Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
1.
Neurotox Res ; 42(1): 13, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38332435

ABSTRACT

Hypoxia plays a significant role in the development of various cerebral diseases, many of which are associated with the potential risk of recurrence due to mitochondrial damage. Conventional drug treatments are not always effective for hypoxia-related brain diseases, necessitating the exploration of alternative compounds. In this study, we investigated the potential of diphenyl diselenide [(PhSe)2] to ameliorate locomotor impairments and mitigate brain mitochondrial dysfunction in zebrafish subjected to hypoxia. Additionally, we explored whether these improvements could confer resistance to recurrent hypoxia. Through a screening process, an appropriate dose of (PhSe)2 was determined, and animals exposed to hypoxia received a single intraperitoneal injection of 100 mg/kg of the compound or vehicle. After 1 h from the injection, evaluations were conducted on locomotor deficits, (PhSe)2 content, mitochondrial electron transport system, and mitochondrial viability in the brain. The animals were subsequently exposed to recurrent hypoxia to assess the latency time to hypoxia symptoms. The findings revealed that (PhSe)2 effectively crossed the blood-brain barrier, attenuated locomotor deficits induced by hypoxia, and improved brain mitochondrial respiration by modulating complex III. Furthermore, it enhanced mitochondrial viability in the telencephalon, contributing to greater resistance to recurrent hypoxia. These results demonstrate the beneficial effects of (PhSe)2 on both hypoxia and recurrent hypoxia, with cerebral mitochondria being a critical target of its action. Considering the involvement of brain hypoxia in numerous pathologies, (PhSe)2 should be further tested to determine its effectiveness as a potential treatment for hypoxia-related brain diseases.


Subject(s)
Brain Diseases , Organoselenium Compounds , Animals , Zebrafish , Mitochondria , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Organoselenium Compounds/pharmacology , Organoselenium Compounds/therapeutic use , Hypoxia/drug therapy
2.
Neurochem Res ; 49(4): 1076-1092, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38267690

ABSTRACT

Neurotoxicity associated with chemotherapy is a debilitating side effect of cancer management in humans which reportedly involves inflammatory and oxidative stress responses. Diphenyl diselenide (DPDS) is an organoselenium compound which exhibits its anti-tumoral, anti-oxidant, anti-inflammatory and anti-mutagenic effects. Nevertheless, its possible effect on chemotherapy-induced neurotoxicity is not known. Using rat model, we probed the behavioral and biochemical effects accompanying administration of antineoplastic agent doxorubicin (7.5 mg/kg) and DPDS (5 and 10 mg/kg). Anxiogenic-like behavior, motor and locomotor insufficiencies associated with doxorubicin were considerably abated by both DPDS doses with concomitant enhancement in exploratory behavior as demonstrated by reduced heat maps intensity and enhanced track plot densities. Moreover, with exception of cerebral glutathione (GSH) level, superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities, biochemical data demonstrated reversal of doxorubicin-mediated decline in cerebral and cerebellar antioxidant status indices and the increase in acetylcholinesterase (AChE) activity by both doses of DPDS. Also, cerebellar and cerebral lipid peroxidation, hydrogen peroxide as well as reactive oxygen and nitrogen species levels were considerably diminished in rats administered doxorubicin and DPDS. In addition, DPDS administration abated myeloperoxidase activity, tumour necrosis factor alpha and nitric oxide levels along with caspase-3 activity in doxorubicin-administered rats. Chemoprotection of doxorubicin-associated neurotoxicity by DPDS was further validated by histomorphometry and histochemical staining. Taken together, DPDS through offsetting of oxido-inflammatory stress and caspase-3 activation elicited neuroprotection in doxorubicin-treated rats.


Subject(s)
Organoselenium Compounds , Temefos , Humans , Rats , Animals , Caspase 3 , Temefos/pharmacology , Acetylcholinesterase , Oxidative Stress , Antioxidants/pharmacology , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Benzene Derivatives/chemistry , Organoselenium Compounds/pharmacology , Organoselenium Compounds/therapeutic use , Glutathione/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Doxorubicin/toxicity
3.
Lancet Gastroenterol Hepatol ; 6(12): 1002-1014, 2021 12.
Article in English | MEDLINE | ID: mdl-34688373

ABSTRACT

BACKGROUND: Data are needed to inform the positioning of biologic therapy in the treatment of moderate-to-severe Crohn's disease, both first line and after previous biologic exposure. We aimed to assess the comparative efficacy and safety of biologics in patients with Crohn's disease. METHODS: We did a systematic review and network meta-analysis of phase 2 and phase 3 randomised controlled trials done in adults (≥18 years) with moderate-to-severe Crohn's disease (Crohn's Disease Activity Index [CDAI] 220-450) treated with tumour necrosis factor (TNF) antagonists, anti-integrin, anti-interleukin (IL)-12 and IL-23p40, or anti-IL23p19 agents, either alone or in combination with immunosuppressants, as their first-line biologic or after previous biologic exposure, compared with placebo or an active comparator. The minimum duration of therapy was 14 days for trials reporting induction of remission in active disease and 22 weeks in trials reporting maintenance of remission. We searched Medline, EMBASE, the Cochrane CENTRAL Register of Controlled Trials, conference proceedings, trial registries, and unpublished data from inception to June 3, 2021, without any language restrictions. Summary estimates of the primary and secondary outcomes were extracted from the published reports; individual patient-level data were not sought. The primary endpoint was induction of clinical remission in patients with active disease (CDAI <150) and maintenance of remission in patients with response to induction therapy, with data extracted from published reports. A network meta-analysis with multivariate consistency model random-effects meta-regression was done, with rankings based on surface under the cumulative ranking curve (SUCRA) values. FINDINGS: The search strategy yielded 18 382 citations, of which 31 trials were eligible for inclusion. On the basis of 15 randomised controlled trials including 2931 biologic-naive patients, infliximab monotherapy (odds ratio [OR] 4·53 [95% CI 1·49-13·79]), infliximab combined with azathioprine (7·49 [2·04-27·49]), adalimumab (3·01 [1·25-7·27]), and ustekinumab (2·63 [1·10-6·28]) were associated with significantly higher odds of inducing remission compared to certolizumab pegol (all moderate confidence); infliximab and azathioprine combination therapy was also associated with significantly higher odds of inducing remission than vedolizumab (3·76 [1·01-14·03]; low confidence). On the basis of ten randomised controlled trials including 2479 patients with previous biologic exposure, adalimumab after loss of response to infliximab (OR 2·82 [95% CI 1·20-6·62]; low confidence), and risankizumab (2·10 [1·12-3·92]; moderate confidence), were associated with higher odds of inducing remission than vedolizumab. No differences between active interventions were observed in maintenance trials. Most trials were at low or uncertain risk of bias. INTERPRETATION: Although biologic treatment choices in patients with moderate-to-severe Crohn's disease must be individualised for each patient, this analysis suggests that either infliximab with azathioprine or adalimumab might be preferred as a first-line therapy, and adalimumab (after infliximab loss of response) or risankizumab might be preferred as a second-line therapy, for induction of clinical remission. FUNDING: None.


Subject(s)
Biological Therapy/adverse effects , Crohn Disease/drug therapy , Drug Therapy, Combination/adverse effects , Placebos/administration & dosage , Adalimumab/administration & dosage , Adalimumab/therapeutic use , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Azathioprine/administration & dosage , Azathioprine/therapeutic use , Benzene Derivatives/administration & dosage , Benzene Derivatives/therapeutic use , Biological Therapy/methods , Carboxylic Acids/administration & dosage , Carboxylic Acids/therapeutic use , Case-Control Studies , Drug Therapy, Combination/methods , Female , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Infliximab/administration & dosage , Infliximab/therapeutic use , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-23 Subunit p19/antagonists & inhibitors , Male , Network Meta-Analysis , Randomized Controlled Trials as Topic , Remission Induction , Safety , Severity of Illness Index , Treatment Outcome , Tumor Necrosis Factor Inhibitors/administration & dosage , Tumor Necrosis Factor Inhibitors/therapeutic use , Ustekinumab/administration & dosage , Ustekinumab/therapeutic use
4.
Acta Pharmacol Sin ; 42(12): 1991-2003, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34531546

ABSTRACT

We have previously shown that roflupram (ROF) protects against MPP+-induced neuronal damage in models of Parkinson's disease (PD). Since impaired degradation of α-synuclein (α-syn) is one of the key factors that lead to PD, here we investigated whether and how ROF affects the degradation of α-syn in rotenone (ROT)-induced PD models in vivo and in vitro. We showed that pretreatment with ROF (10 µM) significantly attenuated cell apoptosis and reduced the level of α-syn in ROT-treated SH-SY5Y cells. Furthermore, ROF significantly enhanced the lysosomal function, as evidenced by the increased levels of mature cathepsin D (CTSD) and lysosomal-associated membrane protein 1 (LAMP1) through increasing NAD+/NADH and the expression of sirtuin 1 (SIRT1). Pretreatment with an SIRT1 inhibitor selisistat (SELI, 10 µM) attenuated the neuroprotection of ROF, ROF-reduced expression of α-syn, and ROF-increased expression levels of LAMP1 and mature CTSD. Moreover, inhibition of CTSD by pepstatin A (20 µM) attenuated ROF-reduced expression of α-syn. In vivo study was conducted in mice exposed to ROT (10 mg·kg-1·d-1, i.g.) for 6 weeks; then, ROT-treated mice received ROF (0.5, 1, or 2 mg·kg-1·d-1; i.g.) for four weeks. ROF significantly ameliorated motor deficits, which was accompanied by increased expression levels of tyrosine hydroxylase, SIRT1, mature CTSD, and LAMP1, and a reduced level of α-syn in the substantia nigra pars compacta. Taken together, these results demonstrate that ROF exerts a neuroprotective action and reduces the α-syn level in PD models. The mechanisms underlying ROF neuroprotective effects appear to be associated with NAD+/SIRT1-dependent activation of lysosomal function.


Subject(s)
Benzene Derivatives/therapeutic use , Furans/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Rotenone/toxicity , alpha-Synuclein/metabolism , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Benzene Derivatives/pharmacology , Cathepsin D/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Furans/pharmacology , Humans , Lysosomes/drug effects , Male , Mice, Inbred C57BL , Movement/drug effects , Neuroprotective Agents/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Sirtuin 1/metabolism
5.
J Ethnopharmacol ; 279: 114235, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34044081

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Trichodesma indicum (L.) R. Br. (family: Boraginaceae) is a medicinal herb largely used to treat arthralgia, rheumatoid arthritis, wound healing, dysentery, etc. It's mechanism of anti-inflammatory activity has not been systematically analyzed yet. AIM OF THE STUDY: The present study was undertaken to examine the anti-inflammatory effects of successive solvent extracts (n-hexane extract (HE), ethyl acetate extract (EA), ethanol extract (EE), aqueous extract (AE) and fractions of HE) from the aerial parts of Trichodesma indicum (TI) against lipopolysaccharide (LPS) stimulated inflammatory reaction using mouse macrophage RAW 264.7 cells. MATERIALS AND METHODS: Cytotoxic effects of the extracts and fractions of TI were assessed by MTT assay. The effect of extracts and fractions on the production of nitric oxide (NO) in RAW 264.7 macrophages were measured using the Griess reagent method. IL - 6, IL - 1ß, TNF-α, iNOS and COX-2 gene expressions were examined by a qRT-PCR method. RESULTS: RAW 264.7 macrophages pretreated with HE, EA, EE and AE of TI showed a significant decrease in the production of proinflammatory cytokines and NO without exhibiting cytotoxicity. The potent HE was fractionated using flash chromatography into FA, FB, FC, FD and FE. Among the five fractions, FE displayed a stronger ability to reduce IL - 1ß, TNF-α, iNOS, COX2 and NO importantly no cytotoxicity was observed. The phytochemical compounds present in FE were further screened by Gas chromatography - Mass spectroscopy (GC-MS). GC-MS analysis revealed that 1,2-benzenedicarboxylic acid diisooctyl ester is the major compound in FE. Molecular docking analysis showed good inhibition of 1,2-benzenedicarboxylic acid diisooctyl ester against TLR-4, NIK and TACE. CONCLUSION: Our results suggested that 1,2-benzenedicarboxylic acid diisooctyl ester could be a potential candidate in alleviating inflammatory reactions in TI.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzene Derivatives/pharmacology , Boraginaceae/chemistry , Carboxylic Acids/pharmacology , Esters/pharmacology , Inflammation/drug therapy , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Benzene Derivatives/isolation & purification , Benzene Derivatives/therapeutic use , Carboxylic Acids/isolation & purification , Carboxylic Acids/therapeutic use , Cytokines/metabolism , Esters/isolation & purification , Esters/therapeutic use , Gas Chromatography-Mass Spectrometry , Inflammation/pathology , Lipopolysaccharides , Macrophages/drug effects , Mice , Molecular Docking Simulation , Nitric Oxide/metabolism , RAW 264.7 Cells
6.
J Med Chem ; 64(8): 4762-4786, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33835811

ABSTRACT

A benzo[6]annulene, 4-(tert-butyl)-N-(3-methoxy-5,6,7,8-tetrahydronaphthalen-2-yl) benzamide (1a), was identified as an inhibitor against Chikungunya virus (CHIKV) with antiviral activity EC90 = 1.45 µM and viral titer reduction (VTR) of 2.5 log at 10 µM with no observed cytotoxicity (CC50 = 169 µM) in normal human dermal fibroblast cells. Chemistry efforts to improve potency, efficacy, and drug-like properties of 1a resulted in a novel lead compound 8q, which possessed excellent cellular antiviral activity (EC90 = 270 nM and VTR of 4.5 log at 10 µM) and improved liver microsomal stability. CHIKV resistance to an analog of 1a, compound 1c, tracked to a mutation in the nsP3 macrodomain. Further mechanism of action studies showed compounds working through inhibition of human dihydroorotate dehydrogenase in addition to CHIKV nsP3 macrodomain. Moderate efficacy was observed in an in vivo CHIKV challenge mouse model for compound 8q as viral replication was rescued from the pyrimidine salvage pathway.


Subject(s)
Antiviral Agents/pharmacology , Benzene Derivatives/chemistry , Chikungunya virus/physiology , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Binding Sites , Cell Line , Cell Survival/drug effects , Chikungunya Fever/drug therapy , Dihydroorotate Dehydrogenase , Disease Models, Animal , Female , Half-Life , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Structure-Activity Relationship
7.
Chem Biol Interact ; 338: 109427, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33639173

ABSTRACT

Oxidative stress and inflammation are implicated in the occurrence and progression of diabetic nephropathy (DN). Diphenyl diselenide (DPDS) is a stable and simple diaryl diselenide with anti-hyperglycemic, anti-inflammatory, and antioxidant activities. However, the effects of DPDS on DN are still unclear to date. Herein, we aimed to explore whether DPDS could improve renal dysfunction in streptozotocin (STZ)-induced diabetic rats and its underlying mechanisms. STZ-induced DN rats were administered with DPDS (5 or 15 mg/kg) or metformin (200 mg/kg) once daily by intragastric gavage for 12 weeks. DPDS supplementation significantly improved hyperglycemia, glucose intolerance, dyslipidemia, and the renal pathological abnormalities, concurrent with significantly reduced serum levels of creatinine, urea nitrogen, urine volume, and urinary levels of micro-albumin, ß2-microglobulin and N-acetyl-glucosaminidase activities. Moreover, DPDS effectively promoted the activities of antioxidant enzymes, and reduced the levels of MDA and pro-inflammatory factors in serum and the kidney. Furthermore, DPDS supplementation activated the renal Nrf2/Keap1 signaling pathway, but attenuated the high phosphorylation levels of NFκB, JNK, p38 and ERK1/2. Altogether, the current study indicated for the first time that DPDS ameliorated STZ-induced renal dysfunction in rats, and its mechanism of action may be attributable to suppressing oxidative stress via activating the renal Nrf2/Keap1 signaling pathway and mitigating inflammation by suppressing the renal NFκB/MAPK signaling pathways, suggesting a potential therapeutic approach for DN.


Subject(s)
Benzene Derivatives/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/drug therapy , Inflammation/drug therapy , Organoselenium Compounds/therapeutic use , Oxidative Stress , Animals , Antioxidants/metabolism , Benzene Derivatives/pharmacology , Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Diabetic Nephropathies/complications , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Dyslipidemias/complications , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Gene Expression Regulation/drug effects , Glucose/metabolism , Inflammation/complications , Inflammation/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Kidney/pathology , Kidney/physiopathology , Lipid Metabolism/drug effects , MAP Kinase Signaling System/drug effects , Male , Models, Biological , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Streptozocin
8.
Acta Pharmacol Sin ; 42(11): 1769-1779, 2021 11.
Article in English | MEDLINE | ID: mdl-33627802

ABSTRACT

NOD-like receptor (NLR) family pyrin domain-containing-3 (NLRP3) inflammasome is implicated in inflammation-associated diseases such as multiple sclerosis, Parkinson's disease, and stroke. Targeting the NLRP3 inflammasome is beneficial to these diseases, but few NLRP3 inflammasome-selective inhibitors are identified to date. Essential oils (EOs) are liquid mixtures of volatile and low molecular-weight organic compounds extracted from aromatic plants, which show various pharmacological activities, including antibacterial, antifungal, antiviral, antioxidant, and anti-inflammatory properties. In this study we screened active ingredients from essential oils, and identified 1,2,4-trimethoxybenzene (1,2,4-TTB) as a selective NLRP3 inflammasome inhibitor. We showed that 1,2,4-TTB (1 mM) markedly suppressed nigericin- or ATP-induced NLRP3 inflammasome activation, thus decreased caspase-1 activation and IL-1ß secretion in immortalized murine bone marrow-derived macrophages (iBMDMs) and in primary mouse microglia. Moreover, 1,2,4-TTB specifically inhibited the activation of NLRP3 inflammasome without affecting absent in melanoma 2 (AIM2) inflammasome activation. We further demonstrated that 1,2,4-TTB inhibited oligomerization of the apoptosis-associated speck-like protein containing a CARD (ASC) and protein-protein interaction between NLRP3 and ASC, thus blocking NLRP3 inflammasome assembly in iBMDMs and in primary mouse macrophages. In mice with experimental autoimmune encephalomyelitis (EAE), administration of 1,2,4-TTB (200 mg · kg-1 · d-1, i.g. for 17 days) significantly ameliorated EAE progression and demyelination. In conclusion, our results demonstrate that 1,2,4-TTB is an NLRP3 inflammasome inhibitor and attenuates the clinical symptom and inflammation of EAE, suggesting that 1,2,4-TTB is a potential candidate compound for treating NLRP3 inflammasome-driven diseases, such as multiple sclerosis.


Subject(s)
Benzene Derivatives/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Benzene Derivatives/pharmacology , Cell Line, Transformed , Female , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Biochem Pharmacol ; 186: 114430, 2021 04.
Article in English | MEDLINE | ID: mdl-33556338

ABSTRACT

Colorectal cancer (CRC) is one of the most malignant cancers in the world. A major cause of death in CRC patients is the limited therapeutic options in its advanced stages. The Farnesoid X receptor (FXR) is a member of the nuclear superfamily, which is effective in slowing the progression of colorectal cancer in addition to its extraordinary role in regulating metabolic disorders. Due to the systemic side-effects caused by non-selective agonists, the intestine-restricted FXR agonists can induce a whole-body benefit without activating the hepatic FXR, suggesting intestinal FXR activation as a potentially safer therapy in the treatment of CRC. This review highlights the effects of FXR on the disturbed bile acid circulation and the carcinogenesis of CRC and with a specific emphasis on listing the functions of several intestinal-restricted FXR agonists.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Intestinal Mucosa/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Antineoplastic Agents/pharmacology , Azetidines/pharmacology , Azetidines/therapeutic use , Benzene Derivatives/pharmacology , Benzene Derivatives/therapeutic use , Bile Acids and Salts/metabolism , Colorectal Neoplasms/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Isonicotinic Acids/pharmacology , Isonicotinic Acids/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism
10.
Pharmacol Res ; 165: 105457, 2021 03.
Article in English | MEDLINE | ID: mdl-33515706

ABSTRACT

Microglia-mediated neuroinflammatory response and neuron damage are considered as a self-propelling progressive cycle, being strongly implicated in the progression of neurodegeneration in amyotrophic lateral sclerosis (ALS). Diphenyl diselenide (DPDS), a simple organoselenium compound, has been known to possess multiple pharmacological properties. The purpose of this study was to explore the neuroprotective effects of DPDS against microglia-mediated neuroinflammatory injury in ALS models. We found that DPDS pretreatment inhibited LPS-induced activation of IκB/NF-κB pathway and subsequent release of proinflammatory factors from activated primary hSOD1G93A microglia. Moreover, DPDS suppressed NLRP3 inflammasome activation by decreasing protein nitration via reduction in NO and ROS levels, whose low levels are related to NF-κB inhibition responsible for iNOS and NOX2 down-regulations, respectively. Notably, DPDS-mediated ROS attenuation was not linked to Nrf2 activation in this cellular model. Furthermore, in the absence of activated microglia, DPDS has no significant effect on the individual hSOD1G93A-NSC34 cells; however, in in vitro neuron-microglia conditional culture and co-culture experiments, DPDS protected motor neurons from neurotoxic damage caused by LPS or BzATP-stimulated microglia activation. Above observations suggest that DPDS-afforded neuroprotection is linked to inhibition of microglia-mediated neuroinflammation in ALS, which was further verified in vivo as shown by improvements of motor deficits, prolonged survival, and reduction of motor neuron loss and reactive microgliosis in hSOD1G93A transgenic mouse. Altogether, our results show that DPDS elicited neuroprotection in ALS models through inactivation of microglia by inhibiting IκB/NF-κB pathway and NLRP3 inflammasome activation, suggesting that DPDS may be a promising candidate for potential therapy for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Benzene Derivatives/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Microglia/drug effects , Motor Neurons/drug effects , Neuroprotective Agents/therapeutic use , Organoselenium Compounds/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Benzene Derivatives/pharmacology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Transgenic , Microglia/metabolism , Motor Neurons/metabolism , Neuroprotective Agents/pharmacology , Organoselenium Compounds/pharmacology , Superoxide Dismutase-1/genetics
11.
Res Vet Sci ; 134: 78-85, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33338952

ABSTRACT

Bovine alphaherpesvirus 2 (BoHV-2) - the agent of bovine herpetic mamillitis (BHM) - is related to Human alphaherpesviruses 1 and 2 (HHV-1, HHV-2) and, as such, has been proposed as a model for vaccine and drug testing. We herein investigated the anti-viral activity in vitro against BoHV-2 of three anti-herpetic drugs: Cidofovir (CDV), Fanciclovir (FAM), Foscarnet (PFA), and diphenyl disselenide (Ph2Se2), a compound that has showed activity against HHV-2. Plaque reduction assays (PRA) revealed a significant reduction in viral plaques (p < 0.05) in cells treated with Ph2Se2 (79.7% reduction) or CDV (62.8%). FAM treatment resulted in a slight decrease in plaque number (22.9%, p < 0.05); PFA showed no activity. The effects of Ph2Se2 and CDV, alone or in combination, were investigated in ewes inoculated with BoHV-2 transdermally and submitted to daily topic treatment. Virus inoculated ewes developed lesions progressing through the stages of hyperemia, large papules or depressed dark areas, followed by scab formation. Treatment with Ph2Se2 resulted in reduction in clinical score from day 10 pi onwards (P < 0.05), shortening of clinical course and reduction in duration of virus shedding (P < 0.05) compared to untreated controls. Combined treatment (Ph2Se2 + CDV) and CDV alone, also led to clinical improvement (P < 0.05), yet less pronounced and delayed. These results are promising towards the use of Ph2Se2, alone or in combination with anti-herpetic drugs, in the treatment of udder and teat lesions produced by BoHV-2 in dairy cows.


Subject(s)
Antiviral Agents/pharmacology , Benzene Derivatives/pharmacology , Cidofovir/pharmacology , Herpes Simplex/veterinary , Herpesvirus 2, Bovine/drug effects , Organoselenium Compounds/pharmacology , Sheep Diseases/drug therapy , Animals , Antiviral Agents/therapeutic use , Benzene Derivatives/therapeutic use , Cidofovir/therapeutic use , Female , Herpes Simplex/drug therapy , Mammary Glands, Animal/immunology , Mammary Glands, Animal/virology , Organoselenium Compounds/therapeutic use , Sheep , Sheep Diseases/virology , Virus Shedding/drug effects
12.
Biochem Pharmacol ; 182: 114221, 2020 12.
Article in English | MEDLINE | ID: mdl-32949583

ABSTRACT

Diabetic peripheral neuropathy (DPN) is one of the most common microvascular complications occurring in both type 1 and type 2 diabetes mellitus patients. Oxidative stress (OS) plays a key role in the pathogenesis of DPN; thus, antioxidant therapy is considered a promising strategy for treating DPN. Diphenyl diselenide (DPDs) is an organic selenium compound with antioxidant pharmacological activities. This study aimed to evaluate its preventive and therapeutic effects on DPN in rats with streptozotocin (STZ)-induced diabetes and explore the underlying mechanisms. In vitro, RSC96 cells were exposed to high glucose (100 mM) and then treated with different concentrations of DPDs (1, 10, 25 and 50 µM). Notably, DPDs markedly suppressed high glucose-induced cytotoxicity and oxidative stress in Schwann cells by decreasing reactive oxygen species (ROS) and malondialdehyde (MDA) levels. Furthermore, the DPDs treatment effectively activated Nrf2 signaling and inhibited Keap1 expression. An in vivo DPN model was established in Sprague-Dawley (SD) rats injected with STZ (60 mg·kg-1, ip) and orally administered either different doses of DPDs (5 and 15 mg· kg-1· d-1) for 12 weeks or alpha lipoic acid (ALA, 100 mg kg-1·d-1) as a positive control. The administration of DPDs significantly increased the motor nerve conduction velocity (MNCV), improved thermal and mechanical hyperalgesia and the sciatic nerve morphology, and ameliorated oxidative stress in the serum and the sciatic nerve of rats with DPN. Mechanistically, DPDs reduced the level of Keap1 and stimulated Nrf2 signaling in the sciatic nerve. Taken together, the results of this study indicate that DPDs ameliorates experimental DPN as an antioxidant by activating the Nrf2/Keap1 signaling pathway. DPDs may represent a new alternative treatment for DPN.


Subject(s)
Benzene Derivatives/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Organoselenium Compounds/therapeutic use , Oxidative Stress/drug effects , Animals , Benzene Derivatives/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Kelch-Like ECH-Associated Protein 1/metabolism , Male , NF-E2-Related Factor 2/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
13.
Neuropharmacology ; 176: 108241, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32712273

ABSTRACT

Cocaine abuse remains a public health threat around the world. There are no pharmacological treatments approved for cocaine use disorder. Cannabis has received growing attention as a treatment for many conditions, including addiction. Most cannabis-based medication development has focused on cannabinoid CB1 receptor (CB1R) antagonists (and also inverse agonists) such as rimonabant, but clinical trials with rimonabant have failed due to its significant side-effects. Here we sought to determine whether a novel and selective CB2R inverse agonist, Xie2-64, has similar therapeutic potential for cocaine use disorder. Computational modeling indicated that Xie2-64 binds to CB2R in a way similar to SR144528, another well-characterized but less selective CB2R antagonist/inverse agonist, suggesting that Xie2-64 may also have CB2R antagonist profiles. Unexpectedly, systemic administration of Xie2-64 or SR144528 dose-dependently inhibited intravenous cocaine self-administration and shifted cocaine dose-response curves downward in rats and wild-type, but not in CB2R-knockout, mice. Xie2-64 also dose-dependently attenuated cocaine-enhanced brain-stimulation reward maintained by optical stimulation of ventral tegmental area dopamine (DA) neurons in DAT-Cre mice, while Xie2-64 or SR144528 alone inhibited optical brain-stimulation reward. In vivo microdialysis revealed that systemic or local administration of Xie2-64 into the nucleus accumbens reduced extracellular dopamine levels in a dose-dependent manner in rats. Together, these results suggest that Xie2-64 has significant anti-cocaine reward effects likely through a dopamine-dependent mechanism, and therefore, deserves further study as a new pharmacotherapy for cocaine use disorder.


Subject(s)
Benzene Derivatives/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Cocaine-Related Disorders/prevention & control , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Drug Inverse Agonism , Receptor, Cannabinoid, CB2/agonists , Sulfonamides/therapeutic use , Animals , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , Cannabinoid Receptor Agonists/chemistry , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Dose-Response Relationship, Drug , Male , Mice , Mice, Knockout , Protein Structure, Secondary , Rats , Rats, Long-Evans , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Rodentia , Self Administration , Sulfonamides/chemistry , Sulfonamides/pharmacology
14.
Eur J Pharmacol ; 879: 173156, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32360838

ABSTRACT

Humans are ubiquitously exposed to bisphenol A (BPA), one of the most used synthetic monomers for manufacturing polycarbonate plastics. BPA exposure leads to abnormal nociceptive perception and neuroinflammation in rodents. This study investigated whether diphenyl diselenide (PhSe)2, a pleiotropic selenium-containing molecule, would be effective against the hypernociceptive behavior induced by the early-life BPA exposure to mice. Three-week-old male and female Swiss mice received intragastrically BPA (5 mg/kg) from 21st to 60th postnatal day. After, the mice received by the intragastric route (PhSe)2 (1 mg/kg) once a day for seven days. After the last day of treatment, the mice performed the hot plate and tail immersion tests. The cerebral cortex samples were used to determine the levels of proteins related to apoptosis and inflammation. The results demonstrated that females were more susceptible than male mice to thermal hypernociception induced by early-life exposure to BPA. (PhSe)2 was effective against the reduction in the latency to paw and tail withdrawal induced by BPA exposure in female mice. Furthermore, (PhSe)2 restored the impairment in the levels of inflammatory proteins (COX-2, IL-1ß, and p-JNK/JNK) but not those of apoptosis in the cerebral cortex of female mice exposed to BPA. Collectively, these data showed that females were more susceptible to thermal hypernociceptive behavior induced by early-life exposure to BPA than male mice. The administration of (PhSe)2 reduced thermal hypernociceptive behavior, a sex independent effect, in BPA-exposed mice. (PhSe)2 modulated inflammatory protein levels in the cerebral cortex of female mice exposed to BPA.


Subject(s)
Benzene Derivatives/therapeutic use , Benzhydryl Compounds/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Organoselenium Compounds/therapeutic use , Phenols/toxicity , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , Hot Temperature/adverse effects , Male , Mice , NF-kappa B/metabolism
15.
Bioorg Med Chem Lett ; 30(2): 126854, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31859157

ABSTRACT

NIR-light-absorbing photosensitizers with the capability of selective localization and activation in tumor regions are of great importance for practical photodynamic therapy (PDT). Here, selenophenol substituted BODIPYs were designed and synthesized as new photosensitizers for PDT. One of these obtained BODIPYs, IBSeOV, possesses an intense and low energy absorption with a high singlet oxygen quantum yield (ΦΔ = 60%). Considering manganese dioxide (MnO2) nanosheets as versatile nanocarriers in cancer theranostics, nanosystem IBSeOV/MnO2 was then fabricated to furnish tumor environment selective activation. Such designed nanoplatform allowed for GSH-controllable 1O2 production and exhibited low cytotoxicity in dark but good photocytotoxicity to cancer cells. The in vivo antitumor outcome suggested the high treatment efficiency of IBSeOV/MnO2 for tumor therapy.


Subject(s)
Benzene Derivatives/therapeutic use , Organoselenium Compounds/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Benzene Derivatives/pharmacology , Humans , Nanostructures , Organoselenium Compounds/pharmacology , Photosensitizing Agents/pharmacology , Tumor Microenvironment
16.
J Immunol ; 203(12): 3407-3415, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31694910

ABSTRACT

Inflammatory bowel disease (IBD) is a complex autoimmune disease with dysfunction in pattern-recognition responses, including within the NLR family. Nucleotide-binding oligomerization domain, leucine rich repeat containing X1 (NLRX1) is a unique NLR with regulatory and anti-inflammatory functions resulting in protection from IBD in mouse models. NX-13 is an orally active, gut-restricted novel drug candidate that selectively targets and activates the NLRX1 pathway locally in the gut. In vitro and in vivo efficacy of NLRX1 activation by NX-13 was examined. Oral treatment with NX-13 alleviates disease severity, colonic leukocytic infiltration, and cytokine markers of inflammation in three mouse models of IBD (dextran sulfate sodium, Mdr1a-/-, and CD45RBhi adoptive transfer). Treatment of naive CD4+ T cells with NX-13 in vitro decreases differentiation into Th1 and Th17 subsets with increased oxidative phosphorylation and decreased NF-κB activation and reactive oxygen species. With stimulation by PMA/ionomycin, TNF-α, or H2O2, PBMCs from ulcerative colitis patients treated with NX-13 had decreased NF-κB activity, TNF-α+ and IFN-γ+ CD4+ T cells and overall production of IL-6, MCP1, and IL-8. NX-13 activates NLRX1 to mediate a resistance to both inflammatory signaling and oxidative stress in mouse models and human primary cells from ulcerative colitis patients with effects on NF-κB activity and oxidative phosphorylation. NX-13 is a promising oral, gut-restricted NLRX1 agonist for treating IBD.


Subject(s)
Benzene Derivatives/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Mitochondrial Proteins/metabolism , Pyridines/therapeutic use , Signal Transduction/drug effects , Administration, Oral , Animals , Benzene Derivatives/administration & dosage , Benzene Derivatives/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/agonists , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Reactive Oxygen Species/metabolism
17.
Oxid Med Cell Longev ; 2019: 2510936, 2019.
Article in English | MEDLINE | ID: mdl-31772702

ABSTRACT

Tellurium is a rare element that has been regarded as a toxic, nonessential element, and its biological role is not clearly established. In addition, the biological effects of elemental tellurium and some of its organic and inorganic derivatives have been studied, leading to a set of interesting and promising applications. Diphenyl ditelluride (DPDT), an organic tellurium derivate, showed antioxidant, antigenotoxic, antimutagenic, and anticancer properties. The antioxidant and prooxidant properties of DPDT are complex and depend on experimental conditions, which may explain the contradictory reports of these properties. In addition, DPDT may exert its effects through different pathways, including distinct ones to those responsible for chemotherapy resistance phenotypes: transcription factors, membrane receptors, adhesion, structural molecules, cell cycle regulatory components, and apoptosis pathways. This review aims to present recent advances in our understanding of the biological effects, therapeutic potential, and safety of DPDT treatment. Moreover, original results demonstrating the cytotoxic effects of DPDT in different mammalian cell lines and systems biology analysis are included, and emerging approaches for possible future applications are inferred.


Subject(s)
Antioxidants/therapeutic use , Benzene Derivatives/therapeutic use , Organometallic Compounds/therapeutic use , Tellurium/chemistry , Antioxidants/pharmacology , Benzene Derivatives/pharmacology , Humans , Organometallic Compounds/pharmacology , Oxidation-Reduction
18.
Toxins (Basel) ; 11(9)2019 09 18.
Article in English | MEDLINE | ID: mdl-31540492

ABSTRACT

Acid-sensing ion channels (ASICs), which are present in almost all types of neurons, play an important role in physiological and pathological processes. The ASIC1a subtype is the most sensitive channel to the medium's acidification, and it plays an important role in the excitation of neurons in the central nervous system. Ligands of the ASIC1a channel are of great interest, both fundamentally and pharmaceutically. Using a two-electrode voltage-clamp electrophysiological approach, we characterized lindoldhamine (a bisbenzylisoquinoline alkaloid extracted from the leaves of Laurus nobilis L.) as a novel inhibitor of the ASIC1a channel. Lindoldhamine significantly inhibited the ASIC1a channel's response to physiologically-relevant stimuli of pH 6.5-6.85 with IC50 range 150-9 µM, but produced only partial inhibition of that response to more acidic stimuli. In mice, the intravenous administration of lindoldhamine at a dose of 1 mg/kg significantly reversed complete Freund's adjuvant-induced thermal hyperalgesia and inflammation; however, this administration did not affect the pain response to an intraperitoneal injection of acetic acid (which correlated well with the function of ASIC1a in the peripheral nervous system). Thus, we describe lindoldhamine as a novel antagonist of the ASIC1a channel that could provide new approaches to drug design and structural studies regarding the determinants of ASIC1a activation.


Subject(s)
Acid Sensing Ion Channel Blockers/therapeutic use , Acid Sensing Ion Channels/physiology , Anti-Inflammatory Agents/therapeutic use , Benzene Derivatives/therapeutic use , Quinolines/therapeutic use , Acetic Acid , Acid Sensing Ion Channel Blockers/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Benzene Derivatives/pharmacology , Female , Freund's Adjuvant , Hot Temperature/adverse effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Inflammation/chemically induced , Inflammation/drug therapy , Male , Mice , Oocytes/physiology , Pain/chemically induced , Pain/drug therapy , Quinolines/pharmacology , Xenopus laevis
19.
Mini Rev Med Chem ; 19(18): 1544-1557, 2019.
Article in English | MEDLINE | ID: mdl-31362687

ABSTRACT

Soluble Guanylate Cyclase (sGC) is the intracellular receptor of Nitric Oxide (NO). The activation of sGC results in the conversion of Guanosine Triphosphate (GTP) to the secondary messenger cyclic Guanosine Monophosphate (cGMP). cGMP modulates a series of downstream cascades through activating a variety of effectors, such as Phosphodiesterase (PDE), Protein Kinase G (PKG) and Cyclic Nucleotide-Gated Ion Channels (CNG). NO-sGC-cGMP pathway plays significant roles in various physiological processes, including platelet aggregation, smooth muscle relaxation and neurotransmitter delivery. With the approval of an sGC stimulator Riociguat for the treatment of Pulmonary Arterial Hypertension (PAH), the enthusiasm in the discovery of sGC modulators continues for broad clinical applications. Notably, through activating the NO-sGC-cGMP pathway, sGC stimulator and activator potentiate for the treatment of various diseases, such as PAH, Heart Failure (HF), Diabetic Nephropathy (DN), Systemic Sclerosis (SS), fibrosis as well as other diseases including Sickle Cell Disease (SCD) and Central Nervous System (CNS) disease. Here, we review the preclinical and clinical studies of sGC stimulator and activator in recent years and prospect for the development of sGC modulators in the near future.


Subject(s)
Enzyme Activators/chemistry , Guanylate Cyclase/metabolism , Benzene Derivatives/chemistry , Benzene Derivatives/metabolism , Benzene Derivatives/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cyclic GMP/metabolism , Enzyme Activators/metabolism , Enzyme Activators/therapeutic use , Guanylate Cyclase/chemistry , Humans , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Nitric Oxide/metabolism
20.
Mini Rev Med Chem ; 19(18): 1531-1543, 2019.
Article in English | MEDLINE | ID: mdl-31288716

ABSTRACT

The search for inhibitors of the Janus kinase family (JAK1, JAK2, JAK3 and TYK2) has been ongoing for several decades and has resulted in a number of JAK inhibitors being approved for use in patients, such as tofacitinib for the treatment of autoimmune diseases such as Rheumatoid Arthritis (RA). Although initially thought to be a JAK3 selective inhibitor, tofacitinib was subsequently found to possess significant activity to inhibit JAK1 and JAK2 which has contributed to some adverse side effects. A selective JAK3 inhibitor should only have an effect within the immune system since JAK3 is solely expressed in lymphoid tissue; this makes JAK3 a target of interest in the search for treatments of autoimmune diseases. A method to obtain selectivity for JAK3 over the other JAK family members, which has attracted more scientific attention recently, is the targeting of the active site cysteine residue, unique in JAK3 within the JAK family, with compounds containing electrophilic warheads which can form a covalent bond with the nucleophilic thiol of the cysteine residue. This review encompasses the historical search for a covalent JAK3 inhibitor and the most recently published research which hasn't been reviewed to date. The most important compounds from the publications reviewed the activity and selectivity of these compounds together with some of the more important biological results are condensed in to an easily digested form that should prove useful for those interested in the field.


Subject(s)
Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Amides/chemistry , Amides/metabolism , Amides/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Benzene Derivatives/chemistry , Benzene Derivatives/metabolism , Benzene Derivatives/therapeutic use , Drug Evaluation, Preclinical , Humans , Janus Kinase 3/metabolism , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...