Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Adhes Dent ; 25(1): 257-266, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38116811

ABSTRACT

PURPOSE: This study evaluated the long-term push-out bond strength (PBS) and nanoleakage (NL) of universal adhesives with different photo-initiator systems in the root canals of teeth in which fiber posts were luted. MATERIALS AND METHODS: One-hundred twenty endodontically treated human premolars were randomly divided into 12 groups based on the following factors: adhesives (Scotchbond Universal [SBU], Ambar Universal [AMB], and Ambar Universal APS [AMB-APS]); adhesive strategy (etch-and-rinse and self-etch), and time of testing (immediately vs after 2 years). The posts were cemented, sectioned into slices, tested for PBS at 0.5 mm/min, and examined for NL using scanning electron microscopy immediately thereafter or after 2 years of water storage. Data were evaluated using a four-way ANOVA (root thirds vs time vs universal adhesive vs adhesive strategies) and Tukey's test (α = 5%). RESULTS: For both evaluation times, AMB-APS demonstrated no significant difference in the PBS or NL when different radicular thirds were compared (p > 0.05). However, for SBU and AMB, the cervical third demonstrated higher PBS and lower NL than those of the apical third at both time points (p < 0.0001). AMB-APS exhibited higher PBS and lower NL in the apical third in comparison with SBU and AMB (p < 0.0001). SBU and AMB displayed a significant decrease in the PBS and increased NL after 2 years (p < 0.0001), whereas AMB-APS demonstrated no significant signs of degradation even after 2 years of water storage (p > 0.05). CONCLUSION: Independent of the root third evaluated, the universal adhesive containing APS photo-initiator system demonstrated bonding stability at the adhesive interface between the root canal and fiber posts, even after 2 years of water storage.


Subject(s)
Dental Bonding , Post and Core Technique , Humans , Dental Cements/chemistry , Methacrylates/chemistry , Resin Cements/chemistry , Water , Dentin , Dental Bonding/methods , Materials Testing , Dentin-Bonding Agents/chemistry , Adhesives
2.
Methods Enzymol ; 689: 332-352, 2023.
Article in English | MEDLINE | ID: mdl-37802577

ABSTRACT

Conjugation of steroids and sterol compounds with a sulfonate group is a major pathway in the regulation of their activity, synthesis and excretion. Three human cytosolic sulfotransferases are highly involved in the sulfonation of sterol compounds. SULT1E1 has a low nM affinity for estrogen sulfonation and also conjugates non-aromatic steroids with a significantly lower affinity. SULT2A1 is responsible for the high levels of fetal and adult dehydroepiandrosterone (DHEA) sulfate synthesis in the adrenal gland as well as many 3α and 3ß-hydroxysteroids and bile acids. SULT2B1b is responsible for the majority of cholesterol sulfation in tissues as well as conjugating 3ß-hydroxysteroids. Although there are multiple methods for assaying cytosolic SULT activity, two relatively simple, rapid and versatile assays for steroid sulfonation are described. The first method utilizes radiolabeled substrates and organic solvent extraction to isolate the radiolabeled product from the aqueous phase. The second assay utilizes 35S-3'-phosphoadenosine 5'-phosphosulfate (PAPS) to generate 35S-conjugated products that are resolved by thin layer chromatography. Both assays useful in situations requiring measurement of SULT activity in a timely manner.


Subject(s)
Steroids , Sulfotransferases , Adult , Humans , Hydroxysteroids , Sulfotransferases/metabolism , Sterols
3.
Plants (Basel) ; 12(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37687413

ABSTRACT

Osmanthus fragrans (Thunb.) Lour. flowers (OF-F) have been traditionally consumed as a functional food and utilized as folk medicine. This study evaluated the antioxidant, anti-inflammatory and cytotoxic effects of OF-F extracts on prostate cancer cells (DU-145) and determined possible protein-ligand interactions of its compounds in silico. The crude OF-F extracts-water (W) and ethanol (E) were tested for phytochemical screening, antioxidant, anti-inflammatory, and anti-cancer. Network and molecular docking analyses of chemical markers were executed to establish their application for anticancer drug development. OF-F-E possessed higher total polyphenols (233.360 ± 3.613 g/kg) and tannin (93.350 ± 1.003 g/kg) contents than OF-F-W. In addition, OF-F-E extract demonstrated effective DPPH scavenging activity (IC50 = 0.173 ± 0.004 kg/L) and contained a high FRAP value (830.620 ± 6.843 g Trolox/kg). In cell culture experiments, OF-F-E significantly reduced NO levels and inhibited cell proliferation of RAW-264.7 and DU-145 cell lines, respectively. Network analysis revealed O. fragrans (Thunb.) Lour. metabolites could affect thirteen molecular functions and thirteen biological processes in four cellular components. These metabolites inhibited key proteins of DU-145 prostate cancer using molecular docking with rutin owning the highest binding affinity with PIKR31 and AR. Hence, this study offered a new rationale for O. fragrans (Thunb.) Lour. metabolites as a medicinal herb for anticancer drug development.

4.
Int J Mol Sci ; 24(18)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37762232

ABSTRACT

Parkinson's disease (PD) is one of the large-scale health issues detrimental to human quality of life, and current treatments are only focused on neuroprotection and easing symptoms. This study evaluated in silico binding activity and estimated the stability of major metabolites in the roots of R. palmatum (RP) with main protein targets in Parkinson's disease and their ADMET properties. The major metabolites of RP were subjected to molecular docking and QSAR with α-synuclein, monoamine oxidase isoform B, catechol o-methyltransferase, and A2A adenosine receptor. From this, emodin had the greatest binding activity with Parkinson's disease targets. The chemical stability of the selected compounds was estimated using density functional theory analyses. The docked compounds showed good stability for inhibitory action compared to dopamine and levodopa. According to their structure-activity relationship, aloe-emodin, chrysophanol, emodin, and rhein exhibited good inhibitory activity to specific targets. Finally, mediocre pharmacokinetic properties were observed due to unexceptional blood-brain barrier penetration and safety profile. It was revealed that the major metabolites of RP may have good neuroprotective activity as an additional hit for PD drug development. Also, an association between redox-mediating and activities with PD-relevant protein targets was observed, potentially opening discussion on electrochemical mechanisms with biological functions.


Subject(s)
Emodin , Neuroprotective Agents , Parkinson Disease , Rheum , Humans , Parkinson Disease/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Emodin/pharmacology , Molecular Docking Simulation , Quality of Life , Monoamine Oxidase
5.
Plants (Basel) ; 12(16)2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37631218

ABSTRACT

Perilla frutescens (L.) Britt. is extensively cultivated in East Asia as a dietary vegetable, and nutraceuticals are reportedly rich in bioactive compounds, especially with anticancer activities. This study explored the in vitro cytotoxic effects of P. frutescens parts' (stems, leaves, and seeds) extracts on prostate cancer cells (DU-145) and possible interactions of putative metabolites to related prostate cancer targets in silico. The ethanol extract of P. frutescens leaves was the most cytotoxic for the prostate cancer cells. From high-performance liquid chromatography analysis, rosmarinic acid was identified as the major metabolite in the leaf extracts. Network analysis revealed interactions from multiple affected targets and pathways of the metabolites. From gene ontology enrichment analysis, P. frutescens leaf metabolites could significantly affect 14 molecular functions and 12 biological processes in five cellular components. Four (4) KEGG pathways, including for prostate cancer, and six (6) Reactome pathways were shown to be significantly affected. The molecular simulation confirmed the interactions of relevant protein targets with key metabolites, including rosmarinic acid. This study could potentially lead to further exploration of P. frutescens leaves or their metabolites for prostate cancer treatment and prevention.

6.
PLoS One ; 18(3): e0283171, 2023.
Article in English | MEDLINE | ID: mdl-36928424

ABSTRACT

BACKGROUND: Often considered an "invisible disability", hearing loss is one of the most prevalent chronic diseases and the third leading cause for years lived with disability worldwide. Hearing loss has substantial impacts on communication, psychological wellbeing, social connectedness, cognition, quality of life, and economic independence. The Hearing impairment in Adults: a Longitudinal Outcomes Study (HALOS) aims to evaluate the: (1) impacts of hearing devices (hearing aids and/or cochlear implants), (2) differences in timing of these interventions and in long-term outcomes between hearing aid and cochlear implant users, and (3) cost-effectiveness of early intervention for adult-onset hearing loss among hearing device users. MATERIALS AND METHODS: HALOS is a mixed-methods study collecting cross-sectional and longitudinal data on health and social outcomes from 908 hearing aid and/or cochlear implant users aged ≥40 years, recruited from hearing service providers across Australia. The quantitative component will involve an online survey at baseline (time of recruitment), 24-months, and 48-months and will collect audiological, health, psychosocial, functional and employment outcomes using validated instruments. The qualitative component will be conducted in a subset of participants at baseline and involve semi-structured interviews to understand the patient journey and perspectives on the Australian hearing service model. ETHICS: This study has been approved by the Macquarie University Human Research Ethics Committee (ID: 11262) and Southern Adelaide Local Health Network (ID: LNR/22/SAC/88). Dissemination of results: Study findings will be disseminated to participants via a one-page summary, and to the public through publications in peer-reviewed journals and presentations at conferences. TRIAL REGISTRATION: Australia New Zealand Clinical Trial Registry (ANZCTR) registration number: ACTRN12622000752763.


Subject(s)
Cochlear Implantation , Deafness , Hearing Aids , Hearing Loss , Adult , Humans , Australia/epidemiology , Cross-Sectional Studies , Hearing Loss/psychology , Quality of Life
7.
Front Oncol ; 12: 925718, 2022.
Article in English | MEDLINE | ID: mdl-35795040

ABSTRACT

We reported previously that the BET inhibitor (BETi) JQ1 decreases levels of the DNA repair protein RAD51 and that this decrease is concomitant with increased levels of DNA damage. Based on these findings, we hypothesized that a BETi would augment DNA damage produced by radiation and function as a radiosensitizer. We used clonogenic assays to evaluate the effect of JQ1 ± ionizing radiation (IR) on three pancreatic cancer cell lines in vitro. We performed immunofluorescence assays to assess the impact of JQ1 ± IR on DNA damage as reflected by levels of the DNA damage marker γH2AX, and immunoblots to assess levels of the DNA repair protein RAD51. We also compared the effect of these agents on the clonogenic potential of transfectants that expressed contrasting levels of the principle molecular targets of JQ1 (BRD2, BRD4) to determine whether levels of these BET proteins affected sensitivity to JQ1 ± IR. The data show that JQ1 + IR decreased the clonogenic potential of pancreatic cancer cells more than either modality alone. This anticlonogenic effect was associated with increased DNA damage and decreased levels of RAD51. Further, lower levels of BRD2 or BRD4 increased sensitivity to JQ1 and JQ1 + IR, suggesting that pre-treatment levels of BRD2 or BRD4 may predict sensitivity to a BETi or to a BETi + IR. We suggest that a BETi + IR merits evaluation as therapy prior to surgery for pancreatic cancer patients with borderline resectable disease.

8.
J Clin Exp Dent ; 14(6): e486-e491, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35765359

ABSTRACT

Background: New protocols are constantly being tested in the search for complete disinfection of root canals without the undesirable effects of sodium hypochlorite. This study evaluated the antimicrobial effect of ozonated water and photodynamic therapy (PDT) with sonic activation in root canals infected with Enterococcus faecalis. Material and Methods: Seventy single-rooted canals of human teeth were prepared and contaminated with E. faecalis for 21 days. The teeth were divided into six experimental groups (n=10): ozonized water without (O + S-) and with sonic activation (O + S +); PDT without (PDT + S-) and with sonic activation (PDT + S +); PDT + ozonized water without (PDT + O + S-) and with sonic activation (PDT + O + S +); and two control groups, one positive (n = 5) and one negative (n = 5). Microbial collections were performed before and shortly after treatment, counted in the log of colony-forming units and tested for significant difference between these counts, if any (Student's t-test). The ANOVA two-way test was applied to evaluate whether the Treatment factor (Ozone, PDT and Ozone + PDT) and the Sonic factor (With and without sonic activation) had any effect on microbial reduction. Results: In all protocols, there was a significant microbial reduction (p=0.025), whereas in the groups in which sonic activation was used, the microbial reduction was significantly greater (p=0.001). Conclusions: The treatments significantly reduced the number of microorganisms in the root canals. Sonic activation helped to increase the microbial reduction in infected root canals. Key words:Endodontics, ozone, photodynamic therapy, enterococcus faecalis.

9.
Med Oncol ; 39(2): 24, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34982270

ABSTRACT

This work describes the effects of immunotherapy with Protein Aggregate Magnesium-Ammonium Phospholinoleate-Palmitoleate Anhydride in the treatment of non-muscle invasive bladder cancer in an animal model. NMIBC was induced by treating female Fischer 344 rats with N-methyl-N-nitrosourea. After treatment with MNU, the rats were distributed into four experimental groups: Control (without MNU) group, MNU (cancer) group, MNU-BCG (Bacillus Calmette-Guerin) group, and MNU-P-MAPA group. P-MAPA intravesical treatment was more effective in histopathological recovery from cancer state in relation to BCG treatment. Western blot assays showed an increase in the protein levels of c-Myc, COUP-TFII, and wild-type p53 in P-MAPA-treated rats in relation to BCG-treated rats. In addition, rats treated with P-MAPA intravesical immunotherapy showed the highest BAX protein levels and the lowest proliferation/apoptotic ratio in relation to BCG-treated rats, pointing out a preponderance of apoptosis. P-MAPA intravesical treatment increased the wild-type p53 levels and enhanced c-Myc/COUP-TFII-induced apoptosis mediated by p53. These alterations were fundamental for histopathological recovery from cancer and for suppress abnormal cell proliferation. This action of P-MAPA on apoptotic pathways may represent a new strategy for treating NMIBC.


Subject(s)
Immunomodulating Agents/administration & dosage , Linoleic Acids/administration & dosage , Oleic Acids/administration & dosage , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Administration, Intravesical , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunotherapy/methods , Neoplasm Invasiveness , Proto-Oncogene Proteins c-myc/metabolism , Rats, Inbred F344 , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , bcl-2-Associated X Protein/metabolism
10.
Cancers (Basel) ; 13(14)2021 Jul 11.
Article in English | MEDLINE | ID: mdl-34298684

ABSTRACT

Gemcitabine is used to treat pancreatic cancer (PC), but is not curative. We sought to determine whether gemcitabine + a BET bromodomain inhibitor was superior to gemcitabine, and identify proteins that may contribute to the efficacy of this combination. This study was based on observations that cell cycle dysregulation and DNA damage augment the efficacy of gemcitabine. BET inhibitors arrest cells in G1 and allow increases in DNA damage, likely due to inhibition of expression of DNA repair proteins Ku80 and RAD51. BET inhibitors (JQ1 or I-BET762) + gemcitabine were synergistic in vitro, in Panc1, MiaPaCa2 and Su86 PC cell lines. JQ1 + gemcitabine was more effective in vivo than either drug alone in patient-derived xenograft models (P < 0.01). Increases in the apoptosis marker cleaved caspase 3 and DNA damage marker γH2AX paralleled antitumor efficacy. Notably, RNA-seq data showed that JQ1 + gemcitabine selectively inhibited HMGCS2 and APOC1 ~6-fold, compared to controls. These proteins contribute to cholesterol biosynthesis and lipid metabolism, and their overexpression supports tumor cell proliferation. IPA data indicated that JQ1 + gemcitabine selectively inhibited the LXR/RXR activation pathway, suggesting the hypothesis that this inhibition may contribute to the observed in vivo efficacy of JQ1 + gemcitabine.

11.
Autophagy ; 17(6): 1330-1348, 2021 06.
Article in English | MEDLINE | ID: mdl-32450052

ABSTRACT

Stroke is a leading cause of death and disability. The pathophysiological mechanisms associated with stroke are very complex and not fully understood. Lysosomal function has a vital physiological function in the maintenance of cellular homeostasis. In neurons, CTSD (cathepsin D) is an essential protease involved in the regulation of proteolytic activity of the lysosomes. Loss of CTSD leads to lysosomal dysfunction and accumulation of different cellular proteins implicated in neurodegenerative diseases. In cerebral ischemia, the role of CTSD and lysosomal function is not clearly defined. We used oxygen-glucose deprivation (OGD) in mouse cortical neurons and the middle cerebral artery occlusion (MCAO) model of stroke to assess the role of CTSD in stroke pathophysiology. Our results show a time-dependent decrease in CTSD protein levels and activity in the mouse brain after stroke and neurons following OGD, with concurrent defects in lysosomal function. We found that shRNA-mediated knockdown of CTSD in neurons is sufficient to cause lysosomal dysfunction. CTSD knockdown further aggravates lysosomal dysfunction and cell death in OGD-exposed neurons. Restoration of CTSD protein levels via lentiviral transduction increases CTSD activity in neurons and, thus, renders resistance to OGD-mediated defects in lysosomal function and cell death. This study indicates that CTSD-dependent lysosomal function is critical for maintaining neuronal survival in cerebral ischemia; thus, strategies focused on maintaining CTSD function in neurons are potentially novel therapeutic approaches to prevent neuronal death in stroke.Abbreviations: 3-MA: 3-methyladenine; ACTB: actin beta; AD: Alzheimer disease; ALS: amyotrophic lateral sclerosis; CQ: chloroquine; CTSB: cathepsin B; CTSD: cathepsin D; CTSL: cathepsin L; FTD: frontotemporal dementia, HD: Huntington disease; LAMP1: lysosomal associated membrane protein 1; LSD: lysosomal storage disease; MCAO: middle cerebral artery occlusion; OGD: oxygen glucose deprivation; OGR: oxygen glucose resupply; PD: Parkinson disease; SQSMT1: sequestosome 1; TCA: trichloroacetic acid; TTC: triphenyl tetrazolium chloride.


Subject(s)
Autophagy/physiology , Cathepsin D/metabolism , Lysosomes/metabolism , Neuroprotection/physiology , Stroke/metabolism , Animals , Brain/metabolism , Brain Ischemia/metabolism , Cell Death/physiology , Mice , Neurons/metabolism
12.
Cancer Drug Resist ; 3: 572-585, 2020.
Article in English | MEDLINE | ID: mdl-33073205

ABSTRACT

AIM: Gemcitabine is a frontline agent for locally-advanced and metastatic pancreatic ductal adenocarcinoma (PDAC), but neither gemcitabine alone nor in combination produces durable remissions of this tumor type. We developed three PDAC patient-derived xenograft (PDX) models with gemcitabine resistance (gemR) acquired in vivo, with which to identify mechanisms of resistance relevant to drug exposure in vivo and to evaluate novel therapies. METHODS: Mice bearing independently-derived PDXs received 100 mg/kg gemcitabine once or twice weekly. Tumors initially responded, but regrew on treatment and were designated gemR. We used immunohistochemistry to compare expression of proteins previously associated with gemcitabine resistance [ribonucleotide reductase subunit M1 (RRM1), RRM2, human concentrative nucleoside transporter 1 (hCNT1), human equilibrative nucleoside transporter 1 (hENT1), cytidine deaminase (CDA), and deoxycytidine kinase (dCK)] in gemR and respective gemcitabine-naive parental tumors. RESULTS: Parental and gemR tumors did not differ in tumor cell morphology, amount of tumor-associated stroma, or expression of stem cell markers. No consistent pattern of expression of the six gemR marker proteins was observed among the models. Increases in RRM1 and CDA were consistent with in vitro-derived gemR models. However, rather than the expected decreases of hCNT1, hENT1, and dCK, gemR tumors expressed no change in or higher levels of these gemR marker proteins than parental tumors. CONCLUSION: These models are the first PDAC PDX models with gemcitabine resistance acquired in vivo. The data indicate that mechanisms identified in models with resistance acquired in vitro are unlikely to be the predominant mechanisms when resistance is acquired in vivo. Ongoing work focuses on characterizing unidentified mechanisms of gemR and on identifying agents with anti-tumor efficacy in these gemR models.

13.
Cancers (Basel) ; 12(5)2020 May 22.
Article in English | MEDLINE | ID: mdl-32456018

ABSTRACT

Pancreatic cancer (PC) is anticipated to be second only to lung cancer as the leading cause of cancer-related deaths in the United States by 2030. Surgery remains the only potentially curative treatment for patients with pancreatic ductal adenocarcinoma (PDAC), the most common form of PC. Multiple recent preclinical studies focus on identifying effective treatments for PDAC, but the models available for these studies often fail to reproduce the heterogeneity of this tumor type. Data generated with such models are of unknown clinical relevance. Patient-derived xenograft (PDX) models offer several advantages over human cell line-based in vitro and in vivo models and models of non-human origin. PDX models retain genetic characteristics of the human tumor specimens from which they were derived, have intact stromal components, and are more predictive of patient response than traditional models. This review briefly describes the advantages and disadvantages of 2D cultures, organoids and genetically engineered mouse (GEM) models of PDAC, and focuses on the applications, characteristics, advantages, limitations, and the future potential of PDX models for improving the management of PDAC.

14.
Pharmacol Res ; 155: 104740, 2020 05.
Article in English | MEDLINE | ID: mdl-32135247

ABSTRACT

Pancreatic cancer is a fatal disease. The five-year survival for patients with all stages of this tumor type is less than 10%, with a majority of patients dying from drug resistant, metastatic disease. Gemcitabine has been a standard of care for the treatment of pancreatic cancer for over 20 years, but as a single agent gemcitabine is not curative. Since the only therapeutic option for the over 80 percent of pancreatic cancer patients ineligible for surgical resection is chemotherapy with or without radiation, the last few decades have seen a significant effort to develop effective therapy for this disease. This review addresses preclinical and clinical efforts to identify agents that target molecular characteristics common to pancreatic tumors and to develop mechanism-based combination approaches to therapy. Some of the most promising combinations include agents that inhibit transcription dependent on BET proteins (BET bromodomain inhibitors) or that inhibit DNA repair mediated by PARP (PARP inhibitors).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Acetylation/drug effects , Animals , Antineoplastic Agents/therapeutic use , DNA Damage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Histones/metabolism , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proteins/antagonists & inhibitors , Gemcitabine
15.
Cancer Lett ; 468: 48-58, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31605774

ABSTRACT

Our previous finding that the BET inhibitor (BETi) JQ1 increases levels of the DNA damage marker γH2AX suggested that JQ1 might enhance the sensitivity of tumor cells to PARP inhibitors (PARPi), which are selectively toxic to cells that harbor relatively high levels of DNA damage. To address this hypothesis, we evaluated the effect of a BETi (JQ1 or I-BET762) combined with a PARPi (olaparib or veliparib) in KKU-055 and KKU-100 cholangiocarcinoma (CCA) cell lines and of JQ1 with olaparib in a xenograft model of CCA. Each combination was more effective than any of the four drugs as single agents. Combination indices ranged from 0.1 to 0.8 at the ED50 for all combinations, indicating synergy and demonstrating that synergy was not limited to a specific combination. Mechanistically, downregulation of BETi molecular targets BRD2 or BRD4 by shRNA sensitized CCA cells to BETi as single agents as well as to the combination of a BETi + a PARPi. Our data indicate that combinations of a BETi with a PARPi merit further evaluation as a promising strategy for CCA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bile Duct Neoplasms/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , Cholangiocarcinoma/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azepines/pharmacology , Azepines/therapeutic use , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
16.
Drug Metab Dispos ; 47(9): 949-953, 2019 09.
Article in English | MEDLINE | ID: mdl-31266751

ABSTRACT

Sulfotransferase 4A1 (SULT4A1), a member of cytosolic sulfotransferases (SULT), is exclusively expressed in neurons with no known function. Severe phenotype and early postnatal death in SULT4A1 knockout mice revealed that SULT4A1 is an essential neuronal protein. Localization of SULT4A1 in different cytosolic compartments, including mitochondria, suggests multiple roles for this protein. We observed that knockdown of SULT4A1 results in the accumulation of reactive oxygen species in primary cortical neurons, suggesting a potential role of SULT4A1 in regulating redox homeostasis. Expression of SULT4A1 in the human neuroblastoma SH-SY5Y cells revealed a defused but nonuniform staining pattern in the cytoplasm, with increased density around mitochondria. Subcellular fractionation of SULT4A1 expressing SH-SY5Y cells confirms the presence of SULT4A1 in mitochondrial fractions. SULT4A1 expressing cells display significant protection against H2O2-mediated defects in mitochondrial function and loss of mitochondrial membrane potential. Expression of SULT4A1 in SH-SY5Y cells also protects against H2O2-induced cell death. These data indicate that SULT4A1 protects mitochondria against oxidative damage and may serve as a potential pharmacological target in neural diseases involving mitochondrial dysfunction and oxidative stress. SIGNIFICANCE STATEMENT: Studies on SULT4A1 knockout mice suggest that SULT4A1 plays a vital role in neuronal function and survival via yet undefined mechanisms. Our data demonstrate that depletion of SULT4A1 induces oxidative stress in neurons and expression of SULT4A1 in SH-SY5Y cells protects against oxidative-stress-induced mitochondrial dysfunction and cell death. These results suggest that SULT4A1 may have a crucial protective function against mitochondrial dysfunction and oxidative stress, and may serve a potential therapeutic target in different neurological diseases involving mitochondrial dysfunction and oxidative stress.


Subject(s)
Mitochondria/pathology , Neurons/pathology , Sulfotransferases/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cerebral Cortex/cytology , Cloning, Molecular , Humans , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Knockout , Mitochondria/metabolism , Neurons/cytology , Oxidative Stress , Primary Cell Culture , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfotransferases/genetics
17.
EBioMedicine ; 44: 419-430, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31126889

ABSTRACT

BACKGROUND: DNA repair deficiency accumulates DNA damage and sensitizes tumor cells to PARP inhibitors (PARPi). Based on our observation that the BET inhibitor JQ1 increases levels of DNA damage, we evaluated the efficacy of JQ1 + the PARPi olaparib in preclinical models of pancreatic ductal adenocarcinoma (PDAC). We also addressed the mechanism by which JQ1 increased DNA damage. METHODS: The effect of JQ1 + olaparib on in vivo tumor growth was assessed with patient-derived xenograft (PDX) models of PDAC. Changes in protein expression were detected by immunohistochemistry and immunoblot. In vitro growth inhibition and mechanistic studies were done using alamarBlue, qRT-PCR, immunoblot, immunofluorescence, ChIP, and shRNA knockdown assays. FINDINGS: Tumors exposed in vivo to JQ1 had higher levels of the DNA damage marker γH2AX than tumors exposed to vehicle only. Increases in γH2AX was concomitant with decreased expression of DNA repair proteins Ku80 and RAD51. JQ1 + olaparib inhibited the growth of PDX tumors greater than either drug alone. Mechanistically, ChIP assays demonstrated that JQ1 decreased the association of BRD4 and BRD2 with promoter loci of Ku80 and RAD51, and shRNA data showed that expression of Ku80 and RAD51 was BRD4- and BRD2-dependent in PDAC cell lines. INTERPRETATION: The data are consistent with the hypothesis that JQ1 confers a repair deficient phenotype and the consequent accumulation of DNA damage sensitizes PDAC cells to PARPi. Combinations of BET inhibitors with PARPi may provide a novel strategy for treating PDAC. FUND: NIH grants R01CA208272 and R21CA205501; UAB CMB T32 predoctoral training grant.


Subject(s)
Azepines/pharmacology , Carcinoma, Pancreatic Ductal/genetics , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triazoles/pharmacology , Acid Anhydride Hydrolases , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Histones/metabolism , Humans , Ku Autoantigen/metabolism , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phthalazines/pharmacology , Piperazines/pharmacology , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
18.
Cancer Immunol Immunother ; 68(1): 1-9, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30564889

ABSTRACT

A collaborative think tank involving panellists from immuno-oncology networks, clinical/translational investigators and the pharmaceutical industry was held in Siena, Italy, in October 2017 to discuss the evolving immune-oncology landscape, identify selected key challenges, and provide a perspective on the next steps required in the translation of current research and knowledge to clinical reality. While there is a trend of combining new agents (e.g., co-stimulator agonists) with a PD-1/PD-L1 treatment backbone, use of alternative combination therapy approaches should also be considered. While the rapid evolution in systems biology provides a deeper understanding of tumor and tumor microenvironment heterogeneity, there remains the need to identify and define genuinely predictive biomarkers to guide treatment and patient selection. Cross-specialty and cross-sector collaboration, along with a broader collective data-sharing approach are key to optimizing immuno-oncology therapy in clinical practice. Continued support of younger research-clinicians is essential for future success in clinical, translational and basic science investigations.


Subject(s)
Immunotherapy/methods , Medical Oncology/methods , Neoplasms/therapy , Translational Research, Biomedical/methods , Biomarkers, Tumor/blood , Diffusion of Innovation , Humans , Immunotherapy/trends , Italy , Medical Oncology/trends , Neoplasms/blood , Neoplasms/immunology , Translational Research, Biomedical/trends
19.
Drug Metab Dispos ; 46(1): 41-45, 2018 01.
Article in English | MEDLINE | ID: mdl-29109113

ABSTRACT

Sulfotransferase 4A1 (SULT4A1) belongs to the cytosolic sulfotransferase (SULT) superfamily of enzymes that catalyze sulfonation reactions with a variety of endogenous and exogenous substrates. Of the SULTs, SULT4A1 was shown to have the highest sequence homology between vertebrate species, yet no known function or enzymatic activity has been identified for this orphan SULT. To better understand SULT4A1 function in mammalian brain, two mutant SULT4A1 mouse strains were generated utilizing clustered regulatory interspaced short palindromic repeats (CRISPR)-content-addressable storage (Cas) 9 technology. The first strain possessed a 28-base pair (bp) deletion (Δ28) in exon 1 that resulted in a frameshift mutation with premature termination. The second strain possessed a 12-bp in-frame deletion (Δ12) immediately preceding an active site histidine111 common to the SULT family. Homozygous pups of both strains present with severe and progressive neurologic symptoms, including tremor, absence seizures, abnormal gait, ataxia, decreased weight gain compared with littermates, and death around postnatal days 21-25. SULT4A1 immunostaining was decreased in brains of heterozygous pups and not detectable in homozygous pups of both SULT4A1 mutants. SULT4A1 localization in subcellular fractions of mouse brain showed SULT4A1 associated with mitochondrial, cytosolic, and microsomal fractions, a novel localization pattern for SULTs. Finally, primary cortical neurons derived from embryonic (E15) CD-1 mice expressed high levels of SULT4A1 throughout the cell except in nuclei. Taken together, SULT4A1 appears to be an essential neuronal protein required for normal brain function, at least in mammals. Mouse models will be valuable in future studies to investigate the regulation and functions of SULT4A1 in the mammalian brain.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Sulfotransferases/metabolism , Animals , Behavior, Animal , Brain/cytology , Brain/growth & development , CRISPR-Cas Systems/genetics , Cytosol/metabolism , Exons/genetics , Female , Frameshift Mutation/genetics , Homozygote , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nerve Tissue Proteins/genetics , Primary Cell Culture , Sulfotransferases/genetics
20.
Mol Cancer Ther ; 17(1): 107-118, 2018 01.
Article in English | MEDLINE | ID: mdl-29142067

ABSTRACT

Cholangiocarcinoma (CCA) is a fatal disease with a 5-year survival of <30%. For a majority of patients, chemotherapy is the only therapeutic option, and virtually all patients relapse. Gemcitabine is the first-line agent for treatment of CCA. Patients treated with gemcitabine monotherapy survive ∼8 months. Combining this agent with cisplatin increases survival by ∼3 months, but neither regimen produces durable remissions. The molecular etiology of this disease is poorly understood. To facilitate molecular characterization and development of effective therapies for CCA, we established a panel of patient-derived xenograft (PDX) models of CCA. We used two of these models to investigate the antitumor efficacy and mechanism of action of the bromodomain inhibitor JQ1, an agent that has not been evaluated for the treatment of CCA. The data show that JQ1 suppressed the growth of the CCA PDX model CCA2 and demonstrate that growth suppression was concomitant with inhibition of c-Myc protein expression. A second model (CCA1) was JQ1-insensitive, with tumor progression and c-Myc expression unaffected by exposure to this agent. Also selective to CCA2 tumors, JQ1 induced DNA damage and apoptosis and downregulated multiple c-Myc transcriptional targets that regulate cell-cycle progression and DNA repair. These findings suggest that c-Myc inhibition and several of its transcriptional targets may contribute to the mechanism of action of JQ1 in this tumor type. We conclude that BET inhibitors such as JQ1 warrant further investigation for the treatment of CCA. Mol Cancer Ther; 17(1); 107-18. ©2017 AACR.


Subject(s)
Azepines/therapeutic use , Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Triazoles/therapeutic use , Animals , Apoptosis , Azepines/pharmacology , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , DNA Damage , Disease Models, Animal , Gene Expression , Humans , Mice , Triazoles/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...