Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38639280

ABSTRACT

Under the umbrella of targeted drug delivery systems, several techniques are unleashed in the market that allow a drug or other pharmacologically active material to be delivered to the target cell to treat a condition or health problem. The improvement of the pharmaceutical delivery systems' effectiveness, safety, and stability is accomplished through the Formulation of the nano-gel-based delivery system. Nanogels are aqueous dispersions of submicronsized, three-dimensional, strongly cross-linked networks of hydrophilic polymers that are inflated by water. Through a variety of delivery routes, such as oral, pulmonary, nasal, parenteral, and intraocular, an active pharmaceutical agent or therapeutic agent with a high or low molecular weight can be easily encapsulated into nanogels. Nanogels have been researched as drug delivery systems due to their beneficial qualities, such as biocompatibility, high stability, flexible particle size, drug loading capacity, and potential surface modification for active targeting by attaching ligands that recognize cognate receptors on target cells or tissues. By responding to internal or external stimuli, including pH, temperature, light, and redox, nano gels can be made to be stimulus-responsive, allowing for regulated drug release. Thus, in the fact of said characteristics' of nano gels, this review manuscript aims to provide an overview of characterization, evaluation, formulation technique, recent applications, and patents of nano gels.

2.
Adv Clin Chem ; 118: 87-110, 2024.
Article in English | MEDLINE | ID: mdl-38280808

ABSTRACT

Osteopontin (OPN) is a heavily post-translationally modified protein with a molecular weight of 44-70 kDa, depending on the degree of glycosylation. OPN is involved in various biological processes, including bone remodeling, immune response, cell adhesion, migration, and survival. It is essential for controlling osteoclast and osteoblast activity for maintaining bone mass and bone strength. Additionally, OPN has been linked to cardiovascular, inflammatory illnesses, as well as the onset and progression of cancer. OPN is a multifunctional protein that can interact with a variety of cell surface receptors, such as integrins, CD44, the urokinase-type plasminogen activator receptor (uPAR), as well as extracellular matrix (ECM) components (e.g. collagen and hydroxyapatite). These interactions contribute to its wide range of biological functions in general and has significant implications for bone biology, immunology and cancer, specifically. In this chapter, we summarize the structure of OPN with a focus on its molecular mechanisms of action in various cancers.


Subject(s)
Neoplasms , Osteopontin , Humans , Bone and Bones/metabolism , Osteopontin/metabolism , Signal Transduction
3.
Biomater Investig Dent ; 9(1): 1-9, 2022.
Article in English | MEDLINE | ID: mdl-35174335

ABSTRACT

AIM: To evaluate the microleakage and dentin shear bond strength of two glass containing restorative materials, Zirconomer and Cention N, and to compare them with a conventional glass ionomer cement (GIC) (GC Fuji II). MATERIALS AND METHODS: Zirconomer (Shofu) and GC Fuji II (GC Corp.) are self-curing GICs whereas Cention N (IvoclarVivadent) also offers a self-curing option as well as the option of light-curing using an adhesive. For evaluating microleakage, standardized class V cavities were prepared on the buccal surface of 30 premolars. The cavities were restored with one of the three restorative materials (n = 10) according to manufacturers' instructions, Cention N being used with an adhesive (Te-EconomBond, IvoclarVivadent) and in the light-curing mode. After restoration and thermocycling, the microleakage assessment was made under a stereomicroscope at 40x magnification following immersing of the teeth in 0.5% methylene blue dye and buccolingual sectioning. For evaluating dentin shear bond strength, the occlusal surface of the 30 premolars was ground flat, and cylinders of the three restorative materials (n = 10) were bonded to the occlusal surface according to manufacturers' instructions, Cention N being used with an adhesive (Te-EconomBond, IvoclarVivadent) and in the light-curing mode. Following 24-h storage at 100% humidity, the dentin shear bond strength was measured and the fracture mode was determined under a stereomicroscope at 10× magnification. Data were statistically analyzed using Mann-Whitney and Scheffé tests (p = .05). RESULTS: Cention N displayed significantly less microleakage than did Zirconomer and GC Fuji II at occlusal as well as the gingival margins. Dentin shear bond strength varied significantly between 5.15 and 9.89 MPa with Cention N showing the highest bond strength and GC Fuji II the lowest. CONCLUSION: In this in vitro evaluation, Cention N consistently performed better than the conventional GIC (GC Fuji II) as well as Zirconomer.

4.
Int J Mol Sci ; 22(24)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34948122

ABSTRACT

The pro-apoptotic tumor suppressor BIN1 inhibits the activities of the neoplastic transcription factor MYC, poly (ADP-ribose) polymerase-1 (PARP1), and ATM Ser/Thr kinase (ATM) by separate mechanisms. Although BIN1 deficits increase cancer-cell resistance to DNA-damaging chemotherapeutics, such as cisplatin, it is not fully understood when BIN1 deficiency occurs and how it provokes cisplatin resistance. Here, we report that the coordinated actions of MYC, PARP1, and ATM assist cancer cells in acquiring cisplatin resistance by BIN1 deficits. Forced BIN1 depletion compromised cisplatin sensitivity irrespective of Ser15-phosphorylated, pro-apoptotic TP53 tumor suppressor. The BIN1 deficit facilitated ATM to phosphorylate the DNA-damage-response (DDR) effectors, including MDC1. Consequently, another DDR protein, RNF8, bound to ATM-phosphorylated MDC1 and protected MDC1 from caspase-3-dependent proteolytic cleavage to hinder cisplatin sensitivity. Of note, long-term and repeated exposure to cisplatin naturally recapitulated the BIN1 loss and accompanying RNF8-dependent cisplatin resistance. Simultaneously, endogenous MYC was remarkably activated by PARP1, thereby repressing the BIN1 promoter, whereas PARP inhibition abolished the hyperactivated MYC-dependent BIN1 suppression and restored cisplatin sensitivity. Since the BIN1 gene rarely mutates in human cancers, our results suggest that simultaneous inhibition of PARP1 and ATM provokes a new BRCAness-independent synthetic lethal effect and ultimately re-establishes cisplatin sensitivity even in platinum-refractory cancer cells.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Protein Kinase Inhibitors/chemistry
5.
J Biol Chem ; 294(14): 5700-5719, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30733337

ABSTRACT

The tumor suppressor bridging integrator 1 (BIN1) is a corepressor of the transcription factor E2F1 and inhibits cell-cycle progression. BIN1 also curbs cellular poly(ADP-ribosyl)ation (PARylation) and increases sensitivity of cancer cells to DNA-damaging therapeutic agents such as cisplatin. However, how BIN1 deficiency, a hallmark of advanced cancer cells, increases cisplatin resistance remains elusive. Here, we report that BIN1 inactivates ataxia telangiectasia-mutated (ATM) serine/threonine kinase, particularly when BIN1 binds E2F1. BIN1 + 12A (a cancer-associated BIN1 splicing variant) also inhibited cellular PARylation, but only BIN1 increased cisplatin sensitivity. BIN1 prevented E2F1 from transcriptionally activating the human ATM promoter, whereas BIN1 + 12A did not physically interact with E2F1. Conversely, BIN1 loss significantly increased E2F1-dependent formation of MRE11A/RAD50/NBS1 DNA end-binding protein complex and efficiently promoted ATM autophosphorylation. Even in the absence of dsDNA breaks (DSBs), BIN1 loss promoted ATM-dependent phosphorylation of histone H2A family member X (forming γH2AX, a DSB biomarker) and mediator of DNA damage checkpoint 1 (MDC1, a γH2AX-binding adaptor protein for DSB repair). Of note, even in the presence of transcriptionally active (i.e. proapoptotic) TP53 tumor suppressor, BIN1 loss generally increased cisplatin resistance, which was conversely alleviated by ATM inactivation or E2F1 reduction. However, E2F2 or E2F3 depletion did not recapitulate the cisplatin sensitivity elicited by E2F1 elimination. Our study unveils an E2F1-specific signaling circuit that constitutively activates ATM and provokes cisplatin resistance in BIN1-deficient cancer cells and further reveals that γH2AX emergence may not always reflect DSBs if BIN1 is absent.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , E2F1 Transcription Factor/metabolism , Neoplasms/metabolism , Nuclear Proteins/deficiency , Transcription, Genetic , Tumor Suppressor Proteins/deficiency , Acid Anhydride Hydrolases , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair/drug effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Histones/genetics , Histones/metabolism , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/drug effects
6.
Biol Reprod ; 99(4): 735-748, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29688260

ABSTRACT

Despite the major negative impact uterine fibroids (UFs) have on female reproductive health, little is known about early events that initiate development of these tumors. Somatic fibroid-causing mutations in mediator complex subunit 12 (MED12), the most frequent genetic alterations in UFs (up to 85% of tumors), are implicated in transforming normal myometrial stem cells (MSCs) into tumor-forming cells, though the underlying mechanism(s) leading to these mutations remains unknown. It is well accepted that defective DNA repair increases the risk of acquiring tumor-driving mutations, though defects in DNA repair have not been explored in UF tumorigenesis. In the Eker rat UF model, a germline mutation in the Tsc2 tumor suppressor gene predisposes to UFs, which arise due to "second hits" in the normal allele of this gene. Risk for developing these tumors is significantly increased by early-life exposure to endocrine-disrupting chemicals (EDCs), suggesting increased UF penetrance is modulated by early drivers for these tumors. We analyzed DNA repair capacity using analyses of related gene and protein expression and DNA repair function in MSCs from adult rats exposed during uterine development to the model EDC diethylstilbestrol. Adult MSCs isolated from developmentally exposed rats demonstrated decreased DNA end-joining ability, higher levels of DNA damage, and impaired ability to repair DNA double-strand breaks relative to MSCs from age-matched, vehicle-exposed rats. These data suggest that early-life developmental EDC exposure alters these MSCs' ability to repair and reverse DNA damage, providing a driver for acquisition of mutations that may promote the development of these tumors in adult life.


Subject(s)
DNA Repair/drug effects , Endocrine Disruptors/toxicity , Leiomyoma/etiology , Myometrium/drug effects , Myometrium/metabolism , Uterine Neoplasms/etiology , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Animals , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , DNA Damage , DNA Repair/genetics , Diethylstilbestrol/toxicity , Disease Models, Animal , Female , Germ-Line Mutation , Humans , Leiomyoma/genetics , Leiomyoma/metabolism , Mediator Complex/genetics , Myometrium/growth & development , Rats , Rats, Mutant Strains , Tuberous Sclerosis Complex 2 Protein/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
7.
Genes (Basel) ; 8(6)2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28590415

ABSTRACT

Cancer is associated with genomic instability and aging. Genomic instability stimulates tumorigenesis, whereas deregulation of oncogenes accelerates DNA replication and increases genomic instability. It is therefore reasonable to assume a positive feedback loop between genomic instability and oncogenic stress. Consistent with this premise, overexpression of the MYC transcription factor increases the phosphorylation of serine 139 in histone H2AX (member X of the core histone H2A family), which forms so-called γH2AX, the most widely recognized surrogate biomarker of double-stranded DNA breaks (DSBs). Paradoxically, oncogenic MYC can also promote the resistance of cancer cells to chemotherapeutic DNA-damaging agents such as cisplatin, clearly implying an antagonistic role of MYC in genomic instability. In this review, we summarize the underlying mechanisms of the conflicting functions of MYC in genomic instability and discuss when and how the oncoprotein exerts the contradictory roles in induction of DSBs and protection of cancer-cell genomes.

8.
Mol Cell Oncol ; 2(2): e981447, 2015.
Article in English | MEDLINE | ID: mdl-27308445

ABSTRACT

The adenovirus E2 promoter-binding factor-1 (E2F1) induces apoptosis in response to DNA damage and serum starvation. After DNA damage, E2F1 is phosphorylated by ataxia telangiectasia-mutated (ATM) kinase to promote apoptosis. However, precisely how serum starvation stimulates E2F1-induced apoptosis is unclear. We recently found that serum starvation reduces E2F1 poly(ADP-ribosyl)ation, thereby releasing a proapoptotic protein, bridging integrator-1 (BIN1), into the cytoplasm.

9.
Mol Cell Biol ; 31(24): 5037-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21986497

ABSTRACT

Aberrant MYC expression is a common oncogenic event in human cancer. Paradoxically, MYC can either drive cell cycle progression or induce apoptosis. The latent ability of MYC to induce apoptosis has been termed "intrinsic tumor suppressor activity," and reactivating this apoptotic function in tumors is widely considered a valuable therapeutic goal. As a transcription factor, MYC controls the expression of many downstream targets, and for the majority of these, it remains unclear whether or not they play direct roles in MYC function. To identify the subset of genes specifically required for biological activity, we conducted a screen for functionally important MYC targets and identified BAG1, which encodes a prosurvival chaperone protein. Expression of BAG1 is regulated by MYC in both a mouse model of breast cancer and transformed human cells. Remarkably, BAG1 induction is essential for protecting cells from MYC-induced apoptosis. Ultimately, the synthetic lethality we have identified between MYC overexpression and BAG1 inhibition establishes a new pathway that might be exploited to reactivate the latent apoptotic potential of MYC as a cancer therapy.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, myc , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Division , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Female , Genetic Loci , Humans , Mice , Mice, Knockout , Plasmids , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/genetics
10.
Genetics ; 185(3): 761-70, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20421597

ABSTRACT

Preventing the formation of dysfunctional telomeres is essential for genomic stability. In most organisms, the ribo-nucleoprotein reverse transcriptase telomerase is responsible for telomere GT-strand elongation. However, in telomerase-negative cells, low-frequency recombination mechanisms can avert lethality by elongating critically short telomeres. This study focuses on the involvement of the budding yeast Mre11 in telomere recombination and homeostasis. We have identified a novel allele of MRE11, mre11-A470T, that, in telomerase-positive cells, confers a semidominant decrease in telomere size and a recessive defect in telomere healing. In addition, mutant cells lack normal telomere size homeostasis. Telomerase-negative mre11-A470T cells display a Rad51-dependent bypass of replicative senescence via induction of a highly efficient type I-related recombination pathway termed type IA. The type IA pathway involves an amplification of subtelomeric Y' elements, coupled with elongated and more heterogeneous telomere tracts relative to the short telomere size of type I survivors. The data have led us to propose the involvement of break-induced replication in telomere expansion. The differing phenotypes elicited by the mre11-A470T mutants in telomerase-positive and telomerase-negative cells have also led us to speculate that the telomere end structure may be modified differentially in mre11-A470T cells, directing the telomere into specific pathways.


Subject(s)
Cellular Senescence , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Mutation/genetics , Recombination, Genetic , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Telomere/genetics , Blotting, Southern , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Telomerase/metabolism
11.
Pancreatology ; 9(4): 360-8, 2009.
Article in English | MEDLINE | ID: mdl-19451745

ABSTRACT

BACKGROUND: The gene promoter region of human telomerase reverse transcriptase (hTERT) contains binding sites for c-myc and E2F1 as well as CpG islands, suggesting regulation by genetic factors and epigenetically by methylation. Hence, the effect of the demethylating agent 5-azacytidine and silencing of c-MYC and E2F1 genes on its expression and consequently on telomerase activity were studied in pancreatic cancer-derived cell lines. METHODS: MIaPaCa-2 and PANC-1 cell lines were transfected with SiRNA against E2F1 and c-MYC genes separately as well as along with 5-azacytidine treatment. The hTERT gene methylation status was determined by methylation-specific PCR and telomerase activity quantitated by TRAP-PCR-ELISA. RESULTS: Demethylation by 5-azacytidine resulted in hTERT inhibition with a reduction in telomerase activity to 37-49% of controls. Silencing of E2F-1 or c-MYC also decreased the hTERT transcript and telomerase activity with a more pronounced effect with respect to c-MYC silencing. There was a synergistic effect of demethylation and gene silencing on the inhibition of hTERT mRNA expression which resulted in undetectable levels of telomerase activity. CONCLUSION: Telomerase activity, which is necessary for cellular immortalization, can be shut down by a combined approach using SiRNA-mediated gene silencing and demethylating agents, which has therapeutic implications.


Subject(s)
Azacitidine/pharmacology , E2F1 Transcription Factor/genetics , Genes, myc/drug effects , Telomerase/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , Gene Silencing , Humans , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference
12.
Ann Surg Oncol ; 16(4): 1051-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19194757

ABSTRACT

AIM: We sought to determine the role of telomerase and its catalytic subunit hTERT in pancreatic cancer and evaluate the epigenetic regulation of hTERT by promoter methylation. METHODS: Thirty paired samples of pancreatic ductal adenocarcinomas and adjacent normal tissue and 12 chronic pancreatitis samples were studied. Reverse transcriptase polymerase chain reaction, telomeric repeat amplification protocol assay, and methylation-specific polymerase chain reaction were performed to analyze hTERT expression, telomerase activity, and methylation status of gene promoters, respectively. RESULT: hTERT and telomerase activity were upregulated in pancreatic cancer compared with paired normal tissues and samples of pancreatitis. hTERT expression correlated with telomerase activity (P \ .05) and in turn correlated positively with hTERT promoter methylation (P \ .001) and p16 promoter methylation. hTERT transcript expression and telomerase activity both conferred a worse outcome by univariate and multivariate analysis (P \ .05). CONCLUSION: hTERT expression and telomerase activity are predictors of poor outcome in pancreatic cancer. hTERT gene expression is positively regulated by promoter methylation.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation , Pancreatic Neoplasms/genetics , Telomerase/genetics , Adenocarcinoma/metabolism , Epigenesis, Genetic , Gene Expression , Gene Expression Regulation , Genes, p16 , Humans , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/genetics , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...