Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Dev Cell ; 56(11): 1617-1630.e6, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34033756

ABSTRACT

Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.


Subject(s)
Histone Deacetylase 1/genetics , Neurogenesis/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Regeneration/genetics , Spinal Cord/growth & development , Zebrafish Proteins/genetics , Animals , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Macrophages/cytology , Macrophages/metabolism , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Spinal Cord/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factor AP-1/genetics , Zebrafish/genetics
2.
J Neurosci ; 39(24): 4694-4713, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30948475

ABSTRACT

Adult zebrafish, in contrast to mammals, regenerate neurons in their brain, but the extent and variability of this capacity is unclear. Here we ask whether the loss of various dopaminergic neuron populations is sufficient to trigger their functional regeneration. Both sexes of zebrafish were analyzed. Genetic lineage tracing shows that specific diencephalic ependymo-radial glial (ERG) progenitor cells give rise to new dopaminergic [tyrosine hydroxylase-positive (TH+)] neurons. Ablation elicits an immune response, increased proliferation of ERG progenitor cells, and increased addition of new TH+ neurons in populations that constitutively add new neurons (e.g., diencephalic population 5/6). Inhibiting the immune response attenuates neurogenesis to control levels. Boosting the immune response enhances ERG proliferation, but not addition of TH+ neurons. In contrast, in populations in which constitutive neurogenesis is undetectable (e.g., the posterior tuberculum and locus ceruleus), cell replacement and tissue integration are incomplete and transient. This is associated with a loss of spinal TH+ axons, as well as permanent deficits in shoaling and reproductive behavior. Hence, dopaminergic neuron populations in the adult zebrafish brain show vast differences in regenerative capacity that correlate with constitutive addition of neurons and depend on immune system activation.SIGNIFICANCE STATEMENT Despite the fact that zebrafish show a high propensity to regenerate neurons in the brain, this study reveals that not all types of dopaminergic neurons are functionally regenerated after specific ablation. Hence, in the same adult vertebrate brain, mechanisms of successful and incomplete regeneration can be studied. We identify progenitor cells for dopaminergic neurons and show that activating the immune system promotes the proliferation of these cells. However, in some areas of the brain this only leads to insufficient replacement of functionally important dopaminergic neurons that later disappear. Understanding the mechanisms of regeneration in zebrafish may inform interventions targeting the regeneration of functionally important neurons, such as dopaminergic neurons, from endogenous progenitor cells in nonregenerating mammals.


Subject(s)
Dopaminergic Neurons/physiology , Immune System Phenomena/physiology , Nerve Regeneration/physiology , Zebrafish/physiology , Aging , Animals , Axons/physiology , Cell Lineage/genetics , Cell Proliferation , Diencephalon/cytology , Diencephalon/physiology , Female , Male , Microglia/physiology , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Sexual Behavior, Animal/physiology
3.
Nat Commun ; 9(1): 4670, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30405119

ABSTRACT

Spinal cord injury leads to a massive response of innate immune cells in non-regenerating mammals, but also in successfully regenerating zebrafish. However, the role of the immune response in successful regeneration is poorly defined. Here we show that inhibiting inflammation reduces and promoting it accelerates axonal regeneration in spinal-lesioned zebrafish larvae. Mutant analyses show that peripheral macrophages, but not neutrophils or microglia, are necessary for repair. Macrophage-less irf8 mutants show prolonged inflammation with elevated levels of Tnf-α and Il-1ß. Inhibiting Tnf-α does not rescue axonal growth in irf8 mutants, but impairs it in wildtype animals, indicating a pro-regenerative role of Tnf-α. In contrast, decreasing Il-1ß levels or number of Il-1ß+ neutrophils rescue functional regeneration in irf8 mutants. However, during early regeneration, interference with Il-1ß function impairs regeneration in irf8 and wildtype animals. Hence, inflammation is dynamically controlled by macrophages to promote functional spinal cord regeneration in zebrafish.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Nerve Regeneration , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/metabolism , Zebrafish/metabolism , Animals , Axons/metabolism , Collagen Type XII/metabolism , Microglia/metabolism , Microglia/pathology , Mutation/genetics , Neutrophils/metabolism , Spinal Cord/pathology , Zebrafish/immunology
4.
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
5.
Neurosci Lett ; 656: 120-125, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28732762

ABSTRACT

Transcriptional deregulation emerges as a key pathogenetic mechanism in ALS pathogenesis, and non-class-specific histone deacetylase (HDACs) inhibitors proved of therapeutic efficacy in preclinical models of ALS. When tested in patients, however, these drugs failed, probably because of a lack of selectivity toward pathogenetic HDACs. Here, we studied the effects of MC1568, an inhibitor of Class-II HDACs which have been reported to contribute to ALS pathogenesis. We focused on transcriptional regulation of glutamate transporter EAAT2, whose reduced expression may contribute to motor neuron degeneration in ALS. We report that MC1568 highly increased EAAT2 transcripts in primary cultures of mouse glia, but these increases did not correlate with increased glutamate uptake capacity. Accordingly, we found that MC1568 augmented protein expression of EAAT2 together with its sumoylation, a post-translational modification typically altering protein function and localization. When tested in SOD1G93A mice, however, MC1568 fully restored the reduced spinal cord expression of EAAT2 and glutamate uptake up to control levels. A prolonged treatment with MC1568 (from onset to end stage) was unable to prolong survival of mice. Data reveal a key role of Class-II HDACs in expression and function of glutamate transporter, further corroborating preclinical and clinical evidence that the sole restoration of glutamate uptake is not of therapeutic relevance to ALS therapy.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Pyrroles/pharmacology , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/mortality , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Excitatory Amino Acid Transporter 2/metabolism , Female , Glutamic Acid/metabolism , Male , Mice, Transgenic , Spinal Cord/drug effects , Spinal Cord/metabolism
6.
Neuropharmacology ; 117: 74-84, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28161373

ABSTRACT

Mitochondrial encephalopathies are fatal, infantile neurodegenerative disorders caused by a deficit of mitochondrial functioning, for which there is urgent need to identify efficacious pharmacological treatments. Recent evidence shows that rapamycin administered both intraperitoneally or in the diet delays disease onset and enhances survival in the Ndufs4 null mouse model of mitochondrial encephalopathy. To delineate the clinical translatability of rapamycin in treatment of mitochondrial encephalopathy, we evaluated the drug's effects on disease evolution and mitochondrial parameters adopting treatment paradigms with fixed daily, oral doses starting at symptom onset in Ndufs4 knockout mice. Molecular mechanisms responsible for the pharmacodynamic effects of rapamycin were also evaluated. We found that rapamycin did not affect disease development at clinically-relevant doses (0.5 mg kg-1). Conversely, an oral dose previously adopted for intraperitoneal administration (8 mg kg-1) delayed development of neurological symptoms and increased median survival by 25%. Neurological improvement and lifespan were not further increased when the dose raised to 20 mg kg-1. Notably, rapamycin at 8 mg kg-1 did not affect the reduced expression of respiratory complex subunits, as well as mitochondrial number and mtDNA content. This treatment regimen however significantly ameliorated architecture of mitochondria cristae in motor cortex and cerebellum. However, reduction of mTOR activity by rapamycin was not consistently found within the brain of knockout mice. Overall, data show the ability of rapamycin to improve ultrastructure of dysfunctional mitochondria and corroborate its therapeutic potential in mitochondrial disorders. The non-clinical standard doses required, however, raise concerns about its rapid and safe clinical transferability.


Subject(s)
Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Sirolimus/therapeutic use , Administration, Oral , Animals , Cerebellum/metabolism , Cerebellum/pathology , DNA, Mitochondrial/metabolism , Disease Progression , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex I/genetics , Female , Male , Mice , Mice, Knockout , Mitochondria/ultrastructure , Motor Cortex/metabolism , Motor Cortex/pathology , Muscle, Skeletal/metabolism , Sirolimus/administration & dosage , Sirolimus/blood , Sirolimus/pharmacokinetics , Survival Analysis , TOR Serine-Threonine Kinases/metabolism
7.
J Alzheimers Dis ; 54(2): 737-50, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27567859

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP1) activation contributes to the cascade of events initiated by amyloid-ß (Aß) peptide eventually leading to cell death in Alzheimer's disease brain. A significant accumulation of PAR polymers and increase of PARP1 expression were detected in the cortex at the early (3.5 months) and intermediate (6 months) stage of Aß deposition in the TgCRND8 mouse model. Our previous data highlighted the beneficial effects of oleuropein aglycone (OLE), the main polyphenol found in the olive oil, against neurodegeneration both in cultured cells and in model organisms. Here we found that 8-week OLE treatment (50 mg/kg of diet) to 6-month-old TgCRND8 mice rescued to control values PARP1 activation and the levels of its product, PAR. In N2a neuroblastoma cells, PARP1 activation and PAR formation upon exposure to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) were abolished by pretreatment for 24 h with either OLE (100µM) or PARP inhibitors. A significant reduction of the NAD+ content, compared to controls, was found in N2a cells exposed to MNNG (100µM) for 90 min; the latter was slightly attenuated by cell treatment for 24 h with PJ-34 or with OLE. In vitro and in vivo, the OLE-induced reduction of PARP1 activation was paralleled by the overexpression of Sirtuin1 (SIRT1), and, in vivo, by a decrease of NF-κB and the pro-apoptotic marker p53. In N2a cells, we also found that OLE potentiates the MNNG-induced increase of Beclin1 levels. In conclusion, our data show that OLE treatment counteracts neuronal damage through modulation of the PARP1-SIRT1 interplay.


Subject(s)
Iridoids/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Polyphenols/pharmacology , Sirtuin 1/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Iridoid Glucosides , Mice , Mice, Transgenic , Vasodilator Agents/pharmacology
8.
Brain Behav Immun ; 50: 78-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26130058

ABSTRACT

Fingolimod affords protection from MS by sequestering lymphocytes in secondary lymphoid organs via down regulation of their sphingosine 1 phosphate receptor (S1P1). Unexpectedly, accumulating evidence indicates that patients who discontinue fingolimod treatment may be at risk of rehearsal of magnetic resonance (MR) and clinical disease activity, sometimes featuring dramatic rebound. We therefore developed in vivo and in vitro models of post-fingolimod MS rebound to unravel its cellular and molecular mechanisms. The impact of fingolimod withdrawal on T regulatory lymphocytes was also investigated by means of cytofluorimetric analysis and antigen-specific lymphocyte proliferation assays. We show that mice with relapsing-remitting experimental autoimmune encephalomyelitis (EAE) undergo extremely severe, chronic disease rebound upon discontinuation of fingolimod. Remarkably, rebound is preceded by a burst of S1P1 overexpression in lymph node-entrapped lymphocytes that correlates with subsequent massive lymphocyte egress and widespread CNS immune infiltration. Also, consistent with the ability of S1P1 to counteract polarization and function of T regulatory lymphocytes their number and suppression of effector T cells is reduced by fingolimod suspension. Data disclose the first pathogenic mechanisms of post-fingolimod rebound that may be targeted for therapeutic intervention.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fingolimod Hydrochloride/administration & dosage , Fingolimod Hydrochloride/adverse effects , Immunosuppression Therapy , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Mice , Mice, Inbred C57BL , Receptors, Lysosphingolipid/agonists , Signal Transduction/immunology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
Clin Exp Otorhinolaryngol ; 8(2): 123-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26045910

ABSTRACT

OBJECTIVES: The nuclear protein high mobility group protein box 1 (HMGB1) is a proinflammatory mediator that belongs to the alarmin family of proinflammatory mediators, and it has recently emerged as a key player in different acute and chronic immune disorders. Several lines of evidence demonstrate that HMGB1 is actively released extracellularly from immune cells or passively released from necrotic cells. Because of the ability of HMGB1 to sustain chronic inflammation, we investigated whether the protein is present in nasal fluids of patients with different forms of rhinitis. METHODS: HMGB1 levels were evaluated in nasal fluids of healthy subjects or rhinitis patients who were treated or not treated with different treatments. RESULTS: We report that the level of HMGB1 was significantly increased in nasal fluids of patients with allergic rhinitis, patients with NARES (nonallergic rhinitis with eosinophiliac syndrome), as well as patients with polyps. We also found that a formulation containing the HMGB1-binding compound glycyrrhizin (GLT) reduced the HMGB1 content in nasal fluids of rhinitis patients to an extent similar to that with nasal budesonide treatment. We also found that among the cultured human leukocyte populations, eosinophils released higher amounts of HMGB1. Based on the ability of HMGB1 to sustain eosinophil survival and the ability of GLT to inactivate HMGB1, we report that GLT selectively killed cultured eosinophils and had no effect on neutrophils, macrophages, and lymphocytes. CONCLUSION: Collectively, these data underscore the role of HMGB1 in rhinitis pathogenesis and the therapeutic potential of GLT formulations in treatment of chronic inflammatory disorders of the nasal mucosa.

10.
Mol Pharmacol ; 87(6): 965-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25788480

ABSTRACT

Mitochondrial disorders are devastating genetic diseases for which efficacious therapies are still an unmet need. Recent studies report that increased availability of intracellular NAD obtained by inhibition of the NAD-consuming enzyme poly(ADP-ribose) polymerase (PARP)-1 or supplementation with the NAD-precursor nicotinamide riboside (NR) ameliorates energetic derangement and symptoms in mouse models of mitochondrial disorders. Whether these pharmacological approaches also improve bioenergetics of human cells harboring mitochondrial defects is unknown. It is also unclear whether the same signaling cascade is prompted by PARP-1 inhibitors and NR supplementation to improve mitochondrial homeostasis. Here, we show that human fibroblasts mutant for the NADH dehydrogenase (ubiquinone) Fe-S protein 1 (NDUFS1) subunit of respiratory complex I have similar ATP, NAD, and mitochondrial content compared with control cells, but show reduced mitochondrial membrane potential. Interestingly, mutant cells also show increased transcript levels of mitochondrial DNA but not nuclear DNA respiratory complex subunits, suggesting activation of a compensatory response. At variance with prior work in mice, however, NR supplementation, but not PARP-1 inhibition, increased intracellular NAD content in NDUFS1 mutant human fibroblasts. Conversely, PARP-1 inhibitors, but not NR supplementation, increased transcription of mitochondrial transcription factor A and mitochondrial DNA-encoded respiratory complexes constitutively induced in mutant cells. Still, both NR and PARP-1 inhibitors restored mitochondrial membrane potential and increased organelle content as well as oxidative activity of NDUFS1-deficient fibroblasts. Overall, data provide the first evidence that in human cells harboring a mitochondrial respiratory defect exposure to NR or PARP-1, inhibitors activate different signaling pathways that are not invariantly prompted by NAD increases, but equally able to improve energetic derangement.


Subject(s)
Fibroblasts/drug effects , Mitochondria/drug effects , NADH Dehydrogenase/genetics , NAD/metabolism , Niacinamide/analogs & derivatives , Energy Metabolism , Fibroblasts/metabolism , Homeostasis , Humans , Infant , Leukoencephalopathies/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mutation , Niacinamide/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Pyridinium Compounds , Signal Transduction
11.
Invest Ophthalmol Vis Sci ; 55(11): 7266-71, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25301877

ABSTRACT

PURPOSE: We evaluated the potential protective effects of Coenzyme Q10 (CoQ10) on human corneal cells and rabbit eyes after ultraviolet B (UVB) exposure and a model of wound healing in rabbit eyes after corneal epithelium removal. METHODS: Human corneal epithelium cells (HCE) were exposed to a source of UVB radiation (312 nM) in the presence of different CoQ10 concentrations or vehicle. The mitochondrial function and cell survival were evaluated by means of 3-(4,5-dimethylthiazole-2-yl)2,5-diphenyl-tetrazolium (MTT) reduction and lactic dehydrogenase (LDH) release. Furthermore, quantitation of oxygen consumption and mitochondrial membrane potential were conducted. In vivo rabbit models were adopted to evaluate the effect of CoQ10 on UVB-induced conjunctival vessel hyperemia and corneal recovery after ethanol induced corneal lesion. RESULTS: In UVB-exposed HCE cells, CoQ10 addition led to an increased survival rate and mitochondrial function. Furthermore, oxygen consumption was maintained at control levels and adenosine triphosphate (ATP) decline was completely prevented in the CoQ10-treated cells. Interestingly, in an in vivo model, CoQ10 was able dose-dependently to reduce UVB-induced vessel hyperemia. Finally, in a model of corneal epithelium removal, 12 hours from surgery, animals treated with CoQ10 showed a reduction of damaged area in respect to vehicle controls, which lasted until 48 hours. CONCLUSIONS: We demonstrated that CoQ10 reduces corneal damages after UVB exposure in vivo and in vitro by preserving mitochondrial function. Also, for the first time to our knowledge we showed that the administration of CoQ10 after corneal epithelium removal promotes corneal wound healing.


Subject(s)
Corneal Diseases/drug therapy , Corneal Injuries/drug therapy , Epithelium, Corneal/pathology , Eye Burns/drug therapy , Mitochondria/physiology , Ubiquinone/analogs & derivatives , Wound Healing/drug effects , Cell Proliferation , Cell Survival , Cells, Cultured , Corneal Diseases/metabolism , Corneal Diseases/pathology , Corneal Injuries/metabolism , Corneal Injuries/pathology , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Eye Burns/metabolism , Eye Burns/pathology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Ophthalmic Solutions , Ubiquinone/administration & dosage , Ultraviolet Rays/adverse effects , Vitamins/administration & dosage
12.
Neurotherapeutics ; 11(3): 651-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24935635

ABSTRACT

Mitochondrial disorders are deadly childhood diseases for which therapeutic remedies are an unmet need. Given that genetic suppression of the nuclear enzyme poly (adenine diphosphate-ribose) polymerase(PARP)-1 improves mitochondrial functioning, we investigated whether pharmacological inhibition of the enzyme affords protection in a mouse model of a mitochondrial disorder. We used mice lacking the Ndufs4 subunit of the respiratory complex I (Ndufs4 knockout [ KO] mice); these mice undergo progressive encephalopathy and die around postnatal day 50. Mice were treated daily with the potent PARP inhibitor N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-(N,N-dimethylamino)acetamide hydrochloride (PJ34); neurological parameters, PARP activity, and mitochondrial homeostasis were evaluated. We found that mice receiving N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-(N,N-dimethylamino)acetamide hydrochloride from postnatal day 30 to postnatal day 50 show reduced neurological impairment, and increased exploratory activity and motor skills compared with vehicle-treated animals. However, drug treatment did not delay or reduce death. We found no evidence of increased PARP activity within the brain of KO mice compared with heterozygous, healthy controls. Conversely, a 10-day treatment with the PARP inhibitor significantly reduced basal poly(ADP-ribosyl)ation in different organs of the KO mice, including brain, skeletal muscle, liver, pancreas, and spleen. In keeping with the epigenetic role of PARP-1, its inhibition correlated with increased expression of mitochondrial respiratory complex subunits and organelle number. Remarkably, pharmacological targeting of PARP reduced astrogliosis in olfactory bulb and motor cortex, but did not affect neuronal loss of KO mice. In light of the advanced clinical development of PARP inhibitors, these data emphasize their relevance to treatment of mitochondrial respiratory defects.


Subject(s)
Disease Progression , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Encephalomyopathies/enzymology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Animals , Brain/drug effects , Mice , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Encephalomyopathies/drug therapy , Motor Activity/drug effects , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Phenanthrenes/therapeutic use , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism
13.
Immunol Cell Biol ; 92(2): 191-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275857

ABSTRACT

NAD biosynthesis is emerging as a key regulator of immune cell functions. Accordingly, inhibitors of the NAD-synthesizing enzyme nicotinamide phosphoribosyltransferase (NAMPT) have anti-inflammatory effects, counteract hematological malignancies and are being tested in clinical trials. Still, their effect on different cell types still waits to be fully investigated. Here we show that the NAMPT inhibitor FK866 induces NAD depletion in various mouse organs but selectively causes dramatic atrophy of the spleen red pulp. Accordingly, in cultured mouse lymphocytes exposed to FK866, NAD contents drop to 50% of basal values within 2 days, a condition sufficient to prompt complete cell death. Cultures of human lymphocytes are more resistant to FK866 and sustain a 50% NAD reduction for 5 days before dying. Death of both cell types can be prevented by different NAD precursors, indicating critical NAD homeostasis in lymphocytes. Indeed, inhibition of the NAD-consuming enzyme poly(ADP-ribose) polimerase-1 suffices to prevent FK866-induced NAD depletion and death of both lymphocyte types. Poly(ADP-ribose) polymerase-1-null lymphocytes also undergo lower NAD depletion and reduced cell death when exposed to the drug. At variance with other cell types, neither apoptosis nor autophagy are exclusively responsible for lymphocyte death by FK866, consistent with a general impairment of lymphocyte homeostasis following NAD depletion. Data demonstrate a unique sensitivity of resting lymphocytes to NAD-depleting agents, providing new hints of relevance to lymphocyte biology and therapeutic interventions with NAMPT inhibitors.


Subject(s)
Apoptosis/immunology , Cytokines/immunology , NAD/immunology , Nicotinamide Phosphoribosyltransferase/immunology , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cytokines/antagonists & inhibitors , Humans , Male , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/immunology
14.
PLoS One ; 8(11): e82180, 2013.
Article in English | MEDLINE | ID: mdl-24312407

ABSTRACT

Limited information is available on the brain expression and role of GPR35, a Gi/o coupled receptor activated by kynurenic acid (KYNA). In mouse cultured astrocytes, we detected GPR35 transcript using RT-PCR and we found that KYNA (0.1 to 100 µM) decreased forskolin (FRSK)-induced cAMP production (p<0.05). Both CID2745687 (3 µM, CID), a recently described GPR35 antagonist, and GPR35 gene silencing significantly prevented the action of KYNA on FRSK-induced cAMP production. In these cultures, we then evaluated whether GPR35 activation was able to modulate intracellular Ca(2+) concentration ([Ca(2+)]i ) and [Ca(2+)]i fluxes. We found that both KYNA and zaprinast, a phosphodiesterase (PDE) inhibitor and GPR35 agonist, did not modify either basal or peaks of [Ca(2+)]i induced by challenging the cells with ATP (30 µM). However, the [Ca(2+)]i plateau phase following peak was significantly attenuated by these compounds in a store-operated Ca(2+) channel (SOC)-independent manner. The activation of GPR35 by KYNA and zaprinast was also studied at the CA3-CA1 synapse in the rat hippocampus. Evoked excitatory post synaptic currents (eEPSCs) were recorded from CA1 pyramidal neurons in acute brain slices. The action of KYNA on GPR35 was pharmacologically isolated by using NMDA and α7 nicotinic receptor blockers and resulted in a significant reduction of eEPSC amplitude. This effect was prevented in the presence of CID. Moreover, zaprinast reduced eEPSC amplitude in a PDE5- and cGMP-independent mechanism, thus suggesting that glutamatergic transmission in this area is modulated by GPR35. In conclusion, GPR35 is expressed in cultured astrocytes and its activation modulates cAMP production and [Ca(2+)]i. GPR35 activation may contribute to KYNA effects on the previously reported decrease of brain extracellular glutamate levels and reduction of excitatory transmission.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Kynurenic Acid/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Mice , Real-Time Polymerase Chain Reaction , Synapses/physiology , Synaptic Transmission
15.
PLoS One ; 8(6): e66527, 2013.
Article in English | MEDLINE | ID: mdl-23840500

ABSTRACT

BACKGROUND: High mobility group box-1 (HMGB1) is a DNA-binding protein that is released from injured cells during inflammation. Advances in targeting HMGB1 represent a major challenge to improve the treatment of acute/chronic inflammation. AIM: This study is aimed at verifying whether the inhibition of HMGB1 through dipotassium glycyrrhizate (DPG) is a good strategy to reduce intestinal inflammation. METHODS: Human colon adenocarcinoma cell line, HT29, human epithelial colorectal adenocarcinoma, Caco2, and murine macrophage cell line, RAW 264.7, were cultured to investigate the effect of DPG on the secretion of HMGB1. Acute colitis was induced in C57BL/6 mice through administration of 3% dextran sodium sulphate (DSS); a combined treatment with DSS and 3 or 8 mg/kg/day DPG was used to investigate the effects of DPG on intestinal inflammation. Animals were euthanized at seventh day and colonic samples underwent molecular and histological analyses. RESULTS: DPG significantly reduces in vitro the release of HMGB1 in the extracellular matrix as well as expression levels of pro-inflammatory cytokines, TNF-alpha, IL-1beta and IL-6, by inhibiting HMGB1. Moreover, DPG significantly decreases the severity of DSS-induced colitis in mice. Murine colonic samples show decreased mRNA levels of pro-inflammatory cytokines TNF-alpha, IL-1beta and IL-6, as well as HMGB1 receptors, RAGE and TLR4. Finally, HMGB1, abundantly present in the feces of mice with DSS-induced colitis, is strongly reduced by DPG. CONCLUSIONS: HMGB1 is an early pro-inflammatory cytokine and an active protagonist of mucosal gut inflammation. DPG exerts inhibitory effects against HMGB1 activity, significantly reducing intestinal inflammation. Thus, we reason that DPG could represent an innovative tool for the management of human intestinal inflammation.


Subject(s)
Colitis/prevention & control , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/antagonists & inhibitors , Inflammation/prevention & control , Animals , Cell Line , Cell Line, Tumor , Colitis/metabolism , Cytokines/metabolism , Female , HMGB1 Protein/metabolism , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
16.
J Cereb Blood Flow Metab ; 33(2): 183-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23093068

ABSTRACT

Therapeutic hypothermia is of relevance to treatment of increased body temperature and brain injury, but drugs inducing selective, rapid, and safe cooling in humans are not available. Here, we show that injections of adenosine 5'-monophosphate (AMP), an endogenous nucleotide, promptly triggers hypothermia in mice by directly activating adenosine A1 receptors (A1R) within the preoptic area (POA) of the hypothalamus. Inhibition of constitutive degradation of brain extracellular AMP by targeting ecto 5'-nucleotidase, also suffices to prompt hypothermia in rodents. Accordingly, sensitivity of mice and rats to the hypothermic effect of AMP is inversely related to their hypothalamic 5'-nucleotidase activity. Single-cell electrophysiological recording indicates that AMP reduces spontaneous firing activity of temperature-insensitive neurons of the mouse POA, thereby retuning the hypothalamic thermoregulatory set point towards lower temperatures. Adenosine 5'-monophosphate also suppresses prostaglandin E2-induced fever in mice, having no effects on peripheral hyperthermia triggered by dioxymetamphetamine (ecstasy) overdose. Together, data disclose the role of AMP, 5'-nucleotidase, and A1R in hypothalamic thermoregulation, as well and their therapeutic relevance to treatment of febrile illness.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine Monophosphate/metabolism , Body Temperature Regulation , Fever/metabolism , Nerve Tissue Proteins/metabolism , Preoptic Area/metabolism , Receptor, Adenosine A1/metabolism , Adenosine Monophosphate/pharmacology , Adrenergic Uptake Inhibitors/adverse effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Dinoprostone/adverse effects , Dinoprostone/pharmacology , Fever/chemically induced , Fever/drug therapy , Humans , Hypothermia, Induced , Male , Mice , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurons/metabolism , Oxytocics/adverse effects , Oxytocics/pharmacology , Preoptic Area/pathology , Prescription Drug Misuse , Rats , Rats, Wistar
17.
J Cereb Blood Flow Metab ; 32(6): 978-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22434066

ABSTRACT

Although treatment of stroke patients with mild hypothermia is a promising therapeutic approach, chemicals inducing prompt and safe reduction of body temperature are an unmet need. We measured the effects of the transient receptor potential vanilloid-1 (TRPV1) agonist rinvanil on thermoregulation and ischemic brain injury in mice. Intraperitoneal or intracerebroventricular injection of rinvanil induces mild hypothermia that is prevented by the receptor antagonist capsazepine. Both intraischemic and postischemic treatments provide permanent neuroprotection in animals subjected to transient middle cerebral artery occlusion (MCAo), an effect lost in mice artificially kept normothermic. Data indicate that TRPV1 receptor agonists are promising candidates for hypothermic treatment of stroke.


Subject(s)
Hypothermia, Induced/methods , Neuroprotective Agents/pharmacology , Stroke/therapy , TRPV Cation Channels/agonists , Animals , Brain Ischemia/therapy , Male , Mice
18.
Trends Mol Med ; 18(2): 92-100, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22078487

ABSTRACT

Despite significant advancement in developing therapies for multiple sclerosis (MS), drugs that cure this devastating disorder are an unmet need. Among the remedies showing efficacy in preclinical MS models, inhibitors of poly(ADP-ribose) polymerase (PARP)-1 have gained great momentum. Emerging evidence demonstrates that PARP-1 inhibitors epigenetically regulate gene expression and finely tune transcriptional activation in immune and neural cells. In this review, we present an appraisal of the effects of PARP-1 and its inhibitors on immune activation, with particular emphasis on the processes taking place during the autoimmune attack directed against the central nervous system. One explanation is that drugs inhibiting PARP-1 activity protect from neuroinflammation in MS models via immunomodulation and direct neuroprotection. PARP-1 inhibitors have already reached the clinical arena as cancer treatments, and observations made in treating these patients could help advance treatments for MS.


Subject(s)
Enzyme Inhibitors/pharmacology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/immunology , Animals , Enzyme Inhibitors/therapeutic use , Humans , Immunomodulation/drug effects , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Transcriptional Activation/drug effects
19.
Mol Med ; 17(5-6): 442-7, 2011.
Article in English | MEDLINE | ID: mdl-21373721

ABSTRACT

Multiple sclerosis (MS) is a devastating autoimmune disorder of the central nervous system (CNS) for which there is no efficacious cure. Thanks to numerous preclinical and clinical studies, drugs able to mitigate the inexorable course of the disease have been made available recently. Still, there is a terrible need for compounds capable of reducing the severity of the autoimmune attack and of blocking progression of the disorder. Also, besides the classic immunosuppressive strategies, it is now appreciated that compounds directly targeting neuronal death can be of relevance to the treatment of MS patients. Acetylation homeostasis is a key regulator of both immune cell activation and neuronal survival. Of note, potent histone deacetylase inhibitors (HDACi) endowed with antiinflammatory and neuroprotective properties have been identified. Efficacy of HDACi in experimental models of MS has been reported consistently. In this review, we provide an appraisal of the literature on HDACi and MS, also discussing the mechanisms by which HDACi can suppress the autoimmune attack to the CNS.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Multiple Sclerosis/drug therapy , Animals , Humans , Multiple Sclerosis/immunology
20.
Ocul Immunol Inflamm ; 19(3): 180-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21426233

ABSTRACT

PURPOSE: High-mobility group proteins are chromatin-binding factors with key roles in nuclear homeostasis. Evidence indicates that extracellularly released high-mobility group box 1 protein (HMGB1) behaves as a cytokine, promoting inflammation and disease pathogenesis. HMGB1 release occurs during endophtalmitis or uveoretinitis. METHODS: The authors investigated the presence of HMGB1 in tear fluid of patients with different inflammatory disorders of the external eye. RESULTS: Data demonstrate that HMGB1 content is close to detection limit in tears of control subjects but highly increased (about 15-fold) in patients with conjunctivitis or blepharitis. The authors also report that 18ß-glycyrrhetic acid impairs antibody recognition of HMGB1, suggesting direct binding to the protein. Accordingly, 18ß-glycyrrhetic acid prevented HMGB1-dependent COX2 expression and cluster formation in primary cultures of human macrophages. CONCLUSION: Together, these findings suggest that HMGB1 contributes to inflammatory disorders of the external eye, and 18ß-glycyrrhetic acid may scavenge the protein and inhibit its detrimental effects.


Subject(s)
Blepharitis/immunology , Conjunctivitis/immunology , Glycyrrhetinic Acid/analogs & derivatives , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/immunology , Tears/metabolism , Adult , Antibodies/drug effects , Antibodies/immunology , Cells, Cultured , Cyclooxygenase 2/metabolism , Enzyme Induction/drug effects , Female , Glycyrrhetinic Acid/pharmacology , HMGB1 Protein/metabolism , Humans , Macrophages/enzymology , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...