Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 252
1.
Sci Rep ; 11(1): 23475, 2021 12 06.
Article En | MEDLINE | ID: mdl-34873208

Fluorescence lifetime imaging microscopy of a fluorescence probe, 3,6-bis(1-methyl-2-vinylpyridinium) carbazole diiodide (o-BMVC), provides an objective method for preoperative diagnosis of fine-needle aspiration (FNA) of thyroid nodules. The key of this o-BMVC test of FNA smears is the measurement of the digital number of o-BMVC foci in the nucleus. Thus, there are three categories classified in the o-BMVC test, which are nondiagnostic for unsatisfactory samples, benign for less numbers of o-BMVC foci, and malignant for more numbers of o-BMVC foci. The discrimination of indeterminate (including atypia, follicular neoplasm, suspicious) cytology into benign or malignant cases can reduce diagnostic uncertainty and benefit clinical decision making. This pilot study strongly suggests that the o-BMVC test is an invaluable method for diagnosing FNA samples. Particularly, the combination of FNA cytology and the o-BMVC test holds great promise to improve the efficacy of diagnosis and reduce the healthcare costs.


Thyroid Nodule/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy, Fine-Needle/methods , Carbazoles/administration & dosage , Cytodiagnosis/methods , Female , Fluorescent Dyes/administration & dosage , Humans , Male , Microscopy, Fluorescence/methods , Middle Aged , Pilot Projects , Pyridinium Compounds/administration & dosage , Thyroid Gland/pathology , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/pathology , Young Adult
2.
Nat Commun ; 12(1): 6767, 2021 11 19.
Article En | MEDLINE | ID: mdl-34799586

Nicotinamide riboside (NR) is one of the orally bioavailable NAD+ precursors and has been demonstrated to exhibit beneficial effects against aging and aging-associated diseases. However, the metabolic pathway of NR in vivo is not yet fully understood. Here, we demonstrate that orally administered NR increases NAD+ level via two different pathways. In the early phase, NR was directly absorbed and contributed to NAD+ generation through the NR salvage pathway, while in the late phase, NR was hydrolyzed to nicotinamide (NAM) by bone marrow stromal cell antigen 1 (BST1), and was further metabolized by the gut microbiota to nicotinic acid, contributing to generate NAD+ through the Preiss-Handler pathway. Furthermore, we report BST1 has a base-exchange activity against both NR and nicotinic acid riboside (NAR) to generate NAR and NR, respectively, connecting amidated and deamidated pathways. Thus, we conclude that BST1 plays a dual role as glycohydrolase and base-exchange enzyme during oral NR supplementation.


ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Glycoside Hydrolases/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/pharmacokinetics , A549 Cells , ADP-ribosyl Cyclase/genetics , Administration, Oral , Aging/drug effects , Animals , Antigens, CD/genetics , Dietary Supplements , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gastrointestinal Microbiome , Glycoside Hydrolases/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/metabolism , Intestine, Small/microbiology , Mice , Mice, Knockout , Niacin/metabolism , Niacinamide/administration & dosage , Niacinamide/metabolism , Niacinamide/pharmacokinetics , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Pyridinium Compounds/administration & dosage
3.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article En | MEDLINE | ID: mdl-34497121

Alzheimer's disease (AD) is a progressive and fatal neurodegenerative disorder. Impaired neuronal bioenergetics and neuroinflammation are thought to play key roles in the progression of AD, but their interplay is not clear. Nicotinamide adenine dinucleotide (NAD+) is an important metabolite in all human cells in which it is pivotal for multiple processes including DNA repair and mitophagy, both of which are impaired in AD neurons. Here, we report that levels of NAD+ are reduced and markers of inflammation increased in the brains of APP/PS1 mutant transgenic mice with beta-amyloid pathology. Treatment of APP/PS1 mutant mice with the NAD+ precursor nicotinamide riboside (NR) for 5 mo increased brain NAD+ levels, reduced expression of proinflammatory cytokines, and decreased activation of microglia and astrocytes. NR treatment also reduced NLRP3 inflammasome expression, DNA damage, apoptosis, and cellular senescence in the AD mouse brains. Activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are associated with DNA damage and senescence. cGAS-STING elevation was observed in the AD mice and normalized by NR treatment. Cell culture experiments using microglia suggested that the beneficial effects of NR are, in part, through a cGAS-STING-dependent pathway. Levels of ectopic (cytoplasmic) DNA were increased in APP/PS1 mutant mice and human AD fibroblasts and down-regulated by NR. NR treatment induced mitophagy and improved cognitive and synaptic functions in APP/PS1 mutant mice. Our findings suggest a role for NAD+ depletion-mediated activation of cGAS-STING in neuroinflammation and cellular senescence in AD.


Alzheimer Disease/complications , Cellular Senescence , Dietary Supplements , Membrane Proteins/metabolism , NAD/administration & dosage , Neuroinflammatory Diseases/drug therapy , Nucleotidyltransferases/metabolism , Animals , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Nucleotidyltransferases/genetics , Pyridinium Compounds/administration & dosage
4.
PLoS One ; 16(6): e0252554, 2021.
Article En | MEDLINE | ID: mdl-34061900

Acute kidney injury (AKI) is associated with a very high mortality and an increased risk for progression to chronic kidney disease (CKD). Ischemia-reperfusion injury (IRI) is a model for AKI, which results in tubular damage, dysfunction of the mitochondria and autophagy, and in decreased cellular nicotinamide adenine dinucleotide (NAD+) with progressing fibrosis resulting in CKD. NAD+ is a co-enzyme for several proteins, including the NAD+ dependent sirtuins. NAD+ augmentation, e.g. by use of its precursor nicotinamide riboside (NR), improves mitochondrial homeostasis and organismal metabolism in many species. In the present investigation the effects of prophylactic administration of NR on IRI-induced AKI were studied in the rat. Bilateral IRI reduced kidney tissue NAD+, caused tubular damage, reduced α-Klotho (klotho), and altered autophagy flux. AKI initiated progression to CKD, as shown by induced profibrotic Periostin (postn) and Inhibin subunit beta-A, (activin A / Inhba), both 24 hours and 14 days after surgery. NR restored tissue NAD+ to that of the sham group, increased autophagy (reduced p62) and sirtuin1 (Sirt1) but did not ameliorate renal tubular damage and profibrotic genes in the 24 hours and 14 days IRI models. AKI induced NAD+ depletion and impaired autophagy, while augmentation of NAD+ by NR restored tissue NAD+ and increased autophagy, possibly serving as a protective response. However, prophylactic administration of NR did not ameliorate tubular damage of the IRI rats nor rescued the initiation of fibrosis in the long-term AKI to CKD model, which is a pivotal event in CKD pathogenesis.


Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , NAD/metabolism , Niacinamide/analogs & derivatives , Protective Agents/administration & dosage , Pyridinium Compounds/administration & dosage , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Acute Kidney Injury/complications , Acute Kidney Injury/metabolism , Animals , Autophagy/drug effects , Disease Progression , Fibrosis , Glucuronidase/metabolism , Kidney/metabolism , Kidney/pathology , Klotho Proteins , Male , Mitochondria/metabolism , Niacinamide/administration & dosage , Random Allocation , Rats , Rats, Wistar , Renal Insufficiency, Chronic/etiology , Sirtuin 1/metabolism , Treatment Outcome
5.
Mol Pharm ; 18(6): 2416-2427, 2021 06 07.
Article En | MEDLINE | ID: mdl-34019427

Oxime-based molecules are used for the treatment of patients to reactivate acetylcholinesterase (AChE) function after organophosphate intoxication. However, their efficacy is limited by low penetration through the blood-brain barrier and fast elimination. In this work, the cucurbit[7]uril (CB[7]) carrier was used for the encapsulation of the clinical agent asoxime to enhance brain bioavailability and the treatment window. We present a pharmacokinetic study of asoxime and the asoxime-CB[7] complex in an in vivo mouse model. Ultrahigh-performance liquid chromatography with electrospray ionization-mass spectrometry detection was developed to determine asoxime and CB[7] in biological fluids and tissues after thorough optimization of chromatographic conditions. The dihydroxypropane-silica stationary phase using hydrophilic interaction liquid chromatography conditions provided the best chromatographic performance. The final method was validated and applied for the pharmacokinetic study of mouse plasma, urine, bile, liver, kidney, and brain samples at different times after administration of asoxime and the asoxime-CB[7] complex. The results showed a greater than 3-fold increase in the area under the curve (AUC) in the brain for asoxime administered as a complex with CB[7] relative to that for the administration of asoxime alone. The effectiveness of the treatment strategy was evaluated using a reactivation study and a functional observatory battery. Protection of brain AChE activity is crucial for saving human lives or reducing the consequences of poisoning. The asoxime administered as a complex increased the brain activity by approximately 30% compared to that with atropine alone. CB[7] coadministration improved the AChE activity by 11%, which agrees with the higher asoxime AUC assessed in the pharmacokinetic study.


Bridged-Ring Compounds/chemistry , Cholinesterase Reactivators/administration & dosage , Drug Carriers/chemistry , Imidazoles/chemistry , Organophosphate Poisoning/drug therapy , Oximes/pharmacokinetics , Pyridinium Compounds/pharmacokinetics , Acetylcholinesterase/metabolism , Animals , Area Under Curve , Blood-Brain Barrier/metabolism , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/toxicity , Cholinesterase Reactivators/pharmacokinetics , Chromatography, High Pressure Liquid , Disease Models, Animal , Enzyme Assays , Humans , Hydrophobic and Hydrophilic Interactions , Mass Spectrometry , Mice , Oximes/administration & dosage , Pyridinium Compounds/administration & dosage , Sarin/administration & dosage , Sarin/toxicity
7.
Aging Cell ; 20(4): e13329, 2021 04.
Article En | MEDLINE | ID: mdl-33734555

Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD+ levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm-/- mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention.


Ataxia Telangiectasia/diet therapy , Ataxia Telangiectasia/metabolism , Dietary Supplements , Membrane Proteins/metabolism , Mitophagy/drug effects , NAD/metabolism , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Senescence-Associated Secretory Phenotype/genetics , Signal Transduction/drug effects , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Case-Control Studies , Cell Line, Tumor , Disease Models, Animal , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , Neurons/drug effects , Neurons/metabolism , Niacinamide/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Transfection , Treatment Outcome
8.
Cell Death Dis ; 12(2): 179, 2021 02 15.
Article En | MEDLINE | ID: mdl-33589591

Human epidermal growth factor receptor 2 gene (HER2) is focally amplified in approximately 20% of breast cancers. HER2 inhibitors alone are not effective, and sensitizing agents will be necessary to move away from a reliance on heavily toxic chemotherapeutics. We recently demonstrated that the efficacy of HER2 inhibitors is mitigated by uniformly low levels of the myeloid cell leukemia 1 (MCL-1) endogenous inhibitor, NOXA. Emerging clinical data have demonstrated that clinically advanced cyclin-dependent kinase (CDK) inhibitors are effective MCL-1 inhibitors in patients, and, importantly, well tolerated. We, therefore, tested whether the CDK inhibitor, dinaciclib, could block MCL-1 in preclinical HER2-amplified breast cancer models and therefore sensitize these cancers to dual HER2/EGFR inhibitors neratinib and lapatinib, as well as to the novel selective HER2 inhibitor tucatinib. Indeed, we found dinaciclib suppresses MCL-1 RNA and is highly effective at sensitizing HER2 inhibitors both in vitro and in vivo. This combination was tolerable in vivo. Mechanistically, liberating the effector BCL-2 protein, BAK, from MCL-1 results in robust apoptosis. Thus, clinically advanced CDK inhibitors may effectively combine with HER2 inhibitors and present a chemotherapy-free therapeutic strategy in HER2-amplified breast cancer, which can be tested immediately in the clinic.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cyclic N-Oxides/pharmacology , Indolizines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridinium Compounds/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclic N-Oxides/administration & dosage , Drug Synergism , Female , Gene Amplification , Humans , Indoles/administration & dosage , Indoles/pharmacology , Indolizines/administration & dosage , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oxazoles/administration & dosage , Oxazoles/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Pyridinium Compounds/administration & dosage , Quinazolines/administration & dosage , Quinazolines/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Random Allocation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
9.
Drug Deliv ; 28(1): 433-444, 2021 Dec.
Article En | MEDLINE | ID: mdl-33605178

Nicotinamide riboside (NR), as a dietary supplement, can be converted to nicotinamide adenine dinucleotide (NAD+) in cells to support mitochondrial energy metabolism. However, the efficacy of oral administrated NR is limited due to its quick degradation in circulation and low bioavailability in targeted organs. In this study, we fabricated nanocrystal self-assembled microspheres by Nano Spray Dryer for oral delivery of NR. The structure of NR and resveratrol (RES) nanocrystal self-assembled microspheres (NR/RESms) is confirmed by the morphology, chemical structure, and crystallization. The NR/RESms displayed restricted NR release at the gastric acid-mimic condition (<15% in the first 8 hours), while achieved accelerated NR release in an enteric-mimic environment (>46% within 8 hours). Oral administration of NR/RESms for 8 hours significantly elevated NAD+ levels in serum (169.88 nM versus 30.93 nM in the NR group, p < .01; and 66.89 nM in the NR + RES group, p < .05), and enhanced NAD+ abundance in multiple organs in mice, exhibiting an improved oral NAD+ bioavailability. In addition, without any serious adverse effects on major organs, oral delivery of NR/RESms attenuated myocardial infarction (15.82% versus 19.38% in the I/R + NR group and 20.76% in the I/R + NR + RES group) in a cardiac ischemia/reperfusion (I/R) injury mouse model. Therefore, our data supported that the NR/RESms is a promising candidate as NAD+ booster for oral administration.


Drug Delivery Systems , NAD/blood , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Reperfusion Injury/drug therapy , Administration, Oral , Animals , Biological Availability , Dietary Supplements , Disease Models, Animal , Drug Liberation , Male , Mice , Mice, Inbred C57BL , Microspheres , Nanoparticles , Niacinamide/administration & dosage , Niacinamide/pharmacology , Pyridinium Compounds/pharmacology , Time Factors
10.
Int J Neuropsychopharmacol ; 24(5): 434-445, 2021 05 18.
Article En | MEDLINE | ID: mdl-33305805

BACKGROUND: N-methyl-D-aspartate (NMDA) receptor activation requires the binding of a co-agonist on the glycine-binding site. D-serine is the main endogenous co-agonist of NMDA receptors, and its availability significantly depends on the activity of the metabolic enzyme D-amino acid oxidase (DAAO). Inhibition of DAAO increases the brain levels of D-serine and modulates a variety of physiological functions, including cognitive behavior. METHODS: Here, we examined the effects of a novel 4-hydroxypyridazin-3(2H)-one derivative DAAO inhibitor, Compound 30 (CPD30), on passive avoidance learning and on neuronal firing activity in rats. RESULTS: D-serine administration was applied as reference, which increased cognitive performance and enhanced hippocampal firing activity and responsiveness to NMDA after both local and systemic application. Similarly to D-serine, CPD30 (0.1 mg/kg) effectively reversed MK-801-induced memory impairment in the passive avoidance test. Furthermore, local iontophoretic application of CPD30 in the vicinity of hippocampal pyramidal neurons significantly increased firing rate and enhanced their responses to locally applied NMDA. CPD30 also enhanced hippocampal firing activity after systemic administration. In 0.1- to 1.0-mg/kg doses, CPD30 increased spontaneous and NMDA-evoked firing activity of the neurons. Effects of CPD30 on NMDA responsiveness emerged faster (at 10 minutes post-injection) when a 1.0-mg/kg dose was applied compared with the onset of the effects of 0.1 mg/kg CPD30 (at 30 minutes post-injection). CONCLUSIONS: The present results confirm that the inhibition of DAAO enzyme is an effective strategy for cognitive enhancement. Our findings further facilitate the understanding of the cellular mechanisms underlying the behavioral effects of DAAO inhibition in the mammalian brain.


Avoidance Learning/drug effects , Behavior, Animal/drug effects , D-Amino-Acid Oxidase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Memory Disorders/drug therapy , Nootropic Agents/pharmacology , Pyramidal Cells/drug effects , Pyridinium Compounds/pharmacology , Action Potentials/drug effects , Animals , Enzyme Inhibitors/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/enzymology , Male , Memory Disorders/enzymology , N-Methylaspartate/pharmacology , Nootropic Agents/administration & dosage , Pyridinium Compounds/administration & dosage , Rats , Rats, Wistar
11.
Curr Opin Clin Nutr Metab Care ; 23(6): 413-420, 2020 11.
Article En | MEDLINE | ID: mdl-32925178

PURPOSE OF REVIEW: The present review aims to address the clinical benefits of using nicotinamide riboside, a precursor to the essential pyridine nucleotide, nicotinamide adenine dinucleotide (NAD+) as a therapeutic agent to attenuate age-related cognitive decline. RECENT FINDINGS: Oral supplementation with nicotinamide riboside can inhibit the accumulation of pathological hallmarks of Alzheimer's disease and improve learning and memory in various murine models for dementia. Nicotinamide riboside can also reduce DNA damage, neuroinflammation, apoptosis, and improved hippocampal synaptic plasticity in diabetic mice, and another Alzheimer's disease mouse model. The cognitive benefits of nicotinamide riboside in Alzheimer's disease models may be modulated in part by upregulation of proliferator-activated-γ coactivator 1α-mediated ß-secretase 1(BACE-1) ubiquitination and degradation, preventing Aß production in the brain. Nicotinamide riboside also maintained blood-brain barrier integrity and maintained the gut microbiota in a mouse model for cerebral small vessel disease and alcohol-induced depression, respectively. Oral nicotinamide riboside has been shown to be bioavailable and well tolerated in humans with limited adverse effects compared to other NAD+ precursors. SUMMARY: Oral nicotinamide riboside may represent a promising stratagem to improve cognitive decline during 'normal' ageing, Alzheimer's disease and other diseases. Results from recent clinical trials are needed to enumerate the preclinical benefits in humans.


Alzheimer Disease/therapy , Cognitive Aging/physiology , Cognitive Dysfunction/therapy , Dietary Supplements , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Cognitive Dysfunction/prevention & control , Disease Models, Animal , Humans , Mice , Niacinamide/administration & dosage
12.
Pharm Res ; 37(10): 196, 2020 Sep 17.
Article En | MEDLINE | ID: mdl-32944844

PURPOSE: Hypoxia-inducible factor (HIF) is one of the critical components of the tumor microenvironment that is involved in tumor development. HIF-1α functionally and physically interacts with CDK1, 2, and 5 and stimulates the cell cycle progression and Cyclin-Dependent Kinase (CDK) expression. Therefore, hypoxic tumor microenvironment and CDK overexpression lead to increased cell cycle progression and tumor expansion. Therefore, we decided to suppress cancer cell expansion by blocking HIF-1α and CDK molecules. METHODS: In the present study, we used the carboxylated graphene oxide (CGO) conjugated with trimethyl chitosan (TMC) and hyaluronate (HA) nanoparticles (NPs) loaded with HIF-1α-siRNA and Dinaciclib, the CDK inhibitor, for silencing HIF-1α and blockade of CDKs in CD44-expressing cancer cells and evaluated the impact of combination therapy on proliferation, metastasis, apoptosis, and tumor growth. RESULTS: The results indicated that the manufactured NPs had conceivable physicochemical properties, high cellular uptake, and low toxicity. Moreover, combination therapy of cancer cells using CGO-TMC-HA NPs loaded with HIF-1α siRNA and Dinaciclib (SCH 727965) significantly suppressed the CDKs/HIF-1α and consequently, decreased the proliferation, migration, angiogenesis, and colony formation in tumor cells. CONCLUSIONS: These results indicate the ability of CGO-TMC-HA NPs for dual drug/gene delivery in cancer treatment. Furthermore, the simultaneous inhibition of CDKs/HIF-1α can be considered as a novel anti-cancer treatment strategy; however, further research is needed to confirm this treatment in vivo. Graphical Abstract The suppression of HIF-1α and CDKs inhibits cancer growth. HIF-1α is overexpressed by the cells present in the tumor microenvironment. The hypoxic environment elevates mitochondrial ROS production and increases p38 MAP kinase, JAK/STAT, ERK, JNK, and Akt/PI3K signaling, resulting in cyclin accumulation and aberrant cell cycle progression. Furthermore, the overexpression of HIF-1α/CDK results in increased expression of genes such as BCL2, Bcl-xl, Ki-67, TGFß, VEGF, FGF, MMP2, MMP9, and, HIF-1α and consequently raise the survival, proliferation, angiogenesis, metastasis, and invasion of tumor cells. In conclusion, HIF-1α-siRNA/Dinaciclib-loaded CGO-TMC-HA NPs can inhibit the tumor expansion by blockage of CDKs and HIF-1α (JAK: Janus kinase, STAT: Signal transducer and activator of transcription, MAPK: mitogen-activated protein kinase, ERK: extracellular signal-regulated kinase, JNK: c-Jun N-terminal kinase, PI3K: phosphatidylinositol 3-kinase).


Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms, Experimental/therapy , Pyridinium Compounds/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chitosan/chemistry , Cyclic N-Oxides , Graphite/chemistry , Hyaluronic Acid/chemistry , Indolizines , Mice , Nanoparticles/chemistry , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacokinetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics
13.
Clin Transl Sci ; 13(6): 1178-1188, 2020 11.
Article En | MEDLINE | ID: mdl-32738099

The combination of drugs targeting Ral and PI3K/AKT signaling has antitumor efficacy in preclinical models of pancreatic cancer. We combined dinaciclib (small molecule cyclin dependent kinase inhibitor with MK-2206 (Akt inhibitor) in patients with previously treated/metastatic pancreatic cancer. Patients were treated with dinaciclib (6-12 mg/m2 i.v.) and MK-2206 (60-135 mg p.o.) weekly. Tumor biopsies were performed to measure pAKT, pERK, and Ki67 at baseline and after one completed cycle (dose level 2 and beyond). Thirty-nine patients participated in the study. The maximum tolerated doses were dinaciclib 9 mg/m2 and MK-2206 135 mg. Treatment-related grade 3 and 4 toxicities included neutropenia, lymphopenia, anemia, hyperglycemia, hyponatremia, and leukopenia. No objectives responses were observed. Four patients (10%) had stable disease as their best response. At the recommended dose, median survival was 2.2 months. Survival rates at 6 and 12 months were 11% and 5%, respectively. There was a nonsignificant reduction in pAKT composite scores between pretreatment and post-treatment biopsies (mean 0.76 vs. 0.63; P = 0.635). The combination of dinaciclib and MK-2206 was a safe regimen in patients with metastatic pancreatic cancer, although without clinical benefit, possibly due to not attaining biologically effective doses. Given the strong preclinical evidence of Ral and AKT inhibition, further studies with better tolerated agents should be considered.


Antineoplastic Combined Chemotherapy Protocols/toxicity , Carcinoma, Pancreatic Ductal/drug therapy , Cyclic N-Oxides/toxicity , Heterocyclic Compounds, 3-Ring/toxicity , Indolizines/toxicity , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/toxicity , Pyridinium Compounds/toxicity , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biopsy , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cyclic N-Oxides/administration & dosage , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Indolizines/administration & dosage , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Pyridinium Compounds/administration & dosage , Survival Rate , Treatment Outcome
14.
BMC Nephrol ; 21(1): 342, 2020 08 13.
Article En | MEDLINE | ID: mdl-32791973

BACKGROUND: Preclinical studies have identified both NAD+ and sirtuin augmentation as potential strategies for the prevention and treatment of AKI. Nicotinamide riboside (NR) is a NAD+ precursor vitamin and pterostilbene (PT) is potent sirtuin activator found in blueberries. Here, we tested the effect of combined NR and PT (NRPT) on whole blood NAD+ levels and safety parameters in patients with AKI. METHODS: We conducted a randomized, double-blind, placebo-controlled study of escalating doses of NRPT in 24 hospitalized patients with AKI. The study was comprised of four Steps during which NRPT (5 subjects) or placebo (1 subject) was given twice a day for 2 days. NRPT dosing was increased in each Step: Step 1250/50 mg, Step 2500/100 mg, Step 3750/150 mg and Step 41,000/200 mg. Blood NAD+ levels were measured by liquid chromatography-mass spectrometry and safety was assessed by history, physical exam, and clinical laboratory testing. RESULTS: AKI resulted in a 50% reduction in whole blood NAD+ levels at 48 h compared to 0 h in patients receiving placebo (p = 0.05). There was a trend for increase in NAD+ levels in all NRPT Steps individually at 48 h compared to 0 h, but only the change in Step 2 reached statistical significance (47%, p = 0.04), and there was considerable interindividual variability in the NAD+ response to treatment. Considering all Steps together, NRPT treatment increased NAD+ levels by 37% at 48 h compared to 0 h (p = 0.002). All safety laboratory tests were unchanged by NRPT treatment, including creatinine, estimated glomerular filtration rate (eGFR), electrolytes, liver function tests, and blood counts. Three of 20 patients receiving NRPT reported minor gastrointestinal side effects. CONCLUSION: NRPT increases whole blood NAD+ levels in hospitalized patients with AKI. In addition, NRPT up to a dose of 1000 mg/200 mg twice a day for 2 days is safe and well tolerated in these patients. Further studies to assess the potential therapeutic benefit of NRPT in AKI are warranted. TRIAL REGISTRATION: NCT03176628 , date of registration June 5th, 2017.


Acute Kidney Injury/drug therapy , Creatinine/blood , NAD/blood , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Stilbenes/administration & dosage , Acute Kidney Injury/blood , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Female , Glomerular Filtration Rate , Humans , Male , Middle Aged , Niacinamide/administration & dosage , Niacinamide/therapeutic use , Pilot Projects , Pyridinium Compounds/therapeutic use , Stilbenes/therapeutic use
15.
CNS Neurosci Ther ; 26(4): 438-447, 2020 04.
Article En | MEDLINE | ID: mdl-31943833

AIMS: Hypertension is a leading cause of cerebral small vessel disease (CSVD). Currently, treatments for CSVD are limited. Nicotinamide riboside (NR) can protect against vascular injury and cognitive impairment in neurodegenerative diseases. In this study, the protective effects of NR against angiotensin - (Ang -)-induced CSVD were evaluated. METHODS: To explore the effects of NR in CSVD, C57BL/6 mice were infused with Ang -, and NR was added to the food of the mice for 28 days. Then, short-term memory, blood-brain barrier (BBB) integrity, and endothelial function were detected. Arteriole injury and glial activation were also evaluated. RESULTS: Our data showed that mice infused with Ang - exhibited decreased short-term memory function and BBB leakage due to decreased claudin-5 expression and increased caveolae-mediated endocytosis after 28 days. Furthermore, Ang - decreased the expression of α-smooth muscle actin (α-SMA) and increased the expression of proliferating cell nuclear antigen (PCNA) in arterioles and decreased the expression of neurofilament 200 (NF200) and myelin basic protein (MBP) in the white matter. These CSVD-related damages induced by Ang - were inhibited by NR administration. Moreover, NR administration significantly reduced glial activation around the vessels. CONCLUSION: Our results indicated that NR administration alleviated Ang --induced CSVD by protecting BBB integrity, vascular remodeling, neuroinflammation, and white matter injury (WMI)-associated cognitive impairment.


Angiotensin II/toxicity , Cerebral Small Vessel Diseases/chemically induced , Cerebral Small Vessel Diseases/drug therapy , Niacinamide/analogs & derivatives , Pyridinium Compounds/administration & dosage , Animals , Cerebral Small Vessel Diseases/pathology , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Niacinamide/administration & dosage
16.
Toxicol Lett ; 320: 64-72, 2020 Mar 01.
Article En | MEDLINE | ID: mdl-31794810

Oxime-based acetylcholinesterase reactivators (briefly oximes) regenerate organophosphate-inactivated acetylcholinesterase and restore its function. Poor blood-brain-barrier passage and fast elimination from blood limit their actual use in treatment of patients exposed to organophosphates. Previous in vitro results implicated further testing of cucurbit[7]uril as a delivery vehicle for bisquaternary oximes. The present paper focuses on cell toxicity, in vivo safety and influence of cucurbit[7]uril on oxime pharmacokinetics and pharmacodynamics. Neither the K027 nor the complex caused any cell toxicity, changes in blood biochemistry or hepato- or nephrotoxicity in tested concentrations. The encapsulation of K027 increased and accelerated the blood-brain-barrier penetration. The peripheral oxime exposure also increased, supporting the suggestion that cucurbit[7]uril protects the circulating oxime from rapid renal clearance. Contrary to the comparable in vitro reactivation power of K027 and the encapsulated K027, we failed to confirm this in vivo. In theory, this might result from the non-specific binding of molecules to the cucurbit[7]uril or the interaction of K027 with cucurbit[7]uril being too strong for acetylcholinesterase reactivation. Precise explanation requires additional in silico, in vitro and also in vivo experiments.


Acetylcholinesterase/blood , Acetylcholinesterase/metabolism , Brain/drug effects , Bridged-Ring Compounds/pharmacokinetics , Cholinesterase Reactivators/pharmacokinetics , Erythrocytes/drug effects , Imidazoles/pharmacokinetics , Oximes/pharmacokinetics , Pyridinium Compounds/pharmacokinetics , A549 Cells , Animals , Brain/enzymology , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/toxicity , Cell Survival/drug effects , Cholinesterase Reactivators/administration & dosage , Cholinesterase Reactivators/toxicity , Dose-Response Relationship, Drug , Erythrocytes/enzymology , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , Hep G2 Cells , Humans , Imidazoles/administration & dosage , Imidazoles/toxicity , Injections, Intramuscular , Male , Maximum Tolerated Dose , Mice, Inbred ICR , Oximes/administration & dosage , Oximes/toxicity , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/toxicity , Risk Assessment , Tissue Distribution
17.
Proc Natl Acad Sci U S A ; 116(47): 23714-23723, 2019 11 19.
Article En | MEDLINE | ID: mdl-31712430

Tumor-associated myeloid cells (TAMCs) are key drivers of immunosuppression in the tumor microenvironment, which profoundly impedes the clinical response to immune-dependent and conventional therapeutic modalities. As a hallmark of glioblastoma (GBM), TAMCs are massively recruited to reach up to 50% of the brain tumor mass. Therefore, they have recently been recognized as an appealing therapeutic target to blunt immunosuppression in GBM with the hope of maximizing the clinical outcome of antitumor therapies. Here we report a nano-immunotherapy approach capable of actively targeting TAMCs in vivo. As we found that programmed death-ligand 1 (PD-L1) is highly expressed on glioma-associated TAMCs, we rationally designed a lipid nanoparticle (LNP) formulation surface-functionalized with an anti-PD-L1 therapeutic antibody (αPD-L1). We demonstrated that this system (αPD-L1-LNP) enabled effective and specific delivery of therapeutic payload to TAMCs. Specifically, encapsulation of dinaciclib, a cyclin-dependent kinase inhibitor, into PD-L1-targeted LNPs led to a robust depletion of TAMCs and an attenuation of their immunosuppressive functions. Importantly, the delivery efficiency of PD-L1-targeted LNPs was robustly enhanced in the context of radiation therapy (RT) owing to the RT-induced up-regulation of PD-L1 on glioma-infiltrating TAMCs. Accordingly, RT combined with our nano-immunotherapy led to dramatically extended survival of mice in 2 syngeneic glioma models, GL261 and CT2A. The high targeting efficiency of αPD-L1-LNP to human TAMCs from GBM patients further validated the clinical relevance. Thus, this study establishes a therapeutic approach with immense potential to improve the clinical response in the treatment of GBM and warrants a rapid translation into clinical practice.


Brain Neoplasms/pathology , Glioblastoma/pathology , Myeloid Cells/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cyclic N-Oxides , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Indolizines , Mice , Myeloid Cells/drug effects , Myeloid Cells/radiation effects , Nanoparticles , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/therapeutic use , Tumor Microenvironment , Xenograft Model Antitumor Assays
18.
Toxicol Appl Pharmacol ; 383: 114776, 2019 11 15.
Article En | MEDLINE | ID: mdl-31629733

The major function of compounds with an oxime moiety attached to a quarternary nitrogen pyridinium ring is to reactivate acetylcholinesterase inhibited by organophosphorus agent (OP). However, other oxime mechanisms (e.g. modulation of cholinergic or glutamatergic receptor) may be involved in the recovery. The main disadvantage of positively charged reactivators is their low ability to penetrate into the brain although crossing the blood brain barrier could be supported via increasing the dose of administered oxime. Thus, this study presents maximal tolerated doses (MTD) for marketed oximes (TMB-4, MMB-4, LüH-6, HI-6, 2-PAM) and the most promising K-oximes (K027, K048, K203) which can be used in OP therapy in the future. No signs of sarin intoxication were observed in mice treated with 100% MTD of HI-6 in contrast to those treated with atropine and only 5% LD50 of HI-6. 100% MTD of HI-6 resulted in levels of 500 µM and 12 µM in plasma and brain, respectively. This concentration is by a far margin safe with respect to direct effects on neuronal cell viability and, on the other hand, does not have any effects on central NMDA receptors or central nACh receptors. However, a weak antimuscarinic activity in case of LüH-6 and a weak peripheral antinicotinic action in case of TMB-4 and 2-PAM could be observed at their respective 100% MTD dose. These high doses, represented by MTD, are, however, irrelevant to clinical practice since they led to mild to moderate toxic side effects. Therefore, we conclude that clinically used doses of marketed oxime reactivators have no significant direct pharmacological effect on the tested receptors.


Cholinesterase Reactivators/administration & dosage , Maximum Tolerated Dose , Organophosphorus Compounds/toxicity , Oximes/administration & dosage , Pyridinium Compounds/administration & dosage , Animals , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Chickens , Cholinesterase Reactivators/toxicity , Cricetinae , Cricetulus , Female , Humans , Male , Mice , Mice, Inbred BALB C , Oximes/toxicity , Pralidoxime Compounds/administration & dosage , Pralidoxime Compounds/toxicity , Pyridinium Compounds/toxicity
19.
Clin Cancer Res ; 25(22): 6839-6851, 2019 11 15.
Article En | MEDLINE | ID: mdl-31439587

PURPOSE: In this study, we addressed the underlying mechanisms for the association between enzalutamide (ENZ) treatment and neuroendocrine prostate cancer (NEPC), and the critical involvement of MYCN, and loss of RB1 function in neuroendocrine differentiation (NED) of prostatic epithelial cells, and the development of NEPC. We further sought to determine whether PARP inhibition could suppress NEPC, and to identify molecular determinants of this therapeutic activity. EXPERIMENTAL DESIGN: We used a novel prostate cancer patient-derived xenograft (PDX) treatment model, prostatic adenocarcinoma and NEPC cell lines, an NEPC organoid line, and NEPC xenograft models to address the mechanistic basis of ENZ-induced NED, and to analyze suppression of NED and NEPC growth by PARP inhibition. RESULTS: We identified an ENZ treatment-associated glucocorticoid receptor (GR)-MYCN-CDK5-RB1-E2F1 signaling pathway that drives NED in prostatic adenocarcinoma PDX and cell line models. Mechanistically, long-term ENZ treatment transcriptionally upregulates signaling of the GR-MYCN axis, leading to CDK5R1 and CDK5R2 upregulation, Rb1 phosphorylation, and N-Myc-mediated and E2F1-mediated NED gene expression. Importantly, olaparib (OLA) or talazoparib (TALA) suppressed these activities, and the combination of OLA and dinaciclib (DINA), an inhibitor of CDK2 and CDK5, which also inhibits Rb1 phosphorylation, suppressed NED and significantly improved therapeutic efficiency in NEPC cells in vitro and in NEPC tumors in vivo. CONCLUSIONS: The results of our study indicate an important role of GR-MYCN-CDK5R1/2-RB1-NED signaling in ENZ-induced and PARP inhibitor-suppressed NEPC. We also demonstrated efficacy for OLA+DINA combination therapy in NEPC xenograft models.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroendocrine Tumors/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Proteins/genetics , Animals , Benzamides , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Cyclic N-Oxides , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indolizines , Male , Mice, Nude , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/analogs & derivatives , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Proteins/metabolism , Pyridinium Compounds/administration & dosage , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Treatment Outcome , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays/methods
20.
Cancer Med ; 8(9): 4348-4358, 2019 08.
Article En | MEDLINE | ID: mdl-31207099

OBJECTIVE: This study aimed to identify and evaluate the mechanism by which apoptosis and cell cycle arrest were induced by dinaciclib in lymphoma Raji cells. METHODS: The colony formation assay was used to detect cell proliferation of Raji cells. Cell cycle arrest and cell apoptosis were determined by flow cytometry and TUNEL assays, respectively. Protein expression related to the Raji cell state was evaluated by Western blot. The Raji/Dinaciclib drug-resistant cell line was established, where the regulating functions of CDK1-involved pathway were verified. In addition, the effect of dinaciclib in vivo was examined in orthotopically implanted tumors in nude mice. RESULTS: Cell apoptosis was induced, and DNA synthesis ability was decreased in a time-dependent manner in dinaciclib-treated lymphoma Raji cells. Furthermore, the cell cycle was found to be blocked in the G2/M Phase. Further study indicated that CDK1-involved pathway played a key regulatory role in this process. It was revealed by cell transfection that the expression of cell cycle proteins was downregulated after treatment with dinaciclib through a CDK1-involved pathway, which eventually led to apoptosis. Knockdown of CDK1 restored the sensitivity of the Raji/Dinaciclib cells to dinaciclib. Xenograft model of nude mice showed that dinaciclib treatment in vivo could effectively inhibit tumor growth, consistent with the experiment results mentioned before. CONCLUSION: In this study, we clarified the mechanisms through which dinaciclib induces Raji cell apoptosis and blocks the cell cycle through a CDK1-involved pathway, which supported that dinaciclib had potential values in the treatment of lymphoma.


Antineoplastic Agents/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , CDC2 Protein Kinase/metabolism , Lymphoma/drug therapy , Pyridinium Compounds/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic N-Oxides , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Female , Humans , Indolizines , Lymphoma/metabolism , Mice , Mice, Nude , Pyridinium Compounds/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
...