Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 301
1.
J Huntingtons Dis ; 13(1): 55-66, 2024.
Article En | MEDLINE | ID: mdl-38489193

Background: Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric dysfunction caused by a mutant huntingtin protein. Compromised metabolic activity resulting from systemic administration of the mitochondrial toxin, 3-nitropropionic acid (3-NP), is known to mimic the pathology of HD and induce HD-like symptoms in rats. N-hexanoic-Tyr-Ile-(6)-amino hexanoic amide (PNB-0408), also known as Dihexa, has been shown to have neuroprotective and procognitive properties in animal models of Alzheimer's and Parkinson's diseases. Given the mechanism of action and success in other neurodegenerative diseases, we felt it an appropriate compound to investigate further for HD. Objective: The present study was designed to test if PNB-0408, an angiotensin IV analog, could attenuate 3-NP-induced HD-like symptoms in rats and serve as a potential therapeutic agent. Methods: Forty male Wistar rats were randomized into three groups consisting of a "vehicle" group, a "3-NP" group, and a "3-NP + PNB-0408" group. PNB-0408 was administered along with chronic exposure to 3-NP. Animal body weight, motor function, and cognitive abilities were measured for five weeks, before euthanasia and histopathological analysis. Results: Exposure to 3-NP decreased the amount of weight rats gained, impaired spatial learning and memory consolidation, and led to marked motor dysfunction. From our observations and analysis, PNB-0408 did not protect rats from the deficits induced by 3-NP neurotoxicity. Conclusions: Our findings suggest that PNB-0408 may not be an efficacious treatment strategy for preventing 3-NP-induced HD-like symptoms in a preclinical model. These data highlight the need for further research of this compound in alternate models and/or alternative approaches to managing this disorder.


Angiotensin II/analogs & derivatives , Huntington Disease , Neuroprotective Agents , Rats , Male , Animals , Rats, Wistar , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitro Compounds/toxicity , Nitro Compounds/therapeutic use , Propionates/toxicity , Propionates/therapeutic use , Disease Models, Animal
2.
Neurochem Res ; 49(4): 1017-1033, 2024 Apr.
Article En | MEDLINE | ID: mdl-38184805

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder characterized by motor, psychiatric and cognitive symptoms. Injection of 3-nitropropionic acid (3-NP) is a widely used experimental model for induction of HD. The current study aimed to inspect the potential neuroprotective properties of azilsartan (Azil), an angiotensin II type 1 receptor blocker (ATR1), in 3-NP-induced striatal neurotoxicity in rats. Rats were randomly allocated into five groups and treated for 14 days as follows: group I received normal saline; group II received Azil (10 mg/kg, p.o.); group III received 3-NP (10 mg/kg, i.p); group IV and V received Azil (5 or 10 mg/kg, p.o, respectively) 1 h prior to 3-NP injection. Both doses of Azil markedly attenuated motor and behavioural dysfunction as well as striatal histopathological alterations caused by 3-NP. In addition, Azil balanced striatal neurotransmitters levels as evidenced by the increase of striatal gamma-aminobutyric acid content and the decrease of glutamate content. Azil also amended neuroinflammation and oxidative stress via modulating IĸB/NF-ĸB and KEAP1/Nrf2 downstream signalling pathways, as well as reducing iNOS and COX2 levels. Moreover, Azil demonstrated an anti-apoptotic activity by reducing caspase-3 level and BAX/BCL2 ratio. In conclusion, the present study reveals the neuroprotective potential of Azil in 3-NP-induced behavioural, histopathological and biochemical changes in rats. These findings might be attributed to inhibition of ATR1/NF-κB signalling, modulation of Nrf2/KEAP1 signalling, anti-inflammatory, anti-oxidant and anti-apoptotic properties.


Benzimidazoles , Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , Oxadiazoles , Rats , Animals , NF-kappa B/metabolism , Rats, Wistar , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Signal Transduction , Neuroprotective Agents/adverse effects , Nitro Compounds/toxicity , Propionates/pharmacology , Huntington Disease/chemically induced
3.
Behav Brain Res ; 461: 114864, 2024 Mar 12.
Article En | MEDLINE | ID: mdl-38220060

Huntington's disease (HD) is a neurodegenerative disorder characterized by degeneration of the striatum; it results in oxidative stress and motor deficits. Thyroid hormones regulate oxidative metabolism. In the present study, we evaluated the effect of administration of levothyroxine (LT-4) on neurobehavioral, oxidative stress, and histological changes in a rat model of HD. Forty-eight Wistar male rats were divided into the following six groups (n = 8): Group 1 (control) received physiological saline intraperitoneally (ip). Groups 2 and 3 received L-T4,30 and L-T4100 (µg/kg, ip, respectively) daily for 7 days. Group 4 (HD) received 3-nitropropionic acid (3-NP) (25 mg/kg, ip) daily for 7 days. Groups 5 and 6 received L-T4,30 and L-T4100 (µg/kg, ip, respectively) 30 min after 3-NP (25 mg/kg, ip) injection for the same duration. On the 8th day, behavioral parameters were evaluated with the Rotarod, Narrow beam walk, and Limb withdrawal tests. Oxidative markers such as Malondialdehyde (MDA) and Glutathione (GSH) levels and Superoxide dismutase (SOD) activity, in striatum tissue were measured. Moreover, striatum tissues were analyzed by Hematoxylin-eosin staining for histological alterations. We found that 3-NP administration caused motor incoordination and induced oxidative stress increased but reduced free radical scavenging. Also, increased amounts of lipid peroxides caused striatal damage as shown by histopathological evaluation. Administration of L-T4 led to increased falling time in the Rotarod, but reduced the time taken in Narrow beam walking and Limb withdrawal test. Furthermore, L-T4 increased antioxidant activity, decreased lipid peroxidation and ameliorated 3-NP-induced degeneration in neurons.


Huntington Disease , Neuroprotective Agents , Rats , Male , Animals , Rats, Wistar , Thyroxine/metabolism , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Motor Activity , Oxidative Stress , Nitro Compounds/toxicity , Propionates/pharmacology , Glutathione/metabolism , Neuroprotective Agents/therapeutic use , Corpus Striatum/metabolism
4.
PLoS One ; 18(11): e0293660, 2023.
Article En | MEDLINE | ID: mdl-37910529

3-nitropropionic acid (3-NP) is a potent mitochondrial inhibitor mycotoxin. Systemic administration of 3-NP can induce Huntington's disease (HD)-like symptoms in experimental animals. Safranal (Safr) that is found in saffron essential oil has antioxidant, anti-inflammatory and anti-apoptotic actions. Candesartan (Cands) is an angiotensin receptor blocker that has the potential to prevent cognitive deficits. The present study aims to investigate the potential neuroprotective efficacy of Safr or Cands in 3-NP-induced rat model of HD. The experiments continued for nine consecutive days. Rats were randomly assigned into seven groups. The first group (Safr-control) was daily intraperitoneally injected with paraffin oil. The second group (Cands- and 3-NP-control) daily received an oral dose of 0.5% carboxymethylcellulose followed by an intraperitoneal injection of 0.9% saline. The third and fourth groups received a single daily dose of 50 mg/kg Safr (intraperitoneal) and 1 mg/kg Cands (oral), respectively. The sixth group was daily treated with 50 mg Safr kg/day (intraperitoneal) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The seventh group was daily treated with 1 mg Cands /kg/day (oral) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The present results revealed that 3-NP injection induced a considerable body weight loss, impaired memory and locomotor activity, reduced striatal monoamine levels. Furthermore, 3-NP administration remarkably increased striatal malondialdehyde and nitric oxide levels, whereas markedly decreased the total antioxidant capacity. Moreover, 3-NP significantly upregulated the activities of inducible nitric oxide synthase and caspase-3 as well as the Fas ligand, in striatum. On the contrary, Safr and Cands remarkably alleviated the above-mentioned 3-NP-induced alterations. In conclusion, Safr and Cands may prevent or delay the progression of HD and its associated impairments through their antioxidant, anti-inflammatory, anti-apoptotic and neuromodulator effects.


Huntington Disease , Neuroprotective Agents , Rats , Animals , Antioxidants/therapeutic use , Rats, Wistar , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/psychology , Motor Activity , Corpus Striatum/pathology , Anti-Inflammatory Agents/pharmacology , Nitro Compounds/pharmacology , Propionates/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Disease Models, Animal
5.
Neurotoxicology ; 97: 120-132, 2023 Jul.
Article En | MEDLINE | ID: mdl-37302585

Copper (Cu) and Zinc (Zn) are required in small concentrations for metabolic functions, but are also toxic. There is a great concern about soil pollution by heavy metals, which may exposure the population to these toxicants, either by inhalation of dust or exposure to toxicants through ingestion of food derived from contaminated soils. In addition, the toxicity of metals in combination is questionable, as soil quality guidelines only assess them separately. It is well known that metal accumulation is often found in the pathologically affected regions of many neurodegenerative diseases, including Huntington's disease (HD). HD is caused by an autosomal dominantly inherited CAG trinucleotide repeat expansion in the huntingtin (HTT) gene. This results in the formation of a mutant huntingtin (mHTT) protein with an abnormally long polyglutamine (polyQ) repeat. The pathology of HD results in loss of neuronal cells, motor changes, and dementia. Rutin is a flavonoid found in various food sources, and previous studies indicate it has protective effects in HD models and acts as a metal chelator. However, further studies are needed to unravel its effects on metal dyshomeostasis and to discern the underlying mechanisms. In the present study, we investigated the toxic effects of long-term exposure to copper, zinc, and their mixture, and the relationship with the progression of neurotoxicity and neurodegeneration in a C. elegans-based HD model. Furthermore, we investigated the effects of rutin post metal exposure. Overall, we demonstrate that chronic exposure to the metals and their mixture altered body parameters, locomotion, and developmental delay, in addition to increasing polyQ protein aggregates in muscles and neurons causing neurodegeneration. We also propose that rutin has protective effects acting through mechanisms involving antioxidant and chelating properties. Altogether, our data provides new indications about the higher toxicity of metals in combination, the chelating potential of rutin in the C. elegans model of HD and possible strategies for future treatments of neurodegenerative diseases caused by the aggregation of proteins related to metals.


Huntington Disease , Neurodegenerative Diseases , Animals , Humans , Huntington Disease/chemically induced , Huntington Disease/prevention & control , Huntington Disease/genetics , Caenorhabditis elegans , Copper/toxicity , Zinc , Rutin/pharmacology , Disease Models, Animal
6.
Toxicol Lett ; 381: 48-59, 2023 May 15.
Article En | MEDLINE | ID: mdl-37116597

Redox homeostasis, mitochondrial functions, and mitochondria-endoplasmic reticulum (ER) communication were evaluated in the striatum of rats after 3-nitropropionic acid (3-NP) administration, a recognized chemical model of Huntington's disease (HD). 3-NP impaired redox homeostasis by increasing malondialdehyde levels at 28 days, decreasing glutathione (GSH) concentrations at 21 and 28 days, and the activities of glutathione peroxidase (GPx), superoxide dismutase (SOD) and glutathione S-transferase at 7, 21, and 28 days, catalase at 21 days, and glutathione reductase at 21 and 28 days. Impairment of mitochondrial respiration at 7 and 28 days after 3-NP administration was also observed, as well as reduced activities of succinate dehydrogenase (SDH) and respiratory chain complexes. 3-NP also impaired mitochondrial dynamics and the interactions between ER and mitochondria and induced ER-stress by increasing the levels of mitofusin-1, and of DRP1, VDAC1, Grp75 and Grp78. Synaptophysin levels were augmented at 7 days but reduced at 28 days after 3-NP injection. Finally, bezafibrate prevented 3-NP-induced alterations of the activities of SOD, GPx, SDH and respiratory chain complexes, DCFH oxidation and on the levels of GSH, VDAC1 and synaptophysin. Mitochondrial dysfunction and synaptic disruption may contribute to the pathophysiology of HD and bezafibrate may be considered as an adjuvant therapy for this disorder.


Huntington Disease , Rats , Animals , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Rats, Wistar , Bezafibrate/adverse effects , Bezafibrate/metabolism , Synaptophysin/metabolism , Models, Chemical , Oxidative Stress , Glutathione/metabolism , Superoxide Dismutase/metabolism , Mitochondria/metabolism , Propionates/toxicity , Nitro Compounds/toxicity , Nitro Compounds/metabolism
7.
Biol Pharm Bull ; 46(2): 177-186, 2023.
Article En | MEDLINE | ID: mdl-36724946

Huntington's disease (HD) is a hereditary neurodegenerative disease that involves an expansion of the CAG repeats of the Huntingtin (HTT) gene, but the disease onset and progression do not necessarily correspond to the extent of CAG repeats. Decreased mitochondrial complex II activity has also been reported to be closely associated with disease pathogenesis. Here, we examined the mechanism of cell death induced by 3-nitropropionic acid (3-NP), a mitochondrial complex II inhibitor, using striatal cells (STHdhQ111 cells) derived from HD model mice with mutant HTT carrying the CAG repeat extended. Treatment with 3-NP (5 mM) enhanced cell death in STHdhQ111 compared to STHdhQ7 cells with normal HTT. Ferrostatin-1, an inhibitor of ferroptosis, and deferoxamine, an iron chelator, markedly inhibited 3-NP-induced cell death in both the STHdh cell lines. On the other hands, cell death was not abrogated by a broad-spectrum caspase inhibitor, Z-VAD-FMK, indicating that this cell death was caspase-independent. Cell death caused by 3-NP is suggested to be due to ferroptosis. Furthermore, 3-NP-induced cell death was markedly inhibited by GSK2795039, a reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) inhibitor, suggesting that cell death is mainly mediated by intracellular superoxide anion (O2-) production through NOX2. Furthermore, a mitochondria-targeted superoxide dismutase mimetic (Mito-TEMPO), partially inhibited 3-NP-induced cell death, suggesting that O2- production in the mitochondria is partially responsible for cell death. These results indicate that 3-NP-induced cell death in the STHdhQ111 cells is caspase-independent, non-apoptotic, and that ferroptotic cell death is mainly induced via NOX2 activation.


Huntington Disease , Neurodegenerative Diseases , Mice , Animals , Reactive Oxygen Species/metabolism , Mice, Transgenic , Nuclear Proteins/genetics , Nerve Tissue Proteins/metabolism , Caspases/metabolism , Huntington Disease/chemically induced , Huntington Disease/genetics , Huntington Disease/metabolism
8.
Molecules ; 28(3)2023 Feb 01.
Article En | MEDLINE | ID: mdl-36771072

BACKGROUND: Previously reported data suggest that hibiscetin, isolated from roselle, contains delphinidin-3-sambubioside and cyanidin-3-sambubioside including anthocyanidins and has a broad range of physiological effects. In this study, we aim to analyze the effect of hibiscetin neuroprotective ability in rats against 3-nitropropionic acid (3-NPA)-induced Huntington's disease (HD). METHODS: To investigate possible toxicities in animals, oral acute toxicity studies of hibiscetin were undertaken, and results revealed the safety of hibiscetin in animals with a maximum tolerated dose. Wistar rats were divided into four groups (n = 6); (group-1) treated with normal saline, (group-2) hibiscetin (10 mg/kg) only, (group-3) 3-NPA only, and (group-4) 3-NPA +10 mg/kg hibiscetin. The efficacy of hibiscetin 10 mg/kg was studied with the administration of 3-NPA doses for the induction of experimentally induced HD symptoms in rats. The mean body weight (MBW) was recorded at end of the study on day 22 to evaluate any change in mean body weight. Several biochemical parameters were assessed to support oxidative stress (GSH, SOD, CAT, LPO, GR, and GPx), alteration in neurotransmitters (DOPAC, HVA, 5-HIAA, norepinephrine, serotonin, GABA, and dopamine), alterations in BDNF and cleaved caspase (caspase 3) activity. Additionally, inflammatory markers, i.e., tumor necrosis factor alpha (TNF-α), interleukins beta (IL-1ß), and myeloperoxidase (MPO) were evaluated. RESULTS: The hibiscetin-treated group exhibits a substantial restoration of MBW than the 3-NPA control group. Furthermore, 3-NPA caused a substantial alteration in biochemical, neurotransmitter monoamines, and neuroinflammatory parameters which were restored successfully by hibiscetin. CONCLUSION: The current study linked the possible role of hibiscetin by offering neuroprotection in experimental animal models.


Huntington Disease , Neuroprotective Agents , Rats , Animals , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats, Wistar , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Oxidative Stress , Nitro Compounds/pharmacology , Propionates/pharmacology , Neurotransmitter Agents/pharmacology , Body Weight , Brain
9.
Neurochem Res ; 48(6): 1631-1647, 2023 Jun.
Article En | MEDLINE | ID: mdl-36738367

Animal models are used to better understand the various mechanisms involved in the pathogenesis of diseases and explore potential pathways that will aid in discovering therapeutic targets. 3-Nitropropionic Acid (3-NPA) is a neurotoxin used to induce Huntington's disease (HD)-like symptoms in experimental animals. The 3-NPA is a fungus toxin that impairs the complex II (succinate dehydrogenase) activity of the mitochondria and reduces ATP synthesis, leading to excessive production of free radicals resulting in the degeneration of GABAergic medium spiny neurons (MSNs) in the striatum. This is characterized by motor impairments a key clinical manifestation of HD. 3-NPA has the potential to alter several cellular processes, including mitochondrial functions, oxidative stress, apoptosis, and neuroinflammation mimicking HD-like pathogenic conditions in animals. This review strives to provide a new insight towards the 3-NPA induced molecular dysfunctioning in developing an animal model of HD. Moreover, we summarise several preclinical studies that support the use of the 3-NPA-induced models for drug discovery and development in HD. This review is a collection of various articles that were published from 1977 to 2022 on Pubmed (1639), Web of Science (2139), and Scopus (2681), which are related to the 3-NPA induced animal model.


Huntington Disease , Animals , Huntington Disease/chemically induced , Huntington Disease/metabolism , Neurotoxins/toxicity , Disease Models, Animal , Nitro Compounds/toxicity , Propionates/toxicity , Drug Discovery
10.
Neurochem Res ; 48(6): 1775-1782, 2023 Jun.
Article En | MEDLINE | ID: mdl-36689085

Huntington's disease (HD) is a progressive, neurodegenerative, and inherited disease. Antioxidants have been shown to be effective in slowing disease progression in animal models of HD and are under investigation in human clinical trials. α-pinene, a member of the monoterpene class, has been shown to exert antioxidant activity. Therefore, this study aimed to investigate the impact of α-pinene on animal model of HD. Thirty-two male Wistar rats received 3-Nitropropionic acid (3-NP) for induction of the disease model or treated with α-pinene + 3-NP in different groups. Motor skill, and biochemical evaluations to detect oxidant/antioxidant markers in rat cortex and striatum were performed in all groups. We found that α-pinene significantly improved 3-NP-induced changes in the body weight, rotarod activity, time taken to cross the narrow beam, and locomotor activity. Biochemical analysis revealed that α-pinene significantly decreased the 3NP-induced elevation in oxidant markers, nitrite, and malondialdehyde in both cortex and striatum. In addition, α-pinene counteracted the 3-NP-induced fall in antioxidant enzymes, including superoxide dismutase, catalase, and glutathione in the cortex and striatum. In conclusion, we found that α-pinene prevented the motor dysfunction induced by 3-NP in the animal model of Huntington's disease. Oxidants-antioxidant balance might be involved in the protective effect of α-pinene.


Huntington Disease , Neuroprotective Agents , Humans , Rats , Male , Animals , Antioxidants/pharmacology , Rats, Wistar , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Motor Activity , Lipid Peroxidation , Models, Animal , Oxidants , Nitro Compounds/pharmacology , Disease Models, Animal , Neuroprotective Agents/therapeutic use , Behavior, Animal
11.
Chem Biol Interact ; 369: 110288, 2023 Jan 05.
Article En | MEDLINE | ID: mdl-36509115

Huntington's disease (HD) is an inherited neurodegenerative disease characterized by progressive motor, behavioral, and cognitive impairments. Intrastriatal injection of 3- nitropropionic acid (3NP) was used to induce HD-like symptoms by inhibiting succinate dehydrogenase enzyme (SDH) in the mitochondrial complex II. The adenosine A1 receptor has long been known to have a crucial role in neuroprotection, mainly by blocking Ca2+ influx, which causes inhibition of glutamate (Glu) and a decline in its excitatory effects at the postsynaptic level. To this end, this study investigated the possible involvement of TrKB/PI3K/Akt/CREB/BDNF pathway in mediating protection afforded by the central N6-cyclohexyladenosine (CHA), an adenosine A1 receptor agonist. A single intrastriatal CHA injection (6.25 nM/1 µL); 45min after 3-NP injection, attenuated neuronal death, and improved cognitive and motor deficits caused by 3-NP neurotoxin. This effect was shown to parallel an enhanced activation of PI3K/Akt/CREB/BDNF axis as well as boosting pERK1/2 levels. Moreover, CHA attenuated neuroinflammatory and oxidative stress status via reducing NFκB p65, TNFα and iNOS contents and increasing SOD. Furthermore, immunohistochemical data showed a reduction in the glial fibrillary acidic protein (GFAP) immunoreactivity to a marker for astrocyte and microglia activation following CHA treatment. The results of this study suggest that CHA may have protective effect against HD via modulating oxidative stress, excitotoxic and inflammatory pathways.


Huntington Disease , Neurodegenerative Diseases , Neuroprotective Agents , Rats , Animals , Brain-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , Neurodegenerative Diseases/drug therapy , Corpus Striatum , Adenosine/metabolism , Nitro Compounds/toxicity , Neuroprotective Agents/therapeutic use
12.
Neurotox Res ; 40(6): 2089-2102, 2022 Dec.
Article En | MEDLINE | ID: mdl-36385437

Striatal neurotoxicity is the pathological hallmark for a heterogeneous group of movement disorders like Tardive dyskinesia (TD) and Huntington's disease (HD). Both diseases are characterized by progressive impairment in motor function. TD and HD share common features at both cellular and subcellular levels. Filgrastim, a recombinant methionyl granulocyte colony-stimulating factor (GCSF), shows neuroprotective properties in in-vivo models of movement disorders. This study seeks to evaluate the neuroprotective effect of filgrastim in haloperidol and 3-NP-induced neurotoxicity in rats. The study was divided into two: in study one, rats were administered with haloperidol for 21 days, filgrastim at the dose of (20, 40, 60 µg/kg,s.c.) was administered once a day before haloperidol treatment and the following parameters (orofacial movements, rotarod, actophotometer) were performed to assess TD. Similarly, in the second study, rats were administered with 3-NP for 21 days, filgrastim at a dose of (20 and 40 µg/kg, s.c.) was administered, and the following parameters (rotarod, narrow beam walk, and open field test) were assessed for HD. On the 22nd day, animals were sacrificed and cortex and striatum isolated for oxidative stress (LPO, GSH, SOD, catalase, and nitrate) marker assessment. Results revealed that haloperidol and 3-NP treatment significantly impaired motor coordination, and oxidative defense inducing TD and HD-like symptoms. Treatment with filgrastim significantly averted haloperidol and 3-NP-induced behavioral and biochemical alterations. Conclusively, the neuroprotective effect of filgrastim is credited to its antioxidant properties. Hence, filgrastim might be a novel therapeutic candidate for the management of TD and HD.


Huntington Disease , Movement Disorders , Neuroprotective Agents , Neurotoxicity Syndromes , Animals , Rats , Filgrastim/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Haloperidol/toxicity , Huntington Disease/chemically induced , Movement Disorders/drug therapy , Movement Disorders/etiology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/prevention & control , Nitro Compounds/toxicity , Propionates , Rats, Wistar
13.
Eur J Pharmacol ; 934: 175316, 2022 Nov 05.
Article En | MEDLINE | ID: mdl-36209926

Huntington disease (HD), an autosomal dominant neurodegenerative disorder characterized by involuntary choreatic movements with cognitive and behavioral disturbances. HD striatum has increased conversion of kynurenine to quinolinic acid (QA) which activates NMDA receptors leading to activation of microglia and increased levels of nuclear factor kappa B (NF-κB) leading to elevated transcription of inducible nitric oxide synthase (iNOS) and various cytokines causing neuronal death via neuroinflammation, oxidative stress, mitochondrial dysfunction and apoptosis. Therefore, inhibiting IKK-NF-κB pathway induced excitotoxicity, oxidative stress and neuroinflammation could be a potential intervention in slowing down the disease progression. QA injection intrastriatally (IS-QA) produce damage mimicking HD where neuroinflammation, oxidative stress and mitochondrial dysfunction play crucial role. Ellagic acid (EA) and vanillic acid (VA) are well reported to possess antioxidant and NF-κB inhibiting effect. Hence, in present study, rats administered IS-QA were treated with EA and VA for 21 days to explore their neuroprotective effects. Behavioral studies, biochemical estimations for oxidative stress and acetylcholinesterase assay were performed. Mitochondrial function was determined by estimating mitochondrial enzyme complexes; inflammatory markers like TNF-α, IL-6, NF-κB by ELISA and apoptosis by caspase-3 levels. Brain damage was determined by histopathology which revealed their neuroprotective effects. Various doses of EA and VA produced improved motor and cognitive functions, oxidative stress and neuroinflammation were also reduced and mitochondrial functioning was improved. In a nutshell, these results signify improved motor and cognitive functions by EA and VA in QA model of HD, along with declined oxidative stress, mitochondrial dysfunction and neuroinflammation.


Huntington Disease , Neuroprotective Agents , Animals , Rats , Quinolinic Acid/adverse effects , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Huntington Disease/metabolism , NF-kappa B/metabolism , Caspase 3/metabolism , Ellagic Acid/pharmacology , Ellagic Acid/therapeutic use , Vanillic Acid/pharmacology , Vanillic Acid/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Acetylcholinesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antioxidants/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Interleukin-6/metabolism , Kynurenine/metabolism , Oxidative Stress , Anti-Inflammatory Agents/pharmacology
14.
Environ Pollut ; 314: 120301, 2022 Dec 01.
Article En | MEDLINE | ID: mdl-36181934

Household insecticide is largely used for insect and ectoparasite control, in city centers as well as in the countryside. The pyrethroids are the most used class of insecticide, these compounds in low doses have low toxicity for mammalians, in comparison to other compounds, with insecticide effects. The contact of these compounds in sublethal doses begins in early life and many cases, in intrauterine life. Considerable diseases still with undefined etiology, such as neurodegenerative conditions, and Huntington's Disease (HD) is one of them. HD is related to overexpression of Polyglutamine (PolyQ40), its aggregation, and non-solubilization, which leads to neural, behavioral, and cognitive damage. In our study, we evaluate the effect of two sublethal doses of a prallethrin-based insecticide (P-BI), in three different Caenorhabditis elegans life stages transgenerational, neonatal, and lifespan. We evaluated the Body bends and pharyngeal pumping rate, and social feeding as behavioral biomarkers. As well as acetylcholinesterase activity (AChE), PolyQ40 aggregation, antioxidant enzymes, and heat shock protein (HSP) expression. We observe that the toxic effect of P-BI is more pronounced on transgenerational and lifespan exposure. Both sublethal doses of P-BI decreased the AChE activity and retard the HSP expression as well as increased the PolyQ40 aggregates indicating a clear biomarker for possible effect in the progression of the HD, by the environmental contamination.


Huntington Disease , Insecticides , Pyrethrins , Animals , Humans , Acetylcholinesterase/metabolism , Antioxidants/metabolism , Caenorhabditis elegans , Heat-Shock Proteins , Huntington Disease/chemically induced , Insecticides/metabolism , Mammals , Pyrethrins/toxicity
15.
Rejuvenation Res ; 25(6): 260-274, 2022 Dec.
Article En | MEDLINE | ID: mdl-36150031

Huntington's disease (HD) is an inherited neurodegenerative disease. 3-Nitropropionic acid (3-NP) causes increased reactive oxygen species production and neuroinflammation. Centella asiatica (CA) is a strong antioxidant. The aim of this study is to investigate the effect of hydroalcoholic extract of C. asiatica (HA-CA) on 3-NP-induced HD in adult zebrafish. Adult zebrafish (∼5-6 months old) weighing 470 to 530 mg was used and treated with 3-NP (5 mg/kg intraperitoneal [i.p.]). The animals received HA-CA (80 and 100 mg/L) daily for up to 28 days in water. Tetrabenazine (3 mg/kg i.p.) was used as a standard drug. We have done an open field test (for locomotor activity), a novel tank diving test (for anxiety), and a light and dark tank test (for memory), followed by biochemical analysis (acetyl-cholinesterase [AchEs], nitrite, lipid peroxidation [LPO], and glutathione [GSH]) and histopathology to further confirm memory dysfunctions. 3-NP-treated zebrafish exhibit reductions in body weight, progressive neuronal damage, cognition, and locomotor activity. The HA-CA group significantly reduced the 3-NP-induced increase in LPO, AchEs, and nitrite levels while decreasing GSH levels. Oral administration of HA-CA (80 or 100 mg/L) significantly reduces 3-NP-induced changes in body weight and behaviors, in addition to neuroinflammation in the brain by lowering tumor necrosis factor-α and interleukin-1ß levels. Moreover, HA-CA significantly decreases the 3-NP-induced neuronal damage in the brain. HA-CA ameliorates neurotoxicity and neurobehavioral deficits in 3-NP-induced HD-like symptoms in adult zebrafish.


Centella , Huntington Disease , Neurodegenerative Diseases , Neuroprotective Agents , Animals , Centella/metabolism , Zebrafish/metabolism , Nitrites/adverse effects , Neuroinflammatory Diseases , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Motor Activity , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Glutathione/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Body Weight , Pain/drug therapy , Oxidative Stress
16.
Biomed Pharmacother ; 155: 113664, 2022 Nov.
Article En | MEDLINE | ID: mdl-36095961

Mitochondrial abnormalities and a defective expression of neurotrophic factors contribute to neuronal damage in Huntington's disease (HD). HD patients showed a reduction in transforming growth factor-ß1 (TGF-ß1) levels in the peripheral blood and in cortical neurons. 3-n-butylphthalide (NBP) is first isolated from the seeds of celery, treats ischemic stroke in China. NBP could attenuate cognitive and motor impairments in the experimental models of Parkinson's disease and Alzheimer's disease, reduce mitochondrial oxidative stress and increase the expression of TGF-ß1 in rats with focal cerebral ischemia. To our knowledge, the effect of NBP on Huntington's disease has not been reported. We proposed the hypothesis that whether NBP could protect mitochondria and regulate TGF-ß1 and its downstream signaling in a HD animal model, further prevents motor dysfunction. Malonic acid is a reversible inhibitor of mitochondrial enzyme complex-II, induces energy crisis and free radical generation. In this study, we used intrastriatal injections of malonic acid in rats to mimic mitochondrial abnormalities and the other HD like symptoms. We found that treatment with NBP significantly attenuated malonic acid-induced motor and cognitive dysfunction in locomotor behaviour test, rotarod test, novel object recognition test and morris water maze test, prevented neurotoxicity and mitochondrial damage, activated TGF-ß1/Akt/Wnt/ß-Catenin pathway in striatum, but didn't regulate mitochondrial fusion and fission. The above effect was partly reversed by a PI3K/Akt inhibitor. Our data support NBP as a potential candidate for the treatment of HD.


Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , Animals , Rats , Transforming Growth Factor beta1 , Huntington Disease/chemically induced , beta Catenin , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Neurotoxicity Syndromes/drug therapy , Nerve Growth Factors/therapeutic use
17.
Neurochem Res ; 47(11): 3285-3297, 2022 Nov.
Article En | MEDLINE | ID: mdl-35876936

Berberine due to its antioxidant properties, has been used around the globe significantly to treat several brain disorders. Also, oxidative stress is a pathological hallmark in neurodegenerative diseases like Huntington's disease (HD) and Tardive dyskinesia (TD). Berberine an alkaloid from plants has been reported to have neuroprotective potential in several animal models of neurodegenerative diseases. Hence, this study aims to evaluate the neuroprotective effect of berberine in the animal model of 3-nitropropionic acid (3-NP) induced HD and haloperidol induced tardive dyskinesia with special emphasis on its antioxidant property. The study protocol was divided into 2 phases, first phase involved the administration of 3-NP and berberine at the dose of (25, 50, and 100 mg/kg) intraperitoneally (i.p) and orally (p.o.) respectively for 21 days, and the following parameters (rotarod, narrow beam walk and photoactometer) as a measure of motor activity and striatal and cortical levels of (LPO, GSH, SOD, catalase, and nitrate) evaluated as a measure of oxidative stress were assessed for HD. Similarly in the second phase, TD was induced by using haloperidol, for 21 days and berberine at the dose of (25, 50, and 100 mg/kg) was administered, and both physical and biochemical parameters were assessed as mentioned for the HD study. The resultant data indicated that berberine attenuate 3-NP and haloperidol-induced behavioral changes and improved the antioxidant capcity in rodents. Hence berberine might be a novel therapeutic candidate to manage TD & HD.


Berberine , Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , Tardive Dyskinesia , Animals , Antioxidants/metabolism , Berberine/pharmacology , Berberine/therapeutic use , Catalase , Haloperidol/toxicity , Huntington Disease/chemically induced , Motor Activity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Nitro Compounds/toxicity , Propionates , Rats , Rats, Wistar , Superoxide Dismutase , Tardive Dyskinesia/drug therapy
18.
Behav Pharmacol ; 33(5): 333-341, 2022 08 01.
Article En | MEDLINE | ID: mdl-35695543

Tetrabenazine, a preferential inhibitor of the vesicular monoamine transporter type 2, depletes the brain monoamines dopamine, serotonin and norepinephrine. Tetrabenazine and deutetrabenazine (Austedo ®) are used to treat chorea associated with Huntington's disease. However, both compounds are known to aggravate Parkinsonism and depression observed in Huntington's disease patients. NLX-112 (a.k.a. befiradol/F13640) is a highly selective, potent and efficacious serotonin 5-HT 1A agonist. In animal models, it has robust efficacy in combating other iatrogenic motor disorders such as L-DOPA-induced dyskinesia and has marked antidepressant-like activity in rodent tests. In the present study, we investigated, in rats, the efficacy of NLX-112 to counteract tetrabenazine-induced catalepsy (a model of Parkinsonism) and tetrabenazine-induced potentiation of immobility in the forced swim test (FST, a model to detect antidepressant-like activity). The prototypical 5-HT 1A agonist, (±)8-OH-DPAT, and the 5-HT 1A partial agonist/dopamine D2 receptor blocker, buspirone, were used as comparators. Both NLX-112 and (±)8-OH-DPAT (0.16-2.5 mg/kg p.o. or s.c., respectively) abolished catalepsy induced by tetrabenazine (2 mg/kg i.p.). In comparison, buspirone (0.63-5.0 mg/kg p.o.) was ineffective and even tended to potentiate tetrabenazine-induced catalepsy at 0.63 mg/kg. In the FST, NLX-112 and (±)8-OH-DPAT (0.63 mg/kg) strongly reduced immobility when administered alone but also significantly opposed potentiation of immobility induced by tetrabenazine (1.5 mg/kg i.p.). Buspirone (0.63 and 2.5 mg/kg p.o.) had no effect by itself or against tetrabenazine. These results strongly suggest that selective and highly efficacious 5-HT 1A agonists, such as NLX-112, may be useful in combating tetrabenazine-induced Parkinsonism and/or depression in Huntington's disease patients.


Huntington Disease , Parkinsonian Disorders , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Antidepressive Agents/pharmacology , Buspirone/pharmacology , Catalepsy/chemically induced , Catalepsy/drug therapy , Depression/chemically induced , Depression/drug therapy , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Piperidines , Pyridines , Rats , Receptor, Serotonin, 5-HT1A , Serotonin , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Tetrabenazine
19.
Eur J Pharmacol ; 927: 175046, 2022 Jul 15.
Article En | MEDLINE | ID: mdl-35623405

Oxidative stress induced neurotoxicity is increasingly perceived as an important neuropathologic mechanism underlying the motor and behavioral phenotypes associated with Huntington's disease (HD). Repeated exposure to 3-nitropropionic acid (3-NP) induces neurotoxic changes which closely simulate the neuropathological and behavioral characteristics of HD. This study aimed at evaluating the prophylactic effects of the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) inhibitor "harmine" against 3-NP-indued neurotoxicity and HD-like symptoms. The potential prophylactic effect of harmine (10 mg/kg/day; intraperitoneal) was investigated on 3-NP-induced motor and cognitive HD-like deficits, nuclear factor erythroid 2 like 2 (NRF2), AMP kinase (AMPK) and p21 protein levels and the gene expression of haem oxygenase-1 (Ho-1), NAD(P)H: quinone oxidoreductase-1 (Nqo-1) and p62 in addition to redox imbalance and histological neurotoxic changes in the striatum, prefrontal cortex, and hippocampus of male Wistar rats. Harmine successfully increased the protein levels of NRF2, AMPK and p21 and the gene expression of Ho-1, Nqo-1 and p62, restored redox homeostasis, and reduced CASPASE-3 level. This was reflected in attenuation of 3-NP-induced neurodegenerative changes and improvement of rats' motor and cognitive performance. This study draws attention to the protective role of harmine against 3-NP-induced motor and cognitive dysfunction that could be mediated via enhancing NRF2-mediated signaling with subsequent amelioration of oxidative stress injury via NRF2 activators, p21 and AMPK, in the striatum, prefrontal cortex, and hippocampus which could offer a promising therapeutic tool to slow the progression of HD.


AMP-Activated Protein Kinases , Cyclin-Dependent Kinase Inhibitor p21 , Harmine , Huntington Disease , Neuroprotective Agents , Neurotoxicity Syndromes , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Harmine/pharmacology , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/prevention & control , Male , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/adverse effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Nitro Compounds/antagonists & inhibitors , Nitro Compounds/pharmacology , Oxidative Stress , Propionates/antagonists & inhibitors , Propionates/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects
20.
Inflammopharmacology ; 30(5): 1745-1758, 2022 Oct.
Article En | MEDLINE | ID: mdl-35639233

BACKGROUND AND AIM: Huntington's disease (HD) is a rare inherited disease portrayed with marked cognitive and motor decline owing to extensive neurodegeneration. NADPH oxidase is considered as an important contributor to the oxidative injury in several neurodegenerative disorders including HD. Thus, the present study explored the possible neuroprotective effects of diapocynin, a specific NADPH oxidase inhibitor, against 3-nitropropionic acid (3-NP) model of HD in rats. METHODS: Animals received diapocynin (10 mg/kg/day, p.o), 30 min before 3-NP (10 mg/kg/day, i.p) over a period of 14 days. RESULTS: Diapocynin administration attenuated 3-NP-induced oxidative stress with significant increase in reduced glutathione, glutathione-S-transferase, nuclear factor erythroid 2-related factor 2, and brain-derived neurotrophic factor striatal contents contrary to NADPH oxidase (NOX2; gp91phox subunit) diminished expression. Moreover, diapocynin mitigated 3-NP-associated neuroinflammation and glial activation with prominent downregulation of nuclear factor-Ðšß p65 and marked decrement of inducible nitric oxide synthase content in addition to decreased immunoreactivity of ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein; markers of microglial and astroglial activation, respectively. Treatment with diapocynin hindered 3-NP-induced apoptosis with prominent decrease in tumor suppressor protein and Bcl-2-associated X protein contents whereas the anti-apoptotic marker; B-cell lymphoma-2 content was noticeably increased. Diapocynin neuroprotective effects could be attributed to silent information regulator 1 upregulation which curbed 3-NP-associated hazards resulting in improved motor functions witnessed during open field, rotarod, and grip strength tests as well as attenuated 3-NP-associated histopathological derangements. CONCLUSION: The present findings indicated that diapocynin could serve as an auspicious nominee for HD management.


Huntington Disease , Neuroprotective Agents , Acetophenones , Animals , Biphenyl Compounds , Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Huntington Disease/chemically induced , Huntington Disease/drug therapy , NADPH Oxidases/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitro Compounds , Propionates , Rats , Signal Transduction , Sirtuin 1/metabolism , Transferases/metabolism , Transferases/pharmacology , Tumor Suppressor Proteins/adverse effects , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein/metabolism
...