Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Cancer ; 23(1): 188, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243032

ABSTRACT

Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFß, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.


Subject(s)
Cell Proliferation , Myeloid-Derived Suppressor Cells , Neoplastic Stem Cells , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Humans , Animals , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Female , Mice , Myeloid-Derived Suppressor Cells/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Prognosis , Cell Movement , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Neoplasm Proteins
2.
J Integr Plant Biol ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39185667

ABSTRACT

WRKY transcription factors play key roles in plant resistance to various stresses, but their roles in fruit ripening remain largely unknown. Here, we report a WRKY gene PpWRKY14 involved in the regulation of fruit ripening in peach. The expression of PpWRKY14 showed an increasing trend throughout fruit development. PpWRKY14 was a target gene of PpNAC1, a master regulator of peach fruit ripening. PpWRKY14 could directly bind to the promoters of PpACS1 and PpACO1 to induce their expression, and this induction was greatly enhanced when PpWRKY14 formed a dimer with PpNAC1. However, the transcription of PpNAC1 could be directly suppressed by two EIN3/EIL1 genes, PpEIL2 and PpEIL3. The PpEIL2/3 genes were highly expressed at the early stages of fruit development, but their expression was programmed to decrease significantly during the ripening stage, thus derepressing the expression of PpNAC1. These results suggested a PpEIL2/3-PpNAC1-PpWRKY14 module that regulates fruit ripening by modulating ethylene production in peach. Our results provided an insight into the regulatory roles of EIN3/EIL1 and WRKY genes in fruit ripening.

3.
J Med Virol ; 95(7): e28957, 2023 07.
Article in English | MEDLINE | ID: mdl-37465969

ABSTRACT

Nucleus accumbens-associated protein 1 (NAC1), a transcriptional cofactor, has been found to play important roles in regulating regulatory T cells, CD8+ T cells, and antitumor immunity, but little is known about its effects on T-cell memory. In this study, we found that NAC1 expression restricts memory formation of CD4+ T cells during viral infection. Analysis of CD4+ T cells from wild-type (WT) and NAC1-deficient (-/- ) mice showed that NAC1 is essential for T-cell metabolism, including glycolysis and oxidative phosphorylation, and supports CD4+ T-cell survival in vitro. We further demonstrated that a deficiency of NAC1 downregulates glycolysis and correlates with the AMPK-mTOR pathway and causes autophagy defective in CD4+ T cells. Loss of NAC1 reduced the expression of ROCK1 and the phosphorylation and stabilization of BECLIN1. However, a forced expression of ROCK1 in NAC1-/- CD4+ T cells restored autophagy and the activity of the AMPK-mTOR pathway. In animal experiments, adoptively transferred NAC1-/- CD4+ T cells or NAC1-/- mice challenged with VACV showed enhanced formation of VACV-specific CD4+ memory T cells compared to adoptively transferred WT CD4+ T cells or WT mice. This memory T-cell formation enhancement was abrogated by forcing expression of ROCK1. Our study reveals a novel role for NAC1 as a suppressor of CD4+ T-cell memory formation and suggests that targeting NAC1 could be a new approach to promoting memory CD4+ T-cell development, which is critical for an effective immune response against pathogens.


Subject(s)
AMP-Activated Protein Kinases , CD8-Positive T-Lymphocytes , Animals , Mice , AMP-Activated Protein Kinases/metabolism , CD4-Positive T-Lymphocytes , Cell Survival , Immunologic Memory , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism
4.
Ann Hum Biol ; 50(1): 82-93, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36714955

ABSTRACT

BACKGROUND: Resistance to antiepileptic drugs (AEDs) remains one of the main challenges to neurologists. Polymorphisms of drug efflux transporters such as multidrug resistance (MDR1) gene and target sites such as the nucleus accumbens-associated 1 (NAC1) gene have been suggested to influence the responsiveness to treatment. AIM: Evaluation of the association of MDR1 and NAC1 polymorphisms with AEDs resistance among Jordanian epileptic patients. SUBJECTS AND METHODS: 86 Jordanian epileptics were included in the study. DNA was extracted and genotyping was conducted by polymerase chain reaction followed by sequencing. Nine single nucleotide polymorphisms (SNPs) on the MDR1 gene and six SNPs on the NAC1 gene were investigated. RESULTS: MDR1 and NAC1 polymorphisms don't seem to influence the resistance to AEDs at the genotype or allele level. However, a strong association was found between MDR1 rs2032588 (OR = 5; 95%CI = [1.3-18.8], p = 0.01) and AEDs resistance among males at the allele level. Also, data revealed an association between MDR1 rs1128503 and AEDs resistance among females at the allele level. CONCLUSION: The data suggest that MDR1 and NAC1 polymorphisms do not influence the AEDs resistance among Jordanian epileptics. However, there is a gender-dependent association between MDR1 polymorphisms and resistance to AEDs at two SNPs (rs2032588 and rs1128503).


Subject(s)
Anticonvulsants , Epilepsy , Male , Female , Humans , Anticonvulsants/therapeutic use , Anticonvulsants/pharmacology , Cross-Sectional Studies , Jordan , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/therapeutic use , Gene Frequency , Drug Resistance, Multiple/genetics , Epilepsy/drug therapy , Epilepsy/genetics , Polymorphism, Single Nucleotide , Genotype
5.
Int J Mol Sci ; 23(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36293114

ABSTRACT

Root meristem is a reserve of undifferentiated cells which guide root development. To maintain root meristem identity and therefore continuous root growth, the rate of cell differentiation must coordinate with the rate of generation of new cells. The E2 promoter-binding factor a (E2Fa) has been shown to regulate root growth through controlling G1/S cell cycle transitions in Arabidopsis thaliana. Here, we found that NAC1, a member of the NAM/ATAF/CUC family of transcription factors, regulated root growth by directly repressing the transcription of E2Fa. Loss of NAC1 triggers an up-regulation of the E2Fa expression and causes a reduced meristem size and short-root phenotype, which are largely rescued by mutation of E2Fa. Further analysis showed that NAC1 was shown to regulate root meristem by controlling endopolyploidy levels in an E2Fa-dependent manner. This study provides evidence to show that NAC1 maintains root meristem size and root growth by directly repressing the transcription of E2Fa in Arabidopsis.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Meristem , Arabidopsis Proteins/metabolism , Gene Expression Regulation, Plant , Transcription Factors/genetics , Transcription Factors/metabolism , Plant Roots , E2F Transcription Factors/genetics
6.
Plant J ; 104(3): 781-799, 2020 11.
Article in English | MEDLINE | ID: mdl-32772407

ABSTRACT

Salvianolic acids (SalAs), a group of secondary metabolites in Salvia miltiorrhiza, are widely used for treating cerebrovascular diseases. Their biosynthesis is modulated by a variety of abiotic factors, including ultraviolet-B (UV-B) irradiation; however, the underlying mechanisms remain largely unknown. Here, an integrated metabolomic, proteomic, and transcriptomic approach coupled with transgenic analyses was employed to dissect the mechanisms underlying UV-B irradiation-induced SalA biosynthesis. Results of metabolomics showed that 28 metabolites, including 12 SalAs, were elevated in leaves of UV-B-treated S. miltiorrhiza. Meanwhile, the contents of several phytohormones, including jasmonic acid and salicylic acid, which positively modulate the biosynthesis of SalAs, also increased in UV-B-treated S. miltiorrhiza. Consistently, 20 core biosynthetic enzymes and numerous transcription factors that are involved in SalA biosynthesis were elevated in treated samples as indicated by a comprehensive proteomic analysis. Correlation and gene expression analyses demonstrated that the NAC1 gene, encoding a NAC transcriptional factor, was positively involved in UV-B-induced SalA biosynthesis. Accordingly, overexpression and RNA interference of NAC1 increased and decreased SalA contents, respectively, through regulation of key biosynthetic enzymes. Furthermore, ChIP-qPCR and Dual-LUC assays showed that NAC1 could directly bind to the CATGTG and CATGTC motifs present in the promoters of the SalA biosynthesis-related genes PAL3 and TAT3, respectively, and activate their expression. Our results collectively demonstrate that NAC1 plays a crucial role in UV-B irradiation-induced SalA biosynthesis. Taken together, our findings provide mechanistic insights into the UV-B-induced SalA biosynthesis in S. miltiorrhiza, and shed light on a great potential for the development of SalA-abundant varieties through genetic engineering.


Subject(s)
Plant Proteins/genetics , Polyphenols/biosynthesis , Salvia miltiorrhiza/metabolism , Salvia miltiorrhiza/radiation effects , Alkenes , Enzymes/metabolism , Gene Expression Regulation, Plant/radiation effects , Metabolomics/methods , Plant Leaves/metabolism , Plant Leaves/radiation effects , Plant Proteins/metabolism , Plants, Genetically Modified , Polyphenols/genetics , Proteomics/methods , RNA Interference , Salvia miltiorrhiza/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Ultraviolet Rays , Up-Regulation
7.
J Biol Chem ; 294(25): 10006-10017, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31101655

ABSTRACT

Nucleus accumbens-associated protein-1 (NAC1) is a transcriptional repressor encoded by the NACC1 gene, which is amplified and overexpressed in various human cancers and plays critical roles in tumor development, progression, and drug resistance. NAC1 has therefore been explored as a potential therapeutic target for managing malignant tumors. However, effective approaches for effective targeting of this nuclear protein remain elusive. In this study, we identified a core unit consisting of Met7 and Leu90 in NAC1's N-terminal domain (amino acids 1-130), which is critical for its homodimerization and stability. Furthermore, using a combination of computational analysis of the NAC1 dimerization interface and high-throughput screening (HTS) for small molecules that inhibit NAC1 homodimerization, we identified a compound (NIC3) that selectively binds to the conserved Leu-90 of NAC1 and prevents its homodimerization, leading to proteasomal NAC1 degradation. Moreover, we demonstrate that NIC3-mediated down-regulation of NAC1 protein sensitizes drug-resistant tumor cells to conventional chemotherapy and enhances the antimetastatic effect of the antiangiogenic agent bevacizumab both in vitro and in vivo These results suggest that small-molecule inhibitors of NAC1 homodimerization may effectively sensitize cancer cells to some anticancer agents and that NAC1 homodimerization could be further explored as a potential therapeutic target in the development of antineoplastic agents.


Subject(s)
Acetamides/pharmacology , Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/chemistry , Protein Multimerization/drug effects , Repressor Proteins/chemistry , Small Molecule Libraries/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , Bevacizumab/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Bioorg Chem ; 80: 1-10, 2018 10.
Article in English | MEDLINE | ID: mdl-29864683

ABSTRACT

The oncogenic transcriptional corepressor Nac1 contains a conserved POZ protein-protein interaction module that mediates homodimerization or heterodimerization with itself or other POZ proteins. The dimerization has been recognized as an attractive target for cancer therapy. Here, we attempted to (i) discover those potential binding partners of Nac1 in the human genome, (ii) derive key peptide segments from the complex interface of Nac1 with its putative partners, and (iii) improve the peptide binding affinity to Nac1 POZ domain. In the procedure, Nac1 POZ dimerization with 136 human POZ domains was modeled, simulated and analyzed at atomic level to elucidate structural basis, energetic property and dynamics behavior. Two hotspot regions, namely α1-helix and α2/α3-hairpin, at the dimerization interface were identified that are responsible for stabilizing the formed POZ-POZ dimer complexes. The α1-helix and α2/α3-hairpin were stripped from the interface to derive their respective isolated SIP peptides, which, however, exhibited a large flexibility and intrinsic disorder in free state, and thus would incur a considerable penalty upon rebinding to Nac1 POZ domain. By carefully examining the natively folded structures of α1-helix and α2/α3-hairpin in protein context and their interaction modes with the domain, we rationally designed a hydrocarbon bridge and a disulfide bond separately for the two peptides in order to constrain their conformational flexibility in free state, thus largely minimizing the flexibility penalty. Consequently, three α1-helix peptides derived from Nac1, Miz1 and Slx4 were stapled by all-hydrocarbon bridge, while four α2/α3-hairpin peptides derived from Nac1, Bacd1, Klh28 and Mynn were cyclized by disulfide bond. Binding affinity analysis revealed that, as designed, these peptides were converted from non- or weak binders to moderate or good binders of Nac1 POZ domain upon the stapling and cyclization.


Subject(s)
Neoplasm Proteins/metabolism , Peptides/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , BTB-POZ Domain , Binding Sites , Cyclization , Dimerization , Humans , Molecular Dynamics Simulation , Neoplasm Proteins/chemistry , Peptides/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Repressor Proteins/chemistry , Thermodynamics
10.
Arch Biochem Biophys ; 606: 10-5, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27424155

ABSTRACT

Nucleus accumbens-associated protein 1 (NAC1) is a cancer-related transcription regulator protein that is also involved in the pluripotency and differentiation of embryonic stem cells. NAC1 is overexpressed in various carcinomas including ovarian, cervical, breast, and pancreatic carcinomas. NAC1 knock-down was previously shown to result in the apoptosis of ovarian cancer cell lines and to rescue their sensitivity to chemotherapy, suggesting that NAC1 may be a potential therapeutic target, but protein complex formation and the dynamics of intranuclear NAC1 in cancer cells remain poorly understood. In this study, analysis of HeLa cell lysates by fast protein liquid chromatography (FPLC) on a sizing column showed that the NAC1 peak corresponded to an apparent molecular mass of 300-500 kDa, which is larger than the estimated molecular mass (58 kDa) of the protein. Furthermore, live cell photobleaching analyses with green fluorescent protein (GFP)-fused NAC1 proteins revealed the intranuclear dynamics of NAC1. Collectively our results demonstrate that NAC1 forms a protein complex to function as a transcriptional regulator in cancer cells.


Subject(s)
Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatography, High Pressure Liquid , Diffusion , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/chemistry , HeLa Cells , Histones/chemistry , Humans , Kinetics , Molecular Weight , Protein Binding , Protein Domains , Protein Multimerization
11.
Biomedicines ; 11(8)2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37626718

ABSTRACT

Nucleus-accumbens-associated protein-1 (NAC1) is a cancer-related transcriptional factor encoded by the NACC1 gene, which is amplified and overexpressed in various human cancers and has been appreciated as one of the top potential cancer driver genes. NAC1 has therefore been explored as a potential therapeutic target for managing malignant tumors. Here, we show that NAC1 is a negative regulator of NF-κB signaling, and NAC1 depletion enhances the level of the nuclear NF-κB in human melanoma. Furthermore, the inhibition of NF-κB signaling significantly potentiates the antineoplastic activity of the NAC1 inhibition in both the cultured melanoma cells and xenograft tumors. This study identifies a novel NAC1-NF-κB signaling axis in melanoma, offering a promising new therapeutic option to treat melanoma.

12.
Mol Plant ; 16(4): 709-725, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36809880

ABSTRACT

Precise spatiotemporal control of the timing and extent of asymmetric cell divisions (ACDs) is essential for plant development. In the Arabidopsis root, ground tissue maturation involves an additional ACD of the endodermis that maintains the inner cell layer as the endodermis and generates the middle cortex to the outside. Through regulation of the cell cycle regulator CYCLIND6;1 (CYCD6;1), the transcription factors SCARECROW (SCR) and SHORT-ROOT (SHR) play critical roles in this process. In the present study, we found that loss of function of NAC1, a NAC transcription factor family gene, causes markedly increased periclinal cell divisions in the root endodermis. Importantly, NAC1 directly represses the transcription of CYCD6;1 by recruiting the co-repressor TOPLESS (TPL), creating a fine-tuned mechanism to maintain proper root ground tissue patterning by limiting production of middle cortex cells. Biochemical and genetic analyses further showed that NAC1 physically interacts with SCR and SHR to restrict excessive periclinal cell divisions in the endodermis during root middle cortex formation. Although NAC1-TPL is recruited to the CYCD6;1 promoter and represses its transcription in an SCR-dependent manner, NAC1 and SHR antagonize each other to regulate the expression of CYCD6;1. Collectively, our study provides mechanistic insights into how the NAC1-TPL module integrates with the master transcriptional regulators SCR and SHR to control root ground tissue patterning by fine-tuning spatiotemporal expression of CYCD6;1 in Arabidopsis.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Division , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation, Plant , Plant Roots/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Biochem Pharmacol ; 211: 115533, 2023 05.
Article in English | MEDLINE | ID: mdl-37019189

ABSTRACT

In this study, we uncovered the nuclear export of nucleus accumbens-associated protein-1 (NAC1) as a novel mechanism involved in ovarian cancer resistance to taxanes, the chemotherapeutic drugs commonly used in treatment of this malignancy. We showed that NAC1, a nuclear factor of the BTB/POZ gene family, has a nuclear export signal (NES) at the N terminus (aa 17-28), and this NES critically contributes to the NAC1 nuclear-cytoplasmic shuttling when tumor cells were treated with docetaxel. Mechanistically, the nuclear-exported NAC1 bound to cullin3 (Cul3) and Cyclin B1 via its BTB and BOZ domains respectively, and the cyto-NAC1-Cul3 E3 ubiquitin ligase complex promotes the ubiquitination and degradation of Cyclin B1, thereby facilitating mitotic exit and leading to cellular resistance to docetaxel. We also showed in in vitro and in vivo experiments that TP-CH-1178, a membrane-permeable polypeptide against the NAC1 NES motif, blocked the nuclear export of NAC1, interfered with the degradation of Cyclin B1 and sensitized ovarian cancer cells to docetaxel. This study not only reveals a novel mechanism by which the NAC1 nuclear export is regulated and Cyclin B1 degradation and mitotic exit are impacted by the NAC1-Cul3 complex, but also provides the nuclear-export pathway of NAC1 as a potential target for modulating taxanes resistance in ovarian cancer and other malignancies.


Subject(s)
Ovarian Neoplasms , Repressor Proteins , Humans , Female , Active Transport, Cell Nucleus , Docetaxel/pharmacology , Cyclin B1/metabolism , Repressor Proteins/metabolism , Ovarian Neoplasms/metabolism
14.
Front Mol Neurosci ; 16: 1115880, 2023.
Article in English | MEDLINE | ID: mdl-37533751

ABSTRACT

Advances in genome sequencing technologies have favored the identification of rare de novo mutations linked to neurological disorders in humans. Recently, a de novo autosomal dominant mutation in NACC1 was identified (NM_052876.3: c.892C > T, NP_443108.1; p.Arg298Trp), associated with severe neurological symptoms including intellectual disability, microcephaly, and epilepsy. As NACC1 had never before been associated with neurological diseases, we investigated how this mutation might lead to altered brain function. We examined neurotransmission in autaptic glutamatergic mouse neurons expressing the murine homolog of the human mutant NACC1, i.e., Nacc1-R284W. We observed that expression of Nacc1-R284W impaired glutamatergic neurotransmission in a cell-autonomous manner, likely through a dominant negative mechanism. Furthermore, by screening for Nacc1 interaction targets in the brain, we identified SynGAP1, GluK2A, and several SUMO E3 ligases as novel Nacc1 interaction partners. At a biochemical level, Nacc1-R284W exhibited reduced binding to SynGAP1 and GluK2A, and also showed greatly increased SUMOylation. Ablating the SUMOylation of Nacc1-R284W partially restored its interaction with SynGAP1 but did not restore binding to GluK2A. Overall, these data indicate a role for Nacc1 in regulating glutamatergic neurotransmission, which is substantially impaired by the expression of a disease-associated Nacc1 mutant. This study provides the first functional insights into potential deficits in neuronal function in patients expressing the de novo mutant NACC1 protein.

15.
Viruses ; 14(8)2022 08 04.
Article in English | MEDLINE | ID: mdl-36016335

ABSTRACT

Nucleus accumbens-associated protein 1 (NAC1) is a transcription co-factor that has been shown to possess multiple roles in stem cell and cancer biology. However, little is known about its roles in regulation of the immune system. In the current study, we observed that expression of NAC1 impacted the survival of CD8+ T cells in vitro. NAC1-/- CD8+ T cells displayed lower metabolism, including reduced glycolysis and oxidative phosphorylation. In vivo, compared with wild-type (WT) mice, NAC1-/- mice produced a lower response to vaccinia virus (VACV) infection, and viral antigen (Ag)-specific CD8+ T cells decreased more slowly. Additionally, we observed that the NAC1-/- mice demonstrated a stronger memory formation of viral Ag-specific CD8+ T cells post-viral infection. Mechanically, we identified that compared with WT CD8+ T cells, the Interferon Regulatory Factor 4 (IRF4), a key transcription factor in T cell development, was highly expressed in NAC1-/- CD8+ T cells, insinuating that IRF4 could be a critical regulatory target of NAC1 in the memory formation of CD8+ T cells. Our results indicate that NAC1 restrains the memory formation of CD8+ T cells by modulating IRF4, and targeting NAC1 may be exploited as a new approach to boosting CD8+ T cell memory.


Subject(s)
CD8-Positive T-Lymphocytes , Virus Diseases , Animals , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccinia virus , Virus Diseases/metabolism
16.
Plant Signal Behav ; 17(1): 2096780, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35822517

ABSTRACT

Lateral root (LR) formation is a vital organogenetic process that determines the root architecture in plants. The number of root branches governs the degree of anchorage, efficiency of nutrients acquisition, and water uptake. The molecular pathways involved in LR formation have been extensively studied in Arabidopsis thaliana (At). A plant hormone, Auxin, is a key regulator of root development and promotes LR formation in plants. A plethora of Arabidopsis genes have been identified to regulate LR initiation, patterning, and emergence processes. Recently, the involvement of flowering time control pathways and circadian clock pathways in LR development has come to light, but the connecting link between these processes is still missing. We have established that GIGANTEA (GI), a key component of photoperiodic flowering, can regulate the formation of LRs in Arabidopsis. GI is known to be involved in red light signaling and circadian clock signaling pathways. Here, we report that over-expression of GI enhances LR formation in red light in At. Real-time PCR analysis shows that GI positively regulates the transcription of TRANSPORT INHIBITOR RESPONSE 1 (TIR1) which is an upstream component of auxin signaling. Furthermore, gi-100 mutant downregulates the LR initiation signaling gene, AUXIN RESPONSE FACTOR 7 (ARF7), and its downstream target gene, LATERAL ORGAN BOUNDARIES-DOMAIN 16 (LBD16). Hence, GI acts as a positive regulator of IAA14-ARF7-LBD16 modules during LR initiation. We have also checked the effect of GI on the expression of NAC1 and AIR3 genes which are controlled by TIR1 during LR formation. Our results show that GI induces the NAC1 transcription and its downstream gene, AIR3 expression, which leads to the enhancement of LR initiation. Taken together, our results suggest that GI controls the expression of TIR1 to govern auxin signaling during LR formation in presence of red light and GI can act as a link between circadian clock signaling, flowering time control pathways, light signaling, and lateral root development pathways.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Gene Expression Regulation, Plant/genetics , Indoleacetic Acids/metabolism , Plant Roots/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Aging (Albany NY) ; 12(10): 9275-9291, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32412910

ABSTRACT

BACKGROUND: Nucleus accumbens-associated protein 1 (NAC1) has multifaceted roles in cancer pathogenesis and progression, including the development of drug resistance, promotion of cytokinesis, and maintenance of "stem cell-like" phenotypes. NAC1 is a transcriptional co-regulator belonging to the bric-a-brac tramtrack broad (BTB) family of proteins, although it lacks the characteristic DNA binding motif of the BTB family. The formation of higher-order transcription complexes likely depends on its interaction with other DNA-binding co-factors. RESULTS: NAC1 interacts with BCL6 via its C-terminal BEN domain and forms a complex that binds the promoter region and activates transcription of the NAC1 target gene, FOXQ1. NAC1 and BCL6 were coordinately upregulated. Our analysis also identified a novel function of NAC1 in attenuating BCL6 auto-downregulation in ovarian cancer. Lastly, we found a significant overlap among NAC1- and BCL6-regulated genes in tumor cells, suggesting that NAC1 and BCL6 coordinately control transcription in cancer. CONCLUSIONS: The results of this study provide a novel mechanistic insight into the oncogenic roles of NAC1 and underline the importance of developing the NAC1/BCL6-targeted cancer therapy. METHODS: Using the Cistrome database and Chromatin Immunoprecipitation (ChIP) analyses, we identified BCL6 as a potential NAC1- interacting molecule. Co-immunoprecipitation (Co-IP), luciferase reporter assay, immunohistochemistry and microarray analysis were performed to analyze the interaction between NAC1 and BCL6 and the mechanisms by which they regulate the downstream genes including FOXQ1.


Subject(s)
Forkhead Transcription Factors , Neoplasm Proteins , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-6 , Repressor Proteins , Cell Line, Tumor , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
Biomedicines ; 8(12)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327466

ABSTRACT

Nucleus accumbens-associated protein 1 (NAC1) is a nuclear protein that harbors an amino-terminal BTB domain and a carboxyl-terminal BEN domain. NAC1 appears to play significant and diverse functions in cancer and stem cell biology. Here we demonstrated that the BEN domain of NAC1 is a sequence-specific DNA-binding domain. We selected the palindromic 6 bp motif ACATGT as a target sequence by using a PCR-assisted random oligonucleotide selection approach. The interaction between NAC1 and target DNA was characterized by gel shift assays, pull-down assays, isothermal titration calorimetry (ITC), chromatin-immunoprecipitation assays, and NMR chemical shifts perturbation (CSP). The solution NMR structure revealed that the BEN domain of human NAC-1 is composed of five conserved α helices and two short ß sheets, with an additional hitherto unknown N-terminal α helix. In particular, ITC clarified that there are two sequential events in the titration of the BEN domain of NAC1 into the target DNA. The ITC results were further supported by CSP data and structure analyses. Furthermore, live cell photobleaching analyses revealed that the BEN domain of NAC1 alone was unable to interact with chromatin/other proteins in cells.

19.
Cancer Med ; 8(14): 6426-6436, 2019 10.
Article in English | MEDLINE | ID: mdl-31490008

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a new promising target for the treatment of ovarian cancer. Our previous study showed that cross-reacting material 197 (CRM197), a specific HB-EGF inhibitor, significantly reverses resistance against paclitaxel in paclitaxel-resistant ovarian cancer cells. However, the mechanism of the effect of CRM197 on the reversion of paclitaxel resistance was unclear. In this study, in vitro and in vivo data suggested that CRM197 treatment sensitized paclitaxel-resistant ovarian cancer cells to paclitaxel, at least in part, via nucleus accumbens-1 (NAC-1) and its downstream pathway, DNA damage-inducible 45-γ interacting protein (Gadd45gip1)/growth arrest and DNA damage-inducible 45 (Gadd45), in A2780/Taxol and SKOV3/Taxol cells. The results also showed that CRM197 activated the proapoptotic JNK/p38MAPK pathway to enhance caspase-3 activity and apoptosis by downregulation of the NAC-1/Gadd45gip1/Gadd45 pathway, leading to reversion of paclitaxel resistance in A2780/Taxol and SKOV3/Taxol cells. This study provides the first mechanism through which CRM197 significantly reverses resistance against paclitaxel by modulating the NAC-1/Gadd45gip1/Gadd45 pathway in paclitaxel-resistant ovarian cancer cells, and the mechanism of HB-EGF inhibition as a novel therapeutic strategy for patients with paclitaxel-resistant ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Proteins/pharmacology , Cell Cycle Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Repressor Proteins/metabolism , Signal Transduction/drug effects , Animals , Caspase 3/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Silencing , Humans , MAP Kinase Signaling System , Mice , Models, Biological , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , RNA, Small Interfering , Xenograft Model Antitumor Assays
20.
Ann Clin Lab Sci ; 48(5): 559-564, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30373858

ABSTRACT

OBJECTIVE: This study aims to investigate the role of NAC1/HMGB1 signaling pathway in the epithelial-mesenchymal transition (EMT), invasion, and metastasis of lung cancer cell line. METHODS: Human lung cancer cell line A549 was used in this study. They were randomly divided into normal control group, sh-NAC1 empty vector group (sh-NAC1 NC), expression empty vector group (NAC1 NC), NAC1-shRNA and NAC1 over-expression group (NAC1). NAC1 and HMGB1 expression levels were detected by qRT-PCR method. Cell proliferation was detected by CCK8 kit. Cell cycles were detected by flow cytometry method. Cell invasion was detected by Transwell method. The expression levels of E-cadherin, N-cadherin, NAC1, and HMGB1 were detected by qRT-PCR and Western blotting methods. RESULTS: Compared with the control group, the expression level of NAC1 and cell proliferation in NAC1-shRNA group decreased, cells in G1 phase increased and cells in S phase decreased. In NAC1-shRNA group, E-cadherin expression levels increased and the expression levels of N-cadherin, HMGB1 and Vimentin decreased. In NAC1 group, the expression level of NAC1 and cell proliferation increased, cells in S phase increased and cells in G1 phase decreased, E-cadherin expression levels decreased and the expression levels of N-cadherin, HMGB1 and Vimentin increased. All these differences are statistically significant. CONCLUSIONS: The expression of NAC1 and HMGB1 in lung cancer cells may affect the occurrence of EMT, the NAC1/ HMGB1 signaling pathway is associated with the EMT, invasion, and metastasis of lung cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , HMGB1 Protein/physiology , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Repressor Proteins/physiology , Signal Transduction/physiology , Antigens, CD/analysis , Cadherins/analysis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/analysis , Repressor Proteins/analysis , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL