagy, unfolded protein response, and endocytic flux, and directly chaperones the proteasome machinery. However, it does not control the NRF1/2-driven proteasome subunit transcriptional bounce-back. Consequently, targeting of NRF1 proves effective in decreasing the viability of cancer cells with the inhibited proteasome and HSP70.


Subject(s)
HSP70 Heat-Shock Proteins , Neoplasms , Proteasome Endopeptidase Complex , Humans , Cell Line, Tumor , HSP70 Heat-Shock Proteins/metabolism , Neoplasms/genetics , NF-E2-Related Factor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteostasis
9.
J Mol Histol ; 53(4): 729-740, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35727472

ABSTRACT

Cisplatin, a first-line chemotherapeutic agent commonly used to treat various solid tumors, induce severe adverse effects, especially nephrotoxicity, which largely limits its clinical application. However, the currently used measures to prevent nephrotoxicity are not ideal owing to the mechanisms underlying cisplatin-induced nephrotoxicity are not comprehensively understood. Herein, we examined the effects of silibinin on cisplatin-induced nephrotoxicity and found that silibinin exerted cytoprotection effects during cisplatin treatment in HEK293 cells and in a cisplatin-induced acute kidney injury (AKI) model. Mechanistically, silibinin ameliorated cisplatin-induced AKI via decreasing ROS-mediated MAPK signaling pathway activation, which was confirmed using the inhibitor N-acetylcysteine. Moreover, the protective effect of silibinin against cisplatin-induced ROS generation through the antioxidant transcription factor nuclear factor-erythroid 2-related factor 1 (Nfe2l1), rather than Nfe2l2, mediates HO1 expression. Furthermore, interference with the abundance of Nfe2l1 using siRNA or an overexpression plasmid enhanced or decreased the effect of cisplatin-induced apoptosis, respectively, in HEK293 cells. Interestingly, Nfe2l1 protein stability was more sensitive to cisplatin than that of Nfe2l2. More importantly, the mechanism that silibinin activates Nfe2l1-mediated antioxidant responses was confirmed in a cisplatin-induced AKI model. Silibinin rescued cisplatin-induced Nfe2l1 inhibition by regulating its transcription and post-translational modifications. Taken together, our results reveal a novel mechanism by which silibinin ameliorates cisplatin-induced AKI via activating Nfe2l1-mediated antioxidative response, which provides a new insights to protect patients receiving cisplatin-based cancer treatment against AKI.


Subject(s)
Acute Kidney Injury , Cisplatin , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Cisplatin/adverse effects , HEK293 Cells , Humans , Kidney/pathology , NF-E2-Related Factor 1/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction , Silybin/metabolism , Silybin/pharmacology
10.
Cell Death Dis ; 13(5): 501, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614059

ABSTRACT

The antioxidant transcription factor NFE2L1 (also called Nrf1) acts as a core regulator of redox signaling and metabolism homeostasis, and thus, its dysfunction results in multiple systemic metabolic diseases. However, the molecular mechanism(s) by which NFE2L1 regulates glycose and lipid metabolism remains elusive. Here, we found that loss of NFE2L1 in human HepG2 cells led to a lethal phenotype upon glucose deprivation and NFE2L1 deficiency could affect the uptake of glucose. Further experiments revealed that glycosylation of NFE2L1 enabled it to sense the energy state. These results indicated that NFE2L1 can serve as a dual sensor and regulator of glucose homeostasis. The transcriptome, metabolome, and seahorse data further revealed that disruption of NFE2L1 could reprogram glucose metabolism to aggravate the Warburg effect in NFE2L1-silenced hepatoma cells, concomitant with mitochondrial damage. Co-expression and Co-immunoprecipitation experiments demonstrated that NFE2L1 could directly interact and inhibit AMPK. Collectively, NFE2L1 functioned as an energy sensor and negatively regulated AMPK signaling through directly interacting with AMPK. The novel NFE2L1/AMPK signaling pathway delineate the mechanism underlying of NFE2L1-related metabolic diseases and highlight the crosstalk between redox homeostasis and metabolism homeostasis.


