Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33.367
Filter
Add more filters

Publication year range
1.
Cell ; 166(3): 637-650, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27471966

ABSTRACT

Most vertebrate oocytes contain a Balbiani body, a large, non-membrane-bound compartment packed with RNA, mitochondria, and other organelles. Little is known about this compartment, though it specifies germline identity in many non-mammalian vertebrates. We show Xvelo, a disordered protein with an N-terminal prion-like domain, is an abundant constituent of Xenopus Balbiani bodies. Disruption of the prion-like domain of Xvelo, or substitution with a prion-like domain from an unrelated protein, interferes with its incorporation into Balbiani bodies in vivo. Recombinant Xvelo forms amyloid-like networks in vitro. Amyloid-like assemblies of Xvelo recruit both RNA and mitochondria in binding assays. We propose that Xenopus Balbiani bodies form by amyloid-like assembly of Xvelo, accompanied by co-recruitment of mitochondria and RNA. Prion-like domains are found in germ plasm organizing proteins in other species, suggesting that Balbiani body formation by amyloid-like assembly could be a conserved mechanism that helps oocytes function as long-lived germ cells.


Subject(s)
Amyloid/metabolism , Organelle Biogenesis , T-Box Domain Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Benzothiazoles , Female , Fluorescent Dyes , Mitochondria/metabolism , Oocytes/cytology , Organelles/metabolism , Prions/chemistry , Protein Domains , Protein Transport , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/genetics , Thiazoles , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus laevis , Zebrafish
2.
Immunity ; 54(8): 1758-1771.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34256013

ABSTRACT

Apoptosis can potently defend against intracellular pathogens by directly killing microbes and eliminating their replicative niche. However, the reported ability of Mycobacterium tuberculosis to restrict apoptotic pathways in macrophages in vitro has led to apoptosis being dismissed as a host-protective process in tuberculosis despite a lack of in vivo evidence. Here we define crucial in vivo functions of the death receptor-mediated and BCL-2-regulated apoptosis pathways in mediating protection against tuberculosis by eliminating distinct populations of infected macrophages and neutrophils and priming T cell responses. We further show that apoptotic pathways can be targeted therapeutically with clinical-stage compounds that antagonize inhibitor of apoptosis (IAP) proteins to promote clearance of M. tuberculosis in mice. These findings reveal that any inhibition of apoptosis by M. tuberculosis is incomplete in vivo, advancing our understanding of host-protective responses to tuberculosis (TB) and revealing host pathways that may be targetable for treatment of disease.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Tuberculosis, Pulmonary/immunology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Dipeptides/therapeutic use , Humans , Indoles/therapeutic use , Lymphocyte Activation/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/microbiology , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes/immunology , Thiazoles/therapeutic use , Tuberculosis, Pulmonary/drug therapy
3.
Nature ; 632(8026): 930-937, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085602

ABSTRACT

The noradrenaline transporter (also known as norepinephrine transporter) (NET) has a critical role in terminating noradrenergic transmission by utilizing sodium and chloride gradients to drive the reuptake of noradrenaline (also known as norepinephrine) into presynaptic neurons1-3. It is a pharmacological target for various antidepressants and analgesic drugs4,5. Despite decades of research, its structure and the molecular mechanisms underpinning noradrenaline transport, coupling to ion gradients and non-competitive inhibition remain unknown. Here we present high-resolution complex structures of NET in two fundamental conformations: in the apo state, and bound to the substrate noradrenaline, an analogue of the χ-conotoxin MrlA (χ-MrlAEM), bupropion or ziprasidone. The noradrenaline-bound structure clearly demonstrates the binding modes of noradrenaline. The coordination of Na+ and Cl- undergoes notable alterations during conformational changes. Analysis of the structure of NET bound to χ-MrlAEM provides insight into how conotoxin binds allosterically and inhibits NET. Additionally, bupropion and ziprasidone stabilize NET in its inward-facing state, but they have distinct binding pockets. These structures define the mechanisms governing neurotransmitter transport and non-competitive inhibition in NET, providing a blueprint for future drug design.


Subject(s)
Bupropion , Models, Molecular , Norepinephrine Plasma Membrane Transport Proteins , Norepinephrine , Piperazines , Humans , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine/metabolism , Bupropion/metabolism , Bupropion/chemistry , Bupropion/pharmacology , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/metabolism , Allosteric Regulation/drug effects , Binding Sites , Biological Transport , Sodium/metabolism , Protein Conformation , Protein Binding , Thiazoles
4.
Cell ; 159(2): 267-80, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303524

ABSTRACT

Discrimination between pathogenic and beneficial microbes is essential for host organism immunity and homeostasis. Here, we show that chemosensory detection of two secondary metabolites produced by Pseudomonas aeruginosa modulates a neuroendocrine signaling pathway that promotes avoidance behavior in the simple animal host Caenorhabditis elegans. Secondary metabolites phenazine-1-carboxamide and pyochelin activate a G-protein-signaling pathway in the ASJ chemosensory neuron pair that induces expression of the neuromodulator DAF-7/TGF-ß. DAF-7, in turn, activates a canonical TGF-ß signaling pathway in adjacent interneurons to modulate aerotaxis behavior and promote avoidance of pathogenic P. aeruginosa. Our data provide a chemical, genetic, and neuronal basis for how the behavior and physiology of a simple animal host can be modified by the microbial environment and suggest that secondary metabolites produced by microbes may provide environmental cues that contribute to pathogen recognition and host survival.


Subject(s)
Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Pseudomonas aeruginosa/metabolism , Animals , Behavior, Animal , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Neurons/metabolism , Neurosecretory Systems/physiology , Phenazines/metabolism , Phenols/metabolism , Species Specificity , Thiazoles/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
Nature ; 618(7963): 102-109, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225985

ABSTRACT

Parasitic nematodes are a major threat to global food security, particularly as the world amasses 10 billion people amid limited arable land1-4. Most traditional nematicides have been banned owing to poor nematode selectivity, leaving farmers with inadequate means of pest control4-12. Here we use the model nematode Caenorhabditis elegans to identify a family of selective imidazothiazole nematicides, called selectivins, that undergo cytochrome-p450-mediated bioactivation in nematodes. At low parts-per-million concentrations, selectivins perform comparably well with commercial nematicides to control root infection by Meloidogyne incognita, a highly destructive plant-parasitic nematode. Tests against numerous phylogenetically diverse non-target systems demonstrate that selectivins are more nematode-selective than most marketed nematicides. Selectivins are first-in-class bioactivated nematode controls that provide efficacy and nematode selectivity.


Subject(s)
Antinematodal Agents , Tylenchoidea , Animals , Humans , Antinematodal Agents/chemistry , Antinematodal Agents/metabolism , Antinematodal Agents/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Tylenchoidea/drug effects , Tylenchoidea/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Thiazoles/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Plant Roots/drug effects , Plant Roots/parasitology , Plant Diseases , Species Specificity , Substrate Specificity
6.
Mol Cell ; 81(15): 3205-3215.e5, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34314699

ABSTRACT

The ß3-adrenergic receptor (ß3AR) is predominantly expressed in adipose tissue and urinary bladder and has emerged as an attractive drug target for the treatment of type 2 diabetes, obesity, and overactive bladder (OAB). Here, we report the cryogenic electron microscopy structure of the ß3AR-Gs signaling complex with the selective agonist mirabegron, a first-in-class drug for OAB. Comparison of this structure with the previously reported ß1AR and ß2AR structures reveals a receptor activation mechanism upon mirabegron binding to the orthosteric site. Notably, the narrower exosite in ß3AR creates a perpendicular pocket for mirabegron. Mutational analyses suggest that a combination of both the exosite shape and the amino-acid-residue substitutions defines the drug selectivity of the ßAR agonists. Our findings provide a molecular basis for ßAR subtype selectivity, allowing the design of more-selective agents with fewer adverse effects.


Subject(s)
Acetanilides/chemistry , Adrenergic beta-3 Receptor Agonists/chemistry , Receptors, Adrenergic, beta-3/chemistry , Receptors, Adrenergic, beta-3/metabolism , Thiazoles/chemistry , Acetanilides/metabolism , Adrenergic beta-3 Receptor Agonists/metabolism , Animals , Binding Sites , Cryoelectron Microscopy , Dogs , Humans , Models, Molecular , Molecular Dynamics Simulation , Receptors, Adrenergic, beta-3/genetics , Thiazoles/metabolism
7.
Mol Cell ; 81(2): 340-354.e5, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450210

ABSTRACT

In addition to its role as an electron transporter, mitochondrial nicotinamide adenine dinucleotide (NAD+) is an important co-factor for enzymatic reactions, including ADP-ribosylation. Although mitochondria harbor the most intra-cellular NAD+, mitochondrial ADP-ribosylation remains poorly understood. Here we provide evidence for mitochondrial ADP-ribosylation, which was identified using various methodologies including immunofluorescence, western blot, and mass spectrometry. We show that mitochondrial ADP-ribosylation reversibly increases in response to respiratory chain inhibition. Conversely, H2O2-induced oxidative stress reciprocally induces nuclear and reduces mitochondrial ADP-ribosylation. Elevated mitochondrial ADP-ribosylation, in turn, dampens H2O2-triggered nuclear ADP-ribosylation and increases MMS-induced ARTD1 chromatin retention. Interestingly, co-treatment of cells with the mitochondrial uncoupler FCCP decreases PARP inhibitor efficacy. Together, our results suggest that mitochondrial ADP-ribosylation is a dynamic cellular process that impacts nuclear ADP-ribosylation and provide evidence for a NAD+-mediated mitochondrial-nuclear crosstalk.


Subject(s)
ADP-Ribosylation , Cell Nucleus/enzymology , Mitochondria/enzymology , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , ADP-Ribosylation/drug effects , Animals , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/metabolism , Electron Transport/drug effects , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Methacrylates/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Oligomycins/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Rotenone/pharmacology , Thiazoles/pharmacology
8.
EMBO Rep ; 25(4): 2045-2070, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38454159

ABSTRACT

Teratozoospermia is a significant cause of male infertility, but the pathogenic mechanism of acephalic spermatozoa syndrome (ASS), one of the most severe teratozoospermia, remains elusive. We previously reported Spermatogenesis Associated 6 (SPATA6) as the component of the sperm head-tail coupling apparatus (HTCA) required for normal assembly of the sperm head-tail conjunction, but the underlying molecular mechanism has not been explored. Here, we find that the co-chaperone protein BAG5, expressed in step 9-16 spermatids, is essential for sperm HTCA assembly. BAG5-deficient male mice show abnormal assembly of HTCA, leading to ASS and male infertility, phenocopying SPATA6-deficient mice. In vivo and in vitro experiments demonstrate that SPATA6, cargo transport-related myosin proteins (MYO5A and MYL6) and dynein proteins (DYNLT1, DCTN1, and DNAL1) are misfolded upon BAG5 depletion. Mechanistically, we find that BAG5 forms a complex with HSPA8 and promotes the folding of SPATA6 by enhancing HSPA8's affinity for substrate proteins. Collectively, our findings reveal a novel protein-regulated network in sperm formation in which BAG5 governs the assembly of the HTCA by activating the protein-folding function of HSPA8.


Subject(s)
Cytoskeletal Proteins , Infertility, Male , Teratozoospermia , Thiazoles , Animals , Humans , Male , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Dyneins/metabolism , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , Infertility, Male/genetics , Infertility, Male/pathology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Folding , Semen/metabolism , Sperm Head/physiology , Spermatogenesis/genetics , Spermatozoa/metabolism , Teratozoospermia/metabolism , Teratozoospermia/pathology
9.
Cell ; 145(6): 863-74, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21640374

ABSTRACT

Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Huntington Disease/drug therapy , Kynurenic Acid/analysis , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Sulfonamides/therapeutic use , Thiazoles/therapeutic use , Administration, Oral , Alzheimer Disease/physiopathology , Animals , Brain Chemistry , Disease Models, Animal , Female , Humans , Kynurenic Acid/blood , Male , Mice , Mice, Transgenic , Sulfonamides/administration & dosage , Thiazoles/administration & dosage
10.
Circulation ; 149(13): 981-988, 2024 03 26.
Article in English | MEDLINE | ID: mdl-37952187

ABSTRACT

BACKGROUND: Device-detected atrial fibrillation (also known as subclinical atrial fibrillation or atrial high-rate episodes) is a common finding in patients with an implanted cardiac rhythm device and is associated with an increased risk of ischemic stroke. Whether oral anticoagulation is effective and safe in this patient population is unclear. METHODS: We performed a systematic review of MEDLINE and Embase for randomized trials comparing oral anticoagulation with antiplatelet or no antithrombotic therapy in adults with device-detected atrial fibrillation recorded by a pacemaker, implantable cardioverter defibrillator, cardiac resynchronization therapy device, or implanted cardiac monitor. We used random-effects models for meta-analysis and rated the quality of evidence using the Grading of Recommendations Assessment, Development and Evaluation framework (GRADE). The review was preregistered (PROSPERO CRD42023463212). RESULTS: From 785 citations, we identified 2 randomized trials with relevant clinical outcome data: NOAH-AFNET 6 (Non-Vitamin K Antagonist Oral Anticoagulants in Patients With Atrial High Rate Episodes; 2536 participants) evaluated edoxaban, and ARTESiA (Apixaban for the Reduction of Thrombo-Embolism in Patients With Device-Detected Sub-Clinical Atrial Fibrillation; 4012 participants) evaluated apixaban. Meta-analysis demonstrated that oral anticoagulation with these agents reduced ischemic stroke (relative risk [RR], 0.68 [95% CI, 0.50-0.92]; high-quality evidence). The results from the 2 trials were consistent (I2 statistic for heterogeneity=0%). Oral anticoagulation also reduced a composite of cardiovascular death, all-cause stroke, peripheral arterial embolism, myocardial infarction, or pulmonary embolism (RR, 0.85 [95% CI, 0.73-0.99]; I2=0%; moderate-quality evidence). There was no reduction in cardiovascular death (RR, 0.95 [95% CI, 0.76-1.17]; I2=0%; moderate-quality evidence) or all-cause mortality (RR, 1.08 [95% CI, 0.96-1.21]; I2=0%; moderate-quality evidence). Oral anticoagulation increased major bleeding (RR, 1.62 [95% CI, 1.05-2.50]; I²=61%; high-quality evidence). CONCLUSIONS: The results of the NOAH-AFNET 6 and ARTESiA trials are consistent with each other. Meta-analysis of these 2 large randomized trials provides high-quality evidence that oral anticoagulation with edoxaban or apixaban reduces the risk of stroke in patients with device-detected atrial fibrillation and increases the risk of major bleeding.


Subject(s)
Anticoagulants , Atrial Fibrillation , Embolism , Ischemic Stroke , Stroke , Humans , Administration, Oral , Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Embolism/etiology , Hemorrhage/prevention & control , Pyridines , Stroke/epidemiology , Stroke/prevention & control , Thiazoles , Treatment Outcome , Randomized Controlled Trials as Topic
11.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35103284

ABSTRACT

The contractile phenotype of smooth muscle cells (SMCs) is transcriptionally controlled by a complex of the DNA-binding protein SRF and the transcriptional co-activator MYOCD. The pathways that activate expression of Myocd and of SMC structural genes in mesenchymal progenitors are diverse, reflecting different intrinsic and extrinsic signaling inputs. Taking the ureter as a model, we analyzed whether Notch signaling, a pathway previously implicated in vascular SMC development, also affects visceral SMC differentiation. We show that mice with a conditional deletion of the unique Notch mediator RBPJ in the undifferentiated ureteric mesenchyme exhibit altered ureter peristalsis with a delayed onset, and decreased contraction frequency and intensity at fetal stages. They also develop hydroureter 2 weeks after birth. Notch signaling is required for precise temporal activation of Myocd expression and, independently, for expression of a group of late SMC structural genes. Based on additional expression analyses, we suggest that a mesenchymal JAG1-NOTCH2/NOTCH3 module regulates visceral SMC differentiation in the ureter in a biphasic and bimodal manner, and that its molecular function differs from that in the vascular system.


Subject(s)
Cell Differentiation , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Ureter/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation/drug effects , Diamines/pharmacology , Female , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Thiazoles/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Ureter/cytology , Ureter/growth & development , Viscera/cytology , Viscera/metabolism
12.
Plant Physiol ; 195(3): 1835-1850, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38535832

ABSTRACT

Plant transporters regulating the distribution of secondary metabolites play critical roles in defending against pathogens, insects, and interacting with beneficial microbes. The phosphorylation of these transporters can alter their activity, stability, and intracellular protein trafficking. However, the regulatory mechanism underlying this modification remains elusive. In this study, we discovered two orthologs of mammalian PKA, PKG, and PKC (AGC) kinases, oxidative signal-inducible 1 (OXI1) and its closest homologue, AGC subclass 2 member 2 (AGC2-2; 75% amino acid sequence identity with OXI1), associated with the extracellular secretion of camalexin and Arabidopsis (Arabidopsis thaliana) resistance to Pseudomonas syringae, and Botrytis cinerea. These kinases can undergo in vitro kinase reactions with three pleiotropic drug resistance (PDR) transporters: PDR6, PDR8, and PDR12. Moreover, our investigation confirmed PDR6 interaction with OXI1 and AGC2-2. By performing LC-MS/MS and parallel reaction monitoring, we identified the phosphorylation sites on PDR6 targeted by these kinases. Notably, chitin-induced PDR6 phosphorylation at specific residues, namely S31, S33, S827, and T832. Additional insights emerged by expressing dephosphorylated PDR6 variants in a pdr6 mutant background, revealing that the target residues S31, S33, and S827 promote PDR6 efflux activity, while T832 potentially contributes to PDR6 stability within the plasma membrane. The findings of this study elucidate partial mechanisms involved in the activity regulation of PDR-type transporters, providing valuable insights for their potential application in future plant breeding endeavors.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Botrytis , Disease Resistance , Plant Diseases , Pseudomonas syringae , Thiazoles , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Botrytis/physiology , Disease Resistance/genetics , Gene Expression Regulation, Plant , Indoles/metabolism , Phosphorylation , Phytoalexins , Plant Diseases/microbiology , Plant Diseases/genetics , Plant Diseases/immunology , Protein Kinases/metabolism , Protein Kinases/genetics , Pseudomonas syringae/pathogenicity , Pseudomonas syringae/physiology , Thiazoles/metabolism
13.
FASEB J ; 38(2): e23413, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38243760

ABSTRACT

Sphingosine-1-phosphate receptor (S1PR) modulators are clinically used to treat relapse-remitting multiple sclerosis (MS) and the early phase of progressive MS when inflammation still prevails. In the periphery, S1PR modulators prevent lymphocyte egress from lymph nodes, hence hampering neuroinflammation. Recent findings suggest a role for S1PR modulation in remyelination. As the Giα-coupled S1P1 subtype is the most prominently expressed S1PR in oligodendrocyte precursor cells (OPCs), selective modulation (functional antagonism) of S1P1 may have direct effects on OPC functionality. We hypothesized that functional antagonism of S1P1 by ponesimod induces remyelination by boosting OPC differentiation. In the cuprizone mouse model of demyelination, we found ponesimod to decrease the latency time of visual evoked potentials compared to vehicle conditions, which is indicative of functional remyelination. In addition, the Y maze spontaneous alternations test revealed that ponesimod reversed cuprizone-induced working memory deficits. Myelin basic protein (MBP) immunohistochemistry and transmission electron microscopy of the corpus callosum revealed an increase in myelination upon ponesimod treatment. Moreover, treatment with ponesimod alone or in combination with A971432, an S1P5 monoselective modulator, significantly increased primary mouse OPC differentiation based on O4 immunocytochemistry. In conclusion, S1P1 functional antagonism by ponesimod increases remyelination in the cuprizone model of demyelination and significantly increases OPC differentiation in vitro.


Subject(s)
Cuprizone , Demyelinating Diseases , Thiazoles , Mice , Animals , Cuprizone/toxicity , Sphingosine-1-Phosphate Receptors/metabolism , Oligodendroglia , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Evoked Potentials, Visual , Cell Differentiation/physiology , Mice, Inbred C57BL , Myelin Sheath/metabolism , Disease Models, Animal
14.
EMBO Rep ; 24(10): e57369, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37501563

ABSTRACT

Nutritional immunity includes sequestration of transition metals from invading pathogens. Yersinia pestis overcomes nutritional immunity by secreting yersiniabactin to acquire iron and zinc during infection. While the mechanisms for yersiniabactin synthesis and import are well-defined, those responsible for yersiniabactin secretion are unknown. Identification of this mechanism has been difficult because conventional mutagenesis approaches are unable to inhibit trans-complementation by secreted factors between mutants. To overcome this obstacle, we utilized a technique called droplet Tn-seq (dTn-seq), which uses microfluidics to isolate individual transposon mutants in oil droplets, eliminating trans-complementation between bacteria. Using this approach, we first demonstrated the applicability of dTn-seq to identify genes with secreted functions. We then applied dTn-seq to identify an AcrAB efflux system as required for growth in metal-limited conditions. Finally, we showed this efflux system is the primary yersiniabactin secretion mechanism and required for virulence during bubonic and pneumonic plague. Together, these studies have revealed the yersiniabactin secretion mechanism that has eluded researchers for over 30 years and identified a potential therapeutic target for bacteria that use yersiniabactin for metal acquisition.


Subject(s)
Plague , Yersinia pestis , Humans , Yersinia pestis/genetics , Plague/genetics , Plague/microbiology , Phenols , Thiazoles/pharmacology , Metals , Bacterial Proteins/genetics
15.
Brain ; 147(4): 1247-1263, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-37935051

ABSTRACT

Missense variants in SCN3A encoding the voltage-gated sodium (Na+) channel α subunit Nav1.3 are associated with SCN3A-related neurodevelopmental disorder (SCN3A-NDD), a spectrum of disease that includes epilepsy and malformation of cortical development. How genetic variation in SCN3A leads to pathology remains unclear, as prior electrophysiological work on disease-associated variants has been performed exclusively in heterologous cell systems. To further investigate the mechanisms of SCN3A-NDD pathogenesis, we used CRISPR/Cas9 gene editing to modify a control human induced pluripotent stem cell (iPSC) line to express the recurrent de novo missense variant SCN3A c.2624T>C (p.Ile875Thr). With the established Ngn2 rapid induction protocol, we generated glutamatergic forebrain-like neurons (iNeurons), which we showed to express SCN3A mRNA and Nav1.3-mediated Na+ currents. We performed detailed whole-cell patch clamp recordings to determine the effect of the SCN3A-p.Ile875Thr variant on endogenous Na+ currents in, and intrinsic excitability of, human neurons. Compared to control iNeurons, variant-expressing iNeurons exhibit markedly increased slowly-inactivating/persistent Na+ current, abnormal firing patterns with paroxysmal bursting and plateau-like potentials with action potential failure, and a hyperpolarized voltage threshold for action potential generation. We then validated these findings using a separate iPSC line generated from a patient harbouring the SCN3A-p.Ile875Thr variant compared to a corresponding CRISPR-corrected isogenic control line. Finally, we found that application of the Nav1.3-selective blocker ICA-121431 normalizes action potential threshold and aberrant firing patterns in SCN3A-p.Ile1875Thr iNeurons; in contrast, consistent with action as a Na+ channel blocker, ICA-121431 decreases excitability of control iNeurons. Our findings demonstrate that iNeurons can model the effects of genetic variation in SCN3A yet reveal a complex relationship between gain-of-function at the level of the ion channel versus impact on neuronal excitability. Given the transient expression of SCN3A in the developing human nervous system, selective blockade or suppression of Nav1.3-containing Na+ channels could represent a therapeutic approach towards SCN3A-NDD.


Subject(s)
Acetamides , Brain Diseases , Induced Pluripotent Stem Cells , Thiazoles , Humans , Action Potentials , NAV1.3 Voltage-Gated Sodium Channel/genetics , Neurons/physiology , Sodium , Sodium Channels/genetics
16.
Brain ; 147(6): 2144-2157, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38667631

ABSTRACT

Recent longitudinal PET imaging studies have established methods to estimate the age at which amyloid becomes abnormal at the level of the individual. Here we recontextualized amyloid levels into the temporal domain to better understand the downstream Alzheimer's disease processes of tau neurofibrillary tangle (NFT) accumulation and cognitive decline. This cohort study included a total of 601 individuals from the Wisconsin Registry for Alzheimer's Prevention and Wisconsin Alzheimer's Disease Research Center that underwent amyloid and tau PET, longitudinal neuropsychological assessments and met clinical criteria for three clinical diagnosis groups: cognitively unimpaired (n = 537); mild cognitive impairment (n = 48); or dementia (n = 16). Cortical 11C-Pittsburgh compound B (PiB) distribution volume ratio (DVR) and sampled iterative local approximation were used to estimate amyloid positive (A+; global PiB DVR > 1.16 equivalent to 17.1 centiloids) onset age and years of A+ duration at tau PET (i.e. amyloid chronicity). Tau PET burden was quantified using 18F-MK-6240 standardized uptake value ratios (70-90 min, inferior cerebellar grey matter reference region). Whole-brain and region-specific approaches were used to examine tau PET binding along the amyloid timeline and across the Alzheimer's disease clinical continuum. Voxel-wise 18F-MK-6240 analyses revealed that with each decade of A+, the spatial extent of measurable tau spread (i.e. progressed) from regions associated with early to late NFT tau stages. Regional analyses indicated that tau burden in the entorhinal cortex was detectable, on average, within 10 years of A+ onset. Additionally, the entorhinal cortex was the region most sensitive to early amyloid pathology and clinical impairment in this predominantly preclinical sample. Among initially cognitively unimpaired (n = 472) individuals with longitudinal cognitive follow-up, mixed effects models showed significant linear and non-linear interactions of A+ duration and entorhinal tau on cognitive decline, suggesting a synergistic effect whereby greater A+ duration, together with a higher entorhinal tau burden, increases the likelihood of cognitive decline beyond their separable effects. Overall, the amyloid time framework enabled a spatiotemporal characterization of tau deposition patterns across the Alzheimer's disease continuum. This approach, which examined cross-sectional tau PET data along the amyloid timeline to make longitudinal disease course inferences, demonstrated that A+ duration explains a considerable amount of variability in the magnitude and topography of tau spread, which largely recapitulated NFT staging observed in human neuropathological studies. By anchoring disease progression to the onset of amyloid, this study provides a temporal disease context, which may help inform disease prognosis and timing windows for anti-amyloid therapies.


Subject(s)
Alzheimer Disease , Brain , Cognitive Dysfunction , Positron-Emission Tomography , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Aged , Male , Female , tau Proteins/metabolism , Positron-Emission Tomography/methods , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Brain/metabolism , Brain/diagnostic imaging , Brain/pathology , Aged, 80 and over , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Disease Progression , Aniline Compounds , Cohort Studies , Amyloid beta-Peptides/metabolism , Middle Aged , Longitudinal Studies , Thiazoles , Neuropsychological Tests , Amyloid/metabolism
17.
Proc Natl Acad Sci U S A ; 119(15): e2122512119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35380904

ABSTRACT

We identified the anti-Mullerian hormone (also known as Müllerian inhibiting substance or MIS) as an inhibitory hormone that induces long-term contraception in mammals. The type II receptor to this hormone, AMHR2 (also known as MISR2), represents a promising druggable target for the modulation of female reproduction with a mechanism of action distinct from steroidal contraceptives. We designed an in vitro platform to screen and validate small molecules that can activate MISR2 signaling and suppress ovarian folliculogenesis. Using a bone morphogenesis protein (BMP)­response element luciferase reporter cell­based assay, we screened 5,440 compounds from a repurposed drug library. Positive hits in this screen were tested for specificity and potency in luciferase dose­response assays, and biological activity was tested in ex vivo Mullerian duct regression bioassays. Selected candidates were further evaluated in ex vivo follicle/ovary culture assays and in vivo in mice and rats. Here, we report that SP600125, CYC-116, gandotinib, and ruxolitinib can specifically inhibit primordial follicle activation and repress folliculogenesis by stimulating the MISR2 pathway.


Subject(s)
Contraceptive Agents , Drug Repositioning , Ovarian Follicle , Receptors, Peptide , Receptors, Transforming Growth Factor beta , Small Molecule Libraries , Animals , Anthracenes/chemistry , Anthracenes/pharmacology , Contraceptive Agents/chemistry , Contraceptive Agents/pharmacology , Drug Evaluation, Preclinical , Female , Humans , Mice , Nitriles/chemistry , Nitriles/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Receptors, Peptide/agonists , Receptors, Transforming Growth Factor beta/agonists , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology
18.
Eur Heart J ; 45(10): 837-849, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-37956458

ABSTRACT

BACKGROUND AND AIMS: Patients with long atrial high-rate episodes (AHREs) ≥24 h and stroke risk factors are often treated with anticoagulation for stroke prevention. Anticoagulation has never been compared with no anticoagulation in these patients. METHODS: This secondary pre-specified analysis of the Non-vitamin K antagonist Oral anticoagulants in patients with Atrial High-rate episodes (NOAH-AFNET 6) trial examined interactions between AHRE duration at baseline and anticoagulation with edoxaban compared with placebo in patients with AHRE and stroke risk factors. The primary efficacy outcome was a composite of stroke, systemic embolism, or cardiovascular death. The safety outcome was a composite of major bleeding and death. Key secondary outcomes were components of these outcomes and electrocardiogram (ECG)-diagnosed atrial fibrillation. RESULTS: Median follow-up of 2389 patients with core lab-verified AHRE was 1.8 years. AHRE ≥24 h were present at baseline in 259/2389 patients (11%, 78 ± 7 years old, 28% women, CHA2DS2-VASc 4). Clinical characteristics were not different from patients with shorter AHRE. The primary outcome occurred in 9/132 patients with AHRE ≥24 h (4.3%/patient-year, 2 strokes) treated with anticoagulation and in 14/127 patients treated with placebo (6.9%/patient-year, 2 strokes). Atrial high-rate episode duration did not interact with the efficacy (P-interaction = .65) or safety (P-interaction = .98) of anticoagulation. Analyses including AHRE as a continuous parameter confirmed this. Patients with AHRE ≥24 h developed more ECG-diagnosed atrial fibrillation (17.0%/patient-year) than patients with shorter AHRE (8.2%/patient-year; P < .001). CONCLUSIONS: This hypothesis-generating analysis does not find an interaction between AHRE duration and anticoagulation therapy in patients with device-detected AHRE and stroke risk factors. Further research is needed to identify patients with long AHRE at high stroke risk.


Subject(s)
Atrial Fibrillation , Pyridines , Stroke , Thiazoles , Humans , Female , Aged , Aged, 80 and over , Male , Atrial Fibrillation/complications , Atrial Fibrillation/drug therapy , Atrial Fibrillation/diagnosis , Heart Atria , Risk Factors , Stroke/etiology , Stroke/prevention & control , Stroke/diagnosis , Anticoagulants/therapeutic use
19.
Annu Rev Biochem ; 78: 569-603, 2009.
Article in English | MEDLINE | ID: mdl-19348578

ABSTRACT

Thiamin is synthesized by most prokaryotes and by eukaryotes such as yeast and plants. In all cases, the thiazole and pyrimidine moieties are synthesized in separate branches of the pathway and coupled to form thiamin phosphate. A final phosphorylation gives thiamin pyrophosphate, the active form of the cofactor. Over the past decade or so, biochemical and structural studies have elucidated most of the details of the thiamin biosynthetic pathway in bacteria. Formation of the thiazole requires six gene products, and formation of the pyrimidine requires two. In contrast, details of the thiamin biosynthetic pathway in yeast are only just beginning to emerge. Only one gene product is required for the biosynthesis of the thiazole and one for the biosynthesis of the pyrimidine. Thiamin can also be transported into the cell and can be salvaged through several routes. In addition, two thiamin degrading enzymes have been characterized, one of which is linked to a novel salvage pathway.


Subject(s)
Thiamine/biosynthesis , Animals , Eukaryotic Cells/metabolism , Fungi/metabolism , Humans , Plants/metabolism , Prokaryotic Cells/metabolism , Pyrimidines/chemistry , Thiamine/chemistry , Thiazoles/chemistry
20.
Breast Cancer Res ; 26(1): 36, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38439079

ABSTRACT

BACKGROUND: Hyperglycemia is an on-target effect of PI3Kα inhibitors. Early identification and intervention of treatment-induced hyperglycemia is important for improving management of patients receiving a PI3Kα inhibitor like alpelisib. Here, we characterize incidence of grade 3/4 alpelisib-related hyperglycemia, along with time to event, management, and outcomes using a machine learning model. METHODS: Data for the risk model were pooled from patients receiving alpelisib ± fulvestrant in the open-label, phase 1 X2101 trial and the randomized, double-blind, phase 3 SOLAR-1 trial. The pooled population (n = 505) included patients with advanced solid tumors (X2101, n = 221) or HR+/HER2- advanced breast cancer (SOLAR-1, n = 284). External validation was performed using BYLieve trial patient data (n = 340). Hyperglycemia incidence and management were analyzed for SOLAR-1. RESULTS: A random forest model identified 5 baseline characteristics most associated with risk of developing grade 3/4 hyperglycemia (fasting plasma glucose, body mass index, HbA1c, monocytes, age). This model was used to derive a score to classify patients as high or low risk for developing grade 3/4 hyperglycemia. Applying the model to patients treated with alpelisib and fulvestrant in SOLAR-1 showed higher incidence of hyperglycemia (all grade and grade 3/4), increased use of antihyperglycemic medications, and more discontinuations due to hyperglycemia (16.7% vs. 2.6% of discontinuations) in the high- versus low-risk group. Among patients in SOLAR-1 (alpelisib + fulvestrant arm) with PIK3CA mutations, median progression-free survival was similar between the high- and low-risk groups (11.0 vs. 10.9 months). For external validation, the model was applied to the BYLieve trial, for which successful classification into high- and low-risk groups with shorter time to grade 3/4 hyperglycemia in the high-risk group was observed. CONCLUSIONS: A risk model using 5 clinically relevant baseline characteristics was able to identify patients at higher or lower probability for developing alpelisib-induced hyperglycemia. Early identification of patients who may be at higher risk for hyperglycemia may improve management (including monitoring and early intervention) and potentially lead to improved outcomes. REGISTRATION: ClinicalTrials.gov: NCT01219699 (registration date: October 13, 2010; retrospectively registered), ClinicalTrials.gov: NCT02437318 (registration date: May 7, 2015); ClinicalTrials.gov: NCT03056755 (registration date: February 17, 2017).


Subject(s)
Breast Neoplasms , Hyperglycemia , Thiazoles , Humans , Female , Breast Neoplasms/drug therapy , Fulvestrant/adverse effects , Hyperglycemia/chemically induced , Hyperglycemia/epidemiology , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL