Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Transpl Int ; 36: 10938, 2023.
Article in English | MEDLINE | ID: mdl-37091963

ABSTRACT

Solid Organ Transplant (SOT) recipients are at significant higher risk for COVID-19 and due to immunosuppressive medication, the immunogenicity after vaccination is suboptimal. In the previous studies, booster method showed significant benefit in this population. In the current study, we compared using a mix-and-match method vs. same vaccine as a third dose in SOT recipients. This was a patient-blinded, single center, randomized controlled trial comparing BNT162b2 vs. JNJ-78436735 vaccine as the third dose after two doses of BNT162b2 vaccine. We included adult SOT recipients with functional graft who had received two doses of BNT162b2 vaccine. Participants were randomly assigned to receive either BNT162b2 or JNJ-78436735 in one-to-one ratio. Primary outcome was SARS-CoV-2 IgG positivity at 1 month after the third dose. Sixty SOT recipients, including 36 kidney, 12 liver, 2 lung, 3 heart, and 5 combined transplants, were enrolled, and 57 recipients were analyzed per protocol. There were no statistically significant differences between the two vaccine protocols for IgG positivity (83.3% vs. 85.2% for BNT162b2 and JNJ-78436735, respectively, p = 0.85, Odds Ratio 0.95, 95% Confidence Interval 0.23-4.00). Comparison of the geometric mean titer demonstrated a higher trend with BNT162b2 (p = 0.09). In this pilot randomized controlled trial comparing mix and match method vs. uniform vaccination in SOT recipients, both vaccines were safely used. Since this was a small sample sized study, there was no statistically significant difference in immunogenicity; though, the mix and match method showed relatively lower geometric mean titer, as compared to uniform vaccine. Further studies need to be conducted to determine duration of this immunogenicity. Clinical Trial Registration: https://clinicaltrials.gov/ct2/show/NCT05047640?term=20210641&draw=2&rank=1, identifier 20210641.


Subject(s)
COVID-19 , Organ Transplantation , Vaccines , Adult , Humans , Ad26COVS1 , BNT162 Vaccine , COVID-19/prevention & control , SARS-CoV-2 , Transplant Recipients , Immunoglobulin G , Antibodies, Viral
2.
Sci Rep ; 11(1): 3438, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33564035

ABSTRACT

ApoEε4 is a major genetic risk factor for Alzheimer's disease (AD), a disease hallmarked by extracellular amyloid-beta (Aß) plaques and intracellular neurofibrillary tangles (NFTs). The presence of the ApoEε4 allele is associated with increased Aß deposition and a role for ApoEε4 in the potentiation of tau pathology has recently emerged. This study focused on comparing the effects of adeno-associated virus (AAV)-mediated overexpression of the three predominant human ApoE isoforms within astrocytes. The isoform-specific effects of human ApoE were evaluated within in vitro models of tau pathology within neuron/astrocyte co-cultures, as well as in a transgenic tau mouse model. Tau aggregation, accumulation, and phosphorylation were measured to determine if the three isoforms of human ApoE had differential effects on tau. Astrocytic overexpression of the human ApoEε4 allele increased phosphorylation and misfolding of overexpressed neuronal tau in multiple models, including the aggregation and accumulation of added tau oligomers, in an isoform-specific manner. The ability of ApoEε4 to increase tau aggregation could be inhibited by an ApoEε4-specific antibody. This study indicates that astrocytic expression of ApoEε4 can potentiate tau aggregation and phosphorylation within neurons and supports a gain of toxic function hypothesis for the effect of hApoEε4 on tau.


Subject(s)
Alleles , Alzheimer Disease/metabolism , Apolipoprotein E4/biosynthesis , Astrocytes/metabolism , Gene Expression Regulation , Protein Aggregates , tau Proteins , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E4/genetics , Astrocytes/pathology , Disease Models, Animal , Rats , Rats, Sprague-Dawley , tau Proteins/genetics , tau Proteins/metabolism
3.
J Pharmacol Exp Ther ; 373(2): 311-324, 2020 05.
Article in English | MEDLINE | ID: mdl-32094294

ABSTRACT

Treatments for cognitive deficits associated with central nervous system (CNS) disorders such as Alzheimer disease and schizophrenia remain significant unmet medical needs that incur substantial pressure on the health care system. The α7 nicotinic acetylcholine receptor (nAChR) has garnered substantial attention as a target for cognitive deficits based on receptor localization, robust preclinical effects, genetics implicating its involvement in cognitive disorders, and encouraging, albeit mixed, clinical data with α7 nAChR orthosteric agonists. Importantly, previous orthosteric agonists at this receptor suffered from off-target activity, receptor desensitization, and an inverted U-shaped dose-effect curve in preclinical assays that limit their clinical utility. To overcome the challenges with orthosteric agonists, we have identified a novel selective α7 positive allosteric modulator (PAM), BNC375. This compound is selective over related receptors and potentiates acetylcholine-evoked α7 currents with only marginal effect on the receptor desensitization kinetics. In addition, BNC375 enhances long-term potentiation of electrically evoked synaptic responses in rat hippocampal slices and in vivo. Systemic administration of BNC375 reverses scopolamine-induced cognitive deficits in rat novel object recognition and rhesus monkey object retrieval detour (ORD) task over a wide range of exposures, showing no evidence of an inverted U-shaped dose-effect curve. The compound also improves performance in the ORD task in aged African green monkeys. Moreover, ex vivo 13C-NMR analysis indicates that BNC375 treatment can enhance neurotransmitter release in rat medial prefrontal cortex. These findings suggest that α7 nAChR PAMs have multiple advantages over orthosteric α7 nAChR agonists for the treatment of cognitive dysfunction associated with CNS diseases. SIGNIFICANCE STATEMENT: BNC375 is a novel and selective α7 nicotinic acetylcholine receptor (nAChR) positive allosteric modulator (PAM) that potentiates acetylcholine-evoked α7 currents in in vitro assays with little to no effect on the desensitization kinetics. In vivo, BNC375 demonstrated robust procognitive effects in multiple preclinical models across a wide exposure range. These results suggest that α7 nAChR PAMs have therapeutic potential in central nervous system diseases with cognitive impairments.


Subject(s)
Benzethonium/pharmacology , Chlorobenzenes/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Allosteric Regulation , Animals , Brain/drug effects , Brain/physiology , Cell Survival/drug effects , Cells, Cultured , Cognition/drug effects , Long-Term Potentiation/drug effects , Macaca mulatta , Male , Neurotransmitter Agents/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Scopolamine/pharmacology
4.
ACS Chem Biol ; 12(2): 519-527, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28032990

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) mediate glutamatergic signaling that is critical to cognitive processes in the central nervous system, and NMDAR hypofunction is thought to contribute to cognitive impairment observed in both schizophrenia and Alzheimer's disease. One approach to enhance the function of NMDAR is to increase the concentration of an NMDAR coagonist, such as glycine or d-serine, in the synaptic cleft. Inhibition of alanine-serine-cysteine transporter-1 (Asc-1), the primary transporter of d-serine, is attractive because the transporter is localized to neurons in brain regions critical to cognitive function, including the hippocampus and cortical layers III and IV, and is colocalized with d-serine and NMDARs. To identify novel Asc-1 inhibitors, two different screening approaches were performed with whole-cell amino acid uptake in heterologous cells stably expressing human Asc-1: (1) a high-throughput screen (HTS) of 3 M compounds measuring 35S l-cysteine uptake into cells attached to scintillation proximity assay beads in a 1536 well format and (2) an iterative focused screen (IFS) of a 45 000 compound diversity set using a 3H d-serine uptake assay with a liquid scintillation plate reader in a 384 well format. Critically important for both screening approaches was the implementation of counter screens to remove nonspecific inhibitors of radioactive amino acid uptake. Furthermore, a 15 000 compound expansion step incorporating both on- and off-target data into chemical and biological fingerprint-based models for selection of additional hits enabled the identification of novel Asc-1-selective chemical matter from the IFS that was not identified in the full-collection HTS.


Subject(s)
Amino Acid Transport System y+/antagonists & inhibitors , High-Throughput Screening Assays , Animals , Bayes Theorem , CHO Cells , Cricetinae , Cricetulus , Humans , Machine Learning
5.
Bioorg Med Chem Lett ; 27(1): 114-120, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27816515

ABSTRACT

The initial structure activity relationships around an isoindoline uHTS hit will be described. Information gleaned from ligand co-crystal structures allowed for rapid refinements in both MARK potency and kinase selectivity. These efforts allowed for the identification of a compound with properties suitable for use as an in vitro tool compound for validation studies on MARK as a viable target for Alzheimer's disease.


Subject(s)
Drug Design , Protein Kinase Inhibitors/pharmacology , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cell Line , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 27(1): 109-113, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27894874

ABSTRACT

Attempts to optimize pharmacokinetic properties in a promising series of pyrrolopyrimidinone MARK inhibitors for the treatment of Alzheimer's disease are described. A focus on physical properties and ligand efficiency while prosecuting this series afforded key tool compounds that revealed a large discrepancy in the rat in vitro-in vivo DMPK (Drug Metabolism/Pharmacokinetics) correlation. These differences prompted an in vivo rat disposition study employing a radiolabeled representative of the series, and the results from this experiment justified the termination of any further optimization efforts.


Subject(s)
Alzheimer Disease/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Alzheimer Disease/metabolism , Animals , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Rats , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 26(17): 4362-6, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27491711

ABSTRACT

Inhibition of microtubule affinity regulating kinase (MARK) represents a potentially attractive means of arresting neurofibrillary tangle pathology in Alzheimer's disease. This manuscript outlines efforts to optimize a pyrazolopyrimidine series of MARK inhibitors by focusing on improvements in potency, physical properties and attributes amenable to CNS penetration. A unique cylcyclohexyldiamine scaffold was identified that led to remarkable improvements in potency, opening up opportunities to reduce MW, Pgp efflux and improve pharmacokinetic properties while also conferring improved solubility.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Heterocyclic Compounds/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Weight , Rats , Solubility
8.
Eur J Appl Physiol ; 115(2): 373-86, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25326727

ABSTRACT

PURPOSE: The aim of this study was to investigate the effect of tyrosine (TYR) ingestion on cognitive and physical performance during soccer-specific exercise in a warm environment. METHODS: Eight male soccer players completed an individualised 90 min soccer-simulation intermittent soccer performance test (iSPT), on a non-motorised treadmill, on two occasions, within an environmental chamber (25 °C, 40 % RH). Participants ingested tyrosine (TYR; 250 mL sugar free drink plus 150 mg kg body mass(-1) TYR) at both 5 h and 1 h pre-exercise or a placebo control (PLA; 250 mL sugar free drink only) in a double-blind, randomised, crossover design. Cognitive performance (vigilance and dual-task) and perceived readiness to invest physical effort (RTIPE) and mental effort (RTIME) were assessed: pre-exercise, half-time, end of half-time and immediately post-exercise. Physical performance was assessed using the total distance covered in both halves of iSPT. RESULTS: Positive vigilance responses (HIT) were significantly higher (12.6 ± 1.7 vs 11.5 ± 2.4, p = 0.015) with negative responses (MISS) significantly lower (2.4 ± 1.8 vs 3.5 ± 2.4, p = 0.013) in TYR compared to PLA. RTIME scores were significantly higher in the TYR trial when compared to PLA (6.7 ± 1.2 vs 5.9 ± 1.2, p = 0.039). TYR had no significant (p > 0.05) influence on any other cognitive or physical performance measure. CONCLUSION: The results show that TYR ingestion is associated with improved vigilance and RTIME when exposed to individualised soccer-specific exercise (iSPT) in a warm environment. This suggests that increasing the availability of TYR may improve cognitive function during exposure to exercise-heat stress.


Subject(s)
Athletic Performance , Cognition/drug effects , Exercise Test , Hot Temperature , Tyrosine/pharmacology , Arousal/drug effects , Dietary Supplements , Humans , Male , Soccer/physiology , Tyrosine/administration & dosage , Young Adult
9.
Neurochem Res ; 35(1): 130-42, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19649707

ABSTRACT

In Parkinson's disease substantia nigra neurons degenerate likely due to oxidative damage interacting with genetic risk factors. Here, SH-SY5Y cells expressing wild-type or A53T alpha-synuclein had increased sensitivity to methyl-4-phenylpyridinium iodide (MPP(+)), which induces mitochondrial dysfunction, and 6-hydroxydopamine (6-OHDA), which causes oxidative stress. Edaravone protected only against MPP(+), and EGCG ((-)-epigallocatechin-3-O-gallate) protected only against 6-OHDA. Thus genomic responses to MPP(+) and 6-OHDA in the presence of these antioxidants were analyzed using microarrays. Pathway analysis indicated that MPP(+) activated p53 (P < 0.001) while 6-OHDA induced the Nrf2 antioxidative stress response (P < 0.0001). EGCG was more effective at blocking 6-OHDA-mediated genomic responses, while edaravone was more effective against MPP(+). We identified 32 genes that responded to both toxins except in the presence of an effective anti-oxidant; eight are transcription factors and potentially constitute a stress-response transcriptional network. These data provide insights into the mechanisms of neurotoxicity and identifies genes that might mediate antioxidant efficacy.


Subject(s)
Antioxidants/metabolism , Genome , Mutation , Neuroblastoma/metabolism , Oligonucleotide Array Sequence Analysis , Parkinson Disease/genetics , alpha-Synuclein/genetics , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology
10.
J Neurochem ; 95(2): 406-17, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16092930

ABSTRACT

The ability of cells to control the balance between the generation and quenching of reactive oxygen species is important in combating potentially damaging effects of oxidative stress. One mechanism that cells use to maintain redox homeostasis is the antioxidant response pathway. Antioxidant response elements (AREs) are cis-acting elements located in regulatory regions of antioxidant and phase II detoxification genes. Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) is a member of the Cap 'n' Collar family of transcription factors that binds to the ARE and regulates the transcription of specific ARE-containing genes such as NAD(P)H:quinone oxidoreductase 1, glutamylcysteine synthetase and heme oxygenase. Activation of Nrf2 results in release from its negative repressor, Kelch-like ECH-associated protein 1 (Keap1), and allows Nrf2 to translocate into the nucleus to induce gene expression. In this study, we demonstrate that increasing Nrf2 activity by various methods, including chemical induction, Nrf2 overexpression or Keap1 siRNA knockdown, protects cells against specific types of oxidative damage. Cells were protected against 6-hydroxydopamine- and 3-morpholinosydnonimine-mediated toxicity but not against 1-methyl-1-4-phenylpyridinium toxicity. As oxidative stress is a hallmark of several neurodegenerative disorders, including Parkinson's disease, pharmacological agents that selectively target the Keap1-Nrf2 pathway may provide a novel neuroprotective strategy for the treatment of these diseases.


Subject(s)
DNA-Binding Proteins/physiology , Oxidative Stress/physiology , Trans-Activators/physiology , 1-Methyl-4-phenylpyridinium/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/toxicity , Blotting, Western , Cell Survival/drug effects , DNA-Binding Proteins/biosynthesis , Genes, Reporter/genetics , Humans , Hydroquinones/pharmacology , Intracellular Signaling Peptides and Proteins , Kelch-Like ECH-Associated Protein 1 , Luciferases/metabolism , Molsidomine/analogs & derivatives , Molsidomine/toxicity , NF-E2-Related Factor 2 , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Plasmids/genetics , Proteins/antagonists & inhibitors , Proteins/physiology , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Trans-Activators/biosynthesis
11.
Neuropharmacology ; 49 Suppl 1: 135-45, 2005.
Article in English | MEDLINE | ID: mdl-16005030

ABSTRACT

Previous reports have shown that activation of N-methyl-D-aspartate (NMDA) receptors potentiates responses to activation of the group I metabotropic glutamate receptor mGluR5 by reversing PKC-mediated desensitization of this receptor. NMDA-induced reversal of mGluR5 desensitization is dependent on activation of protein phosphatases. However, the specific protein phosphatase involved and the precise mechanism by which NMDA receptor activation reduces mGluR desensitization are not known. We have performed a series of molecular, biochemical, and genetic studies to show that NMDA-induced regulation of mGluR5 is dependent on activation of calcium-dependent protein phosphatase 2B/calcineurin (PP2B/CaN). Furthermore, we report that purified calcineurin directly dephosphorylates the C-terminal tail of mGluR5 at sites that are phosphorylated by PKC. Finally, immunoprecipitation and GST fusion protein pull-down experiments reveal that calcineurin interacts with mGluR5, suggesting that these proteins could be colocalized in a signaling complex. Taken together with previous studies, these data suggest that activation of NMDA receptors leads to activation of calcineurin and that calcineurin modulates mGluR5 function by directly dephosphorylating mGluR5 at PKC sites that are involved in desensitization of this receptor.


Subject(s)
Calcineurin/metabolism , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Animals , Autoradiography/methods , Brain/drug effects , Brain/enzymology , CHO Cells/drug effects , CHO Cells/enzymology , Calcineurin/pharmacology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Drug Synergism , Electric Stimulation/methods , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Glutathione Transferase , Hydroxylation/drug effects , Immunoblotting/methods , Immunoprecipitation/methods , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mutagenesis/physiology , Okadaic Acid/pharmacology , Oocytes/drug effects , Oocytes/enzymology , Patch-Clamp Techniques/methods , Phosphatidylinositols/metabolism , Protein Kinase C/pharmacology , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Transfection , Xenopus
12.
J Pharmacol Exp Ther ; 310(2): 488-97, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15051800

ABSTRACT

Antagonists of the B1 bradykinin receptor (B1R) offer the promise of novel therapeutic agents for the treatment of inflammatory and neuropathic pain. However, the in vivo characterization of the pharmacodynamics of B1R antagonists is hindered by the low level of B1R expression in healthy tissue and the profound species selectivity exhibited by many compounds for the human B1R. To circumvent these issues, we generated a transgenic rat expressing the human B1R under the control of the neuron-specific enolase promoter. Membranes prepared from whole brain homogenates of heterozygous transgenic rats indicate a B1R expression level of 30 to 40 fmol/mg; there is no detectable B1R expression in control nontransgenic rats. The pharmacological profile of the B1R expressed in the transgenic rat matches that expected of the human, but not the rat receptor. The mapping of the transgene insertion site to rat chromosome 1 permitted the development of a reliable assay for the identification of homozygous transgenic rats. Significantly, homozygous transgenic rats express 2-fold more B1R than heterozygous animals. Autoradiographic analyses of tissue sections from transgenic rats reveal that the B1R is broadly expressed in both the brain and spinal cord. The human B1R expressed in the transgenic rat functions in an in vitro contractile assay and thus has the potential to elicit a functional response in vivo. Using the humanized B1R transgenic rat, an assay was developed that is suitable for the routine evaluation of a test compound's ability to occupy the human B1R in the central nervous system.


Subject(s)
Animals, Genetically Modified/genetics , Models, Animal , Rats/genetics , Receptor, Bradykinin B1/biosynthesis , Receptor, Bradykinin B1/genetics , Animals , Animals, Genetically Modified/metabolism , Brain/drug effects , Brain/metabolism , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Female , Humans , Ileum/drug effects , Ileum/metabolism , Male , Peptide Fragments/pharmacology , Protein Binding/drug effects , Protein Binding/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...