Subject(s)
AMP-Activated Protein Kinases , NF-E2-Related Factor 1 , AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Glucose , Homeostasis , NF-E2-Related Factor 1/metabolism , Signal Transduction
11.
J Bioenerg Biomembr ; 54(2): 93-108, 2022 04.
Article in English | MEDLINE | ID: mdl-35274224

ABSTRACT

Circular RNAs (circRNAs) are crucial for the pathogenesis of human diseases, including osteoarthritis (OA). Here, we set to elucidate the biological action of circ-LRP1B in OA pathogenesis. Human C28/I2 chondrocytes were stimulated by lipopolysaccharide (LPS). Circ-LRP1B, nuclear factor, erythroid 2 like 1 (NRF1) and microRNA (miR)-34a-5p were quantified by quantitative real-time PCR (qRT-PCR) or immunoblotting. Cell viability, proliferation, and apoptosis abilities were gauged by MTT, 5-Ethynyl-2'-Deoxyuridine (EdU) staining, and flow cytometry assays, respectively. Direct relationship between miR-34a-5p and circ-LRP1B or NRF1 was validated by RNA pull-down and dual-luciferase reporter assays. Circ-LRP1B was found to be underexpressed in OA cartilage and LPS-stimulated C28/I2 chondrocytes. Enforced expression of circ-LRP1B promoted cell proliferation, and repressed apoptosis and oxidative stress, as well as impacted OA-specific hallmarks expression of LPS-stimulated C28/I2 cells. NRF1 was identified as a downstream effector of circ-LRP1B function. Moreover, NRF1 was identified as a miR-34a-5p target in LPS-stimulated C28/I2 cells. Circ-LRP1B acted as a competing endogenous RNA (ceRNA) for miR-34a-5p to involve the post-transcriptional regulation of NRF1 expression. Furthermore, the effects of circ-LRP1B overexpression partly depended on the reduction of available miR-34a-5p. These findings demonstrate that circ-LRP1B functions as a ceRNA to regulate the proliferation, apoptosis and oxidative stress of LPS-stimulated human C28/I2 chondrocytes by miR-34a-5p/NRF1 network.


Subject(s)
Lipopolysaccharides , MicroRNAs , RNA, Circular , Apoptosis , Cell Proliferation/physiology , Chondrocytes/metabolism , Chondrocytes/pathology , Humans , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , NF-E2-Related Factor 1/metabolism , Oxidative Stress , RNA, Circular/genetics , Receptors, LDL/metabolism
12.
Proc Natl Acad Sci U S A ; 119(11): e2118646119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35271393

ABSTRACT

SignificanceFerroptosis is an oxidative form of cell death whose biochemical regulation remains incompletely understood. Cap'n'collar (CNC) transcription factors including nuclear factor erythroid-2-related factor 1 (NFE2L1/NRF1) and NFE2L2/NRF2 can both regulate oxidative stress pathways but are each regulated in a distinct manner, and whether these two transcription factors can regulate ferroptosis independent of one another is unclear. We find that NFE2L1 can promote ferroptosis resistance, independent of NFE2L2, by maintaining the expression of glutathione peroxidase 4 (GPX4), a key protein that prevents lethal lipid peroxidation. NFE2L2 can also promote ferroptosis resistance but does so through a distinct mechanism that appears independent of GPX4 protein expression. These results suggest that NFE2L1 and NFE2L2 independently regulate ferroptosis.


Subject(s)
Ferroptosis , Gene Expression Regulation , NF-E2-Related Factor 1 , Oxidative Stress , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Phospholipid Hydroperoxide Glutathione Peroxidase , Ferroptosis/genetics , Gene Knockout Techniques , Humans , Lipid Peroxidation , Metabolic Networks and Pathways/genetics , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Oxidative Stress/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
13.
Int J Sports Med ; 43(7): 632-641, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35180801

ABSTRACT

Metabolic diseases are associated with hypoestrogenism owing to their lower energy expenditure and consequent imbalance. Physical training promotes energy expenditure through PGC-1α and NRF-1, which are muscle proteins of the oxidative metabolism. However, the influence of physical training on protein expression in individuals with hypoestrogenism remains uncertain. Thus, the aim of this study is to determine the effect of 12 weeks of moderate-intensity swimming training on the muscle expression of PGC-1α, NRF-1, glycogen and triglyceride in ovariectomised rats. OVX and OVX+TR rats were subjected to ovariectomy. The trained animals swam for 30 minutes, 5 days/week, at 80% of the critical load intensity. Soleus was collected to quantify PGC-1α and NRF-1 expressions, while gastrocnemius and gluteus maximus were collected to measure glycogen and triglyceride. Blood glucose was also evaluated. Whereas ovariectomy decreased PGC-1α expression (p<0.05) without altering NRF-1 (p=0.48), physical training increased PGC-1α (p<0.01) and NRF-1 (p<0.05). Ovariectomy reduced glycogen (p<0.05) and triglyceride (p<0.05), whereas physical training increased glycogen (p<0.05) but did not change triglyceride (p=0.06). Ovariectomy increased blood glucose (p<0.01), while physical training reduced it (p<0.01). In summary, 12 weeks of individualized and moderate-intensity training were capable of preventing muscle metabolic consequences caused by ovariectomy.


Subject(s)
Muscle, Skeletal , NF-E2-Related Factor 1 , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Animals , Blood Glucose/metabolism , Female , Glycogen/metabolism , Muscle, Skeletal/metabolism , NF-E2-Related Factor 1/metabolism , Ovariectomy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal/physiology , Rats , Triglycerides/metabolism
14.
Ecotoxicol Environ Saf ; 232: 113268, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35124418

ABSTRACT

Chronic arsenic exposure is associated with the increased risk of several types of cancer, among which, lung cancer is the most deadly one. Nuclear factor erythroid 2 like 1 (NFE2L1), a transcription factor belonging to CNC-bZIP family, regulates multiple important cellular functions in response to acute arsenite exposure. However, the role of NFE2L1 in lung cancer induced by chronic arsenite exposure is unknown. In this study, we firstly showed that chronic arsenite exposure (36 weeks) led to epithelial-mesenchymal transition (EMT) and malignant transformation in human bronchial epithelial cells (BEAS-2B). During the process of malignant transformation, the expression of long isoforms of NFE2L1 (NFE2L1-L) was elevated. Thereafter, BEAS-2B cells with NFE2L1-L stable knockdown (NFE2L1-L-KD) was chronically exposed to arsenite. As expected, silencing of NFE2L1-L gene strikingly inhibited the arsenite-induced EMT and the subsequent malignant transformation. Additionally, NFE2L1-L silencing suppressed the transcription of EMT-inducer SNAIL1 and increased the expression of E-cadherin. Conversely, NFE2L1-L overexpression increased SNAIL1 transcription but decreased E-cadherin expression. Collectively, our data suggest that NFE2L1-L promotes EMT by positively regulating SNAIL1 transcription, and is involved in malignant transformation induced by arsenite.


Subject(s)
Arsenites , Arsenites/metabolism , Arsenites/toxicity , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , NF-E2-Related Factor 1/metabolism , Phenotype , Protein Isoforms/metabolism
15.
Hum Mutat ; 43(4): 471-476, 2022 04.
Article in English | MEDLINE | ID: mdl-35112409

ABSTRACT

The NFE2L1 transcription factor (also known as Nrf1 for nuclear factor erythroid 2-related factor-1) is a broadly expressed basic leucine zipper protein that performs a critical role in the cellular stress response pathway. Here, we identified a heterozygous nonsense mutation located in the last exon of the gene that terminates translation prematurely, resulting in the production of a truncated peptide devoid of the carboxyl-terminal region containing the DNA-binding and leucine-zipper dimerization interface of the protein. Variant derivatives were well expressed in vitro, and they inhibited the transactivation function of wild-type proteins in luciferase reporter assays. Our studies suggest that this dominant-negative effect of truncated variants is through the formation of inactive heterodimers with wild-type proteins preventing the expression of its target genes. These findings suggest the potential role of diminished NFE2L1 function as an explanation for the developmental delay, hypotonia, hypospadias, bifid scrotum, and failure to thrive observed in the patient.


Subject(s)
Failure to Thrive , Muscle Hypotonia , Gene Expression Regulation , Genitalia , Humans , Male , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Mol Metab ; 57: 101436, 2022 03.
Article in English | MEDLINE | ID: mdl-34999280

ABSTRACT

OBJECTIVE: Ferroptosis continues to emerge as a novel modality of cell death with important therapeutic implications for a variety of diseases, most notably cancer and degenerative diseases. While susceptibility, initiation, and execution of ferroptosis have been linked to reprogramming of cellular lipid metabolism, imbalances in iron-redox homeostasis, and aberrant mitochondrial respiration, the detailed mechanisms of ferroptosis are still insufficiently well understood. METHODS AND RESULTS: Here we show that diminished proteasome function is a new mechanistic feature of ferroptosis. The transcription factor nuclear factor erythroid-2, like-1 (NFE2L1) protects from ferroptosis by sustaining proteasomal activity. In cellular systems, loss of NFE2L1 reduced cellular viability after the induction of both chemically and genetically induced ferroptosis, which was linked to the regulation of proteasomal activity under these conditions. Importantly, this was reproduced in a Sedaghatian-type Spondylometaphyseal Dysplasia (SSMD) patient-derived cell line carrying mutated glutathione peroxidase-4 (GPX4), a critical regulator of ferroptosis. Also, reduced proteasomal activity was associated with ferroptosis in Gpx4-deficient mice. In a mouse model for genetic Nfe2l1 deficiency, we observed brown adipose tissue (BAT) involution, hyperubiquitination of ferroptosis regulators, including the GPX4 pathway, and other hallmarks of ferroptosis. CONCLUSION: Our data highlight the relevance of the NFE2L1-proteasome pathway in ferroptosis. Manipulation of NFE2L1 activity might enhance ferroptosis-inducing cancer therapies as well as protect from aberrant ferroptosis in neurodegeneration, general metabolism, and beyond.


Subject(s)
Ferroptosis , NF-E2-Related Factor 1 , Animals , Homeostasis , Humans , Mice , Mitochondria/metabolism , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Proteasome Endopeptidase Complex/metabolism
17.
Blood Adv ; 6(2): 429-440, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34649278

ABSTRACT

Multiple myeloma (MM) cells suffer from baseline proteotoxicity as the result of an imbalance between the load of misfolded proteins awaiting proteolysis and the capacity of the ubiquitin-proteasome system to degrade them. This intrinsic vulnerability is at the base of MM sensitivity to agents that perturb proteostasis, such as proteasome inhibitors (PIs), the mainstay of modern-day myeloma therapy. De novo and acquired PI resistance are important clinical limitations that adversely affect prognosis. The molecular mechanisms underpinning PI resistance are only partially understood, limiting the development of drugs that can overcome it. The transcription factor NRF1 is activated by the aspartic protease DNA damage inducible 1 homolog 2 (DDI2) upon proteasome insufficiency and governs proteasome biogenesis. In this article, we show that MM cells exhibit baseline NRF1 activation and are dependent upon DDI2 for survival. DDI2 knockout (KO) is cytotoxic for MM cells, both in vitro and in vivo. Protein structure-function studies show that DDI2 KO blocks NRF1 cleavage and nuclear translocation, causing impaired proteasome activity recovery upon irreversible proteasome inhibition and, thereby, increasing sensitivity to PIs. Add-back of wild-type, but not of catalytically dead DDI2, fully rescues these phenotypes. We propose that DDI2 is an unexplored promising molecular target in MM by disrupting the proteasome stress response and exacerbating proteotoxicity.


Subject(s)
Aspartic Acid Proteases/metabolism , Multiple Myeloma , NF-E2-Related Factor 1/metabolism , Proteasome Endopeptidase Complex , Humans , NF-E2-Related Factor 1/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis
18.
Food Chem Toxicol ; 158: 112633, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34699923

ABSTRACT

Streptozotocin (STZ) is a pancreatic ß cell-specific toxicant that is widely used to generate models of diabetes in rodents as well as in the treatment of tumors derived from pancreatic ß cells. DNA alkylation, oxidative stress and mitochondrial toxicity have been recognized as the mechanisms for STZ-induced pancreatic ß cell damage. Here, we found that pancreatic ß cell-specific deficiency of nuclear factor erythroid-derived factor 2-related factor 1 (NFE2L1), a master regulator of the cellular adaptive response to a variety of stresses, in mice led to a dramatic resistance to STZ-induced hyperglycemia. Indeed, fifteen days subsequent to last dosage of STZ, the pancreatic ß cell specific Nfe2l1 knockout [Nfe2l1(ß)-KO] mice showed reduced hyperglycemia, improved glucose tolerance, higher plasma insulin and more intact islets surrounded by exocrine acini compared to the Nfe2l1-Flox control mice with the same treatment. Immunohistochemistry staining revealed a greater amount of insulin-positive cells in the pancreas of Nfe2l1(ß)-KO mice than those in Nfe2l1-Flox mice 15 days after the last STZ injection. In line with this observation, both isolated Nfe2l1(ß)-KO islets and Nfe2l1-deficient MIN6 (Nfe2l1-KD) cells were resistant to STZ-induced toxicity and apoptosis. Furthermore, pretreatment of the MIN6 cells with glycolysis inhibitor 2-Deoxyglucose sensitized Nfe2l1-KD cells to STZ-induced toxicity. These findings demonstrated that loss of Nfe2l1 attenuates pancreatic ß cells damage and dysfunction caused by STZ exposure, partially due to Nfe2l1 deficiency-induced metabolic switch to enhanced glycolysis.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , NF-E2-Related Factor 1 , Animals , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Male , Mice , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Streptozocin
19.
Nat Commun ; 12(1): 5270, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489413

ABSTRACT

Following injury, cells in regenerative tissues have the ability to regrow. The mechanisms whereby regenerating cells adapt to injury-induced stress conditions and activate the regenerative program remain to be defined. Here, using the mammalian neonatal heart regeneration model, we show that Nrf1, a stress-responsive transcription factor encoded by the Nuclear Factor Erythroid 2 Like 1 (Nfe2l1) gene, is activated in regenerating cardiomyocytes. Genetic deletion of Nrf1 prevented regenerating cardiomyocytes from activating a transcriptional program required for heart regeneration. Conversely, Nrf1 overexpression protected the adult mouse heart from ischemia/reperfusion (I/R) injury. Nrf1 also protected human induced pluripotent stem cell-derived cardiomyocytes from doxorubicin-induced cardiotoxicity and other cardiotoxins. The protective function of Nrf1 is mediated by a dual stress response mechanism involving activation of the proteasome and redox balance. Our findings reveal that the adaptive stress response mechanism mediated by Nrf1 is required for neonatal heart regeneration and confers cardioprotection in the adult heart.


Subject(s)
Heart/physiology , Myocardial Reperfusion Injury/metabolism , NF-E2-Related Factor 1/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/physiology , Doxorubicin/pharmacology , Female , Heme Oxygenase (Decyclizing)/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Male , Mice, Knockout , Mice, Transgenic , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/physiology , NF-E2-Related Factor 1/genetics , Oxidation-Reduction , Proteostasis , Rats, Sprague-Dawley , Regeneration
20.
Life Sci ; 279: 119697, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34102194

ABSTRACT

AIMS: Vitamin D and rosuvastatin are well-known drugs that mediate beneficial effects in treating type-2 diabetes (T2D) complications; however, their anti-neuropathic potential is debatable. Hence, our study investigates their neurotherapeutic potential and the possible underlying mechanisms using a T2D-associated neuropathy rat model. MAIN METHODS: Diabetic peripheral neuropathy (DPN) was induced with 8 weeks of administration of a high fat fructose diet followed by a single i.p. injection of streptozotocin (35 mg/kg). Six weeks later, DPN developed and rats were divided into five groups; viz., control, untreated DPN, DPN treated with vitamin D (cholecalciferol, 3500 IU/kg/week), DPN treated with rosuvastatin (10 mg/kg/day), or DPN treated with combination vitamin D and rosuvastatin. We determined their anti-neuropathic effects on small nerves (tail flick test); large nerves (electrophysiological and histological examination); neuronal inflammation (TNF-α and IL-18); apoptosis (caspase-3 activity and Bcl-2); mitochondrial function (NRF-1, TFAM, mtDNA, and ATP); and NICD1, Wnt-10α/ß-catenin, and TGF-ß/Smad-7 pathways. KEY FINDINGS: Two-month treatment with vitamin D and/or rosuvastatin regenerated neuronal function and architecture and abated neuronal inflammation and apoptosis. This was verified by the inhibition of the neuronal content of TNF-α, IL-18, and caspase-3 activity, while augmenting Bcl-2 content in the sciatic nerve. These treatments inhibited the protein expressions of NICD1, Wnt-10α, ß-catenin, and TGF-ß; increased the sciatic nerve content of Smad-7; and enhanced mitochondrial biogenesis and function. SIGNIFICANCE: Vitamin D and/or rosuvastatin alleviated diabetes-induced neuropathy by suppressing Notch1 and Wnt-10α/ß-catenin; modulating TGF-ß/Smad-7 signaling pathways; and enhancing mitochondrial function, which lessened neuronal degeneration, demyelination, and fibrosis.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetic Neuropathies/prevention & control , Gene Expression Regulation/drug effects , Rosuvastatin Calcium/pharmacology , Vitamin D/administration & dosage , Animals , Anticholesteremic Agents/pharmacology , Diabetic Neuropathies/etiology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Drug Therapy, Combination , Male , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Rats , Rats, Wistar , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vitamins/administration & dosage , Wnt Proteins/genetics , Wnt Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...