Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Clin Exp Pharmacol Physiol ; 50(1): 96-106, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36208078

RESUMEN

The long-term side effect of the antiarrhythmic drug, amiodarone (AMIO), such as lung toxicity, remains a critical clinical issue. The previous knowledge denotes diverse antioxidant, anti-inflammatory, and antifibrotic properties of the anti-anginal drug, nicorandil (NI). Therefore, we aimed to investigate the possible protective effect of NI on pulmonary tissue remodelling following AMIO-induced lung toxicity. The included rats were assigned into four equal groups (n = 8): (1) control, (2) control group that received NI 10 mg kg-1  day-1 , (3) model group that received AMIO in a dose of 60 mg kg-1  day-1 , and (4) treated group (AMIO-NI) that were treated with AMIO plus NI as shown above. Drug administration continued for 10 weeks. AMIO resulted in deteriorated (p < 0.001) pulmonary functions accompanied by respiratory acidosis. AMIO showed an obvious histological injury score with intense collagen deposition, disturbed nitric oxide synthase enzymes (NOS/iNOS), and increased alpha smooth muscle actin expression. Furthermore, AMIO upregulated the transforming growth factor (TGF-ß1)/phosphoinositide-3 kinase (PI3K)-Akt1-p/mammalian target of rapamycin (mTOR) axis, which determined the possible mechanism of AMIO on pulmonary remodelling. NI treatment significantly (p < 0.001) prevented the AMIO-induced lung toxicity, as well as inhibited the TGF-ß1/PI3K/Akt1-p/mTOR axis in the lung tissue of rats. The results were confirmed by an in-vitro study. CONCLUSION: The current results revealed that NI was effective in preserving the lung structure and functions. Amelioration of the oxidative stress and modulation of TGF-ß1/PI3K/Akt1-p/mTOR have been achieved. This study suggests NI administration as a preventive therapy from the serious pulmonary fibrosis side effect of AMIO.


Asunto(s)
Amiodarona , Fosfatidilinositol 3-Quinasa , Ratas , Animales , Factor de Crecimiento Transformador beta1 , Amiodarona/toxicidad , Fosfatidilinositol 3-Quinasas , Nicorandil/farmacología , Sirolimus , Fibrosis , Pulmón , Mamíferos , Serina-Treonina Quinasas TOR
2.
Nutr Cancer ; 74(1): 278-287, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33533291

RESUMEN

Thymoquinone (TQ) combined with Cisplatin may augment its anticancer effect on hepatocellular carcinoma (HCC), through oxidative stress mitigation and endoplasmic reticulum (ER) protein modulation. Fifty adult male Wistar albino rats were assigned into five equal experimental groups (n = 10); 1) Control, 2) diethylnitrosamine/carbon tetrachloride-induced liver tumorigenesis model (HCC), 3) Cisplatin (2 mg.kg-1ip) treated rats, 4) Thymoquinone treated group (20 mg.kg-1oral), and 5) group treated with both drugs as in Groups 3 and 4. Treatment regimens started following model confirmation and continued for 4 weeks. In the HCC model, we detected elevated ER chaperone glucose-regulated protein-78 (GRP78) and reduced C/EBP-homologous protein (CHOP)-mediated apoptosis that was accompanied by the elevated alpha-fetoprotein (AFP) marker and deteriorated liver functions. Our original results indicated that Thymoquinone potentiated the pro-apoptotic effect of cisplatin by modulating GRP78/CHOP signaling. Cisplatin/TQ reduced the elevated GRP78 and induced CHOP-mediated apoptosis in the diseased liver tissues compared to the HCC and Cisplatin treated groups. Cisplatin/TQ combination normalized AFP levels and improved liver functions compared to both HCC and cisplatin groups alone. In conclusion, Thymoquinone enhanced the efficacy of Cisplatin in HCC treatment by modulating the GRP78/CHOP/caspase-3 pathway. Thymoquinone is recommended to achieve greater therapeutic benefits and reduce the cisplatin hepatotoxicity in HCC management.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Apoptosis , Benzoquinonas , Carcinogénesis , Carcinoma Hepatocelular/patología , Cisplatino/farmacología , Estrés del Retículo Endoplásmico , Glucosa/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Estrés Oxidativo , Proteína C/metabolismo , Proteína C/farmacología , Ratas , Ratas Wistar
3.
Clin Exp Pharmacol Physiol ; 49(3): 406-418, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34796981

RESUMEN

Cardiac dysfunction is one of the leading causes of death in epilepsy. The anti-arrhythmic drug, amiodarone, is under investigation for its therapeutic effects in epilepsy. We aimed to evaluate the possible effects of amiodarone on cardiac injury during status epilepticus, as it can cause prolongation of the QT interval. Five rat groups were enrolled in the study; three control groups (1) Control, (2) Control-lithium and (3) Control-Amio, treated with 150 mg/kg/intraperitoneal amiodarone, (4) Epilepsy model, induced by sequential lithium/pilocarpine administration, and (5) the epilepsy-Amio group. The model group expressed a typical clinical picture of epileptiform activity confirmed by the augmented electroencephalogram alpha and beta spikes. The anticonvulsive effect of amiodarone was prominent, it diminished (p < 0.001) the severity of seizures and hence, deaths and reduced serum noradrenaline levels. In the model group, the electrocardiogram findings revealed tachycardia, prolongation of the corrected QT (QTc) interval, depressed ST segments and increased myocardial oxidative stress. The in-vitro myocardial performance (contraction force and - (df/dt)max ) was also reduced. Amiodarone decreased (p < 0.001) the heart rate, improved ST segment depression, and myocardial contractility with no significant change in the duration of the QTc interval. Amiodarone preserved the cardiac histological structure and reduced the myocardial injury markers represented by