Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Mol Med ; 30(1): 158, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327568

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a progressive neurodegenerative disease that causes motor, cognitive, and psychiatric abnormalities, with no satisfying disease-modifying therapy so far. 3-nitropropionic acid (3NP) induces behavioural deficits, together with biochemical and histological alterations in animals' striata that mimic HD. The role of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in HD pathogenesis remains largely uncharacterized. Parthenolide (PTL), a naturally occurring nuclear factor kappa B (NF-κB) inhibitor, is also known to inhibit NLRP3 inflammasome. Whether PTL is beneficial in HD has not been established yet. AIM: This study evaluated the possible neuroprotective effects of PTL against 3NP-induced behavioural abnormalities, striatal biochemical derangements, and histological aberrations. METHODS: Male Wistar rats received PTL (0.5 mg/kg/day, i.p) for 3 weeks and 3NP (10 mg/kg/day, i.p) was administered alongside for the latter 2 weeks to induce HD. Finally, animals were subjected to open-field, Morris water maze and rotarod tests. Rat striata were examined histologically, striatal protein expression levels of glial fibrillary acidic protein (GFAP), cluster of differentiation 45 (CD45) and neuron-specific enolase (NSE) were evaluated immunohistochemically, while those of interleukin (IL)-1ß, IL-18, ionized calcium-binding adapter molecule-1 (Iba1) and glutamate were determined by ELISA. Striatal nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein (Keap1), NF-κB, NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, S100 calcium-binding protein A10 (S100A10) and complement-3 (C3) were assessed by gene expression analysis. RESULTS: PTL improved motor, locomotor, cognitive and anxiety-like behaviours, restored neuronal integrity, upregulated Nrf2, and inhibited NLRP3 inflammasome, NF-κB and microglial activation. Additionally, PTL induced astrocyte shifting towards the neuroprotective A2 phenotype. CONCLUSION: PTL exhibits neuroprotection against 3NP-induced HD, that might be ascribed, at least in part, to its modulatory effects on Keap1/Nrf2 and NF-κB/NLRP3 inflammasome signaling.


Subject(s)
Astrocytes , Huntington Disease , Inflammasomes , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Nitro Compounds , Propionates , Sesquiterpenes , Animals , Huntington Disease/drug therapy , Huntington Disease/metabolism , Huntington Disease/chemically induced , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Inflammasomes/metabolism , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Propionates/pharmacology , Rats , Astrocytes/metabolism , Astrocytes/drug effects , Microglia/metabolism , Microglia/drug effects , Disease Models, Animal , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats, Wistar , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Behavior, Animal/drug effects
2.
Brain Res Bull ; 216: 111039, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39089590

ABSTRACT

Huntington's disease (HD) is a scarce neurodegenerative disorder defined by chorea (unusual involuntary movements), behavioral presentations, psychiatric features, and cognitive deterioration. Although the precise pathogenic mechanism behind HD has not yet been identified, the most widely acknowledged pathways include excitotoxicity, mitochondrial malfunction, neuroinflammation, neurochemical imbalance, oxidative stress, and apoptosis HD has no efficient therapy. Current medications have drawbacks. Silymarin, a compound made up of standardized extracts obtained from the seeds of the Silybum marianum and polyphenolic flavonolignan, is utilized in therapeutic settings to treat a variety of experimental disorders in animals. Silymarin's key pharmacological activities include anti-cancer, hepatoprotection, antioxidant, cardioprotection, and anti-inflammatory. It also has no adverse side effects on people or animals. The current study aims to provide Silymarin's neuro-pharmacological activities or therapeutic qualities in HD. In this study, Thirty-six male Sprague-Dawley rats (200-220 g, 8 weeks) at the initial of the study were used. Silymarin solution (100 mg/Kg) was administered by oral gavage for 21 days to ameliorate neural damage in rats injected with 3-nitropropionicacid (3-NP) in a preliminary rat model of HD. The results showed that administration of silymarin to HD rats reduced gliosis, improved motor coordination and muscle activity, and increased striatal volume and the number of neurons and glial cells. Our results suggest that silymarin provides a protective environment for nerve cells and can have beneficial effects against the harmful effects of HD.


Subject(s)
Cell Death , Disease Models, Animal , Huntington Disease , Neuroprotective Agents , Rats, Sprague-Dawley , Silymarin , Animals , Huntington Disease/drug therapy , Huntington Disease/metabolism , Silymarin/pharmacology , Male , Rats , Neuroprotective Agents/pharmacology , Cell Death/drug effects , Neuroinflammatory Diseases/drug therapy , Motor Activity/drug effects , Nitro Compounds , Neurons/drug effects , Neurons/metabolism , Propionates/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology
3.
Mol Neurobiol ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085678

ABSTRACT

Huntington's disease (HD) is an incorrigible neuropsychiatric disorder with reduced cognition and motor abnormalities. Piperine (PIP) is an alkaloid with antioxidant, anti-inflammatory, and neuroprotective activities; however, poor therapeutic efficacy limits its further use. The current study focuses on the enhanced therapeutic potential of PIP@CM against an experimental zebrafish model of HD. PIP@CM was fabricated using spray drying technology, followed by solid-state investigations. We performed in vitro release and in vitro antioxidant activity (DPPH assay) of PIP and PIP@CMs. In addition, in vivo studies were conducted on zebrafish using 3-nitropropionic acid (3-NPA) (60 mg/kg) as a neurotoxin and treated with PIP (5 mg/kg) and PIP@CM (25 mg/kg equivalent to 5 mg/kg PIP). After dosing, various in vivo studies (behavioral, biochemical, and histological) were conducted. The solid-state characterization techniques revealed the loss of crystallinity after micelles formation. In vitro release and antioxidant assays showed higher release and enhanced activity of PIP@CM. In vivo studies revealed that 3-NPA administration causes HD, as evidenced by the results of open field test (OFT) and novel tank diving test (NTD) tests. Moreover, 3-NPA causes an increase in oxidative stress, as confirmed by biochemical and histopathological studies. PIP@CM treatment significantly improved behavioral performance in OFT and NTD tests and reduced oxidative stress markers as compared to pure PIP and untreated HD model.

4.
Animals (Basel) ; 14(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38929423

ABSTRACT

3-nitropropionic acid (3NPA) has been proposed as an useful modifier to mitigate ruminal enteric methane emissions. However, few studies investigated the effects of 3NPA on ruminal fermentation characteristics of grazing ruminants in vitro. Rumen fluid from grazing yak and cattle were collected and incubated with additions of 0, 8, and 16 mM 3NPA. The total gas production, CH4 production, and dry matter digestibility significantly decreased with increasing 3NPA doses in both ruminant species (p < 0.05) and methane production decreased to almost 100% in cattle at 8 mM NPA but not yak, while H2 accumulation showed an opposite trend. The total fatty acid (TVFA) production, acetate concentration, and propionate concentration in cattle decreased as 3NPA doses increased at 12 and 24 h incubation. For yak, the H2 accumulation reached its apex at 8 mM NPA (p < 0.05). The TVFA in yak decreased significantly with increasing 3NPA doses at 12 and 72 h incubation. Moreover, the acetate concentration and propionate concentration in yak decreased as 3NPA doses increased at 12 and 24 h incubation. Overall, these findings demonstrated that 3NPA could be used as a strategy to mitigate methane emissions; although, it negatively affected the dry matter degradability in vitro.

5.
Food Chem ; 456: 139983, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-38850609

ABSTRACT

A method for accurately determining 3-nitropropionic acid in sugarcane was established for the first time using gas chromatography-atmospheric pressure chemical ionization-tandem mass spectrometry (GC - APCI-MS/MS). Under acidic conditions, 3-nitropropionic acid is methylated to obtain methyl 3-nitropropionate. The derivative product was purified using dispersive solid-phase extraction (d-SPE) method and analyzed using GC - APCI-MS/MS. The recovery experiments were conducted at three concentrations: low, medium, and high. The recovery rates ranged from 75.1% to 90.2%, the relative standard deviations were <8.2%, and the limit of quantification was 2.0 µg/kg. The method offers the advantage of being accurate, sensitive, and specific, meeting the requirements of the determination of 3-nitropropionic acid in sugarcane.


Subject(s)
Nitro Compounds , Propionates , Saccharum , Solid Phase Extraction , Tandem Mass Spectrometry , Propionates/isolation & purification , Propionates/analysis , Saccharum/chemistry , Nitro Compounds/chemistry , Nitro Compounds/analysis , Nitro Compounds/isolation & purification , Solid Phase Extraction/methods , Gas Chromatography-Mass Spectrometry , Food Contamination/analysis
6.
Neurosci Lett ; 836: 137882, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38909839

ABSTRACT

Huntington's disease (HD) is an autosomal inherited progressive neurodegenerative disorder which is caused by the CAG trinucleotide repeat in the huntingtin gene. The mutation induces mitochondrial dysfunction in neurons, which leads to striatal neuronal loss. The efficacy of the available therapies is limited, thus acquisition of more data about the pathomechanism of HD and development of new strategies is urgent. Sirtuins (Sirt1-7) belong to the histone deacetylase family, and interestingly they have been associated with HD, however, their role in HD is still not fully understood. To clarify the role of sirtuins in HD, we utilized a 3-nitropropionic acid (3-NP) induced HD model and assessed alterations in gene expression using RT-PCR. Moreover, we studied the extension of neurodegeneration in the striatum, and behavioural changes. Furthermore, we involved Sirt3 knockout (Sirt3KO) mice to investigate the impact of Sirt3 deficiency in the expression of the other sirtuins. Our results showed that with 3-NP treatment, the mRNA level of Sirt2,5,7 changed significantly in wild-type (WT) mice, whereas in Sirt3KO animals there was no change. Interestingly, Sirt3 deficiency did not exacerbate 3-NP-mediated striatal neuronal loss, while Sirt3KO animals showed higher mortality than WT littermates. However, the absence of Sirt3 did not affect the behaviour of animals. Finally, we demonstrated that the changes in the expression of sirtuins are age- and sex- dependent. According to our findings, there is evidence that Sirt3 has a major impact on the regulation of other sirtuin isoforms, survival and neuroprotection. However, this neuroprotective effect does not manifest in the behaviour.


Subject(s)
Corpus Striatum , Huntington Disease , Mice, Knockout , Nitro Compounds , Propionates , Sirtuin 3 , Animals , Nitro Compounds/toxicity , Propionates/pharmacology , Propionates/toxicity , Sirtuin 3/genetics , Sirtuin 3/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/chemically induced , Male , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Female , Sirtuins/genetics , Sirtuins/metabolism , Mice , Mice, Inbred C57BL , Gene Expression/drug effects
7.
Cell Biochem Biophys ; 82(2): 1489-1502, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38760648

ABSTRACT

The neurotoxicity of 3-Nitropropionic acid (3-NP) is well known. Herein, the prophylactic versus therapeutic effects of quercetin (QCT) were investigated against 3-NP-induced behavioral anomalies and oxidative neural damage. Thirty male mice were assigned into five groups; the negative control group, the QCT group (25 mg/kg/day, p.o. for 21 days), the 3-NP group (17 days), the prophylactic group (QCT administration for 14 consecutive days, and then 3-NP was administrated), the therapeutic group (3-NP was administrated and then QCT for 21 days). At the end of the animal treatment, behavioral studies were assessed. Subsequently, the brain sample tissues were assessed for oxidative stress-related parameters and histological evaluation. Moreover, the potential interaction between 3-NP and tumor necrosis factor-alpha (TNF-α) was evaluated by using a molecular docking study. 3-NP markedly led to neurotoxicity which was indicated by behavioral deficits (motor behavior, depression-like behavior, memory dysfunction, and passive avoidance) and oxidative damage. Blind and targeted molecular docking results showed good interaction between 3-NP and TNF-α. However, the prophylactic effects of QCT were superior to the therapeutic effects for attenuating 3-NP-induced neurobehavioral and oxidative neural changes in experimental mice, which histological changes of the brain's striatum region approved our findings. Taken together, the antioxidant activity of QCT remarkably could attenuate 3-NP-induced neurobehavioral deficits and mitochondrial dysfunctions in mice.


Subject(s)
Behavior, Animal , Huntington Disease , Molecular Docking Simulation , Nitro Compounds , Oxidative Stress , Propionates , Quercetin , Tumor Necrosis Factor-alpha , Animals , Male , Nitro Compounds/toxicity , Quercetin/pharmacology , Quercetin/therapeutic use , Quercetin/chemistry , Mice , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/drug therapy , Behavior, Animal/drug effects , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/metabolism , Antioxidants/pharmacology , Antioxidants/chemistry , Brain/metabolism , Brain/drug effects , Brain/pathology , Disease Models, Animal
8.
Reprod Sci ; 31(8): 2261-2272, 2024 08.
Article in English | MEDLINE | ID: mdl-38630174

ABSTRACT

Tannic acid (TA) is a polyphenol with antioxidant properties present in various plants. In this study, we explored the protective effect of TA against ovarian oxidative stress in Brandt's voles and its underlying mechanism. At various doses, 3-nitropropionic acid (3-NPA) was intraperitoneally injected into Brandt's voles to simulate ovarian oxidative stress. Thereafter, various doses of TA were intragastrically administered to examine the protective effect of TA against 3-NPA-induced ovarian damage. Changes in inflammation, autophagy, apoptosis, and oxidative stress-related factors were investigated through various biochemical and histological techniques. Ovarian oxidative stress was successfully induced by the intraperitoneal administration of 12.5 mg/kg 3-NPA for 18 days. As a result, the ovarian coefficient decreased and ovarian tissue fibrosis was induced. TA treatment effectively alleviated the increase in luteinizing hormone and follicle-stimulating hormone levels; the decrease in estradiol, progesterone, and anti-Müllerian hormone levels; and the decline in fertility induced by 3-NPA. Compared to that in the 3-NPA group, TA decreased the expression of autophagy-related proteins beclin-1 and LC3, as well as the level of apoptosis. It also activated the AKT/mTOR signaling pathway, downregulated PTEN and p-NF-κB expression, and upregulated Nrf2 expression. In conclusion, our findings indicate that TA could inhibit autophagy via the regulation of AKT/mTOR signaling, suppressing oxidative damage and inflammatory responses through Nrf2 to alleviate 3-NPA-induced ovarian damage. Collectively, the current findings highlight the protective effects of TA in Brandt's vole, where it promotes the maintenance of normal ovarian function.


Subject(s)
Arvicolinae , Nitro Compounds , Ovary , Oxidative Stress , Propionates , Tannins , Animals , Female , Nitro Compounds/toxicity , Propionates/toxicity , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Oxidative Stress/drug effects , Tannins/pharmacology , Autophagy/drug effects , Apoptosis/drug effects , Antioxidants/pharmacology , Signal Transduction/drug effects , Polyphenols
9.
Toxicol Appl Pharmacol ; 485: 116910, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38521372

ABSTRACT

3-nitropropionic acid (3-NPA), a toxic metabolite produced by mold, is mainly found in moldy sugarcane. 3-NPA inhibits the activity of succinate dehydrogenase that can induce oxidative stress injury in cells, reduce ATP production and induce oxidative stress in mouse ovaries to cause reproductive disorders. Ursolic acid (UA) has a variety of biological activities and is a pentacyclic triterpene compound found in many plants. This experiment aimed to investigate the cytotoxicity of 3-NPA during mouse oocyte in vitro maturation and the protective effects of UA on oocytes challenged with 3-NPA. The results showed that UA could alleviate 3-NPA-induced oocyte meiotic maturation failure. Specifically, 3-NPA induced a decrease in the first polar body extrusion rate of oocytes, abnormal distribution of cortical granules, and an increase in the proportion of spindle abnormalities. In addition, 3-NPA caused mitochondrial dysfunction and induced oxidative stress, including decreases in the GSH, mitochondrial membrane potential and ATP levels, and increases in the ROS levels, and these effects led to apoptosis and autophagy. The addition of UA could significantly improve the adverse effects caused by 3-NPA. In general, our data show that 3-NPA affects the normal development of oocytes during the in vitro culture, and the addition of UA can effectively repair the damage caused by 3-NPA to oocytes.


Subject(s)
Meiosis , Nitro Compounds , Oocytes , Oxidative Stress , Propionates , Triterpenes , Ursolic Acid , Animals , Nitro Compounds/toxicity , Propionates/toxicity , Oocytes/drug effects , Oocytes/metabolism , Female , Meiosis/drug effects , Mice , Triterpenes/pharmacology , Oxidative Stress/drug effects , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Autophagy/drug effects , Adenosine Triphosphate/metabolism , Mice, Inbred ICR
10.
Neurotoxicology ; 102: 12-28, 2024 May.
Article in English | MEDLINE | ID: mdl-38453033

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative condition characterized by a severe motor incoordination, cognitive decline, and psychiatric complications. However, a definitive cure for this devastating disorder remains elusive. Agmatine, a biogenic amine, has gain attention for its reported neuromodulatory and neuroprotective properties. The present study was designed to examine the influence of agmatine on the behavioral, biochemical, and molecular aspects of HD in an animal model. A mitochondrial toxin, 3-nitro propionic acid (3-NP), was used to induce HD phenotype and similar symptoms such as motor incoordination, memory impairment, neuro-inflammation, and depressive-like behavior in rats. Rats were pre-treated with 3-NP (10 mg/kg, i.p.) on days 1, 3, 5, 7, and 9 and then continued on agmatine treatment (5 - 20 µg/rat, i.c.v.) from day-8 to day-27 of the treatment protocol. 3-NP-induced cognitive impairment was associated with declined in agmatine levels within prefrontal cortex, striatum, and hippocampus. Further, the 3-NP-treated rats showed an increase in IL-6 and TNF-α and a reduction in BDNF immunocontent within these brain areas. Agmatine treatment not only improved the 3-NP-induced motor incoordination, depression-like behavior, rota-rod performance, and learning and memory impairment but also normalized the GABA/glutamate, BDNF, IL-6, and TNF-α levels in discrete brain areas. Similarly, various agmatine modulators, which increase the endogenous agmatine levels in the brain, such as L-arginine (biosynthetic precursor), aminoguanidine (diamine oxidase inhibitor), and arcaine (agmatinase inhibitor) also demonstrated similar effects exhibiting the importance of endogenous agmatinergic pathway in the pathogenesis of 3-NP-induced HD like symptoms. The present study proposed the possible role of agmatine in the pathogenesis and treatment of HD associated motor incoordination, and psychiatric and cognitive complications.


Subject(s)
Agmatine , Huntington Disease , Nitro Compounds , Propionates , Animals , Nitro Compounds/toxicity , Propionates/toxicity , Agmatine/pharmacology , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/drug therapy , Male , Rats , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Rats, Wistar , Disease Models, Animal , Motor Activity/drug effects , Rats, Sprague-Dawley , Neuroprotective Agents/pharmacology
11.
Chem Biol Interact ; 393: 110957, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38513929

ABSTRACT

Huntington's disease (HD) is an inheritable autosomal-dominant disorder that targets mainly the striatum. 3-Nitropropionic acid (3-NP) induces obvious deleterious behavioral, neurochemical, and histological effects similar to the symptoms of HD. Our study aimed to examine the neuroprotective activity of tropisetron, an alpha-7 neuronal nicotinic acetylcholine receptor (α-7nAChR) agonist, against neurotoxic events associated with 3-NP-induced HD in rats. Forty-eight rats were randomly allocated into four groups. Group I received normal saline, while Groups II, III and IV received 3-NP for 2 weeks. In addition, Group III and IV were treated with tropisetron 1 h after 3-NP administration. Meanwhile, Group IV received methyllycaconitine (MLA), an α-7nAChR antagonist, 30 min before tropisetron administration. Treatment with tropisetron improved motor deficits as confirmed by the behavioral tests and restored normal histopathological features of the striatum. Moreover, tropisetron showed an anti-oxidant activity via increasing the activities of SDH and HO-1 as well as Nrf2 expression along with reducing MDA level. Tropisetron also markedly upregulated the protein expression of p-PI3K and p-Akt which in turn hampered JAK2/NF-κB inflammatory cascade. In addition, tropisetron showed an anti-apoptotic activity through boosting the expression of Bcl-2 and reducing Bax expression and caspase-3 level. Interestingly, all the aforementioned effects of tropisetron were blocked by pre-administration of MLA, which confirms that such neuroprotective effects are mediated via activating of α-7nAChR. In conclusion, tropisetron showed a neuroprotective activity against 3-NP-induced HD via activating PI3K/Akt signaling and suppressing JAK2/NF-κB inflammatory axis. Thus, repositioning of tropisetron could represent a promising therapeutic strategy in management of HD.


Subject(s)
Huntington Disease , Neuroprotective Agents , Receptors, Nicotinic , Animals , Rats , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Huntington Disease/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , NF-kappa B/metabolism , Nitro Compounds/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Propionates/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Nicotinic/metabolism , Signal Transduction , Tropisetron/therapeutic use
12.
Biomedicines ; 12(3)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38540238

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disease that causes progressive motor and cognitive dysfunction. There is no cure for HD, and current therapeutics can only manage the signs and symptoms as well as slowing disease progression. This investigation examines the possible therapeutic advantages of europinidin in 3-nitropropionic acid (3-NPA) injected HD in rats. Wistar rats were randomly assigned to five groups (n = 6): normal control, 3-NPA (10 mg/kg, i.p.), 3-NPA + europinidin-10 (10 mg/kg, p.o.), 3-NPA + europinidin-20 (20 mg/kg, p.o.), and europinidin alone (20 mg/kg, p.o.) for 15-day. Various behavioral and biochemical parameters including antioxidant levels, oxidative stress, pro-inflammatory markers, mitochondrial enzyme complex, and neurotransmitters were assessed. Europinidin restored biochemical, mitochondrial dysfunction, oxidative stress, neurotransmitter, and pro-inflammatory parameters disrupted by 3-NPA. Here we show that europinidin attenuates 3-NPA-induced neurodegeneration in rat models of HD. Europinidin modulates oxidative stress, enhances antioxidants, restores mitochondrial enzyme complex activity, reduces neuroinflammation, and modulates neurotransmitter levels. Our findings reveal the potential of europinidin as a novel therapeutic agent for the treatment of HD. This study also provides new insights into the molecular mechanisms of europinidin-mediated neuroprotection and may have a beneficial role in the management of neurological diseases.

13.
J Huntingtons Dis ; 13(1): 55-66, 2024.
Article in English | MEDLINE | ID: mdl-38489193

ABSTRACT

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.


Subject(s)
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
14.
Molecules ; 29(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38398528

ABSTRACT

Kaempferol, a flavonoid present in many food products, has chemical and cellular antioxidant properties that are beneficial for protection against the oxidative stress caused by reactive oxygen and nitrogen species. Kaempferol administration to model experimental animals can provide extensive protection against brain damage of the striatum and proximal cortical areas induced by transient brain cerebral ischemic stroke and by 3-nitropropionic acid. This article is an updated review of the molecular and cellular mechanisms of protection by kaempferol administration against brain damage induced by these insults, integrated with an overview of the contributions of the work performed in our laboratories during the past years. Kaempferol administration at doses that prevent neurological dysfunctions inhibit the critical molecular events that underlie the initial and delayed brain damage induced by ischemic stroke and by 3-nitropropionic acid. It is highlighted that the protection afforded by kaempferol against the initial mitochondrial dysfunction can largely account for its protection against the reported delayed spreading of brain damage, which can develop from many hours to several days. This allows us to conclude that kaempferol administration can be beneficial not only in preventive treatments, but also in post-insult therapeutic treatments.


Subject(s)
Brain Injuries , Ischemic Stroke , Neuroprotective Agents , Nitro Compounds , Propionates , Stroke , Animals , Kaempferols/pharmacology , Brain , Oxidative Stress , Stroke/drug therapy , Ischemia/drug therapy , Brain Injuries/drug therapy , Reperfusion , Ischemic Stroke/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
15.
Neurochem Res ; 49(4): 1017-1033, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38184805

ABSTRACT

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.


Subject(s)
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
16.
Behav Brain Res ; 461: 114864, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38220060

ABSTRACT

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.


Subject(s)
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
17.
Mol Neurobiol ; 61(2): 609-621, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37648841

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disease characterized by neuropsychiatric disturbance, cognitive impairment, and locomotor dysfunction. In the early stage (chorea) of HD, expression of dopamine D2 receptors (D2R) is reduced, whereas dopamine (DA) levels are increased. Contrary, in the late stage (bradykinesia), DA levels and the expression of D2R and dopamine D1 receptors (D1R) are reduced. 3-Nitropropionic acid (3-NPA) is a toxin that may replicate HD behavioral phenotypes and biochemical aspects. This study assessed the neurotransmitter levels, dopamine receptor gene expression, and the effect of acute exposure to quinpirole (D2R agonist) and eticlopride (D2R antagonist) in an HD model induced by 3-NPA in adult zebrafish. Quinpirole and eticlopride were acutely applied by i.p. injection in adult zebrafish after chronic treatment of 3-NPA (60 mg/kg). 3-NPA treatment caused a reduction in DA, glutamate, and serotonin levels. Quinpirole reversed the bradykinesia and memory loss induced by 3-NPA. Together, these data showed that 3-NPA acts on the dopaminergic system and causes biochemical alterations similar to late-stage HD. These data reinforce the hypothesis that DA levels are linked with locomotor and memory deficits. Thus, these findings may suggest that the use of DA agonists could be a pharmacological strategy to improve the bradykinesia and memory deficits in the late-stage HD.


Subject(s)
Dopamine , Neurodegenerative Diseases , Nitro Compounds , Propionates , Salicylamides , Animals , Dopamine/metabolism , Quinpirole/pharmacology , Zebrafish/metabolism , Hypokinesia , Receptors, Dopamine D2/metabolism , Dopamine Agonists/pharmacology , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Receptors, Dopamine D1/metabolism
18.
Naunyn Schmiedebergs Arch Pharmacol ; 397(4): 2447-2463, 2024 04.
Article in English | MEDLINE | ID: mdl-37847410

ABSTRACT

3-Nitropropionic acid (3-NP) is strongly believed to be an irreversible inhibitor of mitochondrial complex II, leading to neural damage. This study aimed to investigate the neuroprotective effects of silymarin against 3-NP-induced neurotoxicity in male mice. Six-week-old mice received subacute doses of 3-NP intraperitoneally for 17 days. Mice were given silymarin (70 mg/kg/day, P.O.) for 2 weeks before 3-NP administration or for 4 weeks after 3-NP administration. At the end of the treatment schedule, animals were evaluated for behavioral alterations. Subsequently, neuronal damage in the hippocampus region of the brain tissues, oxidative stress-related parameters (lipid peroxidation, nitric oxide, superoxide dismutase, glutathione, and total antioxidant capacity), and pro-inflammatory cytokine (TNF-α, IL-17, and IL-1ß) levels were evaluated. Our results indicated that 3-NP treatment significantly (p < 0.05) tended to reduce motor coordination, memory, and neuronal antioxidant status while increasing pro-inflammatory cytokine levels. However, silymarin in both treatment and pretreatment protocols markedly (p < 0.05) attenuated the behavioral deficits, oxidative stress status, and neuroinflammation. The results of the current study suggest that the neuroprotective effect of silymarin against 3-NP-induced neurotoxicity might be due to the mitigation of oxidative stress status and provide insight into the therapeutic potential of silymarin.


Subject(s)
Neuroprotective Agents , Neurotoxicity Syndromes , Nitro Compounds , Silymarin , Rats , Male , Mice , Animals , Neuroprotective Agents/pharmacology , Antioxidants/pharmacology , Rats, Wistar , Silymarin/pharmacology , Neuroinflammatory Diseases , Body Weight , Motor Activity , Oxidative Stress , Neurotoxicity Syndromes/drug therapy , Propionates/pharmacology , Cytokines
19.
Life Sci ; 338: 122362, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38141855

ABSTRACT

AIMS: Endoplasmic reticulum stress (ERS) with aberrant mitochondrial-ER contact (MERC), mitophagy, and apoptosis are interconnected determinants in neurodegenerative diseases. Previously, we proved the potential of Morin hydrate (MH), a potent antioxidant flavonoid, to mitigate Huntington's disease (HD)-3-nitropropionic acid (3-NP) model by modulating glutamate/calpain/Kidins220/BDNF trajectory. Extending our work, we aimed to evaluate its impact on combating the ERS/MERC, mitophagy, and apoptosis. METHODS: Rats were subjected to 3-NP for 14 days and post-treated with MH and/or the ERS inducer WAG-4S for 7 days. Disease progression was assessed by gross inspection and striatal biochemical, histopathological, immunohistochemical, and transmission electron microscopical (TEM) examinations. A molecular docking study was attained to explore MH binding to mTOR, JNK, the kinase domain of IRE1-α, and IP3R. KEY FINDINGS: MH decreased weight loss and motor dysfunction using open field and rotarod tests. It halted HD degenerative striatal neurons and nucleus/mitochondria ultra-microscopic alterations reflecting neuroprotection. Mechanistically, MH deactivated striatal mTOR/IRE1-α/XBP1s&JNK/IP3R, PINK1/Ubiquitin/Mfn2, and cytochrome c/caspase-3 signaling pathways, besides enhancing p-PGC-1α and p-VDAC1. WAG-4S was able to ameliorate all effects initiated by MH to different extents. Molecular docking simulations revealed promising binding patterns of MH and hence its potential inhibition of the studied proteins, especially mTOR, IP3R, and JNK. SIGNIFICANCE: MH alleviated HD-associated ERS, MERC, mitophagy, and apoptosis. This is mainly achieved by combating the mTOR/IRE1-α signaling, IP3R/VDAC hub, PINK1/Ubiquitin/Mfn2, and cytochrome c/caspase 3 axis to be worsened by WAG-4S. Molecular docking simulations showed the promising binding of MH to mTOR and JNK as novel identified targets.


Subject(s)
Flavones , Huntington Disease , Mitophagy , Animals , Rats , Apoptosis , Cytochromes c , Flavones/pharmacology , Huntington Disease/metabolism , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins , Molecular Docking Simulation , Phosphoproteins , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases , Ubiquitins/metabolism
20.
J Appl Microbiol ; 134(12)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37960923

ABSTRACT

AIMS: This study aimed to investigate the inhibitory effect of tannic acid (TA) on the growth of Apiospora arundinis and 3-Nitropropionic acid (3-NPA) production. METHODS AND RESULTS: To investigate the antifungal mechanism, the effects of TA on the hypha growth, electrical conductivity, hypha morphology, defense-related enzymes, and 3-NPA production of A. arundinis were studied. TA concentrations of 640 and 1280 µg ml-1 exhibited strong antifungal activity against A. arundinis. The results of scanning electron microscopy and transmission electron microscopy showed that the hypha of the A. arundinis was severely deformed after TA treatment, and the cell membrane was blurred and thin, vacuoles were obviously shrunken and smaller, and most of the organelles were decomposed into irregular fragments. The increased electrical conductivity and malondialdehyde content indicated that TA caused peroxidation of unsaturated fatty acids and damaged the structure of the cell membrane. The decrease of intracellular ATPase and succinate dehydrogenase content indicated that TA damaged the function of mitochondria, and participated in the inhibition of respiratory metabolism. In addition, TA significantly reduced 3-NPA production and completely inhibited 3-NPA production at 640 and 1280 µg ml-1. CONCLUSION: TA effectively inhibited both growth of A. arundinis in vitro and 3-NPA production.


Subject(s)
Antifungal Agents , Mitochondria , Antifungal Agents/pharmacology , Propionates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL