Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Life Sci ; 351: 122813, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38857655

ABSTRACT

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.


Subject(s)
Inflammasomes , Kidney Diseases , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Animals , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/etiology , Kidney/pathology , Kidney/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology
2.
Eur J Pharmacol ; 975: 176669, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38795758

ABSTRACT

Methotrexate (MTX)-induced gastrointestinal mucositis is a common adverse effect characterized by redox imbalance and overproduction of inflammatory mediators that perturb intestinal integrity. Currently, there is no definitive treatment for this condition and its prevention is still far beyond comprehension. Because of its pleiotropic pharmacological actions, we aimed to explore the potential mechanisms through which cilostazol (CILO) can protect against MTX-induced intestinal mucositis. Wistar rats were allocated into 4 groups, control, CILO (100 mg/kg, p.o for 14 days), MTX (7.5 mg/kg for 4 successive days), and CILO + MTX. The improving effect of CILO on the morphological structure was confirmed by an upturn in the histopathological and transition electron microscope examinations evidenced by the increased jejunal villus height/width and the crypt depth besides the maintenance of tight junctions. These findings were verified biochemically; on the molecular level, CILO reduced the MTX-induced lipid peroxidation, cleaved caspase-3, p53, and the inflammatory parameters (TLR-2, NF-κB, IL-23, TNF-α, IL-1ß), while increasing the anti-inflammatory marker IL-10 and the antioxidant enzyme SOD. Moreover, CILO decreased the injurious axis AKT/GSK-3ß/cyclin-D1, and CD44+, but increased the immunoexpression of the cell proliferating marker PCNA. CILO also upheld the intestinal barrier by enhancing the tight junction molecules (ZO-1, claudin-4) and the E-cadherin/ß-catenin complex while abating the mesenchymal marker vimentin. In conclusion, CILO protected gut integrity by reducing the epithelial-mesenchymal transition process, the MTX-induced oxidative, apoptotic, and inflammatory mediators, and turning off the CD44/AKT/GSK-3ß/cyclin D1 trajectory and intensifying the expression of PCNA.


Subject(s)
Cyclin D1 , Glycogen Synthase Kinase 3 beta , Methotrexate , Mucositis , NF-kappa B , Proto-Oncogene Proteins c-akt , Rats, Wistar , Toll-Like Receptor 2 , Animals , Glycogen Synthase Kinase 3 beta/metabolism , NF-kappa B/metabolism , Methotrexate/toxicity , Methotrexate/pharmacology , Rats , Toll-Like Receptor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mucositis/chemically induced , Mucositis/pathology , Mucositis/metabolism , Male , Cyclin D1/metabolism , Signal Transduction/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Disease Models, Animal
3.
Food Chem Toxicol ; 187: 114585, 2024 May.
Article in English | MEDLINE | ID: mdl-38490351

ABSTRACT

Although concern persists regarding possible adverse effects of consumption of synthetic azo food dyes, the mechanisms of any such effects remain unclear. We have tested the hypothesis that chronic consumption of the food dye Sunset Yellow (SY) perturbs the composition of the gut microbiota and alters gut integrity. Male rats were administered SY orally for 12 weeks. Analysis of fecal samples before and after dye administration demonstrated SY-induced microbiome dysbiosis. SY treatment reduced the abundance of beneficial taxa such as Treponema 2, Anaerobiospirillum, Helicobacter, Rikenellaceae RC9 gut group, and Prevotellaceae UCG-003, while increasing the abundance of the potentially pathogenic microorganisms Prevotella 2 and Oribacterium. Dysbiosis disrupted gut integrity, altering the jejunal adherens junction complex E-cadherin/ß-catenin and decreasing Trefoil Factor (TFF)-3. SY administration elevated LPS serum levels, activated the inflammatory inflammasome cascade TLR4/NLRP3/ASC/cleaved-activated caspase-1 to mature IL-1ß and IL-18, and activated caspase-11 and gasdermin-N, indicating pyroptosis and increased intestinal permeability. The possibility that consumption of SY by humans could have effects similar to those that we have observed in rats should be examined.


Subject(s)
Azo Compounds , Gastrointestinal Microbiome , Humans , Male , Rats , Animals , Rats, Wistar , Dysbiosis , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Caspases
4.
Eur J Pharmacol ; 969: 176460, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38402931

ABSTRACT

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.


Subject(s)
Acute Kidney Injury , HMGB1 Protein , Indoles , Reperfusion Injury , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Caspases , Antioxidants/pharmacology , Ischemia , Kidney/metabolism , Reperfusion , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
5.
Int Immunopharmacol ; 126: 111289, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38016347

ABSTRACT

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.


Subject(s)
Acute Kidney Injury , Claudin-2 , Rats , Animals , Mercuric Chloride/toxicity , NF-kappa B/metabolism , Interleukin-4 , Kidney/metabolism , Macrophages/metabolism , Receptors, Adrenergic
6.
Life Sci ; 338: 122362, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38141855

ABSTRACT

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


Subject(s)
Flavones , Huntington Disease , Mitophagy , Animals , Rats , Apoptosis , Cytochromes c , Flavones/pharmacology , Huntington Disease/metabolism , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins , Molecular Docking Simulation , Phosphoproteins , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases , Ubiquitins/metabolism
7.
Biomedicines ; 11(12)2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38137376

ABSTRACT

Autism spectrum disorder (ASD) prevalence is emerging with an unclear etiology, hindering effective therapeutic interventions. Recent studies suggest potential renin-angiotensin system (RAS) alterations in different neurological pathologies. However, its implications in ASD are unexplored. This research fulfills the critical gap by investigating dual arms of RAS and their interplay with Notch signaling in ASD, using a valproic acid (VPA) model and assessing astaxanthin's (AST) modulatory impacts. Experimentally, male pups from pregnant rats receiving either saline or VPA on gestation day 12.5 were divided into control and VPA groups, with subsequent AST treatment in a subset (postnatal days 34-58). Behavioral analyses, histopathological investigations, and electron microscopy provided insights into the neurobehavioral and structural changes induced by AST. Molecular investigations of male pups' cortices revealed that AST outweighs the protective RAS elements with the inhibition of the detrimental arm. This established the neuroprotective and anti-inflammatory axes of RAS (ACE2/Ang1-7/MasR) in the ASD context. The results showed that AST's normalization of RAS components and Notch signaling underscore a novel therapeutic avenue in ASD, impacting neuronal integrity and behavioral outcomes. These findings affirm the integral role of RAS in ASD and highlight AST's potential as a promising treatment intervention, inviting further neurological research implications.

8.
Eur J Pharmacol ; 961: 176191, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37967645

ABSTRACT

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.


Subject(s)
Brain Diseases , Endotoxemia , Neurodegenerative Diseases , Humans , Rats , Male , Animals , Exenatide/pharmacology , Rats, Sprague-Dawley , Mechanistic Target of Rapamycin Complex 1 , Necroptosis , Tumor Necrosis Factor-alpha/pharmacology , Necrosis , Sirolimus/pharmacology
9.
Saudi Pharm J ; 31(11): 101818, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37868646

ABSTRACT

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.

10.
Sci Rep ; 13(1): 11899, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488162

ABSTRACT

Endocannabinoid anandamide (AEA) has a physiological role in regulating renal blood flow, whereas its analogs ameliorated renal ischemia/reperfusion injury. Nonetheless, the role of AEA against mercuric chloride (HgCl2)-induced renal toxicity has not been unraveled. Rats were allocated into control, HgCl2, and HgCl2/AEA treated groups. The administration of AEA quelled the HgCl2-mediated increase in inositol trisphosphate (IP3) and nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). The endocannabinoid also signified its anti-inflammatory potential by turning off the inflammatory cascade evidenced by the suppression of high mobility group box protein-1 (HMGB1), receptor of glycated end products (RAGE), nuclear factor-κB p65 (NF-κB), and unexpectedly PPAR-γ. Additionally, the aptitude of AEA to inhibit malondialdehyde and boost glutathione points to its antioxidant capacity. Moreover, AEA by enhancing the depleted renal WNT-5A and reducing cystatin-C and KIM-1 (two kidney function parameters) partly verified its anti-apoptotic capacity, confirmed by inhibiting caspase-3 and increasing B-cell lymphoma-2 (BCL-2). The beneficial effect of AEA was mirrored by the improved architecture and kidney function evidenced by the reduction in cystatin-C, KIM-1, creatinine, BUN, and caspase1-induced activated IL-18. In conclusion, our results verify the reno-protective potential of AEA against HgCl2-induced kidney injury by its anti-inflammatory, antioxidant, and anti-apoptotic capacities by modulating WNT-5A/BCL-2, IP3/NFATC1, HMGB-1/RAGE/NF-κB, caspase-1/IL-18, and caspase-3/BCL-2 cues.


Subject(s)
Acute Kidney Injury , Cystatins , HMGB1 Protein , Animals , Rats , NF-kappa B , Endocannabinoids , Caspase 3 , Interleukin-18 , Mercuric Chloride , Antioxidants , Transcription Factors , Proto-Oncogene Proteins c-bcl-2
14.
Int Immunopharmacol ; 116: 109771, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36736222

ABSTRACT

The neuroprotective capacity of morin hydrate (MH), a potent antioxidant flavonoid, and calpeptin (CP), a calpain inhibitor, was documented against different insults but not Huntington's disease (HD). Accordingly, we aim to assess the neuroprotective potential of MH and/or CP in a 3-nitropropionic acid (3-NP)-induced HD model. The 3-NP-treated rats were post-treated with saline, MH, CP, or MH + CP for a week. Post-treatment with MH and/or CP amended motor function (beam walking test) and short-/ long-term spatial memory (novel object recognition test) and improved cortical microscopic architecture. On the molecular level, MH, and to a lesser extent CP, inhibited the cortical content/expression of glutamate, calpain, and Kidins220 and abated the inflammatory molecules, nuclear factor (NF)-κB, tumor necrosis factor-α, and interleukin-1ß, as well as lipid peroxidation. However, MH, but barely CP, activated the molecules of the neuroprotective trajectory; viz., brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase receptor B (TrkB), protein kinase B (AKT), and cAMP response element-binding protein (CREB). Compared to the single treatments, the combination regimen mediated further reductions in the cortical contents of glutamate, calpain, and Kidins220, effects that extended to entail the anti-inflammatory/anti-oxidant potentials of MH and to a greater extent CP. However, the combination of MH strengthened the fair effect of CP on the survival signaling pathway BDNF/TrkB/AKT/CREB. In conclusion, MH, CP, and especially their combination, afforded neuroprotection against HD through curbing the glutamate/calpain axis, Kidins220, as well as NF-κB-mediated neuroinflammation/oxidative stress, besides activating the BDNF/TrkB/AKT/CREB hub that was partly dependent on calpain inhibition.


Subject(s)
Neuroprotective Agents , Proto-Oncogene Proteins c-akt , Animals , Rats , Antioxidants/therapeutic use , Antioxidants/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Calpain , Cyclic AMP Response Element-Binding Protein/metabolism , Flavonoids , Glutamic Acid , Membrane Proteins , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphoproteins , Proto-Oncogene Proteins c-akt/metabolism
15.
Ann Afr Med ; 22(1): 82-87, 2023.
Article in English | MEDLINE | ID: mdl-36695227

ABSTRACT

Background: Carbapenem resistant Enterobacteriacae (CRE) bloodstream infection (BSI) causes complicated infections, especially in immunocompromised patients. This study aimed to assess the renal toxicity and the efficacy of therapy with colistin in a cohort of pediatric cancer patients with BSIs due to CRE and sensitivity to colistin. Patients and Methods: This was an observational, prospective cohort study from May 2017 to October 2017 in Children's Cancer Hospital Egypt 57,357. All patients who had blood stream infections due to CRE receiving intravenous colistin were prospectively enrolled. We used a standardized case form to record patient characteristics, including age, sex, weight, underlying comorbidities, type of infection, causative organism, and antibiotic susceptibility testing. Daily doses, duration of colistin therapy, and co-administered antibiotics (aminoglycosides, vancomycin) were collected. Furthermore, clinical and microbiological responses to treatment were reported. The dosing schedule was based on a loading dose of 5 MU and a 5-MU twice-daily divided maintenance dose, titrated on renal function. Clinical cure, bacteriological clearance, and daily serum creatinine were recorded. Results: One hundred and forty-one Blood Stream infectious episodes mainly due to Klebsiella Species (pneumoniae and Oxytoca) (27%) and Escherichia coli (68%) were analyzed. All strains were susceptible to colistin with Minimum inhibitory concentration (MICs) of 0.19-1.5 mg/L. Patients were predominantly females (69%), with a mean age of 7 years. It was used as a combination therapy with carbapenems (69.2%) or aminoglycosides (80%). The median duration of treatment was 9 days (Range 1-50 days). Clinical and microbiological cure was observed in 110 cases (80%). Acute kidney injury developed during five treatment courses (4%) in which colistin was used in combination with amikacin. No renal replacement therapy was required and subsided within 7 days from colistin discontinuation. Conclusions: Our study showed that colistin had a high efficacy without significant renal toxicity in severe infections due to CRE Gram-negative bacteria.


Résumé Carbapenem-resistant Gram-negative (CRE) bloodstream infection (BSI) causes complicated infections, especially in immunocompromised patients .This study aimed to assess the renal toxicity and the efficacy of therapy with colistin in a cohort of pediatric cancer patients with BSIs due to CRE and sensitivity to colistin. colistin proved to be effective and safe in managing CRE in children with cancer Mots-clés: Colistin, cancer, children, and Carbapenem-Resistant Enterobacteriaceae.


Subject(s)
Bacteremia , Carbapenem-Resistant Enterobacteriaceae , Neoplasms , Sepsis , Female , Humans , Child , Male , Colistin/adverse effects , Prospective Studies , Cancer Care Facilities , Egypt/epidemiology , Anti-Bacterial Agents/adverse effects , Bacteremia/drug therapy , Bacteremia/chemically induced , Bacteremia/microbiology , Carbapenems/therapeutic use , Carbapenems/pharmacology , Escherichia coli , Aminoglycosides/therapeutic use , Microbial Sensitivity Tests , Neoplasms/complications
16.
Front Pharmacol ; 13: 1008085, 2022.
Article in English | MEDLINE | ID: mdl-36386153

ABSTRACT

Although dysautonomia was documented in inflammatory bowel disease, with activation of the stress-related sympathetic system, the role of agonists/antagonists of the adrenergic receptors is not conclusive. Moreover, ulcerative colitis was recently linked to dementia, but the potential role of the presenilin 1(PS1)/BACE-1/beta-amyloid (Aß) axis has not been evaluated. Hence, we investigated the impact of mirabegron (ß3-agonist) and/or carvedilol (ß1/ß2 antagonist) on iodoacetamide-induced ulcerative colitis with emphasis on the novel pathomechanism of the PS1/BACE-1/Aß axis in ulcerative colitis, and its relation to the inflammatory cascade, fibrotic processes, and the gut barrier dysfunction. Ulcerated rats were either left untreated or treated for 8 days with mirabegron and/or carvedilol. Besides minimizing colon edema and weight loss, and improving colon structure, mirabegron and/or carvedilol abated colonic PS1/BACE-1/Aß axis and the NOTCH1/NICD/HES1 hub besides the inflammatory cascade GSK3-ß/NF-κΒ/TNF-α, and the oxidative stress marker malondialdehyde. The anti-fibrotic effect was verified by boosting SMAD-7 and inhibiting TGF-ß1, α-SMA immunoexpression, and MTC staining. Moreover, the drugs improved the gut barrier function, attested by the increased goblet cells and expression of E-cadherin, and the inhibited expression of p (Y654)-ß-catenin to preserve the E-cadherin/ß-catenin adherens junction (AJ). These signaling pathways may be orchestrated by the replenished PPAR-γ, a transcription factor known for its anti-colitic effect. Conclusion: Besides maintaining the gut barrier, mirabegron and/or carvedilol mediated their anti-colitic effect by their anti-oxidant, anti-inflammatory, and anti-fibrotic capacities. The therapeutic effect of these drugs depends partly on suppressing the harmful signaling pathways PS1/BACE-1/Aß, NOTCH1/NICD/HES1, GSK3-ß/NF-κΒ/TNF-α, and TGF-1ß/α-SMA while enhancing PPAR-γ, SMAD-7, mucus, and AJ.

17.
Eur J Pharmacol ; 931: 175213, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35981604

ABSTRACT

Morin is a bioactive flavonoid with prominent neuroprotective potentials, however, its impact on epilepsy-provoked cognitive dysregulations has not been revealed. Hence, the present investigation aims to divulge the potential anticonvulsant/neuroprotective effects of morin in rats using a pentylenetetrazole (PTZ)-induced kindling model with an emphasis on the possible signaling trajectories involved. Kindling was induced using a sub-convulsive dose of PTZ (35 mg/kg, i.p.), once every other day for 25 days (12 injections). The expression of targeted biomarkers and molecular signals were examined in hippocampal tissues by ELISA, Western blotting, immunohistochemistry, and histopathology. Contrary to PTZ effects, administration of morin (10 mg/kg, i.p., from day 15 of PTZ injection to the end of the experiment) significantly reduced the severity of seizures coupled with a delay in kindling acquisition. It also preserved hippocampal neurons, and diminished astrogliosis to counteract cognitive deficits, exhibited by the enhanced performance in MWM and PA tests. These favorable impacts of morin were mediated via the abrogation of the PTZ-induced necroptotic changes and mitochondrial fragmentation proven by the suppression of p-RIPK-1/p-RIPK-3/p-MLKL and PGAM5/Drp-1 cues alongside the enhancement of caspase-8. Besides, morin inhibited the inflammatory cascade documented by the attenuation of the pro-convulsant receptor/cytokines TNFR-1, TNF-α, I L-1ß, and IL-6 and the marked reduction of hippocampal IL-6/p-JAK2/p-STAT3/GFAP cue. In tandem, morin signified its anti-oxidant capacity by lowering the hippocampal contents of MDA, NOX-1, and Keap-1 with the restoration of the impaired Nrf-2/HO-1 pathway. Together, these versatile neuro-modulatory effects highlight the promising role of morin in the management of epilepsy.


Subject(s)
Epilepsy , Kindling, Neurologic , Animals , Cognition , Epilepsy/chemically induced , Flavonoids , Hippocampus , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Pentylenetetrazole , Protein Kinases/metabolism , Rats , Seizures/chemically induced , Tumor Necrosis Factor-alpha/metabolism
18.
Life Sci ; 307: 120865, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35934057

ABSTRACT

AIMS: Montelukast (MNK), a leukotriene receptor antagonist, has proven its antioxidant/anti-inflammatory capacity to guard against diabetes-induced complications and to enhance metformin antidiabetic effect. Nevertheless, here we evaluated the involvement of endoplasmic reticulum (ER) stress and insulin signaling cascade in the effect of MNK and/or dapagliflozin (DAPA) using the soleus muscle of type 2 diabetic (T2D)/insulin resistant (IR) rats. MAIN METHODS: To induce T2D/IR, rats were fed a westernized diet (WD) for 8 weeks followed by a sub-diabetogenic dose of streptozotocin (STZ). Animals were divided into control (receiving normal diet; ND), diabetic untreated, and diabetic treated for 4 weeks with DAPA, MNK, or their combination (DAPA+MNK). Blood glucose and serum lipid profile were determined, and the soleus muscle was tested for ER stress-induced IR, besides histopathological examination. KEY FINDINGS: Treatment with DAPA, MNK, and especially their combination decreased the fasting plasma levels of glucose and insulin while improving insulin sensitivity and lipid profile. This was achieved via the activation of insulin signaling IRS-1/AKT/GLUT4 pathway in the soleus muscle consequent to the deactivation of the ER stress response elements, namely IRE1α, ATF6, and PERK to suppress p-JNK and p-eIF2α. SIGNIFICANCE: Improved insulin signaling along with the deactivation of the ER stress response by MNK comparable to the DAPA are partly responsible for the enhanced soleus muscle insulin sensitivity, effects that nominate MNK as an add-on to DAPA to enhance its antidiabetic efficacy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin Resistance , Metformin , Acetates/pharmacology , Animals , Antioxidants/pharmacology , Benzhydryl Compounds , Blood Glucose/metabolism , Cyclopropanes , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum , Endoribonucleases/metabolism , Glucose Transporter Type 4/metabolism , Glucosides , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Leukotriene Antagonists/pharmacology , Metformin/pharmacology , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-akt/metabolism , Quinolines , Rats , Response Elements , Streptozocin/pharmacology , Sulfides
19.
Eur J Pharmacol ; 927: 175066, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35643302

ABSTRACT

The phytochemical sulforaphane (SFN) has been studied for its potential anti-obesity effect, but neither its molecular targets nor its interaction with the antimalarial drug chloroquine (CQ) has been fully delineated. Therefore, high-fat diet (HFD) obese rats were randomly allocated into one of five groups and were left untreated or gavaged orally with SFN (0.5 or 1 mg/kg), CQ (5 mg/kg), or their combination (0.5/5 mg/kg) for six successive weeks to assess their potential interaction and the enrolled mechanisms. SFN effectively reduced the HFD-induced weight gain, blood glucose, and serum leptin levels, and improved lipid profile. On the molecular level, SFN inhibited the lipogenesis-related enzymes, namely sterol regulatory element-binding protein (SREBP)-1c, fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC) in both liver and visceral white adipose tissue (vWAT) of HFD obese rats. SFN also turned off the inflammatory pathway conserved Janus kinase/signaling transducers and activators of transcription/suppressor of cytokine signaling (JAK-2/STAT-3/SOCS-3) in these tissues, as well as the inflammatory markers nuclear factor-kappa (NF-κ) B and interleukin (IL)-22 in serum. In contrast, SFN downregulated the gene expression of microRNA (miR-200a), while significantly increasing the autophagic parameters; viz., beclin-1, autophagy-related protein (ATG)-7, and microtubule-associated protein 2 light chain 3 (LC3-II) in both liver and vWAT. On most of the parameters mentioned above, treatment with CQ solely produced a satisfactory effect and intensified the low dose of SFN in the combination regimen. These findings demonstrated the beneficial effects of using CQ as an add-on anti-obesity medicine to SFN.


Subject(s)
Chloroquine , Diet, High-Fat , Isothiocyanates , Janus Kinase 2 , STAT3 Transcription Factor , Sulfoxides , Suppressor of Cytokine Signaling 3 Protein , Animals , Chloroquine/pharmacology , Isothiocyanates/pharmacology , Janus Kinase 2/metabolism , Liver/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Random Allocation , Rats , STAT3 Transcription Factor/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sulfoxides/pharmacology , Suppressor of Cytokine Signaling 3 Protein/metabolism
20.
BMC Neurosci ; 23(1): 11, 2022 03 05.
Article in English | MEDLINE | ID: mdl-35247984

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Forkhead Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mitoxantrone/pharmacology , Multiple Sclerosis/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Rats , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...