Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 22(2): 229-241, 2023 01.
Article in English | MEDLINE | ID: mdl-35980125

ABSTRACT

This study aimed to investigate the effects of scaffold matrix attachment region binding protein 1 (SMAR1) on the development of bladder cancer (BCa). SMAR1 expression in paired tumor and corresponding adjacent normal tissues from 55 BCa patients was detected by quantitative reverse transcription-polymerase chain reaction. BCa cells were transfected to regulate SMAR1 expression. BCa cells were treated with XAV-939, LiCl and 2-deoxyglucose. The effect of SMAR1 on the viability, proliferation, migration, invasion and Warburg effect of BCa cells was researched by counting kit-8, colony formation assay, Transwell and aerobic glycolysis assays. Western blot was performed to detect protein expression. BCa cell growth in vivo was recorded in nude mice. Immunohistochemical staining was performed for clinical and xenografted tumor tissue specimens. SMAR1 expression was down-regulated in BCa patients, associating with worse prognoses. SMAR1 knockdown enhanced the viability, proliferation, migration, invasion, EMT and Warburg effect of BCa cells. The opposite effect was found in the SMAR1 overexpression BCa cells. XAV-939 treatment reversed the elevation of ß-catenin, c-Myc and Cyclin D1 proteins expression and Warburg effect in Bca cells post-SMAR1 knockdown. LiCl treatment abrogated the inhibition of ß-catenin, c-Myc and Cyclin D1 proteins expression and Warburg effect proteins due to SMAR1 overexpression in BCa cells. SMAR1 overexpression inhibited the growth of BCa cells in vivo. SMAR1 might suppress the Wnt/ß-catenin signaling pathway activity to inhibit the progression of BCa. It might be an effective treatment target for BCa.


Subject(s)
Urinary Bladder Neoplasms , Wnt Signaling Pathway , Animals , Mice , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Cyclin D1/metabolism , Mice, Nude , Urinary Bladder Neoplasms/pathology , Cell Proliferation/physiology , Cell Line, Tumor , Cell Movement
2.
Mol Oncol ; 16(7): 1523-1540, 2022 04.
Article in English | MEDLINE | ID: mdl-34689394

ABSTRACT

TOP1-binding arginine/serine-rich protein (TOPORS), a really interesting new gene finger protein, has the ability to bind to a palindromic consensus DNA sequence that enables it to function as a potential transcriptional regulator. However, its role in regulating the transcription of cancer-associated genes is yet to be explored. As Toll-like receptor 4 (TLR4) agonists are known to regress solid tumors, we observed that lipopolysaccharide (LPS) induces TOPORS via a TLR4-TIR domain-containing adapter-inducing interferon-ß-dependent pathway, which in turn modulates the transcription of tumor suppressor scaffold/matrix attachment region-binding protein 1 (SMAR1, also known as BANP). ChIP analysis showed that TOPORS binds on the SMAR1 promoter and its occupancy increases upon LPS treatment. A previous study from our laboratory revealed that SMAR1 acts as a repressor of signal transducer and activator of transcription 3 (STAT3) transcription. Tumor growth, as well as tumor-associated macrophage polarization, depends on the status of the STAT1:STAT3 ratio. LPS-induced SMAR1 expression decreases STAT3 expression and also skews the macrophage polarization toward M1 phenotype. In contrast, LPS failed to polarize tumor-associated macrophages to M1 phenotype in a SMAR1-silenced condition, which shows the involvement of SMAR1 in dictating the fate of colorectal cancer progression. Identification of the molecular mechanism behind LPS-mediated tumor regression would be crucial for designing cancer treatment strategies involving bacterial components.


Subject(s)
Colorectal Neoplasms , Toll-Like Receptor 4 , Adaptor Proteins, Vesicular Transport/metabolism , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
3.
Int J Biochem Cell Biol ; 141: 106085, 2021 12.
Article in English | MEDLINE | ID: mdl-34551340

ABSTRACT

One of the hallmarks of a cancer cell is the ability for indefinite proliferation leading to the immortalization of the cell. Activation of several signaling pathways leads to the immortalization of cancer cells via the reactivation of enzyme telomerase (hTERT). hTERT is active in germ cells, stem cells and also cancer cells. An earlier report from our lab suggests that SMAR1, a tumor suppressor protein, is significantly downregulated in the higher grades of colorectal cancers. Our study identifies SMAR1 as a transcriptional repressor of hTERT. We find that SMAR1 interacts with HDAC1/mSin3a co-repressor complex at the hTERT promoter and brings about HDAC1-mediated transcriptional repression of the promoter. Most solid tumors including colorectal cancer reactivate hTERT expression as it confers several advantages to the cancer cells like increased proliferation and angiogenesis. One of these non-canonical functions of hTERT is inducing the pool of cancer stem cell population. We find that in the CD133HighCD44High cancer stem cells population, SMAR1 expression is highly diminished leading to elevated hTERT expression. We also find that knockdown of SMAR1 promotes total CD133+CD44+ population and impart enhanced sphere-forming ability to the colorectal cancer cells. SMAR1 also inhibits invasion and metastasis in colorectal cancer cell lines via repression of hTERT. Our study provides evidence that downregulation of SMAR1 causes activation of hTERT leading to an increase in the cancer stem cell phenotype in colorectal cancer cells.


Subject(s)
Cell Cycle Proteins , Neoplastic Stem Cells , DNA-Binding Proteins
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(12): 159045, 2021 12.
Article in English | MEDLINE | ID: mdl-34450266

ABSTRACT

Adipogenesis is described as the process of conversion of pre-adipocytes into differentiated lipid-laden adipocytes. Adipogenesis is known to be regulated by a myriad of transcription factors and co-regulators. However, there is a dearth of information regarding the mechanisms that regulate these transcription factors and hence control adipogenesis. PPARγ is the master transcriptional regulator of adipogenesis and its expression is essential for adipocyte differentiation. Herein, we identified that scaffold/matrix attachment region-binding protein 1 (SMAR1) negatively regulates adipogenesis. We observed that SMAR1 gets downregulated during adipocyte differentiation and knockdown of SMAR1 promotes lipid accumulation and adipocyte differentiation. Mechanistically, we have shown that SMAR1 suppresses PPARγ through recruitment of the HDAC1/mSin3a repressor complex to the PPARγ promoter. We further identified cell division cycle 20 (cdc20) mediated proteasomal degradation of SMAR1 during adipogenesis. Moreover, knockdown of cdc20 resulted in stabilization of SMAR1 and a reduction in adipocyte differentiation. Taken together, our observations suggest that SMAR1 functions as a negative regulator of adipogenesis by inhibiting PPARγ expression in differentiating adipocytes.


Subject(s)
Adipogenesis/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Histone Deacetylase 1/genetics , Nuclear Proteins/genetics , PPAR gamma/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cdc20 Proteins/genetics , Cell Differentiation/genetics , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Developmental/genetics , Lipid Metabolism/genetics , Mice
5.
Cancer Metab ; 9(1): 16, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33863392

ABSTRACT

BACKGROUND: Highly proliferating cancer cells exhibit the Warburg effect by regulation of PKM alternative splicing and promoting the expression of PKM2. Majority of the alternative splicing events are known to occur in the nuclear matrix where various MARBPs actively participate in the alternative splicing events. SMAR1, being a MARBP and an important tumor suppressor, is known to regulate the splicing of various cancer-associated genes. This study focuses on the regulation of PKM alternative splicing and inhibition of the Warburg effect by SMAR1. METHODS: Immunohistochemistry was performed in breast cancer patient samples to establish the correlation between SMAR1 and PKM isoform expression. Further, expression of PKM isoforms upon modulation in SMAR1 expression in breast cancer cell lines was quantified by qRT-PCR and western blot. The acetylation status of PTBP1 was estimated by immunoprecipitation along with its enrichment on PKM pre-mRNA by CLIP in SMAR1 knockdown conditions. The role of SMAR1 in tumor metabolism and tumorigenesis was explored by in vitro enzymatic assays and functional assays upon SMAR1 knockdown. Besides, in vivo tumor formation by injecting adeno-SMAR1-transduced MDA-MB-231 cells in NOD/SCID mice was performed. RESULTS: The expression profile of SMAR1 and PKM isoforms in breast cancer patients revealed that SMAR1 has an inverse correlation with PKM2 and a positive correlation with PKM1. Further quantitative PKM isoform expression upon modulation in SMAR1 expression also reflects that SMAR1 promotes the expression of PKM1 over tumorigenic isoform PKM2. SMAR1 deacetylates PTBP1 via recruitment of HDAC6 resulting in reduced enrichment of PTBP1 on PKM pre-mRNA. SMAR1 inhibits the Warburg effect, tumorigenic potential of cancer cells, and in vivo tumor generation in a PKM2-dependent manner. CONCLUSIONS: SMAR1 regulates PKM alternative splicing by causing HDAC6-dependent deacetylation of PTBP1, resulting in reduced enrichment of PTBP1 on PKM pre-mRNA. Additionally, SMAR1 suppresses glucose utilization and lactate production via repression of PKM2 expression. This suggests that tumor suppressor SMAR1 inhibits tumor cell metabolism and tumorigenic properties of cancer cells via regulation of PKM alternative splicing.

6.
Environ Toxicol ; 36(6): 1090-1098, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33543840

ABSTRACT

The promoting roles of the transcriptional regulator SMAR1 have been revealed in several tumors, such as colorectal and breast cancer, however, its roles in osteosarcoma (OS) progression are still confusing. Here, we find that SMAR1 expression is positively correlated with the overall survival of OS patients and negatively correlated with the expression of stemness markers by analyzing the online datasets. Through analyzing different Gene Expression Omnibus (GEO) datasets, SMAR1 is found to be lowly expressed in OS tissues relative to that in adjacent tissues. Functional experiments indicate that SMAR1 overexpression attenuates the stemness of OS cells, characterized as the decrease of stemness marker expression, sphere-formation ability and ALDH activity. Mechanistically, it is shown that SMAR1 increases the deacetylation level of the drug efflux pump ABCG2 via recruiting HDAC2 to the promoter of the gene coding ABCG2, and thus decreases ABCG2 transcriptional activity. Additionally, overexpression of ABCG2 rescues the inhibition of SMAR1 overexpression on the stemness of OS cells. Moreover, this SMAR1/ABCG2 axis positively regulates the chemotherapeutic sensitivity of OS cells. This work indicates that SMAR1 is a critical suppressor for OS progression through transcriptionally regulating ABCG2 expression.


Subject(s)
Bone Neoplasms , Osteosarcoma , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/genetics , Osteosarcoma/genetics
7.
Biochem Biophys Res Commun ; 526(4): 1157-1163, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32321642

ABSTRACT

The pathogenesis of inflammation bowel disease (IBD) involves exaggerated effector T cell responses and impaired regulatory T cell functions. We previously found that sauchinone (SAU) ameliorated experimental colitis via facilitating Th17 cell production of IL-10, but how SAU regulated Th17 cell differentiation remains unknown. MicroRNAs (miR) have been recognized as a crucial regulator of T cell biology and play a considerable role in IBD. Here, we demonstrated that SAU significantly suppressed miR-340 expression in Th17 cells, and enforced miR-340 expression abrogated SAU inhibition of Th17 differentiation. miR-340 itself was found to facilitate Th17 differentiation, especially the pathogenic "Th1-like" subset. In human IBD, miR-340 was intimately correlated with the disease severity. SAU markedly decreased miR-340 in the inflamed mucosa tissues from IBD patients. Scaffold/matrix-associated region-binding protein 1 (SMAR1) was identified as a target gene of miR-340. We revealed that blockade of miR-340 significantly reduced mucosal damage and Th17 responses in the lamina propria in a mouse colitis model. Our findings suggest that miR-340 negatively affects SAU inhibition of Th17 differentiation and might play a crucial role in the regulation of pathogenic "Th1-like" Th17 cell generation, which might serve as a novel therapeutic target of IBD.


Subject(s)
Benzopyrans/pharmacology , Cell Differentiation/drug effects , Dioxoles/pharmacology , Inflammation/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Intestines/pathology , MicroRNAs/metabolism , Th17 Cells/pathology , Base Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Susceptibility , Down-Regulation/drug effects , Forkhead Transcription Factors/metabolism , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Severity of Illness Index , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Trinitrobenzenesulfonic Acid , Up-Regulation/drug effects
8.
Antiviral Res ; 173: 104648, 2020 01.
Article in English | MEDLINE | ID: mdl-31706900

ABSTRACT

The essential role of SMAR1 in HIV-1 transcription and LTR driven gene expression suggests SMAR1 as an HIV dependency factor (HDF) and a potential anti-HIV therapeutic target. Here, we report for the first time, anti-HIV activity of 8 novel isothiocyanate (ITC) derivatives that differentially stabilise SMAR1. Out of 8 novel ITC derivatives, SCS-OCL-381 was observed to inhibit HIV-1 replication most significantly at the noncytotoxic concentration in reporter T-cell line, CEM-GFP. Further, the highly conserved anti-HIV activity of SCS-OCL-381 is a cell type, virus isolate and viral load independent phenomena and is approximately 3 fold more effective than the representative ITC, Sulforaphane (SFN). Further, SCS-OCL-381 does not hamper the activity of viral enzymes reverse transcriptase, integrase and protease. Mechanistically, SCS-OCL-381 stabilises SMAR1 which, otherwise undergoes proteasomal degradation upon HIV-1 infection in T-cells. This stabilisation results in the recruitment of repressor complex on HIV-1 LTR resulting in repression of LTR mediated transcription and gene expression. These inhibitory consequences were further confirmed by reporter based LTR activity assays in different cell lines. Taken together, these findings highlight the anti-HIV potential of novel ITC derivatives by the stabilisation of SMAR1 and strongly support further in vivo characterisation and potential translational applications of SCS-OCL-381.


Subject(s)
Anti-HIV Agents/pharmacology , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral/drug effects , HIV Infections/metabolism , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , Isothiocyanates/pharmacology , Nuclear Proteins/metabolism , Virus Replication/drug effects , Anti-HIV Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , HIV Long Terminal Repeat , HIV-1/isolation & purification , Host-Pathogen Interactions , Humans , Isothiocyanates/chemistry , Molecular Structure , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic , Viral Load
9.
Cell Mol Biol (Noisy-le-grand) ; 64(12): 70-75, 2018 Sep 30.
Article in English | MEDLINE | ID: mdl-30301506

ABSTRACT

This study was aimed at investigating the specific molecular mechanisms involved in the regulation of immune escape of triple-negative breast cancer (TNBC) by SMRI, so as to provide a new clinical treatment target for the disease. Mouse original 4T1 breast cancer cells were inoculated subcutaneously in BALB/C to establish TNBC mouse model. CD8+T cells with immunological effects were selected from mouse thymus glands for primary culture. The CD8+positive T cells were infected with lentivirus interference vectors, and the proliferation of CD8+ T cells were determined by trypan blue staining and flow cytometry. CD8+T cells and 4T1 cells were cultured together so as to determine the cytotoxic effects of SMAR1-downregulated CD8+ T cells on tumor cells and the expression of cytokines (IFN-γ, TNF-α, IL-2, IL-4 and IL-6). The expressions of SMAR1, T-bet and PD-1 were assayed by Western blot. SMAR1-downregulated CD8+T cells were injected into 4T1 tumor-bearing mice through the caudal vein, and the growth of tumor in mice was monitored. Following the infection of CD8+T cells with SMAR1-downregulated lentiviral system, cell apoptosis level was decreased significantly (control vs. sh-SMAR1: 32.23 ± 12.4 % vs. 18.28 ± 8.93 %, p < 0.05). Results from trypan blue staining experiments showed that the proliferation of CD8+ T cells in the SMAR1-downregulated group was significantly increased; SMAR1-downregulated CD8+ T cells promoted the production of IFN-γ, TNF-α, IL-2, IL-4 and IL-6 in 4T1 breast cancer cells (p < 0.05). Western blot showed that SMAR1 down-regulation led to significant upregulation of T-bet, while PD-1 was downregulated, when compared to the control group (p < 0.05). The downregulation of SMAR1 was associated with significant reduction in tumor size in mice (p < 0.05). SMAR1 downregulation enhances the tumor killing effect of CD8+T cells by activating T-bet and down-regulating PD-1.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Box Domain Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred BALB C , Nuclear Proteins/genetics , Programmed Cell Death 1 Receptor/genetics , T-Box Domain Proteins/genetics , Triple Negative Breast Neoplasms/genetics
10.
Oncotarget ; 9(30): 21322-21336, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29765542

ABSTRACT

Reduced expression of Scaffold/Matrix Attachment Region Binding Protein 1 (SMAR1) is associated with various cancers resulting in poor prognosis of the diseases. However, the precise underlying mechanism elucidating the loss of SMAR1 requires ongoing study. Here, we show that SMAR1 is highly downregulated during aberrant Wnt3a signaling due to proteasomal degradation and predicted poor prognosis of colorectal cancer. However, substitution mutation (Arginine and Lysine to Alanine) in the D-box elements of SMAR1 viz. "RCHL" and "RQRL" completely abrogated its proteasomal degradation despite Wnt3a activity. SMAR1 inhibited Wnt/ß-catenin signaling by recruiting Histone deacetylase-5 to ß-catenin promoter resulting in reduced cell migration and invasion. Consequently, reduced tumor sizes in in-vivo NOD-SCID mice were observed that strongly associated with suppression of ß-catenin. However, loss of SMAR1 led to enriched H3K9 Acetylation in the ß-catenin promoter that further increased Wnt/ß-catenin signaling activities and enhanced colorectal cancer progression drastically. Using docking and isothermal titration calorimetric studies we show that small microbial peptides viz. AT-01C and AT-01D derived from Mycobacterium tuberculosis mask the D-box elements of SMAR1. These peptides stabilized SMAR1 expression that further inhibited metastatic SW480 colorectal cancer cell migration and invasion. Drastically reduced subcutaneous tumors were observed in in-vivo NOD-SCID mice upon administration of these peptides (25 mg/kg body weight) intraperitoneally. Taken together our structural studies, in-vitro and in-vivo results strongly suggest that the D-box elements of SMAR1 represent novel druggable targets, where the microbial peptides hold promise as novel colorectal cancer therapeutics.

11.
Nanomedicine (Lond) ; 13(4): 353-372, 2018 02.
Article in English | MEDLINE | ID: mdl-29338617

ABSTRACT

AIM: To investigate anticancer activity of the DNA binding domain of SMAR1 (His 5) in vitro and in vivo. MATERIALS & METHODS: His 5 was conjugated to hydrothermally synthesized carbon nanospheres (CNs). Anticancer activity of CNs-His 5 was evaluated in vitro and in vivo. RESULTS: CNs- His 5 significantly reduced cyclin D1 levels in MDA-MB-231 cells. Tumor bearing Balb/c mice injected with CNs-His 5 showed approximately 62% tumor regression and significantly reduced 18FDG uptake. Caspases assay and IHC staining confirmed tumor growth inhibition, which could be attributed to apoptotic, antiproliferative and antiangiogenic activities of His 5. CONCLUSION: DNA binding domain of the SMAR1 protein (His 5) has potent anticancer activity and its CNs mediated delivery could control breast tumor in mice model.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Carbon/chemistry , Cell Cycle Proteins/administration & dosage , DNA-Binding Proteins/administration & dosage , Drug Carriers/chemistry , Nanospheres/chemistry , Nuclear Proteins/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/metabolism , DNA-Binding Proteins/metabolism , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Nuclear Proteins/metabolism , Protein Domains , Recombinant Proteins/administration & dosage , Tissue Distribution
12.
Front Immunol ; 8: 72, 2017.
Article in English | MEDLINE | ID: mdl-28232831

ABSTRACT

T cell differentiation from naïve T cells to specialized effector subsets of mature cells is determined by the iterative action of transcription factors. At each stage of specific T cell lineage differentiation, transcription factor interacts not only with nuclear proteins such as histone and histone modifiers but also with other factors that are bound to the chromatin and play a critical role in gene expression. In this review, we focus on one of such nuclear protein known as tumor suppressor and scaffold matrix attachment region-binding protein 1 (SMAR1) in CD4+ T cell differentiation. SMAR1 facilitates Th1 differentiation by negatively regulating T-bet expression via recruiting HDAC1-SMRT complex to its gene promoter. In contrast, regulatory T (Treg) cell functions are dependent on inhibition of Th17-specific genes mainly IL-17 and STAT3 by SMAR1. Here, we discussed a critical role of chromatin remodeling protein SMAR1 in maintaining a fine-tuned balance between effector CD4+ T cells and Treg cells by influencing the transcription factors during allergic and autoimmune inflammatory diseases.

13.
Mol Cell Oncol ; 3(1): e1039689, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27308557

ABSTRACT

p53 and its isoforms are integral in modulating transcriptional gene expression programs and maintaining cellular homeostasis. We recently reported that glucose deprivation/caloric restriction induced translational control of p53 mRNA by scaffold/matrix attachment region binding-protein 1 (SMAR1), adding a cytoplasmic role of SMAR1 to its traditional nuclear role as a transcription factor.

14.
Int J Nanomedicine ; 11: 2039-51, 2016.
Article in English | MEDLINE | ID: mdl-27274234

ABSTRACT

Owing to the suppression of immune responses and associated side effects, steroid based treatments for inflammatory encephalitis disease can be detrimental. Here, we demonstrate a novel carbon nanosphere (CNP) based treatment regime for encephalomyelitis in mice by exploiting the functional property of the nuclear matrix binding protein SMAR1. A truncated part of SMAR1 ie, the DNA binding domain was conjugated with hydrothermally synthesized CNPs. When administered intravenously, the conjugate suppressed experimental animal encephalomyelitis in T cell specific conditional SMAR1 knockout mice (SMAR(-/-)). Further, CNP-SMAR1 conjugate delayed the onset of the disease and reduced the demyelination significantly. There was a significant decrease in the production of IL-17 after re-stimulation with MOG. Altogether, our findings suggest a potential carbon nanomaterial based therapeutic intervention to combat Th17 mediated autoimmune diseases including experimental autoimmune encephalomyelitis.


Subject(s)
Carbon/chemistry , Cell Cycle Proteins/administration & dosage , Cell Cycle Proteins/therapeutic use , DNA-Binding Proteins/administration & dosage , DNA-Binding Proteins/therapeutic use , Drug Delivery Systems/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Nanospheres/chemistry , Nuclear Proteins/administration & dosage , Nuclear Proteins/therapeutic use , Animals , Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/pathology , Endocytosis/drug effects , Interleukin-6/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nanospheres/toxicity , Nanospheres/ultrastructure , STAT3 Transcription Factor/metabolism , Th17 Cells/cytology , Th17 Cells/drug effects
15.
Biochem Biophys Res Commun ; 464(2): 647-53, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26168735

ABSTRACT

Treg cells are not only crucial for controlling immune responses to autoantigens but also prevent those directed towards commensal pathogens. Control of effector immune responses by Treg cells depend on their capacity to accumulate at inflammatory site and accordingly accommodate to inflammatory environment. Till date, the factors associated with maintaining these aspects of Treg phenotype is not understood properly. Here we have shown that a known nuclear matrix binding protein SMAR1 is selectively expressed more in colonic Treg cells and is required for their ability to accumulate at inflammatory site and to sustain high levels of Foxp3 and IL-10 expression during acute colitis. Elimination of anti-inflammatory subsets revealed a protective role for IL-10 producing Treg cells in SMAR1(-/-) mice. Moreover, a combined action of Foxp3 and SMAR1 restricts effector cytokine production and enhance the production of IL-10 by colonic Treg cells that controls acute colitis. This data highlights a critical role of SMAR1 in maintaining Treg physiology during inflammatory disorders.


Subject(s)
Cell Cycle Proteins/physiology , Colitis/immunology , DNA-Binding Proteins/physiology , Interleukin-10/physiology , Nuclear Proteins/physiology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , T-Lymphocytes, Regulatory/pathology
16.
Proc Natl Acad Sci U S A ; 112(26): E3374-83, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26080397

ABSTRACT

Pre-mRNA splicing is a complex regulatory nexus modulated by various trans-factors and their posttranslational modifications to create a dynamic transcriptome through alternative splicing. Signal-induced phosphorylation and dephosphorylation of trans-factors are known to regulate alternative splicing. However, the role of other posttranslational modifications, such as deacetylation/acetylation, methylation, and ubiquitination, that could modulate alternative splicing in either a signal-dependent or -independent manner remain enigmatic. Here, we demonstrate that Scaffold/matrix-associated region-binding protein 1 (SMAR1) negatively regulates alternative splicing through histone deacetylase 6 (HDAC6)-mediated deacetylation of RNA-binding protein Sam68 (Src-associated substrate during mitosis of 68 kDa). SMAR1 is enriched in nuclear splicing speckles and associates with the snRNAs that are involved in splice site recognition. ERK-MAPK pathway that regulates alternative splicing facilitates ERK-1/2-mediated phosphorylation of SMAR1 at threonines 345 and 360 and localizes SMAR1 to the cytoplasm, preventing its interaction with Sam68. We showed that endogenously, SMAR1 through HDAC6 maintains Sam68 in a deacetylated state. However, knockdown or ERK-mediated phosphorylation of SMAR1 releases the inhibitory SMAR1-HDAC6-Sam68 complex, facilitating Sam68 acetylation and alternative splicing. Furthermore, loss of heterozygosity at the Chr.16q24.3 locus in breast cancer cells, wherein the human homolog of SMAR1 (BANP) has been mapped, enhances Sam68 acetylation and CD44 variant exon inclusion. In addition, tail-vein injections in mice with human breast cancer MCF-7 cells depleted for SMAR1 showed increased CD44 variant exon inclusion and concomitant metastatic propensity, confirming the functional role of SMAR1 in regulation of alternative splicing. Thus, our results reveal the complex molecular mechanism underlying SMAR1-mediated signal-dependent and -independent regulation of alternative splicing via Sam68 deacetylation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alternative Splicing/physiology , Cell Cycle Proteins/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Histone Deacetylases/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins/physiology , RNA-Binding Proteins/metabolism , Acetylation , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Histone Deacetylase 6 , Humans , Hyaluronan Receptors/genetics , MAP Kinase Signaling System , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Transport
17.
Int J Biochem Cell Biol ; 55: 220-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25239884

ABSTRACT

Interleukin-8 (IL-8) is a pleiotropic chemokine involved in metastasis and angiogenesis of breast tumors. The expression of IL-8 is deregulated in metastatic breast carcinomas owing to aberrant NF-κB activity, which is known to positively regulate IL-8 transcription. Earlier, we have shown that tumor suppressor SMAR1 suppresses NF-κB transcriptional activity by modulating IκBα function. Here, we show that NF-κB target gene IL-8, is a direct transcriptional target of SMAR1. Using chromatin immunoprecipitation and reporter assays, we demonstrate that SMAR1 binds to IL-8 promoter MAR (matrix attachment region) and recruits HDAC1 dependent co-repressor complex. Further, we also show that SMAR1 antagonizes p300-mediated acetylation of RelA/p65, a post-translational modification indispensable for IL-8 transactivation. Thus, we decipher a new role of SMAR1 in NF-κB dependent transcriptional regulation of pro-angiogenic chemokine IL-8.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Interleukin-8/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Acetylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Histone Deacetylase 1/metabolism , Humans , Immunoblotting , MCF-7 Cells , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/metabolism , Transcription, Genetic
18.
Int J Biochem Cell Biol ; 45(11): 2519-29, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23876508

ABSTRACT

Erythropoiesis is controlled by a complex interplay of several signaling pathways and key transcription factors, as well as microRNAs (miRNAs). MicroRNAs function as critical modulators of gene expression for cellular processes. In the present study, we found that miR-320a inhibits erythroid differentiation by targeting Matrix Attachment Region binding protein SMAR1. miR-320a negatively regulates the expression of SMAR1 by directly binding to its 3'UTR. In response to mild DNA damage, miR-320a expression is decreased resulting in enhanced expression of SMAR1 protein, which in turn, reduces its targets, Bax and Puma inhibiting apoptosis. Our data demonstrate that during hemin-induced erythroid differentiation, enhanced expression of SMAR1 negatively correlates with miR-320a expression. Further analysis reveals that SMAR1 regulates erythroid differentiation, by binding to the promoter of miR-221/222, which play a crucial role in early erythropoiesis. Overall, our studies provide an insight into the regulation of hemin mediated erythroid differentiation of K562 cells through post-transcriptional regulation of SMAR1.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Matrix Attachment Regions/genetics , MicroRNAs/metabolism , Nuclear Proteins/metabolism , 3' Untranslated Regions/genetics , Apoptosis/genetics , Base Sequence , Binding Sites , Cell Differentiation/genetics , Cell Line , Computer Simulation , DNA Damage/genetics , Gene Expression Regulation , Humans , MicroRNAs/genetics , Molecular Sequence Data , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL