Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Neurobiol Dis ; 191: 106387, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38142841

ABSTRACT

INTRODUCTION: Purportedly, the progression of multiple sclerosis (MS) occurs when neurodegenerative processes due to derangement of axonal bioenergetics take over the autoimmune response. However, a clear picture of the causative interrelationship between autoimmunity and axonal mitochondrial dysfunction in progressive MS (PMS) pathogenesis waits to be provided. METHODS: In the present study, by adopting the NOD mouse model of PMS, we compared the pharmacological effects of the immunosuppressants dexamethasone and fingolimod with those of mTOR inhibitors rapamycin and everolimus that, in addition to immunosuppression, also regulate mitochondrial functioning. Female Non-Obese Diabetic (NOD) mice were immunized with MOG35-55 and treated with drugs to evaluate functional, immune and mitochondrial parameters during disease evolution. RESULTS: We found that dexamethasone and fingolimod did not affect the pattern of progression as well as survival. Conversely, mTOR inhibitors rapamycin and everolimus delayed disease progression and robustly extended survival of immunized mice. The same effects were obtained when treatment was delayed by 30 days after immunization. Remarkably, dexamethasone and fingolimod prompted the same degree of immunosuppression of rapamycin within both spleen and spinal cord of mice. However, only rapamycin prompted mitochondriogenesis by increasing mitochondrial content, and expression of several mitochondrial respiratory complex subunits, thereby preventing mtDNA reduction in the spinal cords of immunized mice. These pharmacodynamic effects were not reproduced in healthy NOD mice, suggesting a disease context-dependent pharmacodynamic effect. DISCUSSION: Data corroborate the key role of mitochondriogenesis to treatment of MS progression, and for the first time disclose the translational potential of mTOR inhibitors in PMS therapy.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Female , Animals , Mice , Multiple Sclerosis/pathology , MTOR Inhibitors , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Neuroprotection , Everolimus/pharmacology , Everolimus/therapeutic use , Mice, Inbred NOD , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Sirolimus/pharmacology , Sirolimus/therapeutic use , Dexamethasone/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice, Inbred C57BL
2.
Neurobiol Dis ; 178: 106015, 2023 03.
Article in English | MEDLINE | ID: mdl-36702320

ABSTRACT

INTRODUCTION: Derangement of axonal mitochondrial bioenergetics occurs during progressive multiple sclerosis (PMS). However, whether this is a delayed epiphenomenon or an early causative event of disease progression waits to be understood. Answering this question might further our knowledge of mechanisms underlying neurobiology of PMS and related therapy. METHODS: MOG35-55-immunized NOD and PLP139-151-immunized SJL female mice were adopted as models of progressive or relapsing-remitting experimental autoimmune encephalomyelitis (EAE), respectively. Multiple parameters of mitochondrial homeostasis were analyzed in the mouse spinal cord during the early asymptomatic stage, also evaluating the effects of scavenging mitochondrial reactive oxygen species with Mito-TEMPO. RESULTS: Almost identical lumbar spinal cord immune infiltrates consisting of Th1 cells and neutrophils without B and Th17 lymphocytes occurred early upon immunization in both mouse strains. Still, only NOD mice showed axon-restricted dysregulation of mitochondrial homeostasis, with reduced mtDNA contents and increased cristae area. Increased expression of mitochondrial respiratory complex subunits Nd2, Cox1, Atp5d, Sdha also exclusively occurred in lumbar spinal cord of NOD and not SJL mice. Accordingly, in this region genes regulating mitochondrial morphology (Opa1, Mfn1, Mfn2 and Atp5j2) and mitochondriogenesis (Pgc1α, Foxo, Hif-1α and Nrf2) were induced early upon immunization. A reduced extent of mitochondrial derangement occurred in the thoracic spinal cord. Notably, the mitochondrial radical scavenger Mito-TEMPO reduced H2O2 content and prevented both mtDNA depletion and cristae remodeling, having no effects on dysregulation of mitochondrial transcriptome. DISCUSSION: We provide here the first evidence that axonal-restricted derangement of mitochondrial homeostasis already occurs during the asymptomatic state exclusively in a mouse model of PMS. Data further our understanding of mechanisms related to EAE progression, and point to very early axonal mitochondrial dysfunction as central to the neuropathogenesis of MS evolution.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Mice , Female , Animals , Multiple Sclerosis/pathology , Hydrogen Peroxide/metabolism , Mice, Inbred NOD , Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/pathology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Axons/pathology , Mitochondria/metabolism , DNA, Mitochondrial/metabolism
3.
Neonatology ; 119(5): 611-618, 2022.
Article in English | MEDLINE | ID: mdl-36096109

ABSTRACT

BACKGROUND: We recently demonstrated that oxygen-glucose deprivation (OGD) and unconjugated bilirubin (UCB) can damage mature and immature organotypic hippocampal slices and induce an oxidative stress similar to what occurs in jaundiced term and preterm infants with hypoxic-ischemic encephalopathy (HIE). OBJECTIVES: To assess the effects of OGD and UCB on the expression of heme-oxygenase 1 (HO-1) and oxidative stress-related enzymes in an in vitro model of HIE. METHODS: Mature and immature organotypic hippocampal slices were exposed to 30-min OGD and to 24 h UCB or UCB plus human serum albumin (HSA). The expression of HO-1, superoxide dismutase 1 (SOD1), catalase (CAT), glutathione peroxidase (GPX), and nuclear factor erythroid-related factor 2 were analyzed by real-time PCR. RESULTS: In mature slices, OGD did not affect the expression of HO-1 and oxidative stress-induced enzymes. The addition of UCB was associated with the upregulation of HO-1 and Nrf2 that is abolished by the presence of equimolar amount of HSA. In immature slices, OGD induced the downregulation of CAT, GPX, and Nrf2 expression and the addition of UCB further decreased GPX. The addition of UCB and HSA reverted the effects of OGD and UCB on gene expression. CONCLUSIONS: In an in vitro model of HIE in term infants, we did not observe neuroprotective changes of the expression of HO-1 and genes involved in antioxidant defenses. Conversely, in an in vitro model of HIE in preterm infants, we observed a harmful decrease of the expression of genes encoding for antioxidant enzymes.


Subject(s)
Hypoxia-Ischemia, Brain , NF-E2-Related Factor 2 , Antioxidants/pharmacology , Bilirubin , Catalase/genetics , Catalase/metabolism , Catalase/pharmacology , Gene Expression , Glucose , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Peroxidase/pharmacology , Heme/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Infant, Premature , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , Oxidative Stress , Oxygen/metabolism , Serum Albumin, Human/pharmacology , Superoxide Dismutase-1/genetics
4.
Nutr Metab Cardiovasc Dis ; 32(2): 420-428, 2022 02.
Article in English | MEDLINE | ID: mdl-34893418

ABSTRACT

BACKGROUND AND AIM: Left heart remodeling is a well-known pathophysiological effect of arterial hypertension. Right Heart status is not considered in its evaluation. No data are available on right atrium (RA) and its impact on the outcome in hypertension. We wondering to understand whether RA may play a role as a marker of an increased risk for organ damage in well-controlled hypertensives, to probe the clinical significance and whether it could indicate an increased risk. METHODS AND RESULTS: We studied well-controlled hypertensive patients. Heart damage was assessed by echocardiography. Patients were subdivided into those with RA area ≤18 cm2 (normal RA - Group 1) (554 pts, 227 M, aged 60.35 ± 10.48 years) and those >18 cm2 (Increased RA - Group 2) (101 pts, 71 M, age 61.65 ± 9.46 years). Group 2 had a higher left ventricle mass (LVM) and left atrium volume (LAV) both as absolute value (both p < 0.0001) and indexed for body surface area (LVMi p < 0.013; LAVi p = 0.0013). Group 2 showed an increased vascular stiffness (p < 0.0001) and carotid stenosis percentage (p = 0.011). TAPSE (p < 0.0001) resulted significantly increased. In The RA area was significantly correlated directly to LVM and LAV in both groups, but these correlations persisted in indexed values only in Group 2. Moreover, in this group there was a significant direct correlation between RA area and Tricuspid s'wave at echocardiography TDI analysis. Finally, Group 2 had an increased mortality rate compared to Group 1 (Log-Rank p = 0.0006). CONCLUSION: Group 2 hypertensive patients showed more alterations in dimensional and volumetric left heart parameters, and an increased mortality.


Subject(s)
Heart Injuries , Hypertension , Aged , Cardiomegaly/diagnostic imaging , Echocardiography , Heart Atria/diagnostic imaging , Humans , Hypertension/complications , Hypertension/diagnosis , Middle Aged
5.
J Biol Chem ; 297(1): 100855, 2021 07.
Article in English | MEDLINE | ID: mdl-34097876

ABSTRACT

In the last several years, NAD+ supplementation has emerged as an innovative and safe therapeutic strategy for a wide spectrum of disorders, including diabetes and neuropathy. However, critical questions remain as to how NAD+ and its precursors are taken up by cells, as well as the effects of long-lasting intracellular NAD+ (iNAD+) increases. Here, we investigated the kinetics of iNAD+ levels in different cell types challenged with prolonged exposure to extracellular NAD+ (eNAD+). Surprisingly, we found that after the initial increase, iNAD+ contents decreased back to control levels (iNAD+ resetting). Focusing our attention on HeLa cells, we found that oxygen and ATP consumption occurred with similar temporal kinetics after eNAD+ exposure. Using [3H]NAD+ and [14C]NAD+, we determined that NAD+ resetting was not due to increased dinucleotide extrusion but rather due to reduced uptake of cleaved NAD+ products. Indeed, eNAD+ exposure reduced the expression of the ecto-5'-nucleotidase CD73, the nicotinamide adenine mononucleotide transporter solute carrier family 12 member 8, and the nicotinamide riboside kinase. Interestingly, silencing the NAD+-sensor enzyme sirtuin 1 prevented eNAD+-dependent transcriptional repression of ecto-5'-nucleotidase, solute carrier family 12 member 8, and nicotinamide riboside kinase, as well as iNAD+ resetting. Our findings provide the first evidence for a sirtuin 1-mediated homeostatic response aimed at maintaining physiological iNAD+ levels in conditions of excess eNAD+ availability. These data may be of relevance for therapies designed to support the NAD+ metabolome via extracellular supplementation of the dinucleotide or its precursors.


Subject(s)
5'-Nucleotidase/genetics , ADP-ribosyl Cyclase 1/genetics , Energy Metabolism/genetics , Membrane Glycoproteins/genetics , NAD/metabolism , Sirtuin 1/genetics , Adenosine Triphosphate/metabolism , Biological Transport/genetics , HeLa Cells , Homeostasis/genetics , Humans , Kinetics , Oxygen/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Signal Transduction/genetics
6.
Neuroscience ; 465: 38-45, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33862148

ABSTRACT

Drugs able to efficiently counteract progression of multiple sclerosis (MS) are still an unmet need. Several lines of evidence indicate that histone deacetylase inhibitors (HDACi) are clinically-available epigenetic drugs that might be repurposed for immunosuppression in MS therapy. Here, we studied the effects of HDACi on disease evolution in myelin oligodendrocyte glycoprotein (MOG)-immunized NOD mice, an experimental model of progressive experimental autoimmune encephalomyelitis (PEAE). To obtain data of potential clinical relevance, the HDACi panobinostat, givinostat and entinostat were administered orally adopting a daily treatment protocol after disease onset. We report that the 3 drugs efficiently reduced in vitro lymphocyte proliferation in a dose-dependent manner. Notably, however, none of the drugs delayed evolution of PEAE or reduced lethality in NOD mice. In striking contrast with this, however, the lymphocyte proliferation response to MOG as well as Th1 and Th17 spinal cord infiltrates were significantly lower in animals exposed to the HDACi compared to those receiving vehicle. When put into a clinical context, for the first time data cast doubt on the relevance of HDACi to treatment of progressive MS (PMS). Also, our findings further indicate that, akin to PMS, neuropathogensis of PEAE in NOD mice becomes independent from autoimmunity, thereby corroborating the relevance of this model to experimental PMS research.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Multiple Sclerosis/drug therapy , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments
7.
Sleep Med ; 82: 117-124, 2021 06.
Article in English | MEDLINE | ID: mdl-33910161

ABSTRACT

BACKGROUND: Galectin-3, considered as a new inflammatory marker; it is increased in cardiovascular disease. We investigated Galectin-3 in relation to heart damage in patients with OSA and its role in inflammation, based on the Neutrophil-to-Lymphocyte Ratio (NLR). METHODS: Sixty-three consecutive patients (45 males, 18 females, 58.60 ± 12.28 years old) were studied. According to the Apnoea-Hypopnoea Index (AHI) patients were divided into Group 1 - non-severe (AHI <30) (17 males and 10 females, 59.89 ± 10.62 years old) and Group 2 - severe (AHI ≥30) (29 males and 6 females, aged 57.53 ± 13.30 years old) OSA. All patients underwent morning blood gas analysis, laboratory tests, nocturnal polygraphy, and echocardiography. RESULTS: Galectin-3 was significantly increased in Group 2 (p = 0.027) patients. Moreover, it was directly related to left ventricle (LV) mass, left ventricle hypertrophy and LV posterior wall diameter. Tissue Doppler septal velocity (e'), that measures wall motion, was inversely correlated to Galectin-3. Furthermore, a direct association to diastolic dysfunction, evaluated as E/e' ratio, was observed. In line with these data, a direct correlation between Galectin-3 and left atrium volume was also found. Galectin-3 and percentage of total registration time with nocturnal oxygen saturation <90% (TST90) were directly correlated (p = 0.0003), while Galectin-3 and mean nocturnal SpO2 were negatively correlated (p = 0.0045). We found a direct correlation between Galectin-3 and NLR (p = 0.011). Finally, Galectin-3 was able to predict 3-yr mortality with a specificity of 83.33% and a sensitivity of 91.84%. CONCLUSION: Galectin-3 showed a direct association to nocturnal respiratory indices and to cardiac remodelling in patients with OSA. OSA-induced inflammation may play an important role in the pathogenesis of heart damage.


Subject(s)
Galectin 3 , Sleep Apnea, Obstructive , Adult , Aged , Blood Proteins , Female , Galectins , Humans , Lymphocytes , Male , Middle Aged , Neutrophils , Polysomnography , Severity of Illness Index
8.
Mol Neurobiol ; 58(6): 2955-2962, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33566318

ABSTRACT

Dexpramipexole (DEX) has been described as the first-in-class F1Fo ATP synthase activator able to boost mitochondrial bioenergetics and provide neuroprotection in experimental models of ischemic brain injury. Although DEX failed in a phase III trial in patients with amyotrophic lateral sclerosis, it showed favorable safety and tolerability profiles. Recently, DEX emerged as a Nav1.8 Na+ channel and transient outward K+ (IA) conductance blocker, revealing therefore an unexpected, pleiotypic pharmacodynamic profile. In this study, we performed electrophysiological experiments in vitro aimed to better characterize the impact of DEX on voltage-dependent currents and synaptic transmission in the hippocampus. By means of patch-clamp recordings on isolated hippocampal neurons, we found that DEX increases outward K+ currents evoked by a voltage ramp protocol. This effect is prevented by the non-selective voltage-dependent K+ channel (Kv) blocker TEA and by the selective small-conductance Ca2+-activated K+ (SK) channel blocker apamin. In keeping with this, extracellular field recordings from rat hippocampal slices also demonstrated that the compound inhibits synaptic transmission and CA1 neuron excitability. Overall, these data further our understanding on the pharmacodynamics of DEX and disclose an additional mechanism that could underlie its neuroprotective properties. Also, they identify DEX as a lead to develop new modulators of K+ conductances.


Subject(s)
Hippocampus/cytology , Potassium Channels/metabolism , Pramipexole/pharmacology , Action Potentials/drug effects , Animals , Dopamine Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats, Wistar , Receptors, Dopamine/metabolism , Synaptic Transmission/drug effects
9.
Neurotox Res ; 39(3): 658-666, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33475965

ABSTRACT

NOD mice represent a unique strain that recapitulates some aspects of progressive MS when subjected to experimental autoimmune encephalomyelitis (EAE). It is unknown, however, whether a proneness to demyelination and/or defect in remyelination contribute to disease progression in NOD mice. Answering to this question might help deciphering the molecular and cellular events underpinning disease evolution in progressive MS. Here, we compared the cuprizone-dependent demyelination and remyelination responses, as well as their functional correlates, in NOD, C57BL/6, and SJL mice typically adopted to model progressive, chronic or relapsing EAE. We report that demyelination occurred to a similar extent in the three mice strains, and that in none of them there was evidence of axonal degeneration during prolonged demyelination. Moreover, immunostaining for GFAP+ astrocytes, Iba1+ microglia, and NG2+ oligodendrocyte precursor cells similarly increased in the 3 mouse strains after cuprizone exposure. The mice underwent concomitant and complete remyelination 2 weeks after cuprizone withdrawal. On a functional level, NOD mice showed the earliest reduction of spontaneous motility and full recovery, but no impairment of motor skill. Conversely, C57BL/6 animals showed phasic reduction of both spontaneous motility and motor skill. Lastly, SJL mice presented the most severe neurological impairment with long-lasting reduction of spontaneous motility and motor skill. Overall, data suggest that the unique feature of EAE progression in NOD mice is not due to proneness to demyelination or intrinsic defects in myelin formation. Findings also unravel important functional differences in the response of the three mouse stains to cuprizone that can be harnessed to design and interpret future experiments.


Subject(s)
Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Myelin Sheath/drug effects , Remyelination/drug effects , Animals , Chelating Agents/toxicity , Chronic Disease , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Myelin Sheath/genetics , Myelin Sheath/pathology , Remyelination/physiology
10.
Br J Pharmacol ; 177(14): 3342-3356, 2020 07.
Article in English | MEDLINE | ID: mdl-32199028

ABSTRACT

BACKGROUND AND PURPOSE: Drugs able to counteract progressive multiple sclerosis (MS) represent a largely unmet therapeutic need. Even though the pathogenesis of disease evolution is still obscure, accumulating evidence indicates that mitochondrial dysfunction plays a causative role in neurodegeneration and axonopathy in progressive MS patients. Here, we investigated the effects of dexpramipexole, a compound with a good safety profile in humans and able to sustain mitochondria functioning and energy production, in a mouse model of progressive MS. EXPERIMENTAL APPROACH: Female non-obese diabetic mice were immunized with MOG35-55 . Functional, immune and neuropathological parameters were analysed during disease evolution in animals treated or not with dexpramipexole. The compound's effects on bioenergetics and neuroprotection were also evaluated in vitro. KEY RESULTS: We found that oral treatment with dexpramipexole at a dose consistent with that well tolerated in humans delayed disability progression, extended survival, counteracted reduction of spinal cord mitochondrial DNA content and reduced spinal cord axonal loss of mice. Accordingly, the drug sustained in vitro bioenergetics of mouse optic nerve and dorsal root ganglia and counteracted neurodegeneration of organotypic mouse cortical cultures exposed to the adenosine triphosphate-depleting agents oligomycin or veratridine. Dexpramipexole, however, was unable to affect the adaptive and innate immune responses both in vivo and in vitro. CONCLUSION AND IMPLICATION: The present findings corroborate the hypothesis that neuroprotective agents may be of relevance to counteract MS progression and disclose the translational potential of dexpramipexole to treatment of progressive MS patients as a stand-alone or adjunctive therapy.


Subject(s)
Diabetes Mellitus, Experimental , Multiple Sclerosis , Animals , Disease Progression , Female , Humans , Mice , Multiple Sclerosis/drug therapy , Neuroprotection , Pramipexole
11.
Ocul Immunol Inflamm ; 28(5): 839-845, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-31490701

ABSTRACT

PURPOSE: To compare the immunosuppressive and cytotoxic effects of three anti-inflammatory eye drops formulations containing betamethasone plus chloramphenicol (B+C), dexamethasone plus netilmicin (D+N) or dexamethasone plus tobramycin (D+T).Methods: The eye drops formulations have been tested at different dilutions on cytokine synthesis by mouse or human cultured macrophages, as well as proliferation and viability of cultured human corneal cells (HCE).Results: B+C reduced IL6 and TNFα production by cultured mouse or human macrophages more potently than D+N and D+T, with the tree formulations having the same impact on IL-10 expression. We also found that the eye drops preparations reduced proliferation of HCE cells, with D+T showing the higher anti-proliferative potency and B+C showing the lower cytotoxic potential.Conclusion: Our study points out that it may be erroneous to consider routinely-used anti-inflammatory eye drops preparations with analogous formulations as readily interchangeable and of similar potency and tolerability.


Subject(s)
Anti-Bacterial Agents/toxicity , Epithelium, Corneal/drug effects , Glucocorticoids/toxicity , Macrophages/drug effects , Administration, Ophthalmic , Animals , Betamethasone/toxicity , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chloramphenicol/toxicity , Cytokines/metabolism , Dexamethasone/toxicity , Drug Combinations , Epithelium, Corneal/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Mice , Netilmicin/toxicity , Ophthalmic Solutions , Tobramycin/toxicity
12.
Pain ; 161(4): 831-841, 2020 04.
Article in English | MEDLINE | ID: mdl-31815915

ABSTRACT

Selective targeting of sodium channel subtypes Nav1.7, Nav1.8, and Nav1.9, preferentially expressed by peripheral nociceptors, represents a unique opportunity to develop analgesics devoid of central side effects. Several compounds that target Nav1.7 and Nav1.8 with different degrees of selectivity have been developed and are currently being tested in clinical trials for multiple pain indications. Among these chemicals, benzothiazole-like compounds emerged as potent sodium channel blockers. We evaluated the effects of dexpramipexole, a benzothiazole-bearing drug with pleiotypic neuroactive properties and a good safety profile in humans, on sodium conductances of dorsal root ganglia neurons, as well as in multiple nociceptive and neuropathic pain models. Dexpramipexole blocks TTX-resistant sodium conductances in cultured rat dorsal root ganglion neurons with an IC50 of 294.4 nM, suggesting selectivity towards Nav1.8. In keeping with this, dexpramipexole does not affect sodium currents in dorsal root ganglion neurons from Nav1.8 null mice and acquires binding pose predicted to overlap that of the Nav1.8 channel-selective blocker A-8034637. The drug provides analgesia when parenterally, orally, or topically applied in inflammatory and visceral mouse pain models, as well as in mice affected by neuropathic pain induced by oxaliplatin, nerve constriction, or diabetes. Pain reduction in mice occurs at doses consistent with those adopted in clinical trials. The present findings confirm the relevance of selective targeting of peripheral Nav1.8 channels to pain therapy. In light of the excellent tolerability of dexpramipexole in humans, our results support its translational potential for treatment of pain.


Subject(s)
Neuralgia , Analgesia , Animals , Ganglia, Spinal , Mice , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/genetics , Neuralgia/drug therapy , Nociception , Pramipexole/therapeutic use , Rats , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium Channels
13.
Cephalalgia ; 40(4): 330-336, 2020 04.
Article in English | MEDLINE | ID: mdl-31852231

ABSTRACT

BACKGROUND: In spite of the substantial therapeutic efficacy of triptans, their site of action is still debated. Subcutaneous sumatriptan is the most efficacious symptomatic treatment for cluster headache (CH) patients, showing therapeutic onset within a few minutes after injection even in migraine patients. However, whether subcutaneous sumatriptan is able to reach the CNS within this short time frame is currently unknown. METHODS: Here, by means of liquid chromatography/mass spectrometry, we investigated peripheral and brain distribution of subcutaneous sumatriptan soon after injection in rats at a dose equivalent to that used in patients. Tissue sumatriptan contents were compared to those of oxazepam, a prototypical lipophilic, neuroactive drug. RESULTS: We report that sumatriptan accumulated within brain regions of relevance to migraine and CH pathogenesis such as the hypothalamus and the brainstem as soon as 1 and 5 minutes after injection. Notably, sumatriptan brain distribution was faster than that of oxazepam, reaching concentrations exceeding its reported binding affinity for 5HT1B/D receptors, and in the range of those able to inhibit neurotransmitter release in vivo. CONCLUSION: Our findings indicate that sumatriptan distributes within the CNS soon after injection, and are in line with prompt pain relief by parenteral sumatriptan in CH patients.


Subject(s)
Brain/metabolism , Cluster Headache/metabolism , Serotonin 5-HT1 Receptor Agonists/metabolism , Sumatriptan/metabolism , Animals , Brain/drug effects , Chromatography, Liquid/methods , Cluster Headache/drug therapy , Injections, Subcutaneous , Male , Mass Spectrometry/methods , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Sumatriptan/administration & dosage , Time Factors
14.
Exp Neurol ; 322: 113065, 2019 12.
Article in English | MEDLINE | ID: mdl-31536728

ABSTRACT

Progressive multiple sclerosis (PMS) is a devastating disorder sustained by neuroimmune interactions still wait to be identified. Recently, immune-independent, neural bioenergetic derangements have been hypothesized as causative of neurodegeneration in PMS patients. To gather information on the immune and neurodegenerative components during PMS, in the present study we investigated the molecular and cellular events occurring in a Non-obese diabetic (NOD) mouse model of experimental autoimmune encephalomyelitis (EAE). In these mice, we also evaluated the effects of clinically-relevant immunosuppressive (dexamethasone) or bioenergetic drugs (bezafibrate and biotin) on functional, immune and neuropathological parameters. We found that immunized NOD mice progressively accumulated disability and severe neurodegeneration in the spinal cord. Unexpectedly, although CD4 and CD8 lymphocytes but not B or NK cells infiltrate the spinal cord linearly with time, their suppression by different dexamethasone treatment schedules did not affect disease progression. Also, the spreading of the autoimmune response towards additional immunogenic myelin antigen occurred neither in the periphery nor in the CNS of EAE mice. Conversely, we found that altered mitochondrial morphology, reduced contents of mtDNA and decreased transcript levels for respiratory complex subunits occurred at early disease stages and preceded axonal degeneration within spinal cord columns. However, the mitochondria boosting drugs, bezafibrate and biotin, were unable to reduce disability progression. Data suggest that EAE NOD mice recapitulate some features of PMS. Also, by showing that bezafibrate or biotin do not affect progression in NOD mice, our study suggests that this model can be harnessed to anticipate experimental information of relevance to innovative treatments of PMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunosuppressive Agents/pharmacology , Neuroprotective Agents/pharmacology , Animals , Bezafibrate/pharmacology , Biotin/pharmacology , Dexamethasone/pharmacology , Mice , Mice, Inbred NOD , Mitochondria/drug effects , Multiple Sclerosis, Chronic Progressive , Spinal Cord/drug effects , Spinal Cord/pathology
15.
J Clin Med ; 8(7)2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31323969

ABSTRACT

Multiple myeloma (MM) is a genetically heterogeneous disease that includes a subgroup of 10-15% of patients facing dismal survival despite the most intensive treatment. Despite improvements in biological knowledge, MM is still an incurable neoplasia, and therapeutic options able to overcome the relapsing/refractory behavior represent an unmet clinical need. The aim of this review is to provide an integrated clinical and biological overview of high-risk MM, discussing novel therapeutic perspectives, targeting the neoplastic clone and its microenvironment. The dissection of the molecular determinants of the aggressive phenotypes and drug-resistance can foster a better tailored clinical management of the high-risk profile and therapy-refractoriness. Among the current clinical difficulties in MM, patients' management by manipulating the tumor niche represents a major challenge. The angiogenesis and the stromal infiltrate constitute pivotal mechanisms of a mutual collaboration between MM and the non-tumoral counterpart. Immuno-modulatory and anti-angiogenic therapy hold great efficacy, but variable and unpredictable responses in high-risk MM. The comprehensive understanding of the genetic heterogeneity and MM high-risk ecosystem enforce a systematic bench-to-bedside approach. Here, we provide a broad outlook of novel druggable targets. We also summarize the existing multi-omics-based risk profiling tools, in order to better select candidates for dual immune/vasculogenesis targeting.

16.
Neuropharmacology ; 143: 306-316, 2018 12.
Article in English | MEDLINE | ID: mdl-30291939

ABSTRACT

Even though pharmacological approaches able to counteract age-dependent cognitive impairment have been highly investigated, drugs improving cognition and memory are still an unmet need. It has been hypothesized that sustaining energy dynamics within the aged hippocampus can boost memory storage by sustaining synaptic functioning and long term potentiation (LTP). Dexpramipexole (DEX) is the first-in-class compound able to sustain neuronal bioenergetics by interacting with mitochondrial F1Fo-ATP synthase. In the present study, for the first time we evaluated the effects of DEX on synaptic fatigue, LTP induction, learning and memory retention. We report that DEX improved LTP maintenance in CA1 neurons of acute hippocampal slices from aged but not young rats. However, we found no evidence that DEX counteracted two classic parameters of synaptic fatigue such as fEPSP reduction or the train area during the high frequency stimulation adopted to induce LTP. Interestingly, patch-clamp recordings in rat hippocampal neurons revealed that DEX dose-dependently inhibited (IC50 814 nM) the IA current, a rapidly-inactivating K+ current that negatively regulates neuronal excitability as well as cognition and memory processes. In keeping with this, DEX counteracted both scopolamine-induced spatial memory loss in rats challenged in Morris Water Maze test and memory retention in rats undergoing Novel Object Recognition. Overall, the present study discloses the ability of DEX to boost hippocampal synaptic plasticity, learning and memory. In light of the good safety profile of DEX in humans, our findings may have a realistic translational potential to treatment of cognitive disorders.


Subject(s)
Aging/drug effects , Hippocampus/drug effects , Memory/drug effects , Neuronal Plasticity/drug effects , Nootropic Agents/pharmacology , Pramipexole/pharmacology , Aging/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Memory Disorders/drug therapy , Memory Disorders/metabolism , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Random Allocation , Rats, Wistar , Scopolamine , Tissue Culture Techniques
17.
Pain ; 159(10): 1980-1988, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29794878

ABSTRACT

We attempted to gather information on the pathogenesis of medication-overuse headache, as well as on the neurochemical mechanisms through which symptomatic medication overuse concurs to headache chronification. Transcriptional profiles were therefore evaluated as an index of the homeostasis of the trigeminovascular system in the trigeminal ganglion of female rats exposed for 1 month to daily oral doses of eletriptan or indomethacin. We report that both drug treatments change trigeminal ganglion gene expression to a similar extend. Of note, qualitative transcriptomic analysis shows that eletriptan and indomethacin prompt nearly identical, increased expression of genes coding for proteins involved in migraine pathogenesis and central pain sensitization such as neuropeptides, their cognate receptors, prostanoid, and nitric oxide-synthesizing enzymes, as well as TRP channels. These genes, however, were not affected in thoracic dorsal root ganglia. Of note, lowering of orofacial nociceptive thresholds, as well as forepaw hyperalgesia occurred in both indomethacin- and eletriptan-treated rats. Our study reveals that chronic rat exposure to 2 acute headache medications with completely different mechanisms of action prompts pain sensitization with highly similar induction of pronociceptive genes selectively within the trigeminal ganglion. Data further our understanding of medication-overuse headache pathogenesis and provide hints for specific mechanism-based treatment options.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Gene Expression Profiling , Headache Disorders, Secondary/pathology , Headache Disorders, Secondary/physiopathology , Pain Threshold/physiology , Trigeminal Ganglion/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Disease Models, Animal , Female , Gene Expression/drug effects , Headache Disorders, Secondary/chemically induced , Hyperalgesia/radiotherapy , Indomethacin/toxicity , Oligonucleotide Array Sequence Analysis , Pain Threshold/drug effects , Pyrrolidines/toxicity , Rats , Rats, Wistar , Serotonin Receptor Agonists/toxicity , Time Factors , Tryptamines/toxicity
18.
Neuroscience ; 379: 228-238, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29588251

ABSTRACT

Emerging evidence indicates that transcriptome alterations due to epigenetic deregulation concur to ALS pathogenesis. Accordingly, pan-histone deacetylase (HDAC) inhibitors delay ALS development in mice, but these compounds failed when tested in ALS patients. Possibly, lack of selectivity toward specific classes of HDACs weakens the therapeutic effects of pan-HDAC inhibitors. Here, we tested the effects of the HDAC Class II selective inhibitor MC1568 on disease evolution, motor neuron survival as well as skeletal muscle function in SOD1G93A mice. We report that HDACs did not undergo expression changes during disease evolution in isolated motor neurons of adult mice. Conversely, increase in specific Class II HDACs (-4, -5 and -6) occurs in skeletal muscle of mice with severe neuromuscular impairment. Importantly, treatment with MC1568 causes early improvement of motor performances that vanishes at later stages of disease. Notably, motor improvement is not paralleled by reduced motor neuron degeneration but by increased skeletal muscle electrical potentials, reduced activation of mir206/FGFBP1-dependent muscle reinnervation signaling, and increased muscle expression of myogenic genes.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Motor Neurons/drug effects , Muscle, Skeletal/drug effects , Neuroprotective Agents/pharmacology , Pyrroles/pharmacology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Genetically Modified , Cell Survival/drug effects , Cells, Cultured , Female , Histone Deacetylases/metabolism , Male , Motor Activity/drug effects , Motor Activity/physiology , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Random Allocation , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Superoxide Dismutase/metabolism
19.
Cell Chem Biol ; 25(4): 471-482.e7, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29478906

ABSTRACT

Interest in the modulation of nicotinamide adenine dinucleotide (NAD) metabolome is gaining great momentum because of its therapeutic potential in different human disorders. Suppression of nicotinamide salvage by nicotinamide phosphoribosyl transferase (NAMPT) inhibitors, however, gave inconclusive results in neoplastic patients because several metabolic routes circumvent the enzymatic block converging directly on nicotinamide mononucleotide adenylyl transferases (NMNATs) for NAD synthesis. Unfortunately, NMNAT inhibitors have not been identified. Here, we report the identification of Vacor as a substrate metabolized by the consecutive action of NAMPT and NMNAT2 into the NAD analog Vacor adenine dinucleotide (VAD). This leads to inhibition of both enzymes, as well as NAD-dependent dehydrogenases, thereby causing unprecedented rapid NAD depletion, glycolytic block, energy failure, and necrotic death of NMNAT2-proficient cancer cells. Conversely, lack of NMNAT2 expression confers complete resistance to Vacor. Remarkably, Vacor prompts VAD formation and growth suppression in NMNAT2-positive neuroblastoma and melanoma xenografts. Our data show the first evidence of harnessing the entire nicotinamide salvage pathway for antimetabolic strategies.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Phenylurea Compounds/pharmacology , Animals , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Glycolysis/drug effects , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mice, Nude , Models, Molecular , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Niacinamide/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Phenylurea Compounds/metabolism , Phenylurea Compounds/therapeutic use
20.
Molecules ; 21(3): 345, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26985887

ABSTRACT

Biocatalytic membrane reactors (BMR) combining reaction and separation within the same unit have many advantages over conventional reactor designs. Ceramic membranes are an attractive alternative to polymeric membranes in membrane biotechnology due to their high chemical, thermal and mechanical resistance. Another important use is their potential application in a biphasic membrane system, where support solvent resistance is highly needed. In this work, the preparation of asymmetric ceramic hollow fibre membranes and their use in a two-separate-phase biocatalytic membrane reactor will be described. The asymmetric ceramic hollow fibre membranes were prepared using a combined phase inversion and sintering technique. The prepared fibres were then used as support for lipase covalent immobilization in order to develop a two-separate-phase biocatalytic membrane reactor. A functionalization method was proposed in order to increase the density of the reactive hydroxyl groups on the surface of ceramic membranes, which were then amino-activated and treated with a crosslinker. The performance and the stability of the immobilized lipase were investigated as a function of the amount of the immobilized biocatalytst. Results showed that it is possible to immobilize lipase on a ceramic membrane without altering its catalytic performance (initial residual specific activity 93%), which remains constant after 6 reaction cycles.


Subject(s)
Biocatalysis , Bioreactors , Ceramics , Membranes, Artificial , Aluminum Oxide/chemistry , Biotechnology , Blood Proteins , Enzymes, Immobilized , Hydroxylation , Lipase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...