Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Commun Biol ; 7(1): 80, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38200107

ABSTRACT

Vagus nerve signaling is a key component of the gut-brain axis and regulates diverse physiological processes that decline with age. Gut to brain vagus firing patterns are regulated by myenteric intrinsic primary afferent neuron (IPAN) to vagus neurotransmission. It remains unclear how IPANs or the afferent vagus age functionally. Here we identified a distinct ageing code in gut to brain neurotransmission defined by consistent differences in firing rates, burst durations, interburst and intraburst firing intervals of IPANs and the vagus, when comparing young and aged neurons. The aminosterol squalamine changed aged neurons firing patterns to a young phenotype. In contrast to young neurons, sertraline failed to increase firing rates in the aged vagus whereas squalamine was effective. These results may have implications for improved treatments involving pharmacological and electrical stimulation of the vagus for age-related mood and other disorders. For example, oral squalamine might be substituted for or added to sertraline for the aged.


Subject(s)
Sensory Receptor Cells , Sertraline , Cholestanols , Vagus Nerve
2.
J Med Chem ; 66(14): 9519-9536, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37433124

ABSTRACT

Natural aminosterols are promising drug candidates against neurodegenerative diseases, like Alzheimer and Parkinson, and one relevant protective mechanism occurs via their binding to biological membranes and displacement or binding inhibition of amyloidogenic proteins and their cytotoxic oligomers. We compared three chemically different aminosterols, finding that they exhibited different (i) binding affinities, (ii) charge neutralizations, (iii) mechanical reinforcements, and (iv) key lipid redistributions within membranes of reconstituted liposomes. They also had different potencies (EC50) in protecting cultured cell membranes against amyloid-ß oligomers. A global fitting analysis led to an analytical equation describing quantitatively the protective effects of aminosterols as a function of their concentration and relevant membrane effects. The analysis correlates aminosterol-mediated protection with well-defined chemical moieties, including the polyamine group inducing a partial membrane-neutralizing effect (79 ± 7%) and the cholestane-like tail causing lipid redistribution and bilayer mechanical resistance (21 ± 7%), linking quantitatively their chemistry to their protective effects on biological membranes.


Subject(s)
Neurodegenerative Diseases , Protein Aggregates , Humans , Cell Membrane/metabolism , Amyloidogenic Proteins/chemistry , Neurodegenerative Diseases/metabolism , Lipids , Lipid Bilayers/metabolism , Amyloid beta-Peptides/metabolism
3.
Colloids Surf B Biointerfaces ; 222: 113115, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36603410

ABSTRACT

Trodusquemine is an amphipathic aminosterol that has recently shown therapeutic benefit in neurodegenerative diseases altering the binding of misfolded proteins to the cell membrane. To unravel the underlying mechanism, we studied the interactions between Trodusquemine (TRO) and lipid monolayers simulating the outer layer of the plasma membrane. We selected two different compositions of dioleoylphosphatidylcholine (DOPC), sphingomyelin (SM), cholesterol (Chol) and monosialotetrahexosylganglioside (GM1) lipid mixture mimicking either a lipid-raft containing membrane (Ld+So phases) or a single-phase disordered membrane (Ld phase). Surface pressure-area isotherms and surface compressional modulus-area combined with Brewster Angle Microscopy (BAM) provided the thermodynamic and morphological information on the lipid monolayer in the presence of increasing amounts of TRO in the monolayer. Experiments revealed that TRO forms stable spreading monolayers at the buffer-air interface where it undergoes multiple reversible phase transitions to bi- and tri-layers at the interface. When TRO was spread at the interface with the lipid mixtures, we found that it distributes in the lipid monolayer for both the selected lipid compositions, but a maximum TRO uptake in the rafts-containing monolayer was observed for a Lipid/TRO molar ratio equal to 3:2. Statistical analysis of BAM images revealed that TRO induces a decrease in the size of the condensed domains, an increase in their number and in the thickness mismatch between the Ld and So phase. Experiments and MD simulations converge to indicate that TRO adsorbs preferentially at the border of the So domains. Removal of GM1 from the lipid Ld+So mixture resulted in an even greater TRO-mediated reduction of the size of the So domains suggesting that the presence of GM1 hinders the localization of TRO at the So domains boundaries. Taken together these observations suggest that Trodusquemine influences the organization of lipid rafts within the neuronal membrane in a dose-dependent manner whereas it evenly distributes in disordered expanded phases of the membrane model.


Subject(s)
G(M1) Ganglioside , Membranes, Artificial , Cholesterol/chemistry , Membrane Microdomains/chemistry
4.
Ann Intern Med ; 175(12): 1666-1674, 2022 12.
Article in English | MEDLINE | ID: mdl-36343348

ABSTRACT

BACKGROUND: Parkinson disease (PD) is associated with α-synuclein (αS) aggregation within enteric neurons. ENT-01 inhibits the formation of αS aggregates and improved constipation in an open-label study in patients with PD. OBJECTIVE: To evaluate the safety and efficacy of oral ENT-01 for constipation and neurologic symptoms in patients with PD and constipation. DESIGN: Randomized, placebo-controlled phase 2b study. (ClinicalTrials.gov: NCT03781791). SETTING: Outpatient. PATIENTS: 150 patients with PD and constipation. INTERVENTION: ENT-01 or placebo daily for up to 25 days. After baseline assessment of constipation severity, daily dosing was escalated to the prokinetic dose, the maximum dose (250 mg), or the tolerability limit, followed by a washout period. MEASUREMENTS: The primary efficacy end point was the number of complete spontaneous bowel movements (CSBMs) per week. Neurologic end points included dementia (assessed using the Mini-Mental State Examination [MMSE]) and psychosis (assessed using the Scale for the Assessment of Positive Symptoms adapted for PD [SAPS-PD]). RESULTS: The weekly CSBM rate increased from 0.7 to 3.2 in the ENT-01 group versus 0.7 to 1.2 in the placebo group (P < 0.001). Improvement in secondary end points included SBMs (P = 0.002), stool consistency (P < 0.001), ease of passage (P = 0.006), and laxative use (P = 0.041). In patients with dementia, MMSE scores improved by 3.4 points 6 weeks after treatment in the ENT-01 group (n = 14) versus 2.0 points in the placebo group (n = 14). Among patients with psychosis, SAPS-PD scores improved from 6.5 to 1.7 six weeks after treatment in the ENT-01 group (n = 5) and from 6.3 to 4.4 in the placebo group (n = 6). ENT-01 was well tolerated, with no deaths or drug-related serious adverse events. Adverse events were predominantly gastrointestinal, including nausea (34.4% [ENT-01] vs. 5.3% [placebo]; P < 0.001) and diarrhea (19.4% [ENT-01] vs. 5.3% [placebo]; P = 0.016). LIMITATION: Longer treatment periods need to be investigated in future studies. CONCLUSION: ENT-01 was safe and significantly improved constipation. PRIMARY FUNDING SOURCE: Enterin, Inc.


Subject(s)
Dementia , Parkinson Disease , Humans , Treatment Outcome , Constipation , Defecation , Double-Blind Method
5.
Cell Rep ; 38(2): 110090, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021075

ABSTRACT

Alpha-synuclein (αS) is causally involved in the development of Parkinson disease (PD); however, its role in normal vertebrate physiology has remained unknown. Recent studies demonstrate that αS is induced by noroviral infection in the enteric nervous system of children and protects mice against lethal neurotropic viral infection. Additionally, αS is a potent chemotactic activator of phagocytes. In this report, using both wild-type and αS knockout mice, we show that αS is a critical mediator of inflammatory and immune responses. αS is required for the development of a normal inflammatory response to bacterial peptidoglycan introduced into the peritoneal cavity as well as antigen-specific and T cell responses following intraperitoneal immunization. Furthermore, we show that neural cells are the sources of αS required for immune competence. Our report supports the hypothesis that αS accumulates within the nervous system of PD individuals because of an inflammatory/immune response.


Subject(s)
Immunity/physiology , alpha-Synuclein/metabolism , alpha-Synuclein/physiology , Animals , Brain/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/metabolism , Neurons/metabolism , Toll-Like Receptor 4/immunology , alpha-Synuclein/genetics
6.
Nat Prod Rep ; 39(4): 742-753, 2022 04 20.
Article in English | MEDLINE | ID: mdl-34698757

ABSTRACT

Covering: 1993 to 2021 (mainly 2017-2021)Alzheimer's and Parkinson's diseases are neurodegenerative conditions affecting over 50 million people worldwide. Since these disorders are still largely intractable pharmacologically, discovering effective treatments is of great urgency and importance. These conditions are characteristically associated with the aberrant deposition of proteinaceous aggregates in the brain, and with the formation of metastable intermediates known as protein misfolded oligomers that play a central role in their aetiology. In this Highlight article, we review the evidence at the physicochemical, cellular, animal model and clinical levels on how the natural products squalamine and trodusquemine offer promising opportunities for chronic treatments for these progressive conditions by preventing both the formation of neurotoxic oligomers and their interaction with cell membranes.


Subject(s)
Alzheimer Disease , Biological Products , Neurodegenerative Diseases , Alzheimer Disease/drug therapy , Animals , Biological Products/pharmacology , Chemistry, Physical , Cholestanes , Cholestanols , Humans , Neurodegenerative Diseases/drug therapy , Spermine/analogs & derivatives
7.
Sci Rep ; 11(1): 21130, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702901

ABSTRACT

The vagus nerve relays mood-altering signals originating in the gut lumen to the brain. In mice, an intact vagus is required to mediate the behavioural effects of both intraluminally applied selective serotonin reuptake inhibitors and a strain of Lactobacillus with antidepressant-like activity. Similarly, the prodepressant effect of lipopolysaccharide is vagus nerve dependent. Single vagal fibres are broadly tuned to respond by excitation to both anti- and prodepressant agents, but it remains unclear how neural responses encode behaviour-specific information. Here we demonstrate using ex vivo experiments that for single vagal fibres within the mesenteric neurovascular bundle supplying the mouse small intestine, a unique neural firing pattern code is common to both chemical and bacterial vagus-dependent antidepressant luminal stimuli. This code is qualitatively and statistically discernible from that evoked by lipopolysaccharide, a non-vagus-dependent antidepressant or control non-antidepressant Lactobacillus strain and are not affected by sex status. We found that all vagus dependent antidepressants evoked a decrease in mean spike interval, increase in spike burst duration, decrease in gap duration between bursts and increase in intra-burst spike intervals. Our results offer a novel neuronal electrical perspective as one explanation for mechanisms of action of gut-derived vagal dependent antidepressants. We expect that our ex vivo individual vagal fibre recording model will improve the design and operation of new, extant electroceutical vagal stimulation devices currently used to treat major depression. Furthermore, use of this vagal antidepressant code should provide a valuable screening tool for novel potential oral antidepressant candidates in preclinical animal models.


Subject(s)
Action Potentials/drug effects , Antidepressive Agents , Lactobacillus/chemistry , Selective Serotonin Reuptake Inhibitors , Vagus Nerve/physiopathology , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Female , Male , Mice , Mice, Inbred BALB C , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology
8.
ACS Chem Neurosci ; 12(17): 3189-3202, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34382791

ABSTRACT

Many neurodegenerative diseases are associated with the self-assembly of peptides and proteins into fibrillar aggregates. Soluble misfolded oligomers formed during the aggregation process, or released by mature fibrils, play a relevant role in neurodegenerative processes through their interactions with neuronal membranes. However, the determinants of the cytotoxicity of these oligomers are still unclear. Here we used liposomes and toxic and nontoxic oligomers formed by the same protein to measure quantitatively the affinity of the two oligomeric species for lipid membranes. To this aim, we quantified the perturbation to the lipid membranes caused by the two oligomers by using the fluorescence quenching of two probes embedded in the polar and apolar regions of the lipid membranes and a well-defined protein-oligomer binding assay using fluorescently labeled oligomers to determine the Stern-Volmer and dissociation constants, respectively. With both approaches, we found that the toxic oligomers have a membrane affinity 20-25 times higher than that of nontoxic oligomers. Circular dichroism, intrinsic fluorescence, and FRET indicated that neither oligomer type changes its structure upon membrane interaction. Using liposomes enriched with trodusquemine, a potential small molecule drug known to penetrate lipid membranes and make them refractory to toxic oligomers, we found that the membrane affinity of the oligomers was remarkably lower. At protective concentrations of the small molecule, the binding of the oligomers to the lipid membranes was fully prevented. Furthermore, the affinity of the toxic oligomers for the lipid membranes was found to increase and slightly decrease with GM1 ganglioside and cholesterol content, respectively, indicating that physicochemical properties of lipid membranes modulate their affinity for misfolded oligomeric species.


Subject(s)
Cholestanes , Lipid Bilayers , Amyloid beta-Peptides , G(M1) Ganglioside , Spermine/analogs & derivatives
9.
Front Neurosci ; 15: 680026, 2021.
Article in English | MEDLINE | ID: mdl-34220435

ABSTRACT

The aberrant aggregation of proteins is a key molecular event in the development and progression of a wide range of neurodegenerative disorders. We have shown previously that squalamine and trodusquemine, two natural products in the aminosterol class, can modulate the aggregation of the amyloid-ß peptide (Aß) and of α-synuclein (αS), which are associated with Alzheimer's and Parkinson's diseases. In this work, we expand our previous analyses to two squalamine derivatives, des-squalamine and α-squalamine, obtaining further insights into the mechanism by which aminosterols modulate Aß and αS aggregation. We then characterize the ability of these small molecules to alter the physicochemical properties of stabilized oligomeric species in vitro and to suppress the toxicity of these aggregates to varying degrees toward human neuroblastoma cells. We found that, despite the fact that these aminosterols exert opposing effects on Aß and αS aggregation under the conditions that we tested, the modifications that they induced to the toxicity of oligomers were similar. Our results indicate that the suppression of toxicity is mediated by the displacement of toxic oligomeric species from cellular membranes by the aminosterols. This study, thus, provides evidence that aminosterols could be rationally optimized in drug discovery programs to target oligomer toxicity in Alzheimer's and Parkinson's diseases.

10.
Front Cell Dev Biol ; 9: 552549, 2021.
Article in English | MEDLINE | ID: mdl-33829010

ABSTRACT

The aggregation of α-synuclein is a hallmark of Parkinson's disease (PD) and a variety of related neurological disorders. A number of mutations in this protein, including A30P and A53T, are associated with familial forms of the disease. Patients carrying the A30P mutation typically exhibit a similar age of onset and symptoms as sporadic PD, while those carrying the A53T mutation generally have an earlier age of onset and an accelerated progression. We report two C. elegans models of PD (PDA30P and PDA53T), which express these mutational variants in the muscle cells, and probed their behavior relative to animals expressing the wild-type protein (PDWT). PDA30P worms showed a reduced speed of movement and an increased paralysis rate, control worms, but no change in the frequency of body bends. By contrast, in PDA53T worms both speed and frequency of body bends were significantly decreased, and paralysis rate was increased. α-Synuclein was also observed to be less well localized into aggregates in PDA30P worms compared to PDA53T and PDWT worms, and amyloid-like features were evident later in the life of the animals, despite comparable levels of expression of α-synuclein. Furthermore, squalamine, a natural product currently in clinical trials for treating symptomatic aspects of PD, was found to reduce significantly the aggregation of α-synuclein and its associated toxicity in PDA53T and PDWT worms, but had less marked effects in PDA30P. In addition, using an antibody that targets the N-terminal region of α-synuclein, we observed a suppression of toxicity in PDA30P, PDA53T and PDWT worms. These results illustrate the use of these two C. elegans models in fundamental and applied PD research.

11.
Nanoscale ; 12(44): 22596-22614, 2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33150350

ABSTRACT

Trodusquemine is an aminosterol known to prevent the binding of misfolded protein oligomers to cell membranes and to reduce their toxicity in a wide range of neurodegenerative diseases. Its precise mechanism of action, however, remains unclear. To investigate this mechanism, we performed confocal microscopy, fluorescence resonance energy transfer (FRET) and nuclear magnetic resonance (NMR) measurements, which revealed a strong binding of trodusquemine to large unilamellar vesicles (LUVs) and neuroblastoma cell membranes. Then, by combining quartz crystal microbalance (QCM), fluorescence quenching and anisotropy, and molecular dynamics (MD) simulations, we found that trodusquemine localises within, and penetrates, the polar region of lipid bilayer. This binding behaviour causes a decrease of the negative charge of the bilayer, as observed through ζ potential measurements, an increment in the mechanical resistance of the bilayer, as revealed by measurements of the breakthrough force applied with AFM and ζ potential measurements at high temperature, and a rearrangement of the spatial distances between ganglioside and cholesterol molecules in the LUVs, as determined by FRET measurements. These physicochemical changes are all known to impair the interaction of misfolded oligomers with cell membranes, protecting them from their toxicity. Taken together, our results illustrate how the incorporation in cell membranes of sterol molecules modified by the addition of polyamine tails leads to the modulation of physicochemical properties of the cell membranes themselves, making them more resistant to protein aggregates associated with neurodegeneration. More generally, they suggest that therapeutic strategies can be developed to reinforce cell membranes against protein misfolded assemblies.


Subject(s)
Lipid Bilayers , Unilamellar Liposomes , Cell Membrane , Cholestanes , Spermine/analogs & derivatives
12.
J Parkinsons Dis ; 10(4): 1477-1491, 2020.
Article in English | MEDLINE | ID: mdl-32925094

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder thought to be caused by accumulation of α-synuclein (α-syn) within the brain, autonomic nerves, and the enteric nervous system (ENS). Involvement of the ENS in PD often precedes the onset of the classic motor signs of PD by many years at a time when severe constipation represents a major morbidity. Studies conducted in vitro and in vivo, have shown that squalamine, a zwitterionic amphipathic aminosterol, originally isolated from the liver of the dogfish shark, effectively displaces membrane-bound α-syn. OBJECTIVE: Here we explore the electrophysiological effect of squalamine on the gastrointestinal (GI) tract of mouse models of PD engineered to express the highly aggregating A53T human α-syn mutant. METHODS: GI motility and in vivo response to oral squalamine in PD model mice and controls were assessed using an in vitro tissue motility protocol and via fecal pellet output. Vagal afferent response to squalamine was measured using extracellular mesenteric nerve recordings from the jejunum. Whole cell patch clamp was performed to measure response to squalamine in the myenteric plexus. RESULTS: Squalamine effectively restores disordered colonic motility in vivo and within minutes of local application to the bowel. We show that topical squalamine exposure to intrinsic primary afferent neurons (IPANs) of the ENS rapidly restores excitability. CONCLUSION: These observations may help to explain how squalamine may promote gut propulsive activity through local effects on IPANs in the ENS, and further support its possible utility in the treatment of constipation in patients with PD.


Subject(s)
Constipation/drug therapy , Electrophysiological Phenomena/drug effects , Enteric Nervous System/drug effects , Gastrointestinal Motility/drug effects , Myenteric Plexus/drug effects , Neurons, Afferent/drug effects , Parkinson Disease/complications , Vagus Nerve/drug effects , Animals , Cholestanols/administration & dosage , Cholestanols/pharmacology , Constipation/etiology , Disease Models, Animal , Jejunum/innervation , Mice , Mice, Transgenic , Mutant Proteins , Neurons, Afferent/cytology , Patch-Clamp Techniques , alpha-Synuclein/metabolism
13.
J Parkinsons Dis ; 9(s2): S313-S322, 2019.
Article in English | MEDLINE | ID: mdl-31594249

ABSTRACT

The gastrointestinal (GI) tract is equipped with robust immune defenses which protect the organism from infection. Enteric nerves are front and center in this defensive network, even in the most primitive organisms. Neuropeptides exhibit potent antimicrobial activity in the vicinity of the nerve and attract the innate and adaptive immune systems to help confine the invading agent. Alpha-synuclein (αS) has many biophysical characteristics of antimicrobial peptides and binds small vesicles such as those carrying endocytosed viruses. It is induced in nerve cells in response to viral and bacterial infections. It renders the nerve cell resistant to viral infection and propagation. It signals the immune system by attracting neutrophils and macrophages, and by activating dendritic cells. Most remarkably αS is trafficked to the central nervous system (CNS) conferring immunity in advance of an infection. Chronic GI infection or breakdown of the epithelial barrier can cause αS to accumulate and form neurotoxic aggregates. Overproduction of αS in the enteric nervous system (ENS) and its chronic trafficking to the CNS may damage nerves and lead to Parkinson's disease. Targeting the formation of αS aggregates in the ENS may therefore slow the progression of the disease.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Enteric Nervous System/metabolism , Immunity/physiology , Neuropeptides/physiology , Parkinson Disease , alpha-Synuclein/physiology , Animals , Humans , Parkinson Disease/etiology , Parkinson Disease/immunology , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
14.
Clin Park Relat Disord ; 1: 2-7, 2019.
Article in English | MEDLINE | ID: mdl-34316590

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is associated with α-synuclein (αS) aggregation within the enteric nervous system (ENS) and constipation. Squalamine displaces proteins that are electrostatically bound to intracellular membranes and through this mechanism suppresses aggregation of αS monomers into neurotoxic oligomers. OBJECTIVE: We sought to evaluate the safety of ENT-01 oral tablets (a synthetic squalamine salt), its pharmacokinetics, and its effect on bowel function in PD patients with constipation. METHODS: In Stage 1, 10 patients received escalating single doses from 25 to 200 mg/day or maximum tolerated dose (MTD). In Stage 2, 34 patients received daily doses escalating from 75 to a maximum of 250 mg/day, a dose that induced change in bowel function or MTD, followed by a fixed dose for 7 days, and a 2-week washout. Primary efficacy endpoint was defined as an increase of 1 complete spontaneous bowel movement (CSBM)/week, or 3 CSBM/week over the baseline period, as defined by FDA guidelines for prokinetic agents. Safety was also assessed. RESULTS: Over 80% of patients achieved the primary efficacy endpoint, with the mean number of CSBM/week increasing from 1.2 at baseline to 3.6 during fixed dosing (p = 1.2 × 10-7). Common adverse events included nausea in 21/44 (47%) and diarrhea in 18/44 (40%) patients. Systemic absorption was <0.3%. CONCLUSIONS: Orally administered ENT-01 was safe and significantly improved bowel function in PD, suggesting that the ENS is not irreversibly damaged in PD. Minimal systemic absorption suggests that improvements result from local stimulation of the ENS. A double-blind, placebo-controlled study is now ongoing.

15.
ACS Chem Biol ; 13(8): 2308-2319, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29953201

ABSTRACT

The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely associated with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compound trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic reduction in the number of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compound has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.


Subject(s)
Cholestanes/pharmacology , Parkinson Disease/drug therapy , Protein Aggregates/drug effects , Protein Aggregation, Pathological/prevention & control , Spermine/analogs & derivatives , alpha-Synuclein/metabolism , Animals , Caenorhabditis elegans/physiology , Cell Line , Cholestanes/therapeutic use , Disease Models, Animal , Humans , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/metabolism , Protein Aggregation, Pathological/metabolism , Spermine/pharmacology , Spermine/therapeutic use
16.
J Innate Immun ; 9(5): 456-463, 2017.
Article in English | MEDLINE | ID: mdl-28651250

ABSTRACT

BACKGROUND: Alpha-synuclein (αS) is a nerve cell protein associated with Parkinson disease (PD). Accumulation of αS within the enteric nervous system (ENS) and its traffic from the gut to the brain are implicated in the pathogenesis and progression of PD. αS has no known function in humans and the reason for its accumulation within the ENS is unknown. Several recent studies conducted in rodents have linked αS to immune cell activation in the central nervous system. We hypothesized that αS in the ENS might play a role in the innate immune defenses of the human gastrointestinal (GI) tract. METHODS: We immunostained endoscopic biopsies for αS from children with documented gastric and duodenal inflammation and intestinal allograft recipients who contracted norovirus. To determine whether αS exhibited immune-modulatory activity, we examined whether human αS induced leukocyte migration and dendritic cell maturation. FINDINGS: We showed that the expression of αS in the enteric neurites of the upper GI tract of pediatric patients positively correlated with the degree of acute and chronic inflammation in the intestinal wall. In intestinal allograft subjects who were closely monitored for infection, expression of αS was induced during norovirus infection. We also demonstrated that both monomeric and oligomeric αS have potent chemoattractant activity, causing the migration of neutrophils and monocytes dependent on the presence of the integrin subunit, CD11b, and that both forms of αS stimulate dendritic cell maturation. INTERPRETATION: These findings strongly suggest that αS is expressed within the human ENS to direct intestinal inflammation and implicates common GI infections in the pathogenesis of PD.


Subject(s)
Caliciviridae Infections/immunology , Dendritic Cells/immunology , Duodenitis/immunology , Gastritis/immunology , Intestines/immunology , Monocytes/immunology , Nervous System , Neurons/metabolism , Neutrophils/immunology , Norovirus/physiology , Parkinson Disease/immunology , alpha-Synuclein/metabolism , Adolescent , CD11b Antigen/metabolism , Cell Differentiation , Cell Movement , Cells, Cultured , Chemotaxis , Child , Female , Humans , Immunity, Innate , Intestines/virology , Male , Parkinson Disease/virology , Protein Folding , alpha-Synuclein/immunology
17.
Invest Ophthalmol Vis Sci ; 55(6): 3586-93, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24825106

ABSTRACT

PURPOSE: To characterize the analgesic potency and toxicity of topical synthetic neurotensin analogues, and localize neurotensin receptors in the cornea and trigeminal ganglion. METHODS: Cochet-Bonnet esthesiometry was performed on the rabbit cornea to test the analgesic dose response and duration of effect for two synthetic neurotensin analogues: NT71 and NT72. Receptors for neurotensin were localized in the murine cornea and trigeminal ganglion using quantitative PCR (qPCR), Western blotting, and immunohistochemistry. In vitro toxicity of NT71, NT72, and sodium channel blockers was evaluated using cytotoxicity, single-cell migration, and scratch closure assays performed on rabbit corneal epithelial cells. In vivo toxicity of these agents was assessed using a rabbit laser phototherapeutic keratectomy (PTK) model and histology. RESULTS: NT71 and NT72 induced potent analgesic effects on the rabbit cornea at concentrations between 1.0 and 2.5 mg/mL, lasting up to 180 minutes. A site-specific distribution of neurotensin receptors was observed in the murine cornea and trigeminal ganglion. NT71 and NT72 did not cause any significant in vitro or in vivo toxicity, in contrast to sodium channel blockers. CONCLUSIONS: Synthetic neurotensin analogues are potent analgesics that avoid the toxicities associated with established topical analgesic agents. Receptors for neurotensin are present in both the cornea and trigeminal ganglion.


Subject(s)
Analgesics/administration & dosage , Cornea/drug effects , Eye Injuries/drug therapy , Neurotensin/analogs & derivatives , Receptors, Neurotensin/biosynthesis , Trigeminal Ganglion/metabolism , Animals , Blinking/drug effects , Blotting, Western , Cell Movement , Cells, Cultured , Chromatography, High Pressure Liquid , Cornea/pathology , Corneal Injuries , Disease Models, Animal , Eye Injuries/metabolism , Eye Injuries/pathology , Gene Expression Regulation , Immunohistochemistry , Ophthalmic Solutions/administration & dosage , RNA, Messenger/genetics , Rabbits , Receptors, Neurotensin/genetics , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/pathology , Wound Healing
18.
Crit Care Med ; 40(2): 580-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21926589

ABSTRACT

OBJECTIVE: Therapeutic hypothermia initiated with cardiopulmonary resuscitation improves neurologic outcomes and survival after prolonged cardiac arrest. However, the potential mechanism by which hypothermia improves neurologic outcomes remains unclear. In the current study, we investigated the effect of rapid head cooling on 96-hr neurologic outcomes and survival by heart rate variability analysis in a pig model of prolonged cardiac arrest. DESIGN: Prospective randomized controlled animal study. SETTING: University-affiliated research laboratory. SUBJECTS: Yorkshire-X domestic pigs (Sus scrofa). INTERVENTIONS: A protocol of 10 mins of untreated ventricular fibrillation followed by 5 mins of cardiopulmonary resuscitation in a pig model of cardiac arrest was used in this study. Sixteen male domestic pigs weighing between 39 and 45 kg were randomized into two groups, hypothermia (n = 8) and control (n = 8). For the hypothermia group, intranasal-induced head cooling was initiated with cardiopulmonary resuscitation and persisted for 4 hrs after resuscitation. For the control group, cardiopulmonary resuscitation was started with normothermia. MEASUREMENTS AND MAIN RESULTS: Time and frequency domain heart rate variability was calculated in 5-min sections of electrocardiographic recordings at baseline and 4 hrs after resuscitation. Neurologic outcomes were evaluated every 24 hrs during the 96-hr postresuscitation observation period. No differences in the baseline measurement and resuscitation outcome were observed between the groups. However, the 96-hr cerebral performance categories of the hypothermic group were significantly lower than control (1.0 ± 0.0 vs. 4.0 ± 1.9, p = .003). Four hrs after resuscitation, mean RR interval, heart rate variability triangular index, and normalized very-low-frequency power were restored to baseline in the hypothermia group. Square root of the mean squared differences of successive RR intervals and SD of instantaneous RR intervals were significantly improved in the cooled animals compared with controls. A significant correlation between 4-hr heart rate variability and 96-hr cerebral performance category was observed in this study. CONCLUSION: Selective head cooling maintains a certain level of autonomic nervous system function in this pig model of cardiac arrest. The preserved heart rate variability during postresuscitation hypothermia was associated with favorable 96-hr neurologic recovery and survival.


Subject(s)
Cardiopulmonary Resuscitation/methods , Heart Arrest/therapy , Heart Rate/physiology , Hypothermia, Induced/methods , Nervous System/physiopathology , Animals , Disease Models, Animal , Electrocardiography , Heart Arrest/mortality , Hemodynamics/physiology , Logistic Models , Male , Random Allocation , Reference Values , Survival Rate , Sus scrofa , Swine , Time Factors , Treatment Outcome , Ventricular Fibrillation/mortality , Ventricular Fibrillation/therapy
19.
Stroke ; 42(8): 2164-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680904

ABSTRACT

BACKGROUND AND PURPOSE: Hypothermia is neuroprotectant but currently available cooling methods are laborious, invasive, and require whole-body cooling. There is a need for less invasive cooling of the brain. This study was conducted to assess the safety and efficacy of temperature reduction of the RhinoChill transnasal cooling device. METHODS: We conducted a prospective single-arm safety and feasibility study of intubated patients for whom temperature reduction was indicated. After rhinoscopy, the device was activated for 1 hour. Brain, tympanic, and core temperatures along with vital signs and laboratory studies were recorded. All general and device-related adverse events were collected for the entire hypothermia treatment. RESULTS: A total of 15 patients (mean age, 50.3 ± 17.1 years) were enrolled. Brain injury was caused by intracerebral hemorrhage, trauma, and ischemic stroke in equal numbers. Hypothermia was induced for fever control in 9 patients and for neuroprotection/intracranial pressure control in 6. Core temperature, brain temperature, and tympanic temperature were reduced an average of 1.1 ± 0.6°C (range, 0.3 to 2.1°C), 1.4 ± 0.4°C (range, 0.8 to 5.1°C), and 2.2 ± 2°C (range, 0.5 to 6.5°C), respectively. Only 2 patients did not achieve the goal of ≥1°C decrease in temperature. Brain temperature, tympanic temperature, and core temperature reductions were similar between the afebrile and febrile patients. There were no unanticipated adverse events and only 1 anticipated adverse event: hypertension in 1 subject that led to discontinuation of cooling after 30 minutes. There were no nasal complications. CONCLUSIONS: Intranasal cooling with the RhinoChill device appears safe and effectively lowers brain and core temperatures. Further study is warranted to assess the efficacy of hypothermia through intranasal cooling for brain-injured patients.


Subject(s)
Brain Injuries/therapy , Equipment Safety , Fever/therapy , Hypothermia, Induced/adverse effects , Hypothermia, Induced/instrumentation , Stroke/therapy , Adult , Aged , Aged, 80 and over , Brain/physiopathology , Brain Injuries/physiopathology , Feasibility Studies , Female , Fever/physiopathology , Humans , Hypothermia, Induced/methods , Male , Middle Aged , Prospective Studies , Stroke/physiopathology
20.
Circulation ; 122(7): 729-36, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679548

ABSTRACT

BACKGROUND: Transnasal evaporative cooling has sufficient heat transfer capacity for effective intra-arrest cooling and improves survival in swine. The aim of this study was to determine the safety, feasibility, and cooling efficacy of prehospital transnasal cooling in humans and to explore its effects on neurologically intact survival to hospital discharge. METHODS AND RESULTS: Witnessed cardiac arrest patients with a treatment interval

Subject(s)
Emergency Medical Services/methods , Heart Arrest/therapy , Hypothermia, Induced/instrumentation , Hypothermia, Induced/methods , Administration, Intranasal , Aged , Body Temperature/physiology , Cold Temperature , Female , Heart Arrest/physiopathology , Humans , Male , Middle Aged , Prospective Studies , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL