Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Saudi Pharm J ; 31(11): 101818, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37868646

RESUMO

The potential health benefits of phytochemicals in preventing and treating diseases have gained increasing attention. Here, we proved that the methylated isoflavone prunetin possesses a reno-therapeutic effect against renal ischemia/reperfusion (I/R) insult by activating G protein-coupled receptor 30 (GPR30). After choosing the therapeutic dose of prunetin against renal I/R injury in the pilot study, male Sprague Dawley rats were allocated into 5 groups; viz., sham-operated (SO), SO injected with 1 mg/kg prunetin intraperitoneally for three successive days, untreated I/R, I/R treated with prunetin, and I/R treated with G-15, the selective GPR30 blocker, followed by prunetin. Treatment with prunetin reversed the I/R renal injury effect and majorly restored normal renal function and architecture. Mechanistically, prunetin restored the I/R-induced depletion of renal GPR30, an impact that was canceled by the pre-administration of G-15. Additionally, post-administration of prunetin normalized the boosted inflammatory markers indoxyl sulfate, TLR4, and TRIF and abrogated renal cell demise by suppressing necroptotic signaling, verified by the inactivation of p-RIPK1, p-RIPK3, and p-MLKL while normalizing the inhibited caspase-8. Besides, prunetin reversed the I/R-mediated mitochondrial fission by inhibiting the protein expression of PGMA5 and p-DRP-1. All these favorable impacts of prunetin were nullified by G-15. To sum up, prunetin exhibited a significant reno-therapeutic effect evidenced by the enhancement of renal morphology and function, the suppression of the inflammatory cascade indoxyl sulfate/TLR4/TRIF, which turns off the activated/phosphorylated necroptotic trajectory RIPK1/RIPK3/MLKL, while enhancing caspase-8. Additionally, prunetin opposed the mitochondrial fission pathway RIPK3/PGMA5/DRP-1, effects that are mediated via the activation of GPR30.

2.
BMC Neurosci ; 23(1): 11, 2022 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-35247984

RESUMO

BACKGROUND: Mitoxantrone has proved efficacy in treatment of multiple sclerosis (MS). The fact that physical exercise could slow down the progression of disease and improve performance is still a debatable issue, hence; we aimed at studying whether combining mitoxantrone with exercise is of value in the management of MS. METHODS: Thirty-six male rats were divided into sedentary and exercised groups. During a 14-day habituation period rats were subjected to exercise training on a rotarod (30 min/day) before Experimental Autoimmune Encephalomyelitis (EAE) induction and thereafter for 17 consecutive days. On day 13 after induction, EAE groups (exercised &sedentary) were divided into untreated and mitoxantrone treated ones. Disease development was evaluated by motor performance and EAE score. Cerebrospinal fluid (CSF) was used for biochemical analysis. Brain stem and cerebellum were examined histopathological and immunohistochemically. RESULTS: Exercise training alone did not add a significant value to the studied parameters, except for reducing Foxp3 immunoreactivity in EAE group and caspase-3 in the mitoxantrone treated group. Unexpectedly, exercise worsened the mitoxantrone effect on EAE score, Bcl2 and Bax. Mitoxantrone alone decreased EAE/demyelination/inflammation scores, Foxp3 immunoreactivity, and interleukin-6, while increased the re-myelination marker BDNF without any change in tumor necrosis factor-α. It clearly interrupted the apoptotic pathway in brain stem, but worsened EAE mediated changes of the anti-apoptotic Bcl2 and pro-apoptotic marker Bax in the CSF. CONCLUSIONS: The neuroprotective effect of mitoxantrone was related with remyelination, immunosuppressive and anti-inflammatory potentials. Exercise training did not show added value to mitoxantrone, in contrast, it disrupts the apoptotic pathway.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fatores de Transcrição Forkhead , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitoxantrona/farmacologia , Esclerose Múltipla/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2 , Ratos , Proteína X Associada a bcl-2
3.
Saudi Pharm J ; 30(1): 72-90, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35145347

RESUMO

Excessive interleukin (IL)-6 production is a driver for malignancy and drug resistance in colorectal cancer (CRC). Our study investigated a seven-week post-treatment with the anti-inflammatory drug, Diacerein (Diac), alone or in combination with 5-fluorouracil (5-FU), using a 1,2-dimethylhydrazine (DMH) rat model of CRC. Diac alone and 5-FU+Diac reduced serum levels of carcino-embryonic antigen (CEA), while all regimens decreased serum levels of colon cancer-specific antigen (CCSA), a more specific CRC biomarker. Additionally, Diac, 5-FU and their combination suppressed colonic content/gene expression of IL-6, its downstream oncogene, Kirsten rat sarcoma viral oncogene homolog (K-Ras), and consequently Notch intracellular domain and nuclear factor-kappa B (NF-κB) p65. In turn, NF-κB downstream factors, viz., matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF), c-Myc, and B-cell lymphoma-2 (Bcl-2) were also downregulated, while E-cadherin was elevated. Additionally, the drugs reduced the immunoreactivity of CD31 to prove their anti-angiogenic effect, while the TUNEL assay confirmed the apoptotic effect. The apoptotic effect was confirmed by transferase dUTP nick-end labeling assay. Moreover, these drugs inhibited colon content of p-Akt, ß-catenin, and cyclin D1 immunoreactivity. The drugs also activated the tumor suppressor glycogen synthase kinase 3- ß (GSK3-ß) and upregulated the expression of the Nur77 gene, which represents the second arm of IL-6 signaling. However, only 5-FU upregulated miR-200a, another K-Ras downstream factor. The in-vitro cytotoxic and migration/invasion assays verified the molecular trajectories. Accordingly, we evaluated the antineoplastic effect of Diac alone and its possible chemosensitization effect when added to 5-FU. This combination may target critical oncogenic pathways, including the IL-6/K-Ras/Notch/NF-κB p65 axis, p-Akt/GSK3-ß/ß-catenin/cyclin D-1 hub, and Nur77.

4.
Toxicol Appl Pharmacol ; 426: 115635, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34174262

RESUMO

The beneficial role of prasugrel, a P2Y12 receptor blocker, in several neurointerventional procedures has been reviewed clinically. Beyond its antiplatelet capacity, the potential neuroprotective mechanisms of prasugrel are poorly addressed experimentally. Relevant to the imbalance between neuro-inflammation and neuroprotective pathways in cerebral ischemia/reperfusion (I/R), our study evaluated the anti-ischemic potential of prasugrel treatment through tackling novel targets. Male Wistar rats were allocated into 2 sets; set 1 (I/R 60 min/3 days) to assess the neurological deficits/biochemical impact of prasugrel and set 2 (I/R 60 min/5 days) for evaluating short memory/morphological/immunoreactive changes. Each set comprised 4 groups designated as sham, sham + prasugrel, I/R, and I/R + prasugrel. Post-administration of prasugrel for 3 and 5 days reduced neurological deficit scores and improved the spontaneous activity/short term spatial memory using the Y-maze paradigm. On the molecular level, prasugrel turned off SUMO2/3-inhibitory kappa (Iκ)Bα, Ubc9 and nuclear factor kappa (NF-κ)B. Besides, it inhibited malondialdehyde (MDA) and inactivated astrocytes by downregulating the glial fibrillary acidic protein (GFAP) hippocampal immune-expression. Conversely, it activated its target molecule cAMP, protein kinase (PK)A, and cAMP response element-binding protein (CREB) to enhance the brain-derived nuclear factor (BDNF) hippocampal content. Additionally, cAMP/PKA axis increased the hippocampal content of deacetylator silent information regulator 1 (SIRT1) and the micro RNA (miR)-22 gene expression. The crosstalk between these paths partakes in preserving hippocampal cellularity. Accordingly, prasugrel, regardless inhibiting platelets activity, modulated other cellular components; viz., SUMO2/3-IκBα/Ubc9/NF-κB, cAMP/PKA related trajectories, CREB/BDNF and SIRT1/miR-22 signaling, besides inhibiting GFAP and MDA to signify its anti-ischemic potential.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Cloridrato de Prasugrel/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Isquemia Encefálica/sangue , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , MicroRNAs/sangue , Inibidor de NF-kappaB alfa/metabolismo , Fármacos Neuroprotetores/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Cloridrato de Prasugrel/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos Wistar , Sirtuína 1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Memória Espacial/efeitos dos fármacos , Enzimas de Conjugação de Ubiquitina/metabolismo
5.
J Pharmacol Sci ; 146(3): 136-148, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34030796

RESUMO

Despite the documented renoprotective effect of pentoxifylline (PTX), a non-selective phosphodiesterase-4 inhibitor, the studies appraised only its anti-inflammatory/-oxidant/-apoptotic capacities without assessment of the possible involved trajectories. Here, we evaluated the potential role of galectin-3 and the ASK-1/NF-κB p65 signaling pathway with its upstream/downstream signals in an attempt to unveil part of the cascades involved in the renotherapeutic effect using a renal bilateral ischemia/reperfusion (I/R) model. Rats were randomized into sham-operated, renal I/R (45 min/72 h) and I/R + PTX (100 mg/kg; p.o). Post-treatment with PTX improved renal function and abated serum levels of cystatin C, creatinine, BUN and renal KIM-1 content, effects that were reflected on an improvement of the I/R-induced renal histological changes. On the molecular level, PTX reduced renal contents of galectin-3, ASK-1 with its downstream molecule JNK and ERK1/2, as well as NF-κB p65 and HMGB1. This inhibitory effect extended also to suppress neutrophil infiltration, evidenced by diminishing ICAM-1 and MPO, as well as inflammatory cytokines (TNF-α/IL-18), oxidative stress (MDA/TAC), and caspase-3. The PTX novel renotherapeutic effect involved in part the inhibition of galectin-3 and ASK-1/JNK and ERK1/2/NF-κB/HMGB-1 trajectories to mitigate renal I/R injury and to provide basis for its anti-inflammatory, antioxidant, and anti-apoptotic impacts.


Assuntos
Galectina 3/metabolismo , Proteína HMGB1/metabolismo , Rim/irrigação sanguínea , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases , Pentoxifilina/farmacologia , Pentoxifilina/uso terapêutico , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfodiesterase/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Animais , Anti-Inflamatórios , Antioxidantes , Modelos Animais de Doenças , Masculino , Ratos Wistar
6.
Molecules ; 26(16)2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34443654

RESUMO

Since westernized diet-induced insulin resistance is a risk factor in Alzheimer's disease (AD) development, and lipopolysaccharide (LPS) coexists with amyloid ß (Aß)1-42 in these patients, our AD novel model was developed to resemble sporadic AD by injecting LPS into high fat/fructose diet (HFFD)-fed rats. The neuroprotective potential of palonosetron and/or methyllycaconitine, 5-HT3 receptor and α7 nAChR blockers, respectively, was evaluated after 8 days of daily administration in HFFD/LPS rats. All regimens improved histopathological findings and enhanced spatial memory (Morris Water Maze); however, palonosetron alone or with methyllycaconitine promoted animal performance during novel object recognition tests. In the hippocampus, all regimens reduced the expression of glial fibrillary acidic protein and skewed microglia M1 to M2 phenotype, indicated by the decreased M1 markers and the enhanced M2 related parameters. Additionally, palonosetron and its combination regimen downregulated the expression of ASC/TMS1, as well as levels of inflammasome downstream molecules and abated cleaved caspase-1, interleukin (IL)-1ß, IL-18 and caspase-11. Furthermore, ACh and 5-HT were augmented after being hampered by the insult. Our study speculates that blocking 5-HT3 receptor using palonosetron overrides methyllycaconitine to combat AD-induced neuroinflammation and inflammasome cascade, as well as to restore microglial function in a HFFD/LPS novel model for sporadic AD.


Assuntos
Aconitina/análogos & derivados , Doença de Alzheimer/tratamento farmacológico , Inflamação/tratamento farmacológico , Palonossetrom/farmacologia , Aconitina/farmacologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Cognição/efeitos dos fármacos , Dieta Ocidental/efeitos adversos , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/patologia , Resistência à Insulina/genética , Interleucina-18/genética , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Microglia/patologia , Fragmentos de Peptídeos/genética , Piroptose/efeitos dos fármacos , Ratos , Receptores 5-HT3 de Serotonina/genética , Fatores de Risco , Memória Espacial/efeitos dos fármacos
7.
Toxicol Appl Pharmacol ; 409: 115309, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33130049

RESUMO

The antimalarial drug artesunate (Art) has proven its beneficial effects against ischemia/reperfusion (I/R) injury in diverse organs, but its potential role against hepatic I/R is still obscure. This study, hence, examined whether treatment with Art alone or in combination with rapamycin (Rapa), an mTOR inhibitor, can ameliorate hepatic I/R injury via targeting the NLRP3 inflammasome signaling pathway. Rats were divided into hepatic sham- and I/R-operated rats. The latter were either left untreated (I/R group) or treated with Art, Rapa, or their combination. On the molecular level, all treatment regimens succeeded to hinder inflammasome assembly and activation, assessed as NLRP3, ASC, cleaved caspase-1, caspase-11, N-terminal cleaved gasdermin-D (GSDMD-N), IL-1ß, and IL-18. This effect was associated by the inhibition in the harmful signaling pathways HMGB1/RAGE and TLR4/MyD88/TRAF6 to inactivate the transcription factor NF-κB and the production of its pro-inflammatory cytokines IL-1ß, IL-18, IL-6, and TNF-α. Additionally, this effect entailed the inhibition of ICAM-1/MPO/ROS cascade, which in turn hampered cell demise induced by apoptosis, manifested as correction of the imbalanced Bcl2/Bax, as well as pyroptosis (LDH, cleaved caspase-1, caspase-11, GSDMD-N, IL-1ß, and IL-18), and necrosis. The corrected pathways were reflected on the improved liver function (serum ALT, AST, and LDH) and microscopical hepatic architecture. Noteworthy, the effect of Art on all parameters exceeded significantly that of Rapa and even improved the effect of the latter in the combination group. In conclusion, our results suggest novel roles for Art in abating functional and structural I/R-induced hepatic abnormalities via several traversing cross-talking pathways that succeeded to abate NLRP3 inflammasome and cell death.


Assuntos
Artesunato/farmacologia , Inflamassomos/deficiência , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Hepatopatias/metabolismo , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Toxicol Appl Pharmacol ; 394: 114956, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32171571

RESUMO

Proper enterocytic proliferation/differentiation, besides providing adequate adherens junctions (AJ) integrity, are responsible for strengthening of the gut barrier that acts as a first line defense against endotoxemia. However, the preferential role of the underlying PI3K/Akt (PKB) axis in triggering enterocytic proliferation/differentiation signaling and AJ assembly is still obscure in sepsis. Additionally, the potential involvement of dipeptidyl peptidase (DPP)-IV in cholestatic sepsis has not yet been reported. Common bile duct ligation (CBDL) insult was performed in adult male Sprague-Dawley rats except for sham operated animals; three doses of vildagliptin (VLD3, 10 and 30 mg/kg/d; p.o) were administered for 10 consecutive days post CBDL. VLD3/10/30 dose-dependently decreased DPP-IV and elevated GLP-1, IGF-1, PI3K, pS473-Akt (PKB), pS9-GSK-3ß, pS133-CREB and cyclin-D1. VLD3/10 reduced fever, portal/aortic endotoxin and IgG, body weight loss as well as ileal NF-κB, TNF-α, MPO, TBARS, subepithelial/pericryptal and submucosal collagen deposition, vimentin immunoreactivity, N-cadherin, Zeb1 and pY654-ß-catenin but increased E-cadherin, NPSH and colon/spleen indices - effects that were quite the opposite of VLD30. Accordingly, maintaining proper enterocytic proliferation/differentiation and phosphorylation inputs consequent to adequate DPP-IV inhibition is integral to AJ assembly in cholestatic sepsis; however, perturbed signals by excessive suppression of the enzyme activity induce toxic effects manifested as AJ disassembly and EMT, hence gut leakage and overt endotoxemia.


Assuntos
Colestase/patologia , Inibidores da Dipeptidil Peptidase IV/farmacologia , Enterócitos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Sepse/patologia , Animais , Diferenciação Celular/efeitos dos fármacos , Colestase/tratamento farmacológico , Ducto Colédoco/lesões , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Íleo/patologia , Ligadura , Masculino , Ratos , Ratos Sprague-Dawley , Sepse/tratamento farmacológico , Vildagliptina/farmacologia , Vimentina/metabolismo , Redução de Peso/efeitos dos fármacos
9.
J Pediatr Gastroenterol Nutr ; 69(6): 710-718, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31764439

RESUMO

OBJECTIVES: Alanylglutamine (AG) is a dipeptide that fuels enterocytes and has a coadjuvant role during gut healing. The current study aimed to investigate the potential ulcer-healing effect of AG in indomethacin-induced gastropathy. METHODS: Animals (n = 10 rats/group) were randomly allocated into 5 groups. Gastric ulcerated rats were administered AG, AG + dexamethasone, or pantoprazole after indomethacin exposure. RESULTS: Comparable to pantoprazole, AG inhibited H-KATPase pump, and elevated the pH of gastric juice. Moreover, the dipeptide increased the serum/mucosal contents of glucagon-like peptide-1 (GLP-1), pS473-Akt, and cyclin-D1. On the contrary, AG abated serum tumor necrosis factor-α and gastric mucosal content of pS45-ß catenin, pS9-GSK3ß, pS133-CREB, pS536-NF-κB, H2O2, claudin-1, and caspase-3. The administration of dexamethasone before AG hampered its effect on almost all the measured parameters. CONCLUSIONS: AG confers its antiulcerogenic/antisecretory potentials by repressing the proton pump to increase the gastric juice pH via boosting p-CREB, p-Akt, p-GSK-3ß, and GLP-1. Also, it inhibits apoptosis through suppressing nuclear factor-kappa B/tumor necrosis factor-α/H2O2/claudin-1 cue. This trajectory contributes to loosen the tight junction priming AG-mediated GLP-1/ß-catenin/cyclin-D1 that results in pronounced increase in gastric mucosa proliferation. Therefore, the crosstalk between multiple pathways orchestrates the action of AG against gastric ulceration.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Dipeptídeos/farmacologia , Mucosa Gástrica/efeitos dos fármacos , Úlcera Gástrica/patologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Dexametasona/metabolismo , Dexametasona/farmacologia , Dipeptídeos/administração & dosagem , Dipeptídeos/metabolismo , Humanos , Indometacina/efeitos adversos , Indometacina/farmacologia , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Úlcera Gástrica/tratamento farmacológico , Cicatrização/efeitos dos fármacos
10.
Molecules ; 24(10)2019 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-31137621

RESUMO

Insulin resistance is a major risk factor for Alzheimer's disease (AD). Chenodeoxycholic acid (CDCA) and synthetic Farnesoid X receptor (FXR) ligands have shown promising outcomes in ameliorating insulin resistance associated with various medical conditions. This study aimed to investigate whether CDCA treatment has any potential in AD management through improving insulin signaling. Adult male Wistar rats were randomly allocated into three groups and treated for six consecutive weeks; control (vehicle), AD-model (AlCl3 50 mg/kg/day i.p) and CDCA-treated group (AlCl3 + CDCA 90 mg/kg/day p.o from day 15). CDCA improved cognition as assessed by Morris Water Maze and Y-maze tests and preserved normal histological features. Moreover, CDCA lowered hippocampal beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and amyloid-beta 42 (Aß42). Although no significant difference was observed in hippocampal insulin level, CDCA reduced insulin receptor substrate-1 phosphorylation at serine-307 (pSer307-IRS1), while increased protein kinase B (Akt) activation, glucose transporter type 4 (GLUT4), peroxisome proliferator-activated receptor gamma (PPARγ) and glucagon-like peptide-1 (GLP-1). Additionally, CDCA activated cAMP response element-binding protein (CREB) and enhanced brain-derived neurotrophic factor (BDNF). Ultimately, CDCA was able to improve insulin sensitivity in the hippocampi of AlCl3-treated rats, which highlights its potential in AD management.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Ácido Quenodesoxicólico/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Insulina/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Transdução de Sinais , Cloreto de Alumínio , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ácido Quenodesoxicólico/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Modelos Biológicos , Degeneração Neural/complicações , Degeneração Neural/patologia , Síndromes Neurotóxicas/metabolismo , PPAR gama/metabolismo , Ratos Wistar , Memória Espacial
12.
J Biochem Mol Toxicol ; 31(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28902463

RESUMO

Renal toxicity is one of the most severe complications that can occur with cisplatin (CIS) administration in cancer patients. Montelukast (ML) renoprotective outcome contrary to CIS-drawn nephrotoxicity remains obscure. Therefore, adult male Sprague-Dawley rats were orally given ML (10 and 20 mg/kg/day) 5 days before and after single CIS (5 mg/kg; i.p.) treatment. ML returned blood urea nitrogen, as well as serum creatinine and gamma glutamyl transferase that were elevated by CIS to normal level. The improved kidney function tests corroborated the attenuation of CIS renal injury at the microscopical level. It also reduced serum/renal nitric oxide and renal hemeoxygenase-1. Meanwhile, ML hindered the raised levels of serum endothelin-1, serum and renal tumor necrosis factor-α, and monocyte chemoattractant protein-1. These effects were associated by deceased caspase-3 expression in kidney after ML treatment. In conclusion, ML guards against CIS-induced nephrotoxicity via anti-inflammatory and antiapoptotic properties.


Assuntos
Acetatos/farmacologia , Injúria Renal Aguda/prevenção & controle , Cisplatino/toxicidade , Rim/efeitos dos fármacos , Antagonistas de Leucotrienos/farmacologia , Quinolinas/farmacologia , Acetatos/uso terapêutico , Injúria Renal Aguda/induzido quimicamente , Animais , Caspase 3/metabolismo , Ciclopropanos , Avaliação Pré-Clínica de Medicamentos , Rim/enzimologia , Rim/patologia , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Quinolinas/uso terapêutico , Ratos Sprague-Dawley , Sulfetos
13.
J Biochem Mol Toxicol ; 28(10): 472-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044399

RESUMO

Ellagic acid (EA) renoprotective effect against cisplatin (CIS)-induced nephrotoxicity remains elusive. Therefore, male Sprague-Dawley rats received CIS alone or EA (10 and 30 mg/kg, p.o.) for 5 days before and after CIS injection. CIS increased serum levels of blood urea nitrogen, creatinine, γ-glutamyl transferase, and reduced those of albumin and total protein. It also raised serum endothelin-1, as well as serum and renal nitric oxide, tumor necrosis factor-α, and monocyte chemoattractant protein-1. CIS enhanced the renal caspase-3, hemeoxygenase (HO)-1, nuclear factor-κB, and inducible nitric oxide. EA hampered CIS-induced nephrotoxicity manifested by an enhancement of the glomerular filtration rate which was associated by the reduction of inflammatory mediators and the apoptotic marker in the serum and/or kidney. The present study discloses that EA suppresses HO-1 and, its renoprotection is also linked to its anti-inflammatory and antiapoptotic properties, as well as the reduction of nitric oxide and endothelin-1.


Assuntos
Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Cisplatino/toxicidade , Ácido Elágico/farmacologia , Rim/efeitos dos fármacos , Animais , Nitrogênio da Ureia Sanguínea , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Quimiocina CCL2/análise , Quimiocina CCL2/efeitos dos fármacos , Creatinina/sangue , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Endotelina-1/sangue , Heme Oxigenase-1/metabolismo , Masculino , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico/análise , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/efeitos dos fármacos , gama-Glutamiltransferase/sangue
14.
Life Sci ; 351: 122865, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38914304

RESUMO

AIMS: Niacin (NIA) supplementation showed effectiveness against Parkinson's disease (PD) in clinical trials. The depletion of NAD and endoplasmic reticulum stress response (ERSR) are implicated in the pathogenesis of PD, but the potential role for NAD precursors on ERSR is not yet established. This study was undertaken to decipher NIA molecular mechanisms against PD-accompanied ERSR, especially in relation to PKC. METHODS: Alternate-day-low-dose-21 day-subcutaneous exposure to rotenone (ROT) in rats induced PD. Following the 5th ROT injection, rats received daily doses of either NIA alone or preceded by the PKC inhibitor tamoxifen (TAM). Extent of disease progression was assessed by behavioral, striatal biochemical and striatal/nigral histopathological/immunohistochemical analysis. KEY FINDINGS: Via activating PKC/LKB1/AMPK stream, NIA post-treatment attenuated the ERSR reflected by the decline in ATF4, ATF6 and XBP1s to downregulate the apoptotic markers, CHOP/GADD153, p-JNK and active caspase-3. Such amendments congregated in motor activity/coordination improvements in open field and rotarod tasks, enhanced grid test latency and reduced overall PD scores, while boosting nigral/striatal tyrosine hydroxylase immunoreactivity and increasing intact neurons (Nissl stain) in both SNpc and striatum that showed less neurodegeneration (H&E stain). To different extents, TAM reverted all the NIA-related actions to prove PKC as a fulcrum in conveying the drug neurotherapeutic potential. SIGNIFICANCE: PKC activation is a pioneer mechanism in the drug ERSR inhibitory anti-apoptotic modality to clarify NIA promising clinical and potent preclinical anti-PD efficacy. This kinase can be tagged as a druggable target for future add-on treatments that can assist dopaminergic neuronal aptitude against this devastating neurodegenerative disease.


Assuntos
Estresse do Retículo Endoplasmático , Niacina , Animais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ratos , Niacina/farmacologia , Masculino , Proteína Quinase C/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Rotenona/farmacologia , Camundongos , Apoptose/efeitos dos fármacos , Ratos Wistar , Modelos Animais de Doenças
15.
Int Immunopharmacol ; 139: 112774, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39067398

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas , Ferroptose , Inflamassomos , Fator 2 Relacionado a NF-E2 , Proteína 3 que Contém Domínio de Pirina da Família NLR , PPAR gama , Ratos Wistar , Receptor PAR-1 , Transdução de Sinais , Animais , Masculino , Ratos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Ferroptose/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase (Desciclizante)/genética , Inflamassomos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , PPAR gama/metabolismo , Receptor PAR-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
16.
Life Sci ; 351: 122813, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38857655

RESUMO

The cytoplasmic oligomer NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated in most inflammatory and autoimmune diseases. Here, we highlight the significance of NLRP3 in diverse renal disorders, demonstrating its activation in macrophages and non-immune tubular epithelial and mesangial cells in response to various stimuli. This activation leads to the release of pro-inflammatory cytokines, contributing to the development of acute kidney injury (AKI), chronic renal injury, or fibrosis. In AKI, NLRP3 inflammasome activation and pyroptotic renal tubular cell death is driven by contrast and chemotherapeutic agents, sepsis, and rhabdomyolysis. Nevertheless, inflammasome is provoked in disorders such as crystal and diabetic nephropathy, obesity-related renal fibrosis, lupus nephritis, and hypertension-induced renal damage that induce chronic kidney injury and/or fibrosis. The mechanisms by which the inflammatory NLRP3/ Apoptosis-associated Speck-like protein containing a Caspase recruitment domain (ASC)/caspase-1/interleukin (IL)-1ß & IL-18 pathway can turn on renal fibrosis is also comprehended. This review further outlines the involvement of dopamine and its associated G protein-coupled receptors (GPCRs), including D1-like (D1, D5) and D2-like (D2-D4) subtypes, in regulating this inflammation-linked renal dysfunction pathway. Hence, we identify D-related receptors as promising targets for renal disease management by inhibiting the functionality of the NLRP3 inflammasome.


Assuntos
Inflamassomos , Nefropatias , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Animais , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/etiologia , Rim/patologia , Rim/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia
17.
Eur J Pharmacol ; 969: 176460, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38402931

RESUMO

Ropinirole used to treat Parkinson's disease highly targets the dopaminergic receptor D3 over the D2 receptor but although both are expressed in the kidneys the ropinirole potential to treat kidney injury provoked by ischemia/reperfusion (I/R) is undraped. We investigated whether ropinirole can alleviate renal I/R by studying its anti-inflammatory, antioxidant, and anti-pyroptotic effects targeting its aptitude to inhibit the High-mobility group box 1/Toll-like receptor 4/Nuclear factor-kappa B (HMGB1/TLR4/NF-κB) cue and the canonical/non-canonical NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome trajectories. Herein, bilateral I/R surgery was induced in animals to be either untreated or treated with ropinirole for three days after the insult. Ropinirole successfully improved the histopathological picture and renal function which was confirmed by reducing cystatin C and the standard parameters creatinine and blood urea nitrogen (BUN). Ropinirole achieved this through its anti-inflammatory capacity mediated by reducing the HMGB1/TLR4 axis and inactivating NF-κB, which are upstream regulators of the NLRP3 pathway. As a result, the injurious inflammasome markers (NLRP3, apoptosis-associated speck-like protein (ASC), active caspase-1) and their target cytokines interleukin-1 beta (IL-1ß) and IL-18 were decreased. Ropinirole also reduced the pyroptotic cell death markers caspase-11 and gasdermin-D. Furthermore, ropinirole by replenishing antioxidants and decreasing malondialdehyde helped to reduce oxidative stress in the kidneys. The docking findings confirmed that ropinirole highly binds to the dopaminergic D3 receptor more than to the D2 receptor. In conclusion, ropinirole has the potential to be a reno-therapeutic treatment against I/R insult by abating the inflammatory NLRP3 inflammasome signal, pyroptosis, and oxidative stress.


Assuntos
Injúria Renal Aguda , Proteína HMGB1 , Indóis , Traumatismo por Reperfusão , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Caspases , Antioxidantes/farmacologia , Isquemia , Rim/metabolismo , Reperfusão , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
18.
Int Immunopharmacol ; 126: 111289, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38016347

RESUMO

The ß3-adrenergic receptor (ß3-AR) agonism mirabegron is used to treat overactive urinary bladder syndrome; however, its role against acute kidney injury (AKI) is not unveiled, hence, we aim to repurpose mirabegron in the treatment of mercuric chloride (HgCl2)-induced AKI. Rats were allocated into normal, normal + mirabegron, HgCl2 untreated, HgCl2 + mirabegron, and HgCl2 + the ß3-AR blocker SR59230A + mirabegron. The latter increased the mRNA of ß3-AR and miR-127 besides downregulating NF-κB p65 protein expression and the contents of its downstream targets iNOS, IL-4, -13, and -17 but increased that of IL-10 to attest its anti-inflammatory capacity. Besides, mirabegron downregulated the protein expression of STAT-6, PI3K, and ERK1/2, the downstream targets of the above cytokines. Additionally, it enhanced the transcription factor PPAR-α but turned off the harmful hub HNF-4α/HNF-1α and the lipid peroxide marker MDA. Mirabegron also downregulated the CD-163 protein expression, which besides the inhibited correlated cytokines of M1 (NF-κB p65, iNOS, IL-17) and M2 (IL-4, IL-13, CD163, STAT6, ERK1/2), inactivated the macrophage phenotypes. The crosstalk between these parameters was echoed in the maintenance of claudin-2, kidney function-related early (cystatin-C, KIM-1, NGAL), and late (creatinine, BUN) injury markers, besides recovering the microscopic structures. Nonetheless, the pre-administration of SR59230A has nullified the beneficial effects of mirabegron on the aforementioned parameters. Here we verified that mirabegron can berepurposedto treat HgCl2-induced AKI by activating the ß3-AR. Mirabegron signified its effect by inhibiting inflammation, oxidative stress, and the activated M1/M2 macrophages, events that preserved the proximal tubular tight junction claudin-2 via the intersection of several trajectories.


Assuntos
Injúria Renal Aguda , Claudina-2 , Ratos , Animais , Cloreto de Mercúrio/toxicidade , NF-kappa B/metabolismo , Interleucina-4 , Rim/metabolismo , Macrófagos/metabolismo , Receptores Adrenérgicos
19.
Life Sci ; 338: 122362, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38141855

RESUMO

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.


Assuntos
Flavonas , Doença de Huntington , Mitofagia , Animais , Ratos , Apoptose , Citocromos c , Flavonas/farmacologia , Doença de Huntington/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas de Membrana , Simulação de Acoplamento Molecular , Fosfoproteínas , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR , Ubiquitinas/metabolismo
20.
Eur J Pharmacol ; 961: 176191, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37967645

RESUMO

Septic encephalopathy (SE) is a critical mental status associated with potential long-term cognitive deficits and higher mortality rates in ICU patients. The shortfall in comprehending its pathophysiology limits effective treatment options, however, GLP-1 agonists opened an entry point for future neurodegenerative disease management. This work aims to explore the mTORC1 prospective role in the pathogenesis of SE using rapamycin (RAPA) and investigate the involvement of this complex in exendin-4 (EX4) neurotherapeutic potential using cecal ligation and puncturing (CLP)-induced SE model, focusing on necroptosis as a novel intervention besides necrosis and apoptosis. EX4 was administered intranasally alone or preceded by RAPA, which was also solely given to male Sprague-Dawley rats subjected to CLP. First, opposite to the SE effect, RAPA inhibited mTORC1 and blunted TNF-α-induced necroptosis and Drp1, a mitochondrial fission marker. However, RAPA worsened the SE effect on endotoxemia, functional/cortical structures, and apoptotic/necrotic cell deaths. Second, EX4 increased mTORC1 assembly in the cerebral cortex and reduced sepsis-induced endotoxemia and behavioral/cerebral histopathology deficits in an mTOR-dependent manner. EX4 also reduced the inflammatory marker TNF-α and necroptosis as indicated by RIPK-1/RIPK-3/MLKL dephosphorylation and deactivated PGAM/Drp1 axis. Besides, EX4 turned off the apoptotic cue, caspase-3&8/cytochrome-C. However, RAPA pre-administration nullified the EX4 effect on apoptosis and HMGB1-induced necrosis. In conclusion, our research declares that targeting mTORC1 could be a promising approach for managing SE. Additionally, we highlight that the novel neuroprotective effect of EX4 in ameliorating SE is achieved by reducing necroptosis and utilizing the anti-apoptotic and anti-necrotic properties of mTORC1.


Assuntos
Encefalopatias , Endotoxemia , Doenças Neurodegenerativas , Humanos , Ratos , Masculino , Animais , Exenatida/farmacologia , Ratos Sprague-Dawley , Alvo Mecanístico do Complexo 1 de Rapamicina , Necroptose , Fator de Necrose Tumoral alfa/farmacologia , Necrose , Sirolimo/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa