Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 25(1)2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38203506

ABSTRACT

The median survival time of patients with an aggressive brain tumor, glioblastoma, is still poor due to ineffective treatment. The discovery of androgen receptor (AR) expression in 56% of cases offers a potential breakthrough. AR antagonists, including bicalutamide and enzalutamide, induce dose-dependent cell death in glioblastoma and glioblastoma-initiating cell lines (GIC). Oral enzalutamide at 20 mg/kg reduces subcutaneous human glioblastoma xenografts by 72% (p = 0.0027). We aimed to further investigate the efficacy of AR antagonists in intracranial models of human glioblastoma. In U87MG intracranial models, nude mice administered Xtandi (enzalutamide) at 20 mg/kg and 50 mg/kg demonstrated a significant improvement in survival compared to the control group (p = 0.24 and p < 0.001, respectively), confirming a dose-response relationship. Additionally, we developed a newly reformulated version of bicalutamide, named "soluble bicalutamide (Bic-sol)", with a remarkable 1000-fold increase in solubility. This reformulation significantly enhanced bicalutamide levels within brain tissue, reaching 176% of the control formulation's area under the curve. In the U87MG intracranial model, both 2 mg/kg and 4 mg/kg of Bic-sol exhibited significant efficacy compared to the vehicle-treated group (p = 0.0177 and p = 0.00364, respectively). Furthermore, combination therapy with 8 mg/kg Bic-sol and Temozolomide (TMZ) demonstrated superior efficacy compared to either Bic-sol or TMZ as monotherapies (p = 0.00706 and p = 0.0184, respectively). In the ZH-161 GIC mouse model, the group treated with 8 mg/kg Bic-sol as monotherapy had a significantly longer lifespan than the groups treated with TMZ or the vehicle (p < 0.001). Our study demonstrated the efficacy of androgen receptor antagonists in extending the lifespan of mice with intracranial human glioblastoma, suggesting a promising approach to enhance patient outcomes in the fight against this challenging disease.


Subject(s)
Anilides , Benzamides , Glioblastoma , Nitriles , Phenylthiohydantoin , Tosyl Compounds , Humans , Animals , Mice , Glioblastoma/drug therapy , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Mice, Nude , Temozolomide/pharmacology
2.
J Autoimmun ; 124: 102713, 2021 11.
Article in English | MEDLINE | ID: mdl-34390919

ABSTRACT

Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1ß from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.


Subject(s)
Arthritis, Rheumatoid/immunology , Hyaluronan Receptors/genetics , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Multiple Sclerosis/immunology , Peptides/genetics , Serum Amyloid A Protein/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Autoimmunity , Cells, Cultured , Computational Biology , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Peptides/therapeutic use , Serum Amyloid A Protein/genetics
4.
Hum Mol Genet ; 23(15): 4134-41, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24667414

ABSTRACT

Prion diseases, which can manifest by transmissible, sporadic or genetic etiologies, share several common features, such as a fatal neurodegenerative outcome and the aberrant accumulation of proteinase K (PK)-resistant PrP forms in the CNS. In infectious prion diseases, such as scrapie in mice, prions first replicate in immune organs, then invade the CNS via ascending peripheral tracts, finally causing death. Accelerated neuroinvasion and death occurs when activated prion-infected immune cells infiltrate into the CNS, as is the case for scrapie-infected mice induced for experimental autoimmune encephalomyelitis (EAE), a CNS inflammatory insult. To establish whether the immune system plays such a central role also in genetic prion diseases, we induced EAE in TgMHu2ME199K mice, a line mimicking for late onset genetic Creutzfeldt Jacob disease (gCJD), a human prion disease. We show here that EAE induction of TgMHu2ME199K mice neither accelerated nor aggravated prion disease manifestation. Concomitantly, we present evidence that PK-resistant PrP forms were absent from CNS immune infiltrates, and most surprisingly also from lymph nodes and spleens of TgMHu2ME199K mice at all ages and stages of disease. These results imply that the mechanism of genetic prion disease differs widely from that of the infectious presentation, and that the conversion of mutant PrPs into PK resistant forms occurs mostly/only in the CNS. If the absence of pathogenic PrP forms form immune organs is also true for gCJD patients, it may suggest their blood is devoid of prion infectivity.


Subject(s)
Central Nervous System/pathology , Creutzfeldt-Jakob Syndrome/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , PrPSc Proteins/genetics , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Creutzfeldt-Jakob Syndrome/complications , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/immunology , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression , Humans , Immune System , Mice , Mice, Transgenic , PrPSc Proteins/metabolism , Severity of Illness Index
5.
Neuroimmunomodulation ; 23(1): 1-7, 2016.
Article in English | MEDLINE | ID: mdl-26670748

ABSTRACT

OBJECTIVES: The role of nitric oxide (NO) in modulating the blood-brain barrier (BBB) is not entirely clear. We examined the effect of different NO synthase (NOS) inhibitors and NO donors on the permeability of the BBB in animals with normally functioning brain blood vessels, following disruption by hyperosmotic mannitol and during immune inflammation. METHODS: We administered L-NAME, aminoguanidine, S-methyl-thiocitrulline (SMT) and 7-indazole (NOS inhibitors) and NOR-4 (an NO donor) into the cerebral ventricle of rats. Disruption of the BBB was induced by intracarotid injection of mannitol (25%). Experimental autoimmune encephalomyelitis (EAE) was induced by brain homogenate. The extent of disruption was evaluated by Evans blue (2%) dye extravasation. RESULTS: L-NAME (a nonspecific NOS inhibitor) and SMT (a neuronal and endothelial NOS inhibitor) increased mannitol-induced disruption of BBB. This effect was inhibited by NO donors. In animals with a normally functioning BBB, none of these inhibitors or NO donors caused a change in the permeability. 7-indazole (a specific neuronal NOS inhibitor) and aminoguanidine (an inducible NOS inhibitor) had no facilitatory effect on BBB permeability, either alone or in combination with hyperosmotic mannitol. Administration of L-NAME and SMT to rats with EAE significantly aggravated the clinical outcome. In contrast, the administration of NOR-4 diminished clinical signs of EAE. CONCLUSION: The NOS system does not play a role in BBB permeability in naïve animals. Only endothelial NOS takes part in the facilitation of BBB compromised by mannitol and EAE. Extrinsic NO decreases this facilitatory effect.


Subject(s)
Blood-Brain Barrier/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Osmotic Pressure/drug effects , Animals , Blood-Brain Barrier/drug effects , Disease Models, Animal , Diuretics, Osmotic/pharmacology , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Mannitol/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Pyridines/pharmacology , Rats , Rats, Inbred Lew , Time Factors
6.
Mult Scler ; 21(5): 572-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25392324

ABSTRACT

BACKGROUND: Screening of putative autoimmune targets in multiple sclerosis (MS) revealed a proportion of patients carrying antibodies (Abs) against KIR4.1, a potassium channel that shares functional properties with AQP4. Both are localized at the perivascular astrocytic processes. AIMS: To measure anti-KIR4.1 Abs in the serum of MS and neuromyelitis optica (NMO) patients, and to identify the clinical and laboratory characteristics of patients harboring anti-KIR4.1 Abs. METHODS: We measured anti-KIR4.1 Abs in serum, using the peptide KIR4.1 (83-120) enzyme-linked immunosorbent assay (ELISA). RESULTS: Serum levels of anti-KIR4.1 Abs were significantly higher in MS and NMO patients than in healthy controls (HCs); with Abs detected in 21 of 80, 10 of 45, and 2 of 32 individuals, respectively (MS versus HC, p < 0.05). The level of anti-KIR4.1 Abs was significantly higher during MS relapse, versus remission (p = 0.04). The clinical characteristics of our study patients did not vary based on KIR4.1 positivity. CONCLUSIONS: Anti-KIR4.1 Abs were found in similar proportions of patients with MS and NMO, at a significantly higher level than observed in HCs; consequently, the presence of Abs does not discriminate between these demyelinating diseases. However, anti-KIR4.1 Ab levels differed in MS patients during relapse and remission; as such, they may represent a marker of disease exacerbation.


Subject(s)
Autoantibodies/analysis , Biomarkers/analysis , Multiple Sclerosis/immunology , Potassium Channels, Inwardly Rectifying/immunology , Adolescent , Child , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Neuromyelitis Optica/immunology , Prognosis , Recurrence , Reproducibility of Results
7.
Neuroimmunomodulation ; 22(6): 394-9, 2015.
Article in English | MEDLINE | ID: mdl-26352093

ABSTRACT

OBJECTIVE: The amygdala (AMG) plays a facilitatory role in the hypothalamic-pituitary-adrenal (HPA) axis. The effect of the AMG on the negative feedback exerted by glucocorticoids (GC) is not clear. We investigated the effect of repeated electrical stimulation of the AMG on the feedback action of GC upon the adrenocortical (AC) response to stressful stimuli. METHODS: Rats received electrical stimulation into the central amygdalar nucleus once daily for 4 days. At days 5 and 12 after the onset of stimulation, rats were treated with dexamethasone (Dex) or vehicle and were exposed to either photic or acoustic stress stimuli, and serum corticosterone (CS) was measured. In another group of rats, we measured the binding of Dex to the hippocampal cytosol at 5 and 12 days after the AMG stimulation. RESULTS: At 5 and 12 days after the onset of stimulation or a sham control, stress increased the serum CS level. In the sham group, Dex completely inhibited the CS response, but at 5 days after stimulation, it was significantly less effective in doing this. At day 12, Dex was as effective as in the control group. AMG stimulation delayed the return of CS response to basal levels and caused a significant decrease in the binding capacity of Dex to hippocampal cytosol. CONCLUSION: Electrical stimulation of the AMG caused a transient impairment of the feedback action of GC upon the stress response. This effect may be due to the decrease in hippocampal corticosteroid receptors. This suggests that the impaired GC feedback caused by AMG stimulation may be involved in the facilitatory effect of the AMG on the function of the AC axis.


Subject(s)
Central Amygdaloid Nucleus/physiology , Dexamethasone/therapeutic use , Electric Stimulation/methods , Glucocorticoids/therapeutic use , Stress, Psychological , Acoustic Stimulation/adverse effects , Animals , Corticosterone/blood , Disease Models, Animal , Feedback , Glucocorticoids/blood , Hippocampus/metabolism , Male , Photic Stimulation/adverse effects , Radioimmunoassay , Rats , Stress, Psychological/blood , Stress, Psychological/pathology , Stress, Psychological/therapy , Time Factors
8.
J Pathol ; 232(3): 369-81, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24421076

ABSTRACT

Although the incidence of breast cancer metastasis (BCM) in brain has increased significantly in triple-negative breast cancer (TNBC), the mechanisms remain elusive. Using in vivo mouse models for BCM in brain, we observed that TNBC cells crossed the blood-brain barrier (BBB), lodged in the brain microvasculature and remained adjacent to brain microvascular endothelial cells (BMECs). Breaching of the BBB in vivo by TNBCs resulted in increased BBB permeability and changes in ZO-1 and claudin-5 tight junction (TJ) protein structures. Angiopoietin-2 expression was elevated in BMECs and was correlated with BBB disruption. Secreted Ang-2 impaired TJ structures and increased BBB permeability. Treatment of mice with the neutralizing Ang-2 peptibody trebananib prevented changes in the BBB integrity and BMEC destabilization, resulting in inhibition of TNBC colonization in brain. Thus, Ang-2 is involved in initial steps of brain metastasis cascade, and inhibitors for Ang-2 may serve as potential therapeutics for brain metastasis.


Subject(s)
Angiopoietin-2/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Blood-Brain Barrier/pathology , Capillary Permeability/physiology , Female , Heterografts , Humans , Immunohistochemistry , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Tight Junctions/metabolism , Tight Junctions/pathology , Triple Negative Breast Neoplasms/pathology
9.
Nanomedicine ; 10(6): 1353-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24704590

ABSTRACT

Neurodegenerative diseases generate the accumulation of specific misfolded proteins, such as PrP(Sc) prions or A-beta in Alzheimer's diseases, and share common pathological features, like neuronal death and oxidative damage. To test whether reduced oxidation alters disease manifestation, we treated TgMHu2ME199K mice, modeling for genetic prion disease, with Nano-PSO, a nanodroplet formulation of pomegranate seed oil (PSO). PSO comprises large concentrations of a unique polyunsaturated fatty acid, Punicic acid, among the strongest natural antioxidants. Nano-PSO significantly delayed disease presentation when administered to asymptomatic TgMHu2ME199K mice and postponed disease aggravation in already sick mice. Analysis of brain samples revealed that Nano-PSO treatment did not decrease PrP(Sc) accumulation, but rather reduced lipid oxidation and neuronal loss, indicating a strong neuroprotective effect. We propose that Nano-PSO and alike formulations may be both beneficial and safe enough to be administered for long years to subjects at risk or to those already affected by neurodegenerative conditions. FROM THE CLINICAL EDITOR: This team of authors report that a nanoformulation of pomegranade seed oil, containing high levels of a strong antioxidant, can delay disease onset in a mouse model of genetic prion diseases, and the formulation also indicates a direct neuroprotective effect.


Subject(s)
Creutzfeldt-Jakob Syndrome/drug therapy , Emulsions/therapeutic use , Lythraceae/chemistry , Neuroprotective Agents/therapeutic use , Plant Oils/therapeutic use , Animals , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Emulsions/chemistry , Lipid Peroxidation/drug effects , Mice , Neuroprotective Agents/chemistry , Oxidation-Reduction , Plant Oils/chemistry , Prions/metabolism , Seeds/chemistry
10.
Neuroimmunomodulation ; 20(1): 51-6, 2013.
Article in English | MEDLINE | ID: mdl-23183048

ABSTRACT

BACKGROUND: Central nervous system (CNS) irradiation has detrimental effects which become evident within hours to few days and after a long latency of months and years. However, the delayed effect of irradiation on neuroimmune diseases has not been thoroughly examined. OBJECTIVES: We evaluated the delayed effects of irradiation on the course of experimental autoimmune encephalomyelitis (EAE), which is used as a model for neuroimmune inflammation and multiple sclerosis. METHODS: Adult male rats were exposed to a dose of 15 Gy given to the thoracolumbar spinal cord. Six months later, EAE was induced by inoculation of rat spinal cord homogenate in complete Freund's adjuvant (CFA). The disease was evaluated by clinical, histopathological and immunological parameters. RESULTS: Irradiated rats developed clinical signs of EAE earlier than the control group and their disease was much more severe. Unlike the control group, all rats in the EAE-irradiated group died within 5 days after the onset of clinical signs. Sections taken from irradiated rats showed diffuse and large hemorrhagic infiltrates of lymphocytes and granulocytes. In contrast, control rats displayed fewer infiltrates, which were less prominent and not hemorrhagic. CONCLUSIONS: CNS irradiation has a delayed effect that caused a marked aggravation of the clinical and pathological signs of EAE. The severity of the disease may be a consequence of the effect of irradiation on the CNS vascular bed and impaired blood-brain barrier.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Spinal Cord/pathology , Spinal Cord/radiation effects , Animals , Craniospinal Irradiation/adverse effects , Female , Flow Cytometry , Rats , Rats, Inbred F344
11.
Neuroimmunomodulation ; 20(1): 57-64, 2013.
Article in English | MEDLINE | ID: mdl-23183099

ABSTRACT

BACKGROUND: Brain irradiation (BI) in humans may cause behavioral changes, cognitive impairment and neuroendocrine dysfunction. The effect of BI on the hypothalamic-pituitary-adrenal (HPA) axis is not fully understood. OBJECTIVES: To evaluate the effect of BI on HPA axis responses under basal and stressful conditions as well as following pretreatment with dexamethasone (Dex). METHODS: Adult male rats were exposed to whole BI. HPA axis responses were examined at 2, 4, 9 and 20 weeks after BI. Histological evaluations of the irradiated rats and matched controls were conducted at 4 and 20 weeks after BI. RESULTS: In contrast to the control group, the basal and stress-induced corticosterone levels were enhanced at 9 and 20 weeks after BI and the inhibitory effect of Dex was reduced. BI also caused hyposuppression of the adrenocortical response to stress. Histological assessment of the irradiated brains revealed hippocampal atrophy at 20 weeks after BI. The neuronal counts were lower only in the CA1 region of the irradiated brains. BI caused a decrease in the binding capacity of Dex to the hippocampal cytosolic fraction. CONCLUSIONS: Enhanced stress-induced HPA axis responses and the reduced effect of Dex suggest that BI has delayed effects on HPA axis responses as manifested by impairment of the negative feedback exerted by glucocorticoids (GCs). The mechanisms underlying these effects of BI are unknown. It is possible that the marked BI-induced damage in the hippocampus, which plays an important role in the regulation of the feedback effect of GCs, may cause abnormal HPA axis responses following BI.


Subject(s)
Cranial Irradiation/adverse effects , Hippocampus/radiation effects , Hypothalamo-Hypophyseal System/radiation effects , Pituitary-Adrenal System/radiation effects , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Disease Models, Animal , Hippocampus/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Immunohistochemistry , Male , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Radioimmunoassay , Rats , Rats, Inbred F344 , Stress, Physiological/physiology , Stress, Psychological/metabolism
12.
Harefuah ; 152(2): 101-5, 122, 2013 Feb.
Article in Hebrew | MEDLINE | ID: mdl-23513502

ABSTRACT

INTRODUCTION: Inflammatory demyelinative diseases of the central nervous system are mostly idiopathic and represent the major cause of neurological disability in young adults. These diseases differ in terms of clinical symptoms, severity, pathological characteristics and epidemiology. However, there are also significant similarities between these diseases, which sometimes bring to a misleading diagnosis. Neuromyelitis optica (NMO) is a demyelinative disease in which the optic nerve and the spinal cord are predominantly affected. The detection of specific antibodies to aquaporin-4 (NMO-IgG) led to a modification of the diagnostic criteria for NMO. METHODS: We performed a retrospective study on NMO-IgG positive patients referred to the Department of Neurology MS Center (2006-2011) with suspected NMO. Based on the presenting symptomatology of the patients, we identified the cases with optic neuritis and various parameters that may differentiate between NMO and MS. NMO-IgG were evaluated by ELISA. RESULTS: A total of 50% of the 107 patients with NMO-IgG fulfilled the revised criteria of NMO; 38 patients had a single attack of optic neuritis or long lesion in the spinal cord and 15 patients presented with an opticospinal type of MS. The visual acuity following a single attack of optic neuritis remained significantly lower in NMO patients as compared to MS patients. Most of the NMO patients with NMO-IgG had additional attacks of optic neuritis within a short time from the initial event. CONCLUSIONS: The finding of NMO-IgG in patients with optic neuritis foreshadows a bad prognosis and relapses. These patients are at high risk of experiencing a second event in the central nervous system and fulfilling the clinical criteria for NMO. Due to the difference in the severity of inflammation of the optic nerve between NMO and MS, it is highly recommended to seek a laboratory check-up for NMO-IgG in serum, immediately after the first event, in order to determine the necessity and the kind of treatment for the patient.


Subject(s)
Multiple Sclerosis/diagnosis , Neuromyelitis Optica/diagnosis , Optic Neuritis/diagnosis , Adolescent , Adult , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/immunology , Male , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/pathology , Optic Neuritis/immunology , Optic Neuritis/pathology , Retrospective Studies , Severity of Illness Index , Time Factors , Visual Acuity , Young Adult
13.
J Neuroinflammation ; 9: 58, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22436404

ABSTRACT

BACKGROUND: Prions, composed of a misfolded protein designated PrP(Sc), are infectious agents causing fatal neurodegenerative diseases. We have shown previously that, following induction of experimental autoimmune encephalomyelitis, prion-infected mice succumb to disease significantly earlier than controls, concomitant with the deposition of PrP(Sc) aggregates in inflamed white matter areas. In the present work, we asked whether prion disease acceleration by experimental autoimmune encephalomyelitis results from infiltration of viable prion-infected immune cells into the central nervous system. METHODS: C57Bl/6 J mice underwent intraperitoneal inoculation with scrapie brain homogenates and were later induced with experimental autoimmune encephalomyelitis by inoculation of MOG(35-55) in complete Freund's adjuvant supplemented with pertussis toxin. Spleen and lymph node cells from the co-induced animals were reactivated and subsequently injected into naïve mice as viable cells or as cell homogenates. Control groups were infected with viable and homogenized scrapie immune cells only with complete Freund's adjuvant. Prion disease incubation times as well as levels and sites of PrP(Sc) deposition were next evaluated. RESULTS: We first show that acceleration of prion disease by experimental autoimmune encephalomyelitis requires the presence of high levels of spleen PrP(Sc). Next, we present evidence that mice infected with activated prion-experimental autoimmune encephalomyelitis viable cells succumb to prion disease considerably faster than do mice infected with equivalent cell extracts or other controls, concomitant with the deposition of PrP(Sc) aggregates in white matter areas in brains and spinal cords. CONCLUSIONS: Our results indicate that inflammatory targeting of viable prion-infected immune cells to the central nervous system accelerates prion disease propagation. We also show that in the absence of such targeting it is the load of PrP(Sc) in the inoculum that determines the infectivity titers for subsequent transmissions. Both of these conclusions have important clinical implications as related to the risk of prion disease contamination of blood products.


Subject(s)
Central Nervous System , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Lymphocytes/pathology , Prion Diseases/pathology , Prions/metabolism , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/virology , Glycoproteins/adverse effects , Humans , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/adverse effects , Prion Diseases/complications , Prions/pathogenicity
14.
Neuroimmunomodulation ; 18(2): 111-6, 2011.
Article in English | MEDLINE | ID: mdl-21071988

ABSTRACT

OBJECTIVE: Herpes simplex virus-1 (HSV-1) is a common cause of viral encephalitis manifested by activation of the adrenocortical axis, fever and behavioral changes. We investigated the early effects of HSV-1 on constitutive (c) and inducible (i) nitric oxide synthase (NOS) activity in rat brain and in mixed glial cell culture. The effect of glucocorticoids (GCs) on NOS responses to HSV-1 was also determined. METHODS: NOS activity was evaluated by the conversion of ³H-arginine to ³H-citrulline. Nitrites were measured in supernatants of activated glial cells. RESULTS: Under basal conditions, the highest cNOS activity was found in the cerebellum, while activity was much lower in the pons and negligible in the hypothalamus and hippocampus. Forty-eight hours after intracerebral injection of HSV-1, serum corticosterone was increased and NOS activity in the cerebellum and pons was inhibited. Adrenalectomy had no effect on the basal NOS activity but completely abrogated the inhibitory effect of HSV-1. Administration of the iNOS inhibitor aminoguanidine did not significantly change NOS activity, suggesting that the activity found in the cerebellum and pons can be attributed to the cNOS isoform. In mixed glial cell culture infected with HSV-1 and then activated with lipopolysaccharide, NOS activity and nitrite production were inhibited by 77 and 53%, respectively. CONCLUSIONS: These results suggest that brain NOS activity is inhibited in the early stages of HSV-1 infection and requires the presence of circulating GCs. HSV-1-induced brain NOS inhibition may play a role in neuronal viral invasion and in the activation of the adrenocortical axis.


Subject(s)
Brain/metabolism , Encephalitis, Herpes Simplex/metabolism , Glucocorticoids/blood , Herpesvirus 1, Human , Nitric Oxide Synthase/metabolism , Animals , Brain/virology , Corticosterone/blood , Isoenzymes/metabolism , Male , Rats
15.
Vision (Basel) ; 5(2)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207828

ABSTRACT

Nitric oxide (NO) is acknowledged as a vital intercellular messenger in multiple systems in the body. Medicine has focused on its functions and therapeutic applications for decades, especially in cardiovascular and nervous systems, and its role in immunological responses. This review was composed to demonstrate the prevalence of NO in components of the ocular system, including corneal cells and multiple cells in the retina. It discussed NO's assistance during the immune, inflammation and wound-healing processes. NO is identified as a vascular endothelial relaxant that can alter the choroidal blood flow and prompt or suppress vascular changes in age-related macular degeneration and diabetes, as well as the blood supply to the optic nerve, possibly influencing the progression of glaucoma. It will provide a deeper understanding of the role of NO in ocular homeostasis, the delicate balance between overproduction or underproduction and the effect on the processes from aqueous outflow and subsequent intraocular pressure to axial elongation and the development of myopia. This review also recognized the research and investigation of therapies being developed to target the NO complex and treat various ocular diseases.

16.
JACS Au ; 1(12): 2361-2376, 2021 Dec 27.
Article in English | MEDLINE | ID: mdl-34977904

ABSTRACT

Integrins α4ß1/ α9ß1 are important in the pathogenesis and progression of inflammatory and autoimmune diseases by their roles in leukocyte activation and trafficking. Natalizumab, a monoclonal antibody selectively targeting α4ß1 integrin and blocking leukocyte trafficking to the central nervous system, is an immunotherapy for multiple sclerosis (MS). However, due to its adverse effects associated with chronic treatment, alternative strategies using small peptide mimetic inhibitors are being sought. In the present study, we synthesized and characterized visabron c (4-4), a backbone cyclic octapeptide based on the sequence TMLD, a non-RGD unique α4ß1 integrin recognition sequence motif derived from visabres, a proteinous disintegrin from the viper venom. Visabron c (4-4) was selected from a minilibrary with conformational diversity based on its potency and selectivity in functional adhesion cellular assays. Visabron c (4-4)'s serum stability, pharmacokinetics, and therapeutic effects following ip injection were assessed in an experimental autoimmune encephalomyelitis (EAE) animal model. Furthermore, visabron c (4-4)'s lack of toxic effects in mice was verified by blood analysis, tissue pathology, immunogenicity, and "off-target" effects, indicating its significant tolerability and lack of immunogenicity. Visabron c (4-4) can be delivered systemically. The in vitro and in vivo data justify visabron c (4-4) as a safe alternative peptidomimetic lead compound/drug to monoclonal anti-α4 integrin antibodies, steroids, and other immunosuppressant drugs. Moreover, visabron c (4-4) design may pave the way for developing new therapies for a variety of other inflammatory and/or autoimmune diseases.

17.
Neuroimmunomodulation ; 17(5): 298-304, 2010.
Article in English | MEDLINE | ID: mdl-20407281

ABSTRACT

OBJECTIVE: Reciprocal pathways of interaction between the nervous and immune systems during stress may be regulated by stress-induced circulating glucocorticoids that act via type II glucocorticoid receptors (GRs). The aim of the present study was to investigate the effect of restraint stress on GRs in lymphocytes and the role of the sympathetic system in this effect. METHODS: We used male Balb/c mice which were adrenalectomized 3 days before exposure to restraint stress (4 h). Specific binding of 3H-dexamethasone (Dex) and the expression of GR protein were measured in the cytosol of spleen cells. RESULTS: Restraint stress caused a significant increase in the maximal binding of 3H-Dex to GRs in the cytosol of spleen cells but not in the binding affinity. In correlation with this increase in binding, restraint stress caused an increase in the amount of GR protein. To establish the relation of the nervous system in this stress response, we blocked the autonomic innervations to the spleen with the ganglionic blocker chlorisondamine. This blocker abrogated the stress-induced increase in the binding of 3H-Dex to GRs and in the GR protein levels. Abrogation of the stress response was also achieved by blocking beta-adrenergic receptors. CONCLUSION: These results suggest that stress-induced increase in the level of GRs is mediated by the sympathetic nervous system via beta-adrenergic receptors. It is possible that stress modulation of lymphocyte GR levels may be implicated in the bidirectional communication between the nervous and the immune systems.


Subject(s)
Lymphocyte Subsets/metabolism , Neuroimmunomodulation/immunology , Receptors, Glucocorticoid/physiology , Spleen/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Sympathetic Nervous System/immunology , Animals , Cells, Cultured , Lymphocyte Subsets/cytology , Male , Mice , Mice, Inbred BALB C , Receptors, Glucocorticoid/metabolism , Restraint, Physical , Spleen/cytology , Spleen/innervation , Sympathetic Nervous System/metabolism
18.
Mult Scler Relat Disord ; 39: 101881, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31835207

ABSTRACT

BACKGROUND: Neuromyelitis-optica (NMO) and multiple-sclerosis (MS) are inflammatory- demyelinating-diseases of the central-nervous-system (CNS). In a previous study, we identified 17 miRNAs that were significantly upregulated in the peripheral blood of patients with NMO, relative to healthy controls (HCs). Target gene analysis have demonstrated that QKI is targeted by 70% of the upregulated miRNAs. QKI gene encodes for a RNA-binding-protein that plays a central role in myelination. QKI variants 5, 6, 7 (QKI-V5, QKI-V6, QKI-V7) are generated via alternative splicing. Given the role played by QKI in myelination we aimed to study the expression levels of QKI variants in the circulation of patients with NMO and MS and in the circulation and brain tissue of mice-model to CNS-inflammatory-demyelinating-disease. METHODS: RNA and protein expression levels of QKI variants QKI-V5, QKI-V6 and QKI-V7 were determined in the blood of patients with NMO (n = 23) or MS (n = 13). The effect of sera from patients on the expression of QKI in normal peripheral-blood-mononuclear-cells (PBMCs) or glial cells was explored. The mog-experimental-autoimmune-encephalomyelitis (EAE) mouse model was used to study the correlation between the changes in the expression levels of QKI in the blood to those in the brain. RESULTS: RNA and protein expression of QKI-V5 was decreased in the peripheral blood of patients with NMO and multiple-sclerosis. Incubation of normal peripheral-blood-mononuclear-cells or glial cells with sera of patients significantly reduced the expression of QKI-V5. The blood and brain of EAE mice exhibited a corresponding decrease in QKI-V5 expression. CONCLUSION: The downregulation in the expression of QKI-V5 in the blood of patients with CNS-inflammatory-demyelinating-diseases and in the brain and blood of EAE mice is likely caused by a circulating factor and might promote re-myelination by regulation of myelin-associated genes. KEY WORDS: QKI variants, Multiple sclerosis (MS), Neuromyelitis optica (NMO), Astrocytes, Demyelination.

19.
Exp Biol Med (Maywood) ; 234(8): 940-51, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19491375

ABSTRACT

Several microbial species, including probiotic lactic acid bacteria, have the ability to irreversibly bind a large variety of polyphenols (flavonoids) and anthocyanidins found in many colored fruits and vegetables and to enhance their total oxidant-scavenging capacities (TOSC). The binding of flavonoids to microbial surfaces was further increased by the cationic polyelectrolytes ligands poly-L-histidine, chlorhexidine and Copaxone. This phenomenon was confirmed visually, by the FRAP, DPPH, cyclic voltammetry, Folin-Ciocalteu as well as by luminol-dependent chemiluminescence techniques employed to assay TOSC. The possibility is considered that clinically, microbial cells in the oral cavity and in the gastro intestinal tract, complexed with antioxidant polyphenols from nutrients and with cationic ligands, might increase the protection of mammalian cells against damage induced by excessive generation of reactive oxygen species during infections and inflammation.


Subject(s)
Bacteria/metabolism , Flavonoids/metabolism , Free Radical Scavengers/metabolism , Oxidants/metabolism , Phenols/metabolism , Beverages , Biphenyl Compounds/metabolism , Electrochemistry , Fluorescence Recovery After Photobleaching , Fruit/chemistry , Glucose Oxidase/metabolism , Luminescent Measurements , Luminol/metabolism , Molybdenum , Picrates/metabolism , Polyamines/metabolism , Polyelectrolytes , Polyphenols , Probiotics/metabolism , Tungsten Compounds
20.
BMC Clin Pathol ; 8: 9, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18803814

ABSTRACT

BACKGROUND: S100B protein is a potential biomarker of central nervous system insult. This study quantitatively compared two methods for assessing serum concentration of S100B. METHODS: A prospective, observational study performed in a single tertiary medical center. Included were fifty two consecutive adult patients undergoing surgery for meningioma that provided blood samples for determination of S100B concentrations. Eighty samples (40 pre-operative and 40 postoperative) were randomly selected for batch testing. Each sample was divided into two aliquots. These were analyzed by ELISA (Sangtec) and a commercial kit (Roche Elecsys(R)) for S100B concentrations. Statistical analysis included regression modelling and Bland-Altman analysis. RESULTS: A parsimonious linear model best described the prediction of commercial kit values by those determined by ELISA (y = 0.045 + 0.277*x, x = ELISA value, R2 = 0.732). ELISA measurements tended to be higher than commercial kit measurements. This discrepancy increased linearly with increasing S100B concentrations. At concentrations above 0.7 microg/L the paired measurements were consistently outside the limits of agreement in the Bland-Altman display. Similar to other studies that used alternative measurement methods, sex and age related differences in serum S100B levels were not detected using the Elecsys(R) (p = 0.643 and 0.728 respectively). CONCLUSION: Although a generally linear relationship exists between serum S100B concentrations measured by ELISA and a commercially available kit, ELISA values tended to be higher than commercial kit measurements particularly at concentrations over 0.7 microg/L, which are suggestive of brain injury. International standardization of commercial kits is required before the predictive validity of S100B for brain damage can be effectively assessed in clinical practice.

SELECTION OF CITATIONS
SEARCH DETAIL