Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Invest New Drugs ; 2024 May 25.
Article in English | MEDLINE | ID: mdl-38789849

ABSTRACT

Worldwide, pancreatic cancer (PC) is a major health problem and almost 0.5 million people were diagnosed with PC in 2020. In the United States, more than 64,000 adults will be diagnosed with PC in 2023. PC is highly resistant to currently available treatments and standard of care chemotherapies cause serious side effects. Most PC patients are resistant to clinical therapies. Combination therapy has showed superior efficacy over single-agent treatment. However, most therapy has failed to show a significant improvement in overall survival due to treatment-related toxicity. Developing efficacious clinically useful PC therapies remains a challenge. Herein, we show the efficacy of an innovative pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) against tumors arising from human pancreatic cancer stem cells (i.e., hPCSCs, FGß3 cells). PAWI-2 is a potent inhibitor of tumor growth. In the present study, we showed PAWI-2 potently inhibited growth of tumors from hPCSCs in orthopic xenograft models of both male and female mice. PAWI-2 worked in a non-toxic manner to inhibit tumors. Compared to vehicle-treated animals, PAWI-2 modulated molecular regulators of tumors. Anti-cancer results showed PAWI-2 in vivo efficacy could be correlated to in vitro potency to inhibit FGß3 cells. PAWI-2 represents a safe, new approach to combat PC.

2.
J AOAC Int ; 107(4): 649-662, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38467139

ABSTRACT

BACKGROUND: Antibiotic residues in milk are a well-known hazard in the dairy food chain. Detection methods for these residues, such as nonspecific microbiological inhibitor tests or group-specific receptor tests, are relatively inexpensive, easy to use, and widely applied to ensure food safety. In contrast, specific detection by liquid chromatography-tandem mass spectrometry (LC-MS/MS)-although a critical, complimentary method to confirm the results of nonspecific testing-is relatively costly, time-consuming, and laborious. Furthermore, sample processing before LC-MS/MS analysis requires unique preparation procedures for different groups of antibiotic compounds. OBJECTIVE: To simplify and speed up specific antibiotic residue detection, a low-cost, passive, and single-step method to fractionate analytes in raw milk was developed. METHODS: Untreated raw milk was fractionated into its water and fat/protein phases using a Fractionation of Milk for Trace Analysis of Contaminants and Residues for Antibiotics (FraMiTrACR® AB) fractionation unit. The water fraction was then analyzed by LC-MS/MS. The analyte fractionation method was evaluated against a Quick Easy Cheap Effective Rugged and Safe (QuEChERS)-based method for sample preparation. RESULTS: Our method allows qualitative and quantitative detection of substances from the penicillin, cephalosporin, macrolide, lincosamide, sulfonamide, tetracycline, and fluoroquinolone groups of antibiotics. Detection limits are below the legally prescribed maximum residue levels, allowing reliable, specific, and rapid validation of a positive result in nonspecific microbiological inhibitor tests. CONCLUSION: Analyte fractionation by FraMiTrACR AB is a faster alternative to QuEChERS-based sample preparation for the detection of antibiotic substances in milk. HIGHLIGHT: This method describes a low-cost, environmentally friendly, passive, and single-step milk analyte fractionation. As an alternative to QuEChERS-based preparation, this fractionation method simplifies and speeds up the process for specific antibiotic residue detection.


Subject(s)
Anti-Bacterial Agents , Drug Residues , Milk , Tandem Mass Spectrometry , Milk/chemistry , Anti-Bacterial Agents/analysis , Animals , Drug Residues/analysis , Tandem Mass Spectrometry/methods , Chromatography, Liquid/methods , Food Contamination/analysis , Chemical Fractionation/methods
3.
J Pediatr Orthop ; 44(4): e357-e360, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38273462

ABSTRACT

OBJECTIVE: Despite idiopathic toe walking (ITW) being a significant source of stress and anxiety for children and parents alike, little is known about the effect on health-related quality of life (HRQoL). The primary research question for this study was "Is ITW associated with impaired HRQoL, and is the degree of equinus contracture related to the degree of impairment?" METHODS: Twelve pediatric orthopaedic centers across the United Kingdom participated in this prospective, cross-sectional observational study of children younger than 18 years with ITW. Data were collected between May 2022 and July 2022. Using a standardized, piloted proforma, data collected included: demographics, toe-walking duration, passive ankle range of motion (Silfverskiold test), associated autism spectrum disorder or attention deficit hyperactivity disorder, previous and planned treatments, and Oxford Ankle Foot Questionnaire for Children scores. Domain scores were compared with a healthy control group and correlation was made to plantarflexion contracture using standard nonparametric statistical methods. RESULTS: Data were collected from 157 children. Significant reductions in physical, school and play, and emotional domain scores were noted compared with healthy controls. A significant moderate correlation was noted between passive ankle dorsiflexion and physical domain scores. There were no significant differences in Oxford Ankle Foot Questionnaire for Children scores among patient groups by treatment. CONCLUSIONS: ITW in children is associated with an impairment in HRQoL, not only across the physical domain but also the school and play and emotional domains. The more severe the equinus contracture, the worse the physical domain scores. LEVEL OF EVIDENCE: Level II-prospective cross-sectional observational study.


Subject(s)
Autism Spectrum Disorder , Equinus Deformity , Movement Disorders , Child , Humans , Walking , Cross-Sectional Studies , Quality of Life , Prospective Studies , Toes , Gait
4.
J Virol Methods ; 310: 114628, 2022 12.
Article in English | MEDLINE | ID: mdl-36209765

ABSTRACT

Enrichment of viral infectious titers following its propagation by cell culture is desirable for various experimental studies. The performance of an ultrafiltration (UF) process to concentrate infectious titers of non-enveloped Canine parvovirus 2 (CPV-2) and enveloped Feline coronavirus (FCoV) obtained from cell culture supernatants was evaluated in this study, and compared with ultracentrifugation (UC) process. A mean gain of > 1.0 log10 TCID50/mL was obtained for CPV-2 with UF, which was comparable with the gain obtained by UC. On the other hand, the gain was lower (0.7-1.0 log10 TCID50/mL) for FCoV with UF in contrast to UC (> 2.0 log10 TCID50/mL). However, the lower retentate volume following UC (∼120 fold) compared to that following UF (∼10 fold) for either of the viruses suggests a trend of increased infectious titer retention in UF concentrates relative to UC concentrates. The simplistic UF process evaluated here thus has the potential for use in applications requiring increased infectious titers of CPV-2 and FCoV.


Subject(s)
Coronavirus, Feline , Parvovirus, Canine , Viruses , Cats , Dogs , Animals , Ultrafiltration , Cell Culture Techniques
5.
Life (Basel) ; 11(12)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34947837

ABSTRACT

Multibody optimisation approaches have not seen much use in routine clinical applications despite evidence of improvements in modelling through a reduction in soft tissue artifacts compared to the standard gait analysis technique of direct kinematics. To inform clinical use, this study investigated the consistency with which both approaches predicted post-surgical outcomes, using changes in Gait Profile Score (GPS) when compared to a clinical assessment of outcome that did not include the 3D gait data. Retrospective three-dimensional motion capture data were utilised from 34 typically developing children and 26 children with cerebral palsy who underwent femoral derotation osteotomies as part of Single Event Multi-Level Surgeries. Results indicated that while, as expected, the GPS estimated from the two methods were numerically different, they were strongly correlated (Spearman's ρ = 0.93), and no significant differences were observed between their estimations of change in GPS after surgery. The two scores equivalently classified a worsening or improvement in the gait quality in 93% of the cases. When compared with the clinical classification of responders versus non-responders to the intervention, an equivalent performance was found for the two approaches, with 27/41 and 28/41 cases in agreement with the clinical judgement for multibody optimisation and direct kinematics, respectively. With this equivalent performance to the direct kinematics approach and the benefit of being less sensitive to skin artefact and allowing additional analysis such as estimation of musculotendon lengths and joint contact forces, multibody optimisation has the potential to improve the clinical decision-making process in children with cerebral palsy.

6.
Pharmacol Res Perspect ; 9(4): e00828, 2021 08.
Article in English | MEDLINE | ID: mdl-34327875

ABSTRACT

Prolongation of the cardiac action potential (AP) and early after depolarizations (EADs) are electrical anomalies of cardiomyocytes that can lead to lethal arrhythmias and are potential liabilities for existing drugs and drug candidates in development. For example, long QT syndrome-3 (LQTS3) is caused by mutations in the Nav 1.5 sodium channel that debilitate channel inactivation and cause arrhythmias. We tested the hypothesis that a useful drug (i.e., mexiletine) with potential liabilities (i.e., potassium channel inhibition and adverse reactions) could be re-engineered by dynamic medicinal chemistry to afford a new drug candidate with greater efficacy and less toxicity. Human cardiomyocytes were generated from LQTS3 patient-derived induced pluripotent stem cells (hIPSCs) and normal hIPSCs to determine beneficial (on-target) and detrimental effects (off-target) of mexiletine and synthetic analogs, respectively. The approach combined "drug discovery" and "hit to lead" refinement and showed that iterations of medicinal chemistry and physiological testing afforded optimized compound 22. Compared to mexiletine, compound 22 showed a 1.85-fold greater AUC and no detectable CNS toxicity at 100 mg/kg. In vitro hepatic metabolism studies showed that 22 was metabolized via cytochrome P-450, as previously shown, and by the flavin-containing monooxygenase (FMO). Deuterated-22 showed decreased metabolism and showed acceptable cardiovascular and physicochemical properties.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Mexiletine/analogs & derivatives , Mexiletine/pharmacokinetics , Myocytes, Cardiac/metabolism , Animals , Behavior, Animal/drug effects , Cells, Cultured , Female , Humans , Liver/metabolism , Long QT Syndrome , Male , Mexiletine/adverse effects , Mice, Inbred BALB C , Rats, Sprague-Dawley , Seizures/chemically induced
7.
Bioorg Med Chem Lett ; 46: 128162, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34062251

ABSTRACT

In the United States, approximately one million individuals are hospitalized every year for arrhythmias, making arrhythmias one of the top causes of healthcare expenditures. Mexiletine is currently used as an antiarrhythmic drug but has limitations. The purpose of this work was to use normal and Long QT syndrome Type 3 (LQTS3) patient-derived human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to identify an analog of mexiletine with superior drug-like properties. Compared to racemic mexiletine, medicinal chemistry optimization of substituted racemic pyridyl phenyl mexiletine analogs resulted in a more potent sodium channel inhibitor with greater selectivity for the sodium over the potassium channel and for late over peak sodium current.


Subject(s)
Cardiac Conduction System Disease/pathology , Induced Pluripotent Stem Cells/chemistry , Long QT Syndrome/pathology , Mexiletine/pharmacology , Myocytes, Cardiac/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Pyridines/pharmacology , Dose-Response Relationship, Drug , Humans , Mexiletine/chemistry , Molecular Structure , Pyridines/chemistry , Structure-Activity Relationship
8.
J Med Chem ; 64(9): 5384-5403, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33942619

ABSTRACT

Ventricular cardiac arrhythmia (VA) arises in acquired or congenital heart disease. Long QT syndrome type-3 (LQT3) is a congenital form of VA caused by cardiac sodium channel (INaL) SCN5A mutations that prolongs cardiac action potential (AP) and enhances INaL current. Mexiletine inhibits INaL and shortens the QT interval in LQT3 patients. Above therapeutic doses, mexiletine prolongs the cardiac AP. We explored structure-activity relationships (SAR) for AP shortening and prolongation using dynamic medicinal chemistry and AP kinetics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using patient-derived LQT3 and healthy hiPSC-CMs, we resolved distinct SAR for AP shortening and prolongation effects in mexiletine analogues and synthesized new analogues with enhanced potency and selectivity for INaL. This resulted in compounds with decreased AP prolongation effects, increased metabolic stability, increased INaL selectivity, and decreased avidity for the potassium channel. This study highlights using hiPSC-CMs to guide medicinal chemistry and "drug development in a dish".


Subject(s)
Anti-Arrhythmia Agents/chemistry , Cardiac Conduction System Disease/pathology , Long QT Syndrome/pathology , Mexiletine/analogs & derivatives , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Behavior, Animal/drug effects , Cardiac Conduction System Disease/metabolism , Cells, Cultured , Drug Design , Drug Stability , Half-Life , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome/metabolism , Male , Mexiletine/pharmacology , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Cell Chem Biol ; 28(5): 625-635.e5, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33503403

ABSTRACT

Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through ß-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.


Subject(s)
Molecular Probes/pharmacology , Protein Serine-Threonine Kinases/metabolism , Small Molecule Libraries/pharmacology , TCF Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Cells, Cultured , Humans , Male , Molecular Probes/chemistry , Small Molecule Libraries/chemistry , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , Xenopus , Zebrafish , beta Catenin/genetics , beta Catenin/metabolism
10.
Invest New Drugs ; 39(1): 131-141, 2021 02.
Article in English | MEDLINE | ID: mdl-32915418

ABSTRACT

Today, pancreatic cancer (PC) is a major health problem in the United States. It remains a challenge to develop efficacious clinically useful PC therapies. New avenues, based on translational approaches and innovative validated biomarkers could be a preclinical option to evaluate PC drug candidates or drug combinations before clinical trials. Herein, we describe evaluation of combination therapies by incorporating a novel pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) with other FDA-approved cancer drugs that have been used in PC clinical trials. PAWI-2 is a potent inhibitor of drug-resistant PC cells that has been shown to selectively ameliorate human pancreatic cancer stem cells (i.e., hPCSCs, FGß3 cells). In the present study, we showed PAWI-2 produced therapeutic synergism with certain types of anti-cancer drugs. These drugs themselves oftentimes do not ameliorate PC cells (especially PCSCs) due to high levels of drug-resistance. PAWI-2 has the ability to rescue the potency of drugs (i.e., erlotinib, trametinib) and inhibit PC cell growth. Key molecular regulators of PAWI-2 could be used to predict synergistic/antagonistic effects between PAWI-2 and other anti-cancer drugs. Anti-cancer results showed potency could be quite accurately correlated to phosphorylation of optineurin (OPTN) in PC cells. Synergism/antagonism was also associated with inhibition of PCSC marker SOX2 that was observed in FGß3 cells. Synergism broadens the potential use of PAWI-2 as an adjunct chemotherapy in patients with PC that have developed resistance to first-line targeted therapies or chemotherapies.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Pancreatic Neoplasms/pathology , Quinoxalines/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Membrane Transport Proteins/drug effects , Neoplastic Stem Cells/drug effects , Quinoxalines/administration & dosage , SOXB1 Transcription Factors/drug effects
11.
J Anal Toxicol ; 2020 Dec 26.
Article in English | MEDLINE | ID: mdl-33367644

ABSTRACT

Organophosphate (OP) pesticides are commonly utilized worldwide for agricultural purposes and pose a health threat through air, ground, and water contamination. Here, we present a convenient method for diagnosing exposure to OP pesticides in humans. This immunoprecipitation method relies on extraction of butyrylcholinesterase (BChE), a biomarker of OP poisoning that adducts OP compounds, from human serum using agarose beads conjugated to anti-BChE antibodies. Extracted BChE was then digested with pepsin and analyzed for unadducted and OP-adducted peptides by high performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS). To characterize and validate this method, pooled human plasma was exposed to parathion and dichlorvos to form diethoxyphospho, aged ethoxyphospho and dimethoxyphospho adducts with BChE. Untreated plasma was also analyzed for unadducted peptides. Additionally, samples were analyzed using Ellman's assay to measure BChE functional activity. The percent inhibition of BChE was 53.5±5.76 and 95.2±0.37%, respectively, for plasma treated with parathion for 1 hour and 24 hours. The percent inhibition was 97.2±0.98 for plasma treated with dichlorvos for 1 hour. The percent inhibition was 97.9±0.41% when the plasma treated with parathion for 1 hour, parathion for 24 hour and dichlorvos for 1 hour were mixed. Individual adducts were quantified in a single chromatographic run. Untreated plasma contained 26.4±1.87 ng/mL of unadducted BChE and no adducted peptides. In contrast, the plasma sample treated with both pesticides contained no unadducted BChE, but did contain 9.46±1.10, 10.9±0.98 and 14.1±1.10 ng/mL of diethoxyphospho, aged-ethoxy, and dimethoxyphospho peptides, respectively. The ability to identify and measure BChE and BChE adducts to parathion and dichlorvos is expected to be useful for diagnosing human exposure to multiple OP pesticides.

12.
Tissue Eng Part C Methods ; 26(11): 577-589, 2020 11.
Article in English | MEDLINE | ID: mdl-33086948

ABSTRACT

Despite considerable research effort, there is a significant need for safe agents that stimulate bone formation. Treatment of large or complex bone defects remains a challenge. Implantation of small molecule-induced human bone marrow-derived mesenchymal stromal cells (hBMSCs) on an appropriate tricalcium phosphate (TCP) scaffold offers a robust system for noninvasive therapy for spinal fusion. To show the efficacy of this approach, we identified a small molecule curcuminoid that when combined with TCP ceramic in the presence of hBMSCs selectively induced growth of bone cells: after 8- or 25-day incubations, alkaline phosphatase was elevated. Treatment of hBMSCs with curcuminoid 1 and TCP ceramic increased osteogenic target gene expression (i.e., Runx2, BMP2, Osteopontin, and Osteocalcin) over time. In the presence of curcuminoid 1 and TCP ceramic, osteogenesis of hBMSCs, including proliferation, differentiation, and mineralization, was observed. No evidence of chondrogenic or adipogenic potential using this protocol was observed. Transplantation of curcuminoid 1-treated hBMSC/TCP mixtures into the spine of immunodeficient rats showed that it achieved spinal fusion and provided greater stability of the spinal column than untreated hBMSC-TCP implants or TCP alone implants. On the basis of histological analysis, greater bone formation was associated with curcuminoid 1-treated hBMSC implants manifested as contiguous growth plates with extensive hematopoietic territories. Stimulation of hBMSCs by administration of small molecule curcuminoid 1 in the presence of TCP ceramic afforded an effective noninvasive strategy that increased spinal fusion repair and provided greater stability of the spinal column after 8 weeks in immunodeficient rats. Impact statement Bone defects only slowly regenerate themselves in humans. Current procedures to restore spinal defects are not always effective. Some have side effects. In this article, a new method to produce bone growth within 8 weeks in rats is presented. In the presence of tricalcium phosphate ceramic, curcuminoid-1 small molecule-stimulated human bone marrow-derived mesenchymal stromal cells showed robust bone cell growth in vitro. Transplantation of this mixture into the spine showed efficient spinal fusion in rats. The approach presented herein provides an efficient biocompatible scaffold for delivery of a potentially clinically useful system that could be applicable in patients.


Subject(s)
Calcium Phosphates/pharmacology , Ceramics/pharmacology , Diarylheptanoids/pharmacology , Spinal Fusion , Alkaline Phosphatase/metabolism , Animals , Calcium/metabolism , Calcium Phosphates/chemistry , Cell Differentiation/drug effects , Cell Survival/drug effects , Collagen/pharmacology , Diarylheptanoids/chemistry , Gene Expression Regulation/drug effects , Humans , Implants, Experimental , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Minerals/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Rats, Nude , Tissue Scaffolds/chemistry , Wnt Proteins/metabolism
13.
Cell Stem Cell ; 27(5): 813-821.e6, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32931730

ABSTRACT

Modeling cardiac disorders with human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes is a new paradigm for preclinical testing of candidate therapeutics. However, disease-relevant physiological assays can be complex, and the use of hiPSC-cardiomyocyte models of congenital disease phenotypes for guiding large-scale screening and medicinal chemistry have not been shown. We report chemical refinement of the antiarrhythmic drug mexiletine via high-throughput screening of hiPSC-CMs derived from patients with the cardiac rhythm disorder long QT syndrome 3 (LQT3) carrying SCN5A sodium channel variants. Using iterative cycles of medicinal chemistry synthesis and testing, we identified drug analogs with increased potency and selectivity for inhibiting late sodium current across a panel of 7 LQT3 sodium channel variants and suppressing arrhythmic activity across multiple genetic and pharmacological hiPSC-CM models of LQT3 with diverse backgrounds. These mexiletine analogs can be exploited as mechanistic probes and for clinical development.


Subject(s)
Induced Pluripotent Stem Cells , Action Potentials , Anti-Arrhythmia Agents/pharmacology , Humans , Myocytes, Cardiac , Patch-Clamp Techniques
14.
Curr Top Med Chem ; 20(26): 2344-2361, 2020.
Article in English | MEDLINE | ID: mdl-32819246

ABSTRACT

Embryonic stem cells (ESCs) are stem cells (SCs) that can self-renew and differentiate into a myriad of cell types. The process of developing stemness is determined by signaling molecules that drive stem cells to a specific lineage. For example, ESCs can differentiate into mature cells (e.g., cardiomyocytes) and mature cardiomyocytes can be characterized for cell beating, action potential, and ion channel function. A goal of this Perspective is to show how small molecules can be used to differentiate ESCs into cardiomyocytes and how this can reveal novel aspects of SC biology. This approach can also lead to the discovery of new molecules of use in cardiovascular disease. Human induced pluripotent stem cells (hiPSCs) afford the ability to produce unlimited numbers of normal human cells. The creation of patient-specific hiPSCs provides an opportunity to study cell models of human disease. The second goal is to show that small molecules can stimulate hiPSC commitment to cardiomyocytes. How iPSCs can be used in an approach to discover new molecules of use in cardiovascular disease will also be shown in this study. Adult SCs, including mesenchymal stem cells (MSCs), can likewise participate in self-renewal and multilineage differentiation. MSCs are capable of differentiating into osteoblasts, adipocytes or chondrocytes. A third goal of this Perspective is to describe differentiation of MSCs into chondrogenic and osteogenic lineages. Small molecules can stimulate MSCs to specific cell fate both in vitro and in vivo. In this Perspective, some recent examples of applying small molecules for osteogenic and chondrogenic cell fate determination are summarized. Underlying molecular mechanisms and signaling pathways involved are described. Small molecule-based modulation of stem cells shows insight into cell regulation and potential approaches to therapeutic strategies for MSC-related diseases.


Subject(s)
Bone and Bones/metabolism , Chondrocytes/metabolism , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Small Molecule Libraries/metabolism , Adipocytes/metabolism , Animals , Ascorbic Acid/metabolism , Bone and Bones/cytology , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Dimethyl Sulfoxide/metabolism , Drug Evaluation, Preclinical , Humans , Hydrazones/metabolism , Hyperbaric Oxygenation , Induced Pluripotent Stem Cells/cytology , Ion Channels/metabolism , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/cytology , Osteoblasts/metabolism , Serine/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
15.
Sci Rep ; 10(1): 9162, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32514015

ABSTRACT

Today, pancreatic cancer (PC) remains a major health problem in the US. The fact that cancer stem cells (CSCs) become enriched in humans following anti-cancer therapy implicates CSCs as key contributors to tumor dormancy, metastasis, and relapse in PC. A highly validated CSC model (FGß3 cells) was used to test a novel compound (PAWI-2) to eradicate CSCs. Compared to parental bulk FG cells, PAWI-2 showed greater potency to inhibit cell viability and self-renewal capacity of FGß3 cells. For FGß3 cells, dysregulated integrin ß3-KRAS signaling drives tumor progression. PAWI-2 inhibited ß3-KRAS signaling independent of KRAS. This is clinically relevant. PAWI-2 targeted the downstream TBK1 phosphorylation cascade that was negatively regulated by optineurin phosphorylation via a feedback mechanism. This was confirmed by TBK1 genetic knockdown or co-treatment with TBK1-specific inhibitor (MRT67307). PAWI-2 also overcame erlotinib (an EGFR inhibitor) resistance in FGß3 cells more potently than bortezomib. In the proposed working model, optineurin acts as a key regulator to link inhibition of KRAS signaling and cell cycle arrest (G2/M). The findings show PAWI-2 is a new approach to reverse tumor stemness that resensitizes CSC tumors to drug inhibition.


Subject(s)
Cell Cycle Checkpoints/drug effects , Drug Resistance, Neoplasm/drug effects , Integrin beta3/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Quinoxalines/pharmacology , Antineoplastic Agents , Humans , NF-kappa B/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured
16.
Drug Metab Dispos ; 48(2): 106-115, 2020 02.
Article in English | MEDLINE | ID: mdl-31727673

ABSTRACT

Oxycodone is used as a potent analgesic medication. Oxycodone is extensively metabolized. To fully describe its metabolism, the oxygenation of oxycodone to oxycodone N-oxide was investigated in hepatic preparations. The hypothesis tested was that oxycodone N-oxygenation was enzymatic and the amount of N-oxide detected was a consequence of both oxygenation and retro-reduction. Methods for testing the hypothesis included both in vitro and in vivo studies. Results indicated that oxycodone was N-oxygenated by the flavin-containing monooxygenase. Oxycodone N-oxide is chemically quite stable but in the presence of hepatic preparations and NADPH was retro-reduced to its parent compound oxycodone. Subsequently, oxycodone was metabolized to other metabolites including noroxycodone, noroxymorphone, and oxymorphone via cytochrome P-450. Retro-reduction of oxycodone N-oxide to oxycodone was facilitated by quinone reductase, aldehyde oxidase, and hemoglobin but not to a great extent by cytochrome P-450 or the flavin-containing monooxygenase. To confirm the in vitro observations, oxycodone was administered to rats and humans. In good agreement with in vitro results, substantial oxycodone N-oxide was observed in urine after oxycodone administration to rats and humans. Administration of oxycodone N-oxide to rats showed substantial amount of recovered oxycodone N-oxide. In vivo, noroxycodone was formed as a major rat urinary metabolite from oxycodone N-oxide presumably after retro-reduction to oxycodone and oxidative N-demethylation. To a lesser extent, oxycodone, noroxymorphone, and oxymorphone were observed as urinary metabolites. SIGNIFICANCE STATEMENT: This manuscript describes the N-oxygenation of oxycodone in vitro as well as in small animals and humans. A new metabolite was quantified as oxycodone N-oxide. Oxycodone N-oxide undergoes extensive retro-reduction to oxycodone. This re-establishes the metabolic profile of oxycodone and introduces new concepts about a metabolic futile cycle related to oxycodone metabolism.


Subject(s)
Oxides/metabolism , Oxycodone/metabolism , Analgesics, Opioid/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Female , Hemoglobins/metabolism , Humans , Male , Mixed Function Oxygenases/metabolism , Morphinans/metabolism , NADP/metabolism , Oxymorphone/metabolism , Rats
17.
Biochem J ; 476(22): 3505-3520, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31682720

ABSTRACT

The mosquitoes of the Anopheles and Aedes genus are some of the most deadly insects to humans because of their effectiveness as vectors of malaria and a range of arboviruses, including yellow fever, dengue, chikungunya, West Nile and Zika. The use of insecticides from different chemical classes is a key component of the integrated strategy against An. gambiae and Ae. aegypti, but the problem of insecticide resistance means that new compounds with different modes of action are urgently needed to replace chemicals that fail to control resistant mosquito populations. We have previously shown that feeding inhibitors of peptidyl dipeptidase A to both An. gambiae and Ae. aegypti mosquito larvae lead to stunted growth and mortality. However, these compounds were designed to inhibit the mammalian form of the enzyme (angiotensin-converting enzyme, ACE) and hence can have lower potency and lack selectivity as inhibitors of the insect peptidase. Thus, for the development of inhibitors of practical value in killing mosquito larvae, it is important to design new compounds that are both potent and highly selective. Here, we report the first structures of AnoACE2 from An. gambiae in its native form and with a bound human ACE inhibitor fosinoprilat. A comparison of these structures with human ACE (sACE) and an insect ACE homologue from Drosophila melanogaster (AnCE) revealed that the AnoACE2 structure is more similar to AnCE. In addition, important elements that differ in these structures provide information that could potentially be utilised in the design of chemical leads for selective mosquitocide development.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Anopheles/enzymology , Insect Proteins/chemistry , Peptidyl-Dipeptidase A/chemistry , Aedes/chemistry , Aedes/enzymology , Aedes/genetics , Animals , Anopheles/chemistry , Anopheles/genetics , Anopheles/growth & development , Drosophila melanogaster/chemistry , Drosophila melanogaster/enzymology , Fosinopril/analogs & derivatives , Fosinopril/chemistry , Humans , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Insect Proteins/metabolism , Insecticides/chemistry , Larva/chemistry , Larva/enzymology , Larva/genetics , Larva/growth & development , Models, Molecular , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism
18.
J Pharmacol Exp Ther ; 371(3): 703-712, 2019 12.
Article in English | MEDLINE | ID: mdl-31582422

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-related death for men in the United States. Approximately 35% of PCa recurs and is often transformed to castration-resistant prostate cancer (CRPCa), the most deadly and aggressive form of PCa. However, the CRPCa standard-of-care treatment (enzalutamide with abiraterone) usually has limited efficacy. Herein, we report a novel molecule (PAWI-2) that inhibits cellular proliferation of androgen-sensitive and androgen-insensitive cells (LNCaP and PC-3, respectively). In vivo studies in a PC-3 xenograft model showed that PAWI-2 (20 mg/kg per day i.p., 21 days) inhibited tumor growth by 49% compared with vehicle-treated mice. PAWI-2 synergized currently clinically used enzalutamide in in vitro inhibition of PCa cell viability and resensitized inhibition of in vivo PC-3 tumor growth. Compared with vehicle-treated mice, PC-3 xenograft studies also showed that PAWI-2 (20 mg/kg per day i.p., 21 days) and enzalutamide (5 mg/kg per day i.p., 21 days) inhibited tumor growth by 63%. Synergism was mainly controlled by the imbalance of prosurvival factors (e.g., Bcl-2, Bcl-xL, Mcl-1) and antisurvival factors (e.g., Bax, Bak) induced by affecting mitochondrial membrane potential/mitochondria dynamics. Thus, PAWI-2 utilizes a distinct mechanism of action to inhibit PCa growth independently of androgen receptor signaling and overcomes enzalutamide-resistant CRPCa. SIGNIFICANCE STATEMENT: Castration-resistant prostate cancer (CRPCa) is the most aggressive human prostate cancer (PCa) but standard chemotherapies for CRPCa are largely ineffective. PAWI-2 potently inhibits PCa proliferation in vitro and in vivo regardless of androgen receptor status and uses a distinct mechanism of action. PAWI-2 has greater utility in treating CRPCa than standard-of-care therapy. PAWI-2 possesses promising therapeutic potency in low-dose combination therapy with a clinically used drug (e.g., enzalutamide). This study describes a new approach to address the overarching challenge in clinical treatment of CRPCa.


Subject(s)
Antineoplastic Agents/pharmacology , Phenylthiohydantoin/analogs & derivatives , Piperazines/pharmacology , Prostatic Neoplasms/drug therapy , Quinoxalines/pharmacology , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Benzamides , Cell Line, Tumor , Drug Synergism , Humans , Male , Mice , Nitriles , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , Xenograft Model Antitumor Assays
19.
Sci Rep ; 9(1): 10811, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346210

ABSTRACT

NOTCH plays a pivotal role during normal development and in congenital disorders and cancer. γ-secretase inhibitors are commonly used to probe NOTCH function, but also block processing of numerous other proteins. We discovered a new class of small molecule inhibitor that disrupts the interaction between NOTCH and RBPJ, which is the main transcriptional effector of NOTCH signaling. RBPJ Inhibitor-1 (RIN1) also blocked the functional interaction of RBPJ with SHARP, a scaffold protein that forms a transcriptional repressor complex with RBPJ in the absence of NOTCH signaling. RIN1 induced changes in gene expression that resembled siRNA silencing of RBPJ rather than inhibition at the level of NOTCH itself. Consistent with disruption of NOTCH signaling, RIN1 inhibited the proliferation of hematologic cancer cell lines and promoted skeletal muscle differentiation from C2C12 myoblasts. Thus, RIN1 inhibits RBPJ in its repressing and activating contexts, and can be exploited for chemical biology and therapeutic applications.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors , Receptors, Notch/metabolism , Signal Transduction/drug effects , Cell Differentiation/drug effects , Cell Line , Gene Expression Regulation/drug effects , Humans , Myoblasts/drug effects , Myoblasts/metabolism
20.
Am J Cancer Res ; 9(2): 390-405, 2019.
Article in English | MEDLINE | ID: mdl-30906636

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), constitutes >90% of pancreatic cancers (PC) and is one of the most aggressive human tumors. Standard chemotherapies for PDAC (e.g., gemcitabine, FOLFIRINOX, etc.) has proven to be largely ineffective. Herein, we report a novel molecule (i.e., compound 1) that potently inhibits proliferation and induces apoptosis of PDAC cells. As we observed in other cancer types (i.e., colorectal, breast cancer), the effect of 1 against PDAC cells is also related to microtubule destabilization and DNA damage checkpoint activation. However, in PDAC cells, the inhibitory effect of 1 was mainly controlled by mitochondrial p53-dependent apoptosis. Compound 1 worked with cells of different p53 mutant status and affected p53 activation/phosphorylation not simply by stabilizing p53 protein but through antagonizing anti-apoptotic effects of Bcl-xL and restoring p53 to activate mitochondrial-apoptotic pathways (i.e., cytochrome c release, caspase activation and PARP cleavage). Compound 1 was more efficient than a typical PDAC combination therapy (i.e., gemcitabine with paclitaxel) and showed synergism in inhibiting PDAC cell proliferation with gemcitabine (or gemcitabine with paclitaxel). This synergism varied between different types of PDAC cells and was partially controlled by the phosphorylation of p53 on Serine15 (phospho-Ser15-p53). In vivo studies in an orthotopic syngeneic murine model showed that 1 (20 mg/kg/day, 28 days, i.p.) inhibited tumor growth by 65% compared to vehicle-treated mice. No apparent acute or chronic toxicity was observed. Thus, compound 1 utilizes a distinct mechanism of action to inhibit PC growth in vitro and in vivo and is a novel anti-PDAC compound.

SELECTION OF CITATIONS
SEARCH DETAIL