Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biomaterials ; 311: 122688, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38943821

ABSTRACT

CD40 agonist antibodies (αCD40) have shown promising anti-tumor response in both preclinical and early clinical studies. However, its systemic administration is associated with immune- and hepato-toxicities which hampers its clinical usage. In addition, αCD40 showed low tumor retention and induced PD-L1 expression which makes tumor microenvironment (TME) immunosuppressive. To overcome these issues, in this study, we have developed a multifunctional Immunosome where αCD40 is conjugated on the surface and RRX-001, a small molecule immunomodulator was encapsulated inside it. Immunosomes showed higher tumor accumulation till 96 h of administration and displayed sustained release of αCD40 in vivo. Immunosomes significantly delayed tumor growth and showed tumor free survival in mice bearing GL-261 glioblastoma by increasing the population of CD45+CD8+ T cells, CD45+CD20+ B cells, CD45+CD11c+ DCs and F4/80+CD86+ cells in TME. Immunosome significantly reduced the population of T-regulatory cells, M2 macrophage, and MDSCs and lowered the PD-L1 expression. Moreover, Immunosomes significantly enhanced the levels of Th1 cytokines (IFN-γ, IL-6, IL-2) over Th2 cytokines (IL-4 and IL-10) which supported anti-tumor response. Most interestingly, Immunosomes averted the in vivo toxicities associated with free αCD40 by lowering the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), IL-6, IL-1α and reduced the degree of liver damage. In addition, Immunosomes treated long-term surviving mice showed tumor specific immune memory response which prevented tumor growth upon rechallenge. Our results suggested that this novel formulation can be further explored in clinics to improve in vivo anti-tumor efficacy of αCD40 with long-lasting tumor specific immunity while reducing the associated toxicities.

2.
ACS Biomater Sci Eng ; 9(11): 6409-6424, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37870457

ABSTRACT

Dendritic cell-derived exosomes (Dex) have overcome the disadvantages associated with dendritic cell (DC) vaccines, such as cost effectiveness, stability, and sensitivity to the systemic microenvironment. However, in clinical trials, Dex failed to provide satisfactory results because of many reasons, including inadequate maturation of DC as well as the immunosuppressive tumor microenvironment (TME). Hence, culturing DCs in the presence of a maturation cocktail showed an induced expression of MHCs and co-stimulatory molecules. Additionally, targeting the colony stimulating factor-1 (CSF-1)/CSF-1 receptor (CSF-1R) signaling pathway by a CSF-1R inhibitor could deplete tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) which are responsible for immunosuppressive TME. Hence, in this study, mDexTA were isolated from bone marrow-derived DC cultured in the presence of a novel maturation cocktail and tumor antigen. mDexTA showed elevated expression of major histocompatibility complexes (MHCs) and co-stimulatory molecules and was found capable of activating naïve DC and T cells in vitro more efficiently when compared to imDexTA isolated from immature DCs. In addition, PLX-3397, a small molecule inhibitor of CSF-1/CSF-1R, was used in combination to enhance the antitumor efficacy of mDexTA. PLX-3397 showed dose-dependent toxicity against bone marrow-derived macrophages (BMDMs). In the B16-F10 murine melanoma model, we found that the combination treatment delayed tumor growth and improved survival compared to the mice treated with mDexTA alone by enhancing the CD8 T cells infiltration in TME. mDexTA when combined with PLX-3397 modulated the TME by shifting the Th1/Th2 toward a dominant Th1 population and depleting the TAMs and MDSCs. Interestingly, PLX-3397-induced FoxP3 expression was diminished when it was used in combination with mDexTA. Combination treatment also induced favorable systemic antitumor immunity in the spleen and lymph node. In conclusion, our findings provide insights into the synergy between mDexTA-based immunotherapy and PLX-3397 as the combination overcame the disadvantages associated with monotherapy and offer a therapeutic strategy for the treatment of solid tumors including melanoma.


Subject(s)
Exosomes , Melanoma , Mice , Animals , Macrophage Colony-Stimulating Factor/pharmacology , Tumor Microenvironment , Antigens, Neoplasm , Dendritic Cells
3.
Br J Cancer ; 129(4): 586-600, 2023 09.
Article in English | MEDLINE | ID: mdl-37400677

ABSTRACT

BACKGROUND: Dysregulation of histone deacetylases has been linked to diverse cancers. HDAC5 is a histone deacetylase belonging to Class IIa family of histone deacetylases. Limited substrate repertoire restricts the understanding of molecular mechanisms underlying its role in tumorigenesis. METHODS: We employed a biochemical screen to identify SATB1 as HDAC5-interacting protein. Coimmunoprecipitation and deacetylation assay were performed to validate SATB1 as a HDAC5 substrate. Proliferation, migration assay and xenograft studies were performed to determine the effect of HDAC5-SATB1 interaction on tumorigenesis. RESULTS: Here we report that HDAC5 binds to and deacetylates SATB1 at the conserved lysine 411 residue. Furthermore, dynamic regulation of acetylation at this site is determined by TIP60 acetyltransferase. We also established that HDAC5-mediated deacetylation is critical for SATB1-dependent downregulation of key tumor suppressor genes. Deacetylated SATB1 also represses SDHA-induced epigenetic remodeling and anti-proliferative transcriptional program. Thus, SATB1 spurs malignant phenotype in a HDAC5-dependent manner. CONCLUSIONS: Our study highlights the pivotal role of HDAC5 in tumorigenesis. Our findings provide key insights into molecular mechanisms underlying SATB1 promoted tumor growth and metastasis.


Subject(s)
Adenocarcinoma of Lung , Matrix Attachment Region Binding Proteins , Humans , Matrix Attachment Region Binding Proteins/genetics , Transcription Factors , Adenocarcinoma of Lung/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Carcinogenesis
4.
ACS Pharmacol Transl Sci ; 6(2): 281-289, 2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36798474

ABSTRACT

Cancer treatment by inhibiting the PD-1/PD-L1 pathway using monoclonal antibodies has made great advances as it showed long-lasting antitumor responses in a wide range of cancers. However, antibodies exhibit several disadvantages, which include low permeability, immune-related adverse effects, complex synthetic procedures, and high treatment costs. Hence, small-molecule inhibitors can be used as alternatives; however, no small molecule with in vivo activity has been reported. In addition, there are many challenges in developing a new drug, including the timeline and escalating cost. Therefore, repurposing an approved drug offers advantages over the development of an entirely new drug. Herein, we identify an FDA-approved small-molecule drug, Ponatinib, as a PD-L1 inhibitor via virtual drug screening of the ZINC database. Ponatinib showed stable binding with PD-L1, with the highest binding energy among all of the screened FDA-approved drugs. The binding of Ponatinib with PD-L1 was supported by a fluorescence quenching assay and immunofluorescence study. Further, we compared the in vivo antitumor efficacy of Ponatinib with a commercially available anti-PD-L1 antibody in the murine melanoma model. Ponatinib was found to be more efficient in delaying tumor growth than the anti-PD-L1 antibody. Furthermore, Ponatinib also reduced the expression of PD-L1 in tumors and increased the T-cell population. Interestingly, splenocytes isolated from Ponatinib-treated mice showed enhanced cytotoxic T-cell (CTL) activity against B16-F10 cells. However, Ponatinib itself did not have any direct toxic effect on cancer cells in vitro. These findings suggest that Ponatinib can be used as a potent small-molecule inhibitor of PD-L1 to overcome the disadvantages associated with antibodies.

5.
J Assoc Physicians India ; 70(3): 11-12, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35438276

ABSTRACT

COVID-19 is the disease caused by SARS-CoV-2. The present hospital based study was performed to find out prevalence of Urinary Tract Infection among COVID 19 patients. The cross sectional study was performed with seven hundred fifty three laboratory confirmed COVID 19 cases over six months (from 1st July to 31st December, 2020). Urine samples collected from laboratory confirmed COVID-19 cases in appropriate sterile manner and were screened for pus cells and bacteria. This was followed by plating on Mac-conkey's agar media and 5% Sheep Blood agar media. Inoculated plates were incubated overnight in aerobic condition at 37°C. Discrete colonies were further studied by Gram staining, tests for motility, battery of biochemical tests. Antibiogram was performed by disk diffusion method as per CLSI guidelines. Species confirmation and MIC (Minimum Inhibitory Concentration) values of the tested antibiotics were detected by automation. Results were analyzed according to standard statistical methods. Ninety urine samples were culture positive (11.95%). Escherichia coli was found to be the commonest pathogen, isolated in forty three cases (47.78%) followed by Enterococcus faecalis in twenty nine (32.22%) and Klebsiella pneumoniae subspp. pneumonia in eighteen occasions (20%). Enterococcus faecalis isolates were sensitive to Vancomycin, Linezolid and Nitrofurantoin and nineteen isolates were resistant to fluroquinolones (65.51%). Majority of the Gram Negative isolates were susceptible to nitrofurantoin (80.32%) where as fifteen carbapenemase producers, thirteen AmpC Betalactamase producers and twenty one Extended Spectrum Beta Lactamase (ESBL) producers have been recorded. Constant awareness regarding the antibiotic guidelines for COVID-19 cases is the need of the hour.


Subject(s)
COVID-19 , Urinary Tract Infections , Agar , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cross-Sectional Studies , Escherichia coli , Female , Humans , Male , Microbial Sensitivity Tests , Nitrofurantoin , Prevalence , SARS-CoV-2 , Urinary Tract Infections/drug therapy , Urinary Tract Infections/epidemiology , Urinary Tract Infections/microbiology , beta-Lactamases
6.
Sci Rep ; 12(1): 2548, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169159

ABSTRACT

E2F1 induces hundreds of protein-coding genes influencing diverse signaling pathways but much less is known about its non-coding RNA targets. For identifying E2F1-dependent oncogenic long non-coding RNAs (lncRNAs), we carried out genome-wide transcriptome analysis and discovered an lncRNA, EMSLR, which is induced both in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). EMSLR depletion blocks the cells in G1 phase and inhibits the clonogenic ability indicating that it is essential for the tumor-related phenotypes. We discovered that EMSLR represses the promoter activity of another lncRNA, LncPRESS1, which is located 6.9 kb upstream of EMSLR and they display an inverse expression pattern in lung cancer cell lines. Depletion of C-MYC results in downregulation of EMSLR and simultaneous upregulation of EMSLR target LncPRESS1, exemplifying how C-MYC and E2F1 signal transduction pathways control the network of lncRNA genes to modulate cell proliferation and differentiation.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinoma, Squamous Cell/metabolism , E2F1 Transcription Factor/metabolism , Lung Neoplasms/metabolism , Gene Expression Regulation, Neoplastic
7.
Cell Mol Life Sci ; 79(3): 136, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35181820

ABSTRACT

Sertoli cells (Sc) are the sole target of follicle-stimulating hormone (FSH) in the testis and attain functional maturation post-birth to significantly augment germ cell (Gc) division and differentiation at puberty. Despite having an operational microRNA (miRNA) machinery, limited information is available on miRNA-mediated regulation of Sc maturation and male fertility. We have shown before that miR-92a-3p levels decline in pubertal rat Sc. In response to FSH treatment, the expressions of FSH Receptor, Claudin11 and Klf4 were found to be elevated in pubertal rat Sc coinciding with our finding of FSH-induced decline in miR-92a-3p levels. To investigate the association of miR-92a-3p and spermatogenesis, we generated transgenic mice where such pubertal decline of miR-92a-3p was prevented by its overexpression in pubertal Sc under proximal Rhox5 promoter, which is known to be activated specifically at puberty, in Sc. Our in vivo observations provided substantial evidence that FSH-induced decline in miR-92a-3p expression during Sc maturation acts as an essential prerequisite for the pubertal onset of spermatogenesis. Elevated expression of miR-92a-3p in post-pubertal testes results into functionally compromised Sc, leading to impairment of the blood-testis barrier formation and apoptosis of pre-meiotic Gc, ultimately culminating into infertility. Collectively, our data suggest that regulation of miR-92a-3p expression is crucial for Sc-mediated induction of active spermatogenesis at puberty and regulation of male fertility.


Subject(s)
Cell Differentiation , Fertility , Follicle Stimulating Hormone/pharmacology , Germ Cells/cytology , MicroRNAs/genetics , Sertoli Cells/cytology , Testis/cytology , Animals , Female , Germ Cells/drug effects , Germ Cells/metabolism , Hormones/pharmacology , Male , Mice , Mice, Transgenic , Rats , Rats, Wistar , Receptors, FSH/genetics , Receptors, FSH/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sexual Maturation , Spermatogenesis , Testis/drug effects , Testis/metabolism
8.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34593639

ABSTRACT

PRAMEF2 is a member of the PRAME multigene family of cancer testis antigens, which serve as prognostic markers for several cancers. However, molecular mechanisms underlying its role in tumorigenesis remain poorly understood. Here, we report that PRAMEF2 is repressed under conditions of altered metabolic homeostasis in a FOXP3-dependent manner. We further demonstrate that PRAMEF2 is a BC-box containing substrate recognition subunit of Cullin 2-based E3 ubiquitin ligase complex. PRAMEF2 mediates polyubiquitylation of LATS1 kinase of the Hippo/YAP pathway, leading to its proteasomal degradation. The site for ubiquitylation was mapped to the conserved Lys860 residue in LATS1. Furthermore, LATS1 degradation promotes enhanced nuclear accumulation of the transcriptional coactivator YAP, resulting in increased expression of proliferative and metastatic genes. Thus, PRAMEF2 promotes malignant phenotype in a YAP-dependent manner. Additionally, elevated PRAMEF2 levels correlate with increased nuclear accumulation of YAP in advanced grades of breast carcinoma. These findings highlight the pivotal role of PRAMEF2 in tumorigenesis and provide mechanistic insight into YAP regulation.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle Proteins/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , MCF-7 Cells , Mice , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Ubiquitination/genetics
9.
J Biol Chem ; 297(2): 100997, 2021 08.
Article in English | MEDLINE | ID: mdl-34302808

ABSTRACT

Long noncoding RNAs (lncRNAs) have been reported to drive key cancer pathways but the functions of majority of lncRNAs are unknown making a case for comprehensive functional evaluation of lncRNAs. With an aim to identify lncRNAs dysregulated in human cancers, we analyzed the cancer patient database of lung adenocarcinoma (LUAD), which revealed an upregulated lncRNA, LINC02381 (renamed HOXC10mRNA stabilizing factor or HMS in this study), whose depletion results in proliferation defects and inhibition of colony formation of human cancer cells. In order to identify the binding targets of HMS, we screened for cis-genes and discovered that HOXC10, an oncogene, is downregulated in the absence of HMS. Depletion of HMS does not affect the HOXC10 promoter activity but inhibits the HOXC10 3'-UTR-linked luciferase reporter activity. Since lncRNAs have been known to associate with RNA-binding proteins (RBPs) to stabilize mRNA transcripts, we screened for different RBPs and discovered that HuR, an ELAV family protein, stabilizes HOXC10 mRNA. Using RNA pull-down and deletion mapping experiments, we show that HuR physically interacts with the cytosine-rich stretch of HMS and HOXC10 3'-UTR to stabilize HOXC10 mRNA. HOXC10 is overexpressed in many human cancers, and our discovery highlights that lncRNA HMS sustains the HOXC10 mRNA levels to maintain the invasive phenotypes of cancer cells.


Subject(s)
ELAV-Like Protein 1/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Lung Neoplasms/pathology , RNA, Long Noncoding/genetics , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation , Computational Biology/methods , Databases, Genetic , Homeodomain Proteins/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Up-Regulation
10.
Nat Commun ; 10(1): 4255, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534141

ABSTRACT

Caspase-10 belongs to the class of initiator caspases and is a close homolog of caspase-8. However, the lack of caspase-10 in mice and limited substrate repertoire restricts the understanding of its physiological functions. Here, we report that ATP-citrate lyase (ACLY) is a caspase-10 substrate. Caspase-10 cleaves ACLY at the conserved Asp1026 site under conditions of altered metabolic homeostasis. Cleavage of ACLY abrogates its enzymatic activity and suppresses the generation of acetyl-CoA, which is critical for lipogenesis and histone acetylation. Thus, caspase-10-mediated ACLY cleavage results in reduced intracellular lipid levels and represses GCN5-mediated histone H3 and H4 acetylation. Furthermore, decline in GCN5 activity alters the epigenetic profile, resulting in downregulation of proliferative and metastatic genes. Thus caspase-10 suppresses ACLY-promoted malignant phenotype. These findings expand the substrate repertoire of caspase-10 and highlight its pivotal role in inhibiting tumorigenesis through metabolic and epigenetic mechanisms.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , Carcinogenesis/pathology , Caspase 10/metabolism , Epigenesis, Genetic/genetics , Neoplasms/pathology , A549 Cells , Acetyl Coenzyme A/biosynthesis , Acetylation , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , HCT116 Cells , HEK293 Cells , Histones/metabolism , Humans , Lipogenesis/physiology , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous , p300-CBP Transcription Factors/metabolism
11.
Cancer Res ; 78(22): 6386-6398, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30254149

ABSTRACT

Cyclin F is a substrate recognition subunit of Skp1-Cul1-F-box protein (SCF) E3 ubiquitin ligase complex. Although there have been reports describing the role of cyclin F in the genotoxic stress response, its function under conditions of altered metabolic homeostasis remain unexplored. Here we report that cyclin F is induced upon metabolic stress in a FOXO1-dependent manner. Under metabolic stress conditions, cyclin F mediated polyubiquitylation of RBPJ at Lys315, leading to its proteasomal degradation. RBPJ regulated the expression of IDH1, which is often mutated to an oncogenic form IDH1R132H in cancers. Thus, metabolic stress-induced cyclin F attenuated the oncogenic functions of IDH1R132H in an RBPJ-dependent manner. Studies in mouse tumor models indicated that abrogation of cyclin F expression facilitates IDH1R132H-mediated tumorigenesis and metastasis. In addition, increased IDH1R132H levels correlated with reduced cyclin F levels in increasing grades of glioma. These findings highlight a novel aspect of cyclin F functions in inhibiting tumorigenesis and provide mechanistic insights into regulation of IDH1R132H Significance: These findings reveal mechanistic insights into the key role of the cyclin F-RBPJ axis in response to metabolic stress in cancer cells. Cancer Res; 78(22); 6386-98. ©2018 AACR.


Subject(s)
Cyclins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Isocitrate Dehydrogenase/metabolism , Animals , Brain Neoplasms/metabolism , Carcinogenesis , Cell Line, Tumor , Epigenesis, Genetic , Female , Forkhead Box Protein O1/metabolism , Gene Silencing , Homeostasis , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Proteasome Endopeptidase Complex/metabolism , Stress, Physiological
12.
Cancer Res ; 78(3): 645-658, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29217762

ABSTRACT

UBE3A is an E3 ubiquitin ligase well known for its role in the proteasomal degradation of p53 in human papillomavirus (HPV)-associated cancers. Here we report that UBE3A ubiquitylates and triggers degradation of the tumor-suppressive sirtuin SIRT6 in hepatocellular carcinoma. UBE3A ubiquitylated the highly conserved Lys160 residue on SIRT6. FOXO1-mediated transcriptional repression of UBE3A was sufficient to stabilize SIRT6 and to epigenetically repress ANXA2, a key mediator of UBE3A oncogenic function. Thus, UBE3A-mediated SIRT6 degradation promoted the proliferative capacity, migration potential, and invasiveness of cells. In mouse models of hepatocellular carcinoma, SIRT6 downregulation and consequent induction of ANXA2 were critical for UBE3A-mediated tumorigenesis. Furthermore, in clinical specimens, increased UBE3A levels correlated with reduced SIRT6 levels and elevated ANXA2 levels in increasing tumor grades. Overall, our findings show how the tumor suppressor SIRT6 is regulated in hepatocellular carcinoma and establish the mechanism underlying UBE3A-mediated tumorigenesis in this disease.Significance: These findings provide mechanistic insights into regulation of the tumor suppressive sirtuin SIRT6 and its implications for the development of hepatocellular carcinoma. Cancer Res; 78(3); 645-58. ©2017 AACR.


Subject(s)
Annexin A2/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Sirtuins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Annexin A2/genetics , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Nude , Signal Transduction , Sirtuins/genetics , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 113(5): E538-47, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787900

ABSTRACT

SIRT6 (sirtuin 6) is a member of sirtuin family of deacetylases involved in diverse processes including genome stability, metabolic homeostasis, and tumorigenesis. However, the role of SIRT6 deacetylase activity in its tumor-suppressor functions is not well understood. Here we report that SIRT6 binds to and deacetylates nuclear PKM2 (pyruvate kinase M2) at the lysine 433 residue. PKM2 is a glycolytic enzyme with nonmetabolic nuclear oncogenic functions. SIRT6-mediated deacetylation results in PKM2 nuclear export. We further have identified exportin 4 as the specific transporter mediating PKM2 nuclear export. As a result of SIRT6-mediated deacetylation, PKM2 nuclear protein kinase and transcriptional coactivator functions are abolished. Thus, SIRT6 suppresses PKM2 oncogenic functions, resulting in reduced cell proliferation, migration potential, and invasiveness. Furthermore, studies in mouse tumor models demonstrate that PKM2 deacetylation is integral to SIRT6-mediated tumor suppression and inhibition of metastasis. Additionally, reduced SIRT6 levels correlate with elevated nuclear acetylated PKM2 levels in increasing grades of hepatocellular carcinoma. These findings provide key insights into the pivotal role of deacetylase activity in SIRT6 tumor-suppressor functions.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , Membrane Proteins/metabolism , Oncogenes , Sirtuins/metabolism , Thyroid Hormones/metabolism , Acetylation , Animals , Hep G2 Cells , Humans , Mice , Protein Transport , Sirtuins/physiology , Thyroid Hormone-Binding Proteins
14.
Mol Cell Oncol ; 1(3): e969653, 2014.
Article in English | MEDLINE | ID: mdl-27308356

ABSTRACT

p53, the revered savior of genomic integrity, receives signals from diverse stress sensors and strategizes to maintain cellular homeostasis. However, the predominance of p53 overshadows the fact that this herculean task is no one-man show; rather, there is a huge army of regulators that reign over p53 at various levels to avoid an unnecessary surge in its levels and sculpt it dynamically to favor one cellular outcome over another. This governance starts right at the time of p53 translation, which is gated by proteins that bind to p53 mRNA and keep a stringent check on p53 protein levels. The same effect is also achieved by ubiquitylases and deubiquitylases that fine-tune p53 turnover and miRNAs that modulate p53 levels, adding precision to this entire scheme. In addition, extensive covalent modifications and differential protein interactions allow p53 to trigger a tailor-made response for a given circumstance. To magnify the marvel, these various tiers of regulation operate simultaneously and in various combinations. In this review, we have tried to provide a glimpse into this bewildering labyrinth. We believe that further studies will result in a better understanding of p53 regulation and that new insights will help unravel many aspects of cancer biology.

15.
Mol Cell ; 52(3): 406-20, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24120667

ABSTRACT

Despite being one of the most well-studied transcription factors, the temporal regulation of p53-mediated transcription is not very well understood. Recent data suggest that target specificity of p53-mediated transactivation is achieved by posttranslational modifications of p53. K120 acetylation is a modification critical for recruitment of p53 to proapoptotic targets. Our data reveal that histone deacetylase 5 (HDAC5) binds to p53 and abrogates K120 acetylation, resulting in preferential recruitment of p53 to proarrest and antioxidant targets at early phases of stress. However, upon prolonged genotoxic stress, HDAC5 undergoes nuclear export. Concomitantly, p53 is acetylated at the K120 residue and selectively transactivates proapoptotic target genes, leading to onset of apoptosis. Furthermore, upon genotoxic stress in mice where HDAC5 expression is downregulated, the onset of apoptosis is accelerated in the highly vulnerable tissues. These findings suggest that HDAC5 is a key determinant of p53-mediated cell fate decisions in response to genotoxic stress.


Subject(s)
Acetylation/drug effects , Apoptosis/genetics , DNA Damage/genetics , Histone Deacetylases/genetics , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/genetics , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Animals , Apoptosis/drug effects , Etoposide/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HCT116 Cells , Histone Deacetylases/metabolism , Humans , Lysine/metabolism , Mice , Protein Binding , Reactive Oxygen Species/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics
16.
Int J Cancer ; 133(12): 2759-68, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23436247

ABSTRACT

E3 ubiquitin ligases and deubiquitylating enzymes (DUBs) are the key components of ubiquitin proteasome system which plays a critical role in cellular protein homeostasis. Any shortcoming in their biological roles can lead to various diseases including cancer. The dynamic interplay between ubiquitylation and deubiquitylation determines the level and activity of several proteins including p53, which is crucial for cellular stress response and tumor suppression pathways. In this review, we describe the different types of E3 ubiquitin ligases including those targeting tumor suppressor p53, SCF ligases and RING type ligases and accentuate on biological functions of few important E3 ligases in the cellular regulatory networks. Tumor suppressor p53 level is tightly regulated by multiple E3 ligases including Mdm2, COP1, Pirh2, etc. SCF ubiquitin ligase complexes are key regulators of cell cycle and signal transduction. BRCA1 and VHL RING type ligases function as tumor suppressors and play an important role in DNA repair and hypoxia response respectively. Further, we discuss the biological consequences of deregulation of the E3 ligases and the implications for cancer development. We also describe deubiquitylases which reverse the process of ubiquitylation and regulate diverse cellular pathways including metabolism, cell cycle control and chromatin remodelling. As the E3 ubiquitin ligases and DUBs work in a substrate specific manner, an improved understanding of them can lead to better therapeutics for cancer.


Subject(s)
Neoplasms/etiology , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Animals , BRCA1 Protein/physiology , Humans , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/physiology , Thiolester Hydrolases/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/physiology , Ubiquitin-Specific Peptidase 7 , Ubiquitination
17.
Transcription ; 3(3): 119-23, 2012.
Article in English | MEDLINE | ID: mdl-22771946

ABSTRACT

Metabolic reprogramming is an integral part of tumorigenesis. Tumor suppressor p53 is a well studied transcription factor intimately linked with the control of cell cycle progression and apoptosis. Here, we discuss the emerging role of p53 in the transcriptional regulation of metabolism. This activity is a key component of p53 tumor suppression function.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/metabolism , Apoptosis , Cell Cycle , Cell Transformation, Neoplastic/genetics , Humans , Transcription Factors , Tumor Suppressor Protein p53/genetics
18.
Mol Cell ; 44(4): 621-34, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099309

ABSTRACT

Metabolic stress results in p53 activation, which can trigger cell-cycle arrest, ROS clearance, or apoptosis. However, what determines the p53-mediated cell fate decision upon metabolic stress is not very well understood. We show here that PGC-1α binds to p53 and modulates its transactivation function, resulting in preferential transactivation of proarrest and metabolic target genes. Thus glucose starvation results in p53-dependent cell-cycle arrest and ROS clearance, but abrogation of PGC-1α expression results in extensive apoptosis. Additionally, prolonged starvation results in PGC-1α degradation concomitant with induction of apoptosis. We have also identified RNF2, a Polycomb group (PcG) protein, as the cognate E3 ubiquitin ligase. Starvation of mice where PGC-1α expression is abrogated results in loss of p53-mediated ROS clearance, enhanced p53-dependent apoptosis, and consequent severe liver atrophy. These findings provide key insights into the role of PGC-1α in regulating p53-mediated cell fate decisions in response to metabolic stress.


Subject(s)
Apoptosis/drug effects , Glucose/deficiency , Liver/metabolism , Repressor Proteins/metabolism , Stress, Physiological/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Flow Cytometry , Gene Silencing/drug effects , Humans , Liver/drug effects , Liver/pathology , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polycomb Repressive Complex 1 , Protein Binding , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcription Factors , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases
19.
Ultramicroscopy ; 110(7): 783-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20356677

ABSTRACT

The medium carbon (0.5 wt% C) steels containing various boron contents were studied to observe the distribution of boron using atom probe tomography and electron energy loss spectroscopy. APT revealed the segregation of boron atoms at retained austenite for 100 ppm boron added steels and the trapped carbon atoms at micro-twins for 50 ppm boron treated steels. Moreover, it was also found that boron was randomly distributed for 20 ppm boron added steels regardless of the interactions between carbon and boron.

20.
Cell Cycle ; 7(2): 154-7, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18212532

ABSTRACT

A crucial unresolved issue about the genotoxic stress response is how the activation of the p53 tumor suppressor can lead either to cell cycle arrest and DNA repair or to apoptosis. p53 is one of the most important tumor suppressor proteins in the cell to prevent to heritable transfer of damaged DNA. In response to different stress conditions p53 rapidly accumulates and functions as a sequence specific DNA-binding transcription factor to regulate a large number of target genes. Activation of p53 has two major outcomes: cell cycle arrest or apoptosis. In this review we attempt to enumerate the different modifications and co-factors that influence p53 promoter selection and demonstrate how p53 chooses life or death for the cell.


Subject(s)
Promoter Regions, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Cycle , DNA Damage , Humans , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL