Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.171
Filter
1.
Neurotherapeutics ; 21(5): e00444, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39353831

ABSTRACT

This study aims to investigate whether butylphthalide can inhibit ferroptosis and ameliorate cerebral ischaemia-reperfusion (I/R) injury in rats by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) / heme oxygenase-1 (HO-1) signalling pathway, known for its antioxidative and cytoprotective properties. Middle cerebral artery occlusion reperfusion (MCAO/R) rat models were established. Male rats were randomly divided into five groups: a sham-operated group (sham), MCAO/R group, MCAO/R â€‹+ â€‹ML385 (Nrf2-specific inhibitor) group, MCAO/R â€‹+ â€‹NBP (butylphthalide) group and MCAO/R â€‹+ â€‹ML385 â€‹+ â€‹NBP group. The effect of butylphthalide on cerebral I/R injury was evaluated using neurological deficit scores. The expression levels of Nrf2, HO-1, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long-chain family member 4 (ACSL4) and transferrin receptor 1 (TfR1) protein were detected using Western blot. Moreover, the expression levels of GPX4, HO-1 and TfR1 mRNA were determined through real-time fluorescence quantitative reverse transcription polymerase chain reaction. The distribution of Nrf2, HO-1, GPX4 and TfR1 was detected using immunohistochemical staining. The levels of iron and related lipid peroxidation indexes, such as reduced glutathione, reactive oxygen species, malondialdehyde and nitric oxide, were measured using a kit. The changes in mitochondria were observed through transmission electron microscopy. Butylphthalide treatment significantly improved neurological dysfunction, reduced cerebral infarction volume and mitigated histopathological damage in MCAO/R rats. It induced the nuclear translocation of Nrf2 and upregulated HO-1 expression, which was attenuated by ML385. Butylphthalide also attenuated lipid peroxidation, iron accumulation and mitochondrial damage induced by MCAO/R. The expression of GPX4, ACSL4 and TfR1 proteins, as well as their mRNA levels, was modulated through butylphthalide treatment, with improvements observed in mitochondrial morphology. Butylphthalide exerts neuroprotective effects by attenuating neurological dysfunction and ferroptosis in MCAO/R rats through the activation of the Nrf2/HO-1 pathway and inhibition of lipid peroxidation and iron accumulation.


Subject(s)
Benzofurans , Ferroptosis , NF-E2-Related Factor 2 , Rats, Sprague-Dawley , Reperfusion Injury , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Male , Benzofurans/pharmacology , Benzofurans/therapeutic use , Ferroptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Rats , Signal Transduction/drug effects , Heme Oxygenase-1/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Heme Oxygenase (Decyclizing)/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism
2.
Clin Exp Pharmacol Physiol ; 51(11): e13922, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39348933

ABSTRACT

Neuropathic pain arises from impairments or malfunctions within the nervous system, resulting in atypical transmission and interpretation of pain signals. In the present study, we examined the neuroprotective effects of agomelatine (AGM) and agomelatine-loaded nanostructured lipid carriers (AGM-NLCs) in neuropathic animal models induced by chronic constriction injury (CCI) of the sciatic nerve. Male Sprague Dawley rats were divided into seven experimental groups to compare the effects of AGM and AGM-NLCs, which were administered at 20 mg/kg for 14 consecutive days after CCI. Our finding demonstrated that CCI triggered the onset of analgesia in these animals, corroborated by mechanical allodynia and thermal hyperalgesia. Furthermore, CCI induced an elevation in inflammatory mediators such as interleukin (IL)-1ß and inducible nitric oxide synthase (iNOS), and downregulated heme oxygenase-1 (HO-1) and nuclear factor E2-related factor (Nrf2). Treatment with AGM and AGM-NLCs reversed inflammatory cascades and elevated antioxidant enzyme levels, leading to a reduction in paw withdrawal latency and threshold in rats. To further investigate the effect of AGM and AGM-NLCs, all-trans retinoic acid (ATRA) was administered, which antagonizes Nrf2. ATRA substantially downregulated Nrf2 expression and exacerbated thermal hyperalgesia, whereas Nrf2 and HO-1 expressions were significantly upregulated after AGM-NLCs administration. Overall, the results demonstrated that AGM-NLCs offer promising antinociceptive and anti-inflammatory properties in alleviating neuropathic pain symptoms, which can be attributed to improved drug delivery and therapeutic outcomes compared with AGM alone.


Subject(s)
Acetamides , Drug Carriers , Lipids , NF-E2-Related Factor 2 , Nanostructures , Neuralgia , Rats, Sprague-Dawley , Signal Transduction , Animals , Neuralgia/drug therapy , Neuralgia/metabolism , Male , NF-E2-Related Factor 2/metabolism , Rats , Acetamides/pharmacology , Acetamides/administration & dosage , Signal Transduction/drug effects , Nanostructures/chemistry , Drug Carriers/chemistry , Hyperalgesia/drug therapy , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Naphthalenes
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167496, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39237046

ABSTRACT

Liver ischemia-reperfusion (I/R) injury is a detrimental complication of organ transplantation, shock, and sepsis. However, the available drugs to mitigate I/R injury remain limited. Jujuboside A (JuA) is renowned for its antioxidant, anti-inflammatory, and anti-apoptotic properties; nevertheless, its potential in liver I/R injury remains unknown. Thus, this study aimed to explore the role and underlying mechanisms of JuA in liver I/R injury. Mouse models of I/R and AML12 cell models of hypoxia/reoxygenation (H/R) were constructed. Haematoxylin and eosin staining, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) detection, and cell viability analysis were used to assess liver injury. To evaluate oxidative stress, inflammation, apoptosis, and mitochondrial damage, immunofluorescence staining, transmission electron microscopy analysis, enzyme-linked immunosorbent assay, and flow cytometry were conducted. Moreover, molecular docking techniques and western blot were employed to identify downstream target molecules and pathways affected by JuA. The results showed that JuA pretreatment effectively attenuated liver necrosis and ALT and AST level elevations induced by I/R while enhancing AML12 cell viability following H/R. Furthermore, JuA pretreatment suppressed oxidative stress triggered by I/R and H/R, thereby inhibiting the level of pro-inflammatory factors and NLRP3 inflammasome activation. Notably, JuA pretreatment alleviated mitochondrial damage and apoptosis. Mechanistically, JuA pretreatment resulted in the activation of the AKT/NRF2/HO-1 signalling pathways, whereas MK2206, the inhibitor of AKT, partially reversed the hepatoprotective effects of JuA during liver I/R. Collectively, our findings illustrated that JuA mitigated oxidative stress, inflammation, apoptosis, and mitochondrial damage by facilitating the AKT/NRF2/HO-1 signalling pathway, thereby alleviating liver I/R injury.


Subject(s)
Apoptosis , Liver , NF-E2-Related Factor 2 , Proto-Oncogene Proteins c-akt , Reperfusion Injury , Signal Transduction , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/drug therapy , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Signal Transduction/drug effects , Male , Liver/pathology , Liver/metabolism , Liver/drug effects , Apoptosis/drug effects , Oxidative Stress/drug effects , Mice, Inbred C57BL , Heme Oxygenase-1/metabolism , Cell Line , Membrane Proteins/metabolism , Heme Oxygenase (Decyclizing)/metabolism
4.
Mar Drugs ; 22(9)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39330276

ABSTRACT

Cyclophosphamide (CP) is an anticancer drug that causes infertility disorders. This study was designed to evaluate a nanoformulation of chitosan with an ethanolic extract from Spirulina platensis in terms of its protection against cyclophosphamide-induced ovarian toxicity. Nine groups of female Wistar rats were randomly assigned as follows: 1: control vehicle, 2: chitosan polymer, 3: telmisartan, 4: Spirulina platensis extract, 5: nanoformulation of the Spirulina platensis, and 6: single injection of CP; groups 7, 8, and 9 received the same treatments as those used in groups 3, 4, and 5, respectively, with a single dose of CP (200 mg/kg, I.P). The results displayed that the CP treatment decreased estradiol, progesterone, anti-mullerian hormone, and GSH content, and it downregulated PPAR-γ, Nrf-2, and HO-1 gene expression. In addition, the CP treatment caused an increase in the FSH, LH, and MDA levels. In the same manner, the protein expression of caspase-3, NF-kB, and TNF-α was upregulated in response to the CP treatment, while PPAR-γ was downregulated in comparison with the control. The rats treated with SPNPs exhibited a substantial reduction in the detrimental effects of oxidative stress and inflammation of the ovarian tissue. This study's conclusions showed that SPNPs counteracted the effects of CP, preventing the death of ovarian follicles and restoring the gonadotropin hormone balance and normal ovarian histological appearance.


Subject(s)
Chitosan , Cyclophosphamide , NF-E2-Related Factor 2 , NF-kappa B , Ovary , PPAR gamma , Tumor Necrosis Factor-alpha , Animals , Female , Rats , Chitosan/chemistry , Chitosan/pharmacology , Cyclophosphamide/toxicity , Ethanol/chemistry , Heme Oxygenase (Decyclizing)/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Ovary/drug effects , Ovary/pathology , Ovary/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Rats, Wistar , Signal Transduction/drug effects , Spirulina , Tumor Necrosis Factor-alpha/metabolism
5.
Int Immunopharmacol ; 142(Pt A): 113113, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39276459

ABSTRACT

Severe acute pancreatitis (SAP) is a prevalent acute inflammatory disease that is clinically manifested by systemic inflammation dysregulation, resulting in a significantly elevated mortality rate. Bufalin has been verified to have potent pharmacological properties, including analgesic, anti-tumor and anti-inflammatory effects. However, it remains unclear whether bufalin inhibits SAP. Thus, we aim to explore the impact of bufalin in SAP rats and to evaluate the potential mechanisms of action. In addition to analyzing serum biochemistry and pancreatic tissue pathology, we elucidated its mechanisms of action through enzyme-linked immunosorbent assay (ELISA), immunohistochemical analysis, Western blot, and quantitative real-time PCR. The results demonstrated that bufalin dose-dependently reversed the elevation of serum Amylase (Amy) and Lipase (LPS) levels in SAP rats, alleviating pancreatic tissue pathological damage. Bufalin exhibited potent antioxidant effects by reducing malondialdehyde (MDA) levels, decreasing Superoxide dismutase (SOD) and glutathione(GSH) consumption, inhibiting the interaction of Keap1-Nrf2, and increasing HO-1 expression. Furthermore, bufalin inhibited TNF-α, IL-6, IL-1ß, p-NF-κB-p65, p-IκBα, and NF-κB-p65 expression, while enhancing IκBα expression, ultimately confirming its anti-inflammatory effects on SAP. In summary, our findings suggest that bufalin exerts anti-inflammatory and antioxidant actions in NaT-SAP rats by inhibiting NF-κB and activating the Keap1-Nrf2/HO-1 pathway. This study represents the inaugural application of bufalin in NaT-induced SAP rats, indicating its potential as an effective therapeutic agent for SAP patients.


Subject(s)
Anti-Inflammatory Agents , Bufanolides , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Pancreatitis , Rats, Sprague-Dawley , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Pancreatitis/drug therapy , Pancreatitis/chemically induced , Pancreatitis/pathology , Oxidative Stress/drug effects , NF-kappa B/metabolism , Male , Bufanolides/pharmacology , Bufanolides/therapeutic use , Signal Transduction/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Rats , Cytokines/metabolism , Pancreas/pathology , Pancreas/drug effects , Pancreas/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Disease Models, Animal , Humans , Amylases/blood
6.
Int Immunopharmacol ; 142(Pt B): 113227, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39321704

ABSTRACT

Ferroptosis, characterized by iron-dependent accumulation of lipid peroxides, plays an important role in spinal cord injury (SCI). Berberine (BBR), as a lipid peroxide scavenger, has been widely used in treating other diseases; however, its role in ferroptosis has not been fully elucidated. Therefore, here, to test our hypothesis that BBR can reduce the severity of SCI and promote motor function recovery by inhibiting neuronal ferroptosis, we evaluated the changes in ferroptosis-related indicators after BBR administration by establishing a cellular ferroptosis model and an SCI contusion model. We found that BBR administration significantly reduces lipid peroxidation damage, maintains normal mitochondrial function, reduces excessive accumulation of iron ions, enhances antioxidant capacity, and activates the ferroptosis defense system in vivo and in vitro. Mechanistically, BBR alleviates neuronal ferroptosis by inducing adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and up-regulating nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) protein expression to promote glutathione production. BBR administration also significantly improves motor function recovery in SCI rats. Meanwhile, applying the AMPK inhibitor Compound C blocks the neuroprotective and all other effects of BBR. Collectively, our findings demonstrate that BBR can attenuate neuronal ferroptosis after SCI by activating the AMPK-NRF2-HO-1 pathway.


Subject(s)
AMP-Activated Protein Kinases , Berberine , Ferroptosis , NF-E2-Related Factor 2 , Neurons , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord Injuries , Animals , Berberine/pharmacology , Berberine/therapeutic use , Ferroptosis/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Rats , AMP-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Heme Oxygenase-1/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Disease Models, Animal
7.
Int Immunopharmacol ; 142(Pt B): 113222, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39321702

ABSTRACT

Obstructive sleep apnea (OSA) is a common clinical condition linked to cognitive impairment, mainly characterized by chronic intermittent hypoxia (CIH). GLP-1 receptor agonist, known for promoting insulin secretion and reducing glucose levels, has demonstrated neuroprotective effects in various experimental models such as stroke, Alzheimer's disease, and Parkinson's disease. This study aims to investigate the potential role and mechanisms of the GLP-1 receptor agonist liraglutide in ameliorating OSA-induced cognitive deficits. CIH exposure, a well-established and mature OSA pathological model, was used both in vitro and in vivo. In vitro, CIH significantly activated oxidative stress, inflammation, and apoptosis in SH-SY5Y cells. Liraglutide enhanced the nuclear translocation of Nrf2, activating its downstream pathways, thereby mitigating CIH-induced injury in SH-SY5Y cells. Additionally, liraglutide modulated the MAPK/NF-κB signaling pathway, reducing the expression of inflammatory factors and proteins. In vivo, we subjected mice to an intermittent hypoxia incubator to mimic the pathogenesis of human OSA. The Morris water maze test revealed that CIH exposure substantially impaired spatial memory. Subsequent western blot analyses and histopathological examinations indicated that liraglutide could activate the Nrf2/HO-1 axis and inhibit the MAPK/NF-κB signaling pathway, thereby alleviating OSA-associated cognitive dysfunction in mice. These findings suggest that GLP-1 receptor agonists may offer a promising preventive strategy for OSA-associated cognitive impairment. By refining these findings, we provide new insights into GLP-1's protective mechanisms in combating cognitive deficits associated with CIH, underscoring its potential as a therapeutic agent for conditions linked to OSA.


Subject(s)
Apoptosis , Cognitive Dysfunction , Hypoxia , Liraglutide , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Signal Transduction , Sleep Apnea, Obstructive , Liraglutide/pharmacology , Liraglutide/therapeutic use , Animals , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Humans , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Apoptosis/drug effects , Male , NF-kappa B/metabolism , Mice , Sleep Apnea, Obstructive/drug therapy , Sleep Apnea, Obstructive/complications , Hypoxia/drug therapy , Hypoxia/complications , Hypoxia/metabolism , Signal Transduction/drug effects , Mice, Inbred C57BL , Disease Models, Animal , Cell Line, Tumor , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Heme Oxygenase-1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Membrane Proteins
8.
J Biochem Mol Toxicol ; 38(10): e23863, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39318027

ABSTRACT

Arsenic is a toxic environmental pollutant heavy metal, and one of its critical target tissues in the body is the liver. Carvacrol is a natural phytocompound that stands out with its antioxidant, anti-inflammatory, and antiapoptotic properties. The current study aims to investigate the protective feature of carvacrol against sodium arsenite-induced liver toxicity. Thirty-five Sprague-Dawley male rats were divided into five groups: Control, Sodium arsenite (SA), CRV, SA + CRV25, and SA + CRV50. Sodium arsenite was administered via oral gavage at a dose of 10 mg/kg for 14 days, and 30 min later, CRV 25 or 50 mg/kg was administered via oral gavage. Oxidative stress, inflammation, apoptosis, autophagy damage pathways parameters, and liver tissue integrity were analyzed using biochemical, molecular, western blot, histological, and immunohistological methods. Carvacrol decreased sodium arsenite-induced oxidative stress by suppressing malondialdehyde levels and increasing superoxide dismutase, catalase, glutathione peroxidase activities, and glutathione levels. Carvacrol reduced inflammation damage by reducing sodium arsenite-induced increased levels of NF-κB and the cytokines (TNF-α, IL-1ß, IL-6, RAGE, and NLRP3) it stimulates. Carvacrol also reduced sodium arsenite-induced autophagic (Beclin-1, LC3A, and LC3B) and apoptotic (P53, Apaf-1, Casp-3, Casp-6, Casp-9, and Bax) parameters. Carvacrol preserved sodium arsenite-induced impaired liver tissue structure. Carvacrol alleviated toxic damage by reducing sodium arsenite-induced increases in oxidative stress, inflammation, apoptosis, and autophagic damage parameters in rat liver tissues. Carvacrol was also beneficial in preserving liver tissue integrity.


Subject(s)
Arsenites , Caspase 3 , Chemical and Drug Induced Liver Injury , Cymenes , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Sodium Compounds , Animals , Male , Rats , Sodium Compounds/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cymenes/pharmacology , Arsenites/toxicity , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 3/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Beclin-1/metabolism , Receptor for Advanced Glycation End Products/metabolism , Heme Oxygenase (Decyclizing)/metabolism , bcl-2-Associated X Protein/metabolism , Signal Transduction/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Oxidative Stress/drug effects
9.
Int Immunopharmacol ; 142(Pt B): 113179, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39298824

ABSTRACT

Triphala is renowned for its curative attributes and has been utilized for centuries to address diverse health ailments. Moreover, the active component of Triphala, polyphenols, is widely recognized for its excellent pharmacological activities, such as anti-inflammatory properties, and has been utilized as a potential natural remedy. However, the precise mechanism through which Triphala alleviates cognitive dysfunction and anxiety induced by chronic sleep deprivation (SD) remains restricted. The objective of this investigation is to examine and clarify the potential mechanism of action that underlies the therapeutic benefits of Triphala in addressing cognitive dysfunction and anxiety induced by chronic SD. Our results demonstrated that Triphala significantly alleviates chronic SD-induced behavioral abnormalities. Additionally, Triphala was highly effective at preventing histopathological or morphological damage to neurons located in the hippocampus. The therapeutic effects of Triphala in treating cognitive dysfunction and anxiety induced by chronic SD involve the modulation of several biological pathways, including inflammation and immune responses, oxidative stress, cell growth and differentiation, metabolism, and neurotransmitter communication. Moreover, our study illustrated that Triphala increased the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and significantly activated the Nrf2/hemeoxygenase-1 (HO-1) axis. Additionally, the neuroprotective properties of Triphala were found to be counteracted by the Nrf2 inhibitor ML385. Our study represented the first to unveil that Triphala exerts therapeutic benefits in alleviating chronic SD-induced cognitive deficits and anxiety by activation of the Nrf2/HO-1 axis. Triphala emerges as a promising nutraceutical ingredient for mitigating cognitive deficits and anxiety linked to chronic SD.


Subject(s)
Anxiety , Cognitive Dysfunction , NF-E2-Related Factor 2 , Sleep Deprivation , Animals , NF-E2-Related Factor 2/metabolism , Anxiety/drug therapy , Cognitive Dysfunction/drug therapy , Sleep Deprivation/drug therapy , Male , Mice , Signal Transduction/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice, Inbred C57BL , Membrane Proteins/metabolism , Oxidative Stress/drug effects , Disease Models, Animal , Heme Oxygenase-1/metabolism , Plant Extracts/therapeutic use , Plant Extracts/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Humans
10.
Mol Biol Rep ; 51(1): 1015, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39325219

ABSTRACT

BACKGROUND: The adverse effects of radiotherapy (RT) primarily occur through oxidative stress, and attempts are being made to mitigate these effects. L-Carnitine (L-Car) involved in physiological functions, possesses antioxidant and tissue-protective properties. The goal of this investigation is to appraise the radioprotective efficacy of L-Car supplementation. METHODS AND RESULTS: The groups were established by dividing thirty-two rats as: control, RT (10 Gy), RT + L-Car (200 mg/kg/d), L-Car. Upon completion of the experiment, the livers were harvested for histopathological, immunostaining [tumor necrosis factor-alpha (TNF-α), Caspase-3], spectrophotometric [total oxidant status (TOS), total antioxidant status (TAS), oxidative stress index (OSI)], and mRNA expression [(Nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein 1 (Keap-1), Heme Oxygenase (HO-1), Transforming growth factor beta 1 (TGF-ß1)] analyses. In the damage group, decreased Keap-1, Nrf2, HO-1, and TAS values, along with increased histopathological findings, alanine transferase, aspartate transferase, TNF-α, Caspase-3, TOS, OSI, TGF-ß1 levels were found. All findings were improved with L-Car treatment. CONCLUSIONS: Considering these findings, it can be inferred that L-Car exhibits tissue-protective effects against organ damage predominantly induced by RT-related oxidative stress. Additionally, it has prevented the development of inflammation, apoptosis, and fibrosis. Therefore, L-Car may be considered as a supplement to reduce complications associated with RT.


Subject(s)
Antioxidants , Carnitine , Dietary Supplements , Liver , NF-E2-Related Factor 2 , Oxidative Stress , Animals , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Carnitine/pharmacology , Rats , Liver/drug effects , Liver/metabolism , Liver/radiation effects , Liver/pathology , NF-E2-Related Factor 2/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Male , Radiation-Protective Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Caspase 3/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Heme Oxygenase-1/metabolism , Rats, Wistar , Apoptosis/drug effects , Apoptosis/radiation effects
12.
J Ethnopharmacol ; 335: 118671, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39103024

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rheumatoid arthritis (RA) is a multifactorial, polygenic inflammatory disease. Mesua assamica (King & Prain) Kosterm. (MA) is an endangered medicinal plant indigenous to South Asia, primarily to Assam in India. The tree bark is claimed to possess anti-inflammatory, anti-diabetic, anti-cancer, and anti-malarial properties; nevertheless, its role in RA has not been elucidated. Hence, this study aims to investigate the in-vitro and in-vivo anti-arthritic effects of Mesua assamica bark ethanolic extract (MAE). AIM OF THE STUDY: This study aims to investigate the anti-rheumatic potential of MAE in-vitro on RAW 264.7 cells for its anti-oxidant and anti-inflammatory activities and in-vivo on the CFA-induced adjuvant arthritis in the rat model. MATERIALS AND METHODS: We investigated the possible therapeutic effects of MAE in-vitro using RAW 264.7 cells triggered by LPS. Meanwhile, adult Wistar rats were injected intradermally with 100 µl of CFA to induce arthritis, and they were given MAE orally at doses of 100 and 200 mg/kg for up to 28 days. Paw volume analysis, X-ray radiography, anti-oxidant levels analysis, gene and protein expression studies, and histological analysis were carried out to assess the effects of MAE in-vivo. RESULTS: MAE significantly mitigated the inflammation by reducing ROS levels and dropped the nitrite, PGE2, and COX-2 levels enhanced by LPS in-vitro. At the same time, MAE treatment reduced the paw and joint inflammation and increased the immune organ index in the CFA rats. Histopathology data revealed that MAE mitigated the CFA-induced lesions of the ankle joints and synovial tissues. Similarly, MAE significantly abated the secretion of pro-inflammatory cytokines, inhibited the protein expression of TLR4, NF-кB, COX-2, and iNOS, as well as improved the Nrf2 and HO-1 levels in-vitro and in-vivo. CONCLUSION: All the results highlighted the anti-rheumatic potential of MAE in RA in-vitro and in-vivo by inhibiting the TLR4/NF-кB/COX-2/iNOS and promoting the Nrf2/HO-1 signaling axis.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Cyclooxygenase 2 , Ethanol , NF-E2-Related Factor 2 , NF-kappa B , Plant Bark , Plant Extracts , Toll-Like Receptor 4 , Animals , Plant Extracts/pharmacology , RAW 264.7 Cells , Mice , Plant Bark/chemistry , Toll-Like Receptor 4/metabolism , NF-E2-Related Factor 2/metabolism , Arthritis, Rheumatoid/drug therapy , NF-kappa B/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Ethanol/chemistry , Cyclooxygenase 2/metabolism , Male , Rats , Rats, Wistar , Down-Regulation/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Antioxidants/pharmacology , Signal Transduction/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Antirheumatic Agents/pharmacology , Antirheumatic Agents/isolation & purification , Heme Oxygenase-1/metabolism , Membrane Proteins
13.
Phytomedicine ; 133: 155941, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128305

ABSTRACT

BACKGROUND: Ulcerative colitis (UC), a chronic idiopathic inflammatory bowel disease (IBD), presents with limited current drug treatment options. Consequently, the search for safe and effective drug for UC prevention and treatment is imperative. Our prior studies have demonstrated that the phenolic compound p-Hydroxybenzaldehyde (HD) from Nostoc commune, effectively mitigates intestinal inflammation. However, the mechanisms underlying HD's anti-inflammatory effects remain unclear. PURPOSE: This study delved into the pharmacodynamics of HD and its underlying anti-inflammation mechanisms. METHODS: For in vivo experiments, dextran sodium sulfate (DSS)-induced colitis mouse model was established. In vitro inflammation model was established using lipopolysaccharide (LPS)-induced RAW264.7 and bone marrow-derived macrophages (BMDMs). The protective effect of HD against colitis was determined by monitoring clinical symptoms and histological morphology in mice. The levels of inflammatory factors and oxidative stress markers were subsequently analyzed with enzyme-linked immunosorbent assay (ELISA) and biochemical kits. Furthermore, western blotting (WB), immunofluorescence (IF), luciferase reporter gene, drug affinity reaction target stability (DARTS) assay, molecular docking, and molecular dynamics (MD) simulation were used to determine the potential target and molecular mechanism of HD. RESULTS: Our findings indicate that HD significantly alleviated the clinical symptoms and histological morphology of colitis in mice, and curtailed the production of pro-inflammatory cytokines, including TNF-α, IL-6, IFN-γ, COX-2, and iNOS. Furthermore, HD stimulated the production of SOD, CAT, and GSH-px, enhanced total antioxidant capacity (T-AOC), and reduced MDA levels. Mechanically, HD augmented the expression of Nrf2, HO-1, and NQO-1, while concurrently downregulating the phosphorylation of p65, IκBα, c-Jun, and c-Fos. ML385 and siNrf2 largely attenuated the protective effect of HD in enteritis mice and RAW 264.7 cells, as well as the promotion of HO-1 expression levels. ZnPP-mediated HO-1 knockdown reversed HD-induced inhibition of colonic inflammation. Luciferase reporter assay and IF assay confirmed the transcriptional activation of Nrf2 by HD. DARTS analysis, molecular docking, and MD results showed high binding strength, interaction efficiency and remarkable stability between Nrf2 and HD. CONCLUSION: These outcomes extend our previous research results that HD can combat oxidative stress through the Nrf2/HO-1/NQO-1/NF-κB/AP-1 pathways, effectively alleviating colitis, and propose new targets for HD to protect against intestinal barrier damage.


Subject(s)
Benzaldehydes , Dextran Sulfate , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Transcription Factor AP-1 , Animals , NF-E2-Related Factor 2/metabolism , Mice , Benzaldehydes/pharmacology , Oxidative Stress/drug effects , NF-kappa B/metabolism , RAW 264.7 Cells , Transcription Factor AP-1/metabolism , Male , Anti-Inflammatory Agents/pharmacology , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/metabolism , Colitis/drug therapy , Colitis/chemically induced , Disease Models, Animal , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Signal Transduction/drug effects , Heme Oxygenase-1/metabolism , Lipopolysaccharides , Heme Oxygenase (Decyclizing)/metabolism , Membrane Proteins/metabolism
14.
J Biochem Mol Toxicol ; 38(9): e23817, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39177155

ABSTRACT

Heavy metal contamination is an alarming concern on a global scale, as drinking tainted water significantly increases human susceptibility to heavy metals. In a realistic scenario, humans are often exposed to a combination of harmful chemicals rather than a single toxicant. Phloretin (PHL), biochanin-A (BCA), and coenzyme Q10 (CoQ10) are bioactive compounds owning plentiful pharmacological properties. Henceforth, the current research explored the putative energizing effects of selected nutraceuticals in combined chromium (Cr) and arsenic (As) intoxicated Swiss albino mice. Potassium dichromate (75 ppm) and sodium meta-arsenite (100 ppm) were given in the drinking water to induce hepatotoxicity, conjugated with PHL and BCA (50 mg/kg each), and CoQ10 (10 mg/kg) intraperitoneally for 2 weeks. After the statistical evaluation, it was observed that the hepato-somatic index, metal load, and antioxidant activity (lipid peroxidation and protein carbonyl content) increased along with the concomitant decrease in the antioxidants (catalase, glutathione-S-transferase, superoxide dismutase, reduced glutathione, and total thiol) in the Cr and As intoxicated mice. Additionally, light microscopy observations, DNA breakages, decreased silent information regulator 1 (SIRT1), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), heme oxygenase (HO-1), and NAD(P)H quinone dehydrogenase 1 (NQO1) gene expressions, together with stimulated apoptotic cell death manifested by the increased expressions of caspase 8 and caspase 3, thus, proved consistency with the aforementioned outcomes. Importantly, the treatment with nutraceuticals not only restored the antioxidant activity but also favorably altered the expressions of SIRT1, Nrf2, HO-1, and NQO1 signaling and apoptosis markers. These findings highlight the crucial role of the PHL, BCA, and CoQ10 combination in reducing Cr and As-induced hepatotoxicity in mice. By averting the triggered apoptosis in conjunction with oxidative stress, this combination increases the SIRT1, Nrf2, HO-1, and NQO1 signaling, thereby reassuringly maintaining the cellular equilibrium.


Subject(s)
Apoptosis , Chromium , Genistein , Liver , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Oxidative Stress , Phloretin , Signal Transduction , Sirtuin 1 , Ubiquinone , Animals , Sirtuin 1/metabolism , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Genistein/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Chromium/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Phloretin/pharmacology , Signal Transduction/drug effects , Male , Arsenic/toxicity , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins
15.
Dokl Biochem Biophys ; 518(1): 442-451, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39196533

ABSTRACT

Acute kidney injury (AKI), formerly known as acute renal failure, refers to a sudden and often reversible decline in kidney function. Inflammatory reaction and oxidative stress play a crucial role in the expansion of renal disease. In this experimental study, we scrutinized the renal protective effect of umbelliferone against gentamicin induced renal injury in the rats and explore the mechanism. Wistar rats were used in this study and Gentamicin was used for the induction the AKI in the rats and rats were received the oral administration of umbelliferone. The body weight, organ weight, renal, oxidative stress, cytokines, inflammatory parameters were estimated. The mRNA expression caspase-3, Bax, Bcl-2, TNF-α, IL-1ß, IL-6, IL-10, HO-1, and Nrf2 were estimated. Umbelliferone remarkably improved the body weight and altered the absolute and relative weight of hepatic and renal tissue. Umbelliferone significantly suppressed the level of BUN, Scr, magnesium, calcium, phosphorus, sodium, and potassium along with altered the level of oxidative stress parameters like CAT, SOD, GSH, LPO, and GPx. Umbelliferone altered the level of cytokines viz., TNF-α, Il-1ß, IL-6, IL-10; inflammatory parameters like PGE2, COX-2, TGF-ß, NF-κB, respectively. Umbelliferone significantly altered the mRNA expression of caspase-3, Bax, Bcl-2, TNF-α, IL-1ß, IL-6, IL-10, HO-1, and Nrf2. The result showed the renal protective effect of umbelliferone against gentamycin induced renal disease via alteration of HO-1/Nrf2 and NF-κB Signaling Pathway.


Subject(s)
Acute Kidney Injury , NF-E2-Related Factor 2 , NF-kappa B , Rats, Wistar , Signal Transduction , Umbelliferones , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/pathology , Umbelliferones/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Rats , Male , Oxidative Stress/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Cytokines/metabolism , Cytokines/genetics , Gentamicins/adverse effects
16.
J Biochem Mol Toxicol ; 38(9): e23809, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39148263

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is an alarming ailment that leads to severe liver damage and increases the risk of serious health conditions. The prevalence of NAFLD due to oxidative stress could be mitigated by plant-derived antioxidants. This study aims to investigate the effects of syringic acid (SA) on NAFLD in a high-fat diet (HFD) rat model. Twenty-four rats were randomly divided into four groups (n = 6): normal control, HFD, SA-administered HFD, and positive control SA on a normal diet. Rats in the normal control and positive control groups received a normal diet, and the remaining groups received an HFD for 8 weeks. SA (20 mg/kg b.w.) was orally (gavage) administered for 8 weeks. Lipid profiles were controlled by SA against HFD-fed rats (p < 0.05). SA reduced the serum aspartate aminotransferase and alanine aminotransferase levels by 70%-190%. SA also suppressed pro-inflammatory cytokines and attenuated histopathological and immunohistochemical changes against HFD-fed rats. SA reversed oxidative stress by suppressing the malondialdehyde formation by 82% and replenished the nonenzymatic and enzymatic antioxidant activities (p < 0.05). Gene expressions of nuclear factor-erythroid 2-related factor/heme oxygenase 1 (Nrf2/HO-1) were elevated in SA-treated rats. Ameliorative effects of SA on NAFLD induced by an HFD in rats were prominent through the reversal of oxidative stress and inflammation, regulated by an intrinsic mechanism of defense against oxidative stress, the Nrf2/HO-1 pathway.


Subject(s)
Gallic Acid , Heme Oxygenase (Decyclizing) , NF-E2-Related Factor 2 , Non-alcoholic Fatty Liver Disease , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/prevention & control , Rats , Male , Signal Transduction/drug effects , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Oxidative Stress/drug effects , Heme Oxygenase-1/metabolism , Diet, High-Fat/adverse effects , Rats, Sprague-Dawley , Antioxidants/pharmacology , Liver/metabolism , Liver/drug effects , Liver/pathology
17.
Peptides ; 180: 171279, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39053647

ABSTRACT

AIMS: It has been reported that some peptides released by the gastro-intestinal tract play important roles in the prevention of myocardial ischemia/reperfusion injury (MIRI). Bombesin (BN) is a biologically active peptide released by non-adrenergic non-cholinergic nerves on the gastric antrum mucosa controlled by the vagus nerve. However, there is a lack of reports on the impact of BN on MIRI. This study aimed to explore the influence of BN on MIRI and its underlying mechanism. MATERIALS AND METHODS: MIRI was induced by either 30 min of global ischemia in Langendorff perfused rat hearts, or by ligation of the descending coronary artery for 30 min in anesthetized Spraque-Dawley rats, and both were followed by 120 min reperfusion. Infarct size and left ventricular function were assessed, and lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH) levels were measured spectrophotometrically, while cardiomyocyte apoptosis was detected by TUNEL assay. The content of BN in plasma was measured with enzyme-linked immunosorbent assays (ELISA). The expression of caspase 3, Kelch-like ECH-associated protein 1 (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) were quantified. KEY FINDINGS: BN and vagus nerve stimulation improved cardiac contractile function and reduced myocardial infarct size, attenuated oxidative stress damage and myocardial cell apoptosis, increased the expression of Keap1, Nrf2, and HO-1. and these effects were blocked by using a BN receptor antagonist. SIGNIFICANCE: BN provides protection against MIRI, and its underlying mechanism is through activation of the Keap1/Nrf2/HO-1 pathway. This research provides more reliable evidence for the "gut-heart axis dialogue" and explores potential therapeutic approaches for MIRI.


Subject(s)
Bombesin , Kelch-Like ECH-Associated Protein 1 , Myocardial Reperfusion Injury , NF-E2-Related Factor 2 , Rats, Sprague-Dawley , Signal Transduction , Animals , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Rats , Signal Transduction/drug effects , Male , Bombesin/pharmacology , Bombesin/analogs & derivatives , Heme Oxygenase (Decyclizing)/metabolism , Apoptosis/drug effects , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects
18.
Arch Biochem Biophys ; 758: 110084, 2024 08.
Article in English | MEDLINE | ID: mdl-38971420

ABSTRACT

Nuclear factor erythroid factor 2 (Nrf2) is the key regulatory of the antioxidant response elements. Also, Nrf2 interacts with nuclear factor kappa B (NF-ĸB) to inhibit subsequent inflammatory cascade. Activation of Nrf2 signaling ameliorates drug-induced liver injury. Sodium valproate (SVP) is an anti-epilepsy drug with a hepatotoxic adverse effect that restricts its clinical use. In this study, coadministration of Dihydromyricetin (DHM), a natural flavonoid, with SVP to rats upregulated gene expression of Nrf2 and its downstream gene, heme oxygenase 1 (HO-1), while suppressed the Nrf2 repressor, Keap-1. Additionally, DHM led to downregulation of proinflammatory factors in liver tissues, including NF-ĸB, interleukin 1 beta (IL-1ß), and tumor necrosis factor alpha (TNF-α). This was accompanied by a decrease in the proapoptotic protein (cleaved caspase-3) expression level. Furthermore, biochemical and histopathological studies showed that DHM treatment improved liver function and lipid profile while decreased inflammatory cell infiltration, congestion, and hepatocellular damage. According to our knowledge, prior research has not examined the protective effect of DHM on the liver injury induced by SVP. Consequently, this study provides DHM as a promising herbal medication that, when used with SVP, can prevent its induced hepatotoxicity owing to its potential anti-oxidative, anti-inflammatory, and anti-apoptotic properties.


Subject(s)
Caspase 3 , Chemical and Drug Induced Liver Injury , Flavonols , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , NF-kappa B , Signal Transduction , Valproic Acid , Animals , NF-E2-Related Factor 2/metabolism , Male , Signal Transduction/drug effects , Flavonols/pharmacology , NF-kappa B/metabolism , Valproic Acid/pharmacology , Rats , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Caspase 3/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Rats, Sprague-Dawley , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism
19.
Cell Tissue Res ; 397(3): 193-204, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38953985

ABSTRACT

Cisplatin nephrotoxicity is a well-known emergency clinical condition caused by oxidative stress and inflammation. Naringin (NAR) is considered an antioxidant agent with renoprotective effects capable of removing reactive oxygen species. Adipose tissue-derived mesenchymal stem cells (AD-MSCs) are reported to have anti-inflammatory and antioxidant properties. The present research examined the renoprotective effect of the combination of NAR and AD-MSCs as opposed to each one alone on cisplatin-induced nephrotoxicity through SIRT-1/Nrf-2/HO-1 pathway. This study included five groups (n = 8 each) of male Sprague-Dawley rats (200 - 220 g): sham, cisplatin: rats receiving cisplatin (6.5 mg/kg, i.p.) on the 4th day; NAR+cisplatin: rats pretreated with NAR (1 week, i.p.) + cisplatin on the 4th day; AD-MSCs: rats receiving AD-MSCs (1 × 106) by injection through the tail vein on the 5th day + cisplatin on the 4th day; and NAR+AD-MSCs+cisplatin. On the 8th day, the animals were anesthetized to obtain tissue and blood samples. Biochemical factors, inflammation, oxidative stress, and gene expression were explored. Cisplatin increased blood urea nitrogen, creatinine, inflammation, and oxidative stress. Moreover, mRNA expression of Sirtuin1, nuclear factor erythroid 2-related factor 2 (Nrf-2), and heme oxygenase-1 (HO-1) remarkably reduced. Furthermore, cisplatin led to a disturbance in kidney structure (glomerular atrophy, cell infiltrations, and tubular dysfunction) as confirmed by histology findings. However, NAR pretreatment, AD-MSC administration, or a combination of both significantly reversed these changes. Overall, when used together, NAR and AD-MSCs had stronger cisplatin-induced effects on kidney dysfunction by inhibiting inflammation, reducing oxidative stress, and increasing the Sirtuin1/Nrf-2/HO-1 pathway.


Subject(s)
Adipose Tissue , Cisplatin , Flavanones , Mesenchymal Stem Cells , NF-E2-Related Factor 2 , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1 , Animals , Male , Rats , Adipose Tissue/metabolism , Cisplatin/pharmacology , Cisplatin/toxicity , Flavanones/pharmacology , Flavanones/therapeutic use , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Sirtuin 1/metabolism
20.
Int Immunopharmacol ; 139: 112774, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39067398

ABSTRACT

Repetitive traumatic brain injury (RTBI) is acknowledged as a silent overlooked public health crisis, with an incomplete understanding of its pathomechanistic signaling pathways. Mounting evidence suggests the involvement of thrombin and its receptor, the protease-activated receptor (PAR)1, in the development of secondary injury in TBI; however, the consequences of PAR1 modulation and its impact on ferroptosis-redox signaling, and NLRP3 inflammasome activation in RTBI, remain unclear. Further, the utilitarian function of PAR1 as a therapeutic target in RTBI has not been elucidated. To study this crosstalk, RTBI was induced in Wistar rats by daily weight drops on the right frontal region for five days. Three groups were included: normal control, untreated RTBI, and RTBI+SCH79797 (a PAR1 inhibitor administered post-trauma at 25 µg/kg/day). The concomitant treatment of PAR1 antagonism improved altered behavior function, cortical histoarchitecture, and neuronal cell survival. Moreover, the receptor blockade downregulated mRNA expression of PAR1 but upregulatedthat of the neuroprotective receptor PPAR-γ. The anti-inflammatory impact of SCH79797 was signified by the low immune expression/levels of NF-κB p65,TNF-α, IL-1ß, and IL-18. Consequently, the PAR1 blocker hindered the formation of inflammasome components NLRP3, ASC, and activated caspase-1. Ultimately, SCH79797 treatment abated ferroptosis-dependent iron redox signaling through the activation of the antioxidant Nrf2/HO-1 axis and its subsequent antioxidant machinery (GPX4, SOD) to limit lipid peroxidation, iron accumulation, and transferrin serum increment. Collectively, SCH79797 offered putative preventive mechanisms against secondary RTBI consequences in rats by impeding ferroptosis and NLRP3 inflammasome through activating the PPAR-γ/Nrf2 antioxidant cue.


Subject(s)
Brain Injuries, Traumatic , Ferroptosis , Inflammasomes , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , PPAR gamma , Rats, Wistar , Receptor, PAR-1 , Signal Transduction , Animals , Male , Rats , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Ferroptosis/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/genetics , Inflammasomes/metabolism , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , PPAR gamma/metabolism , Receptor, PAR-1/metabolism , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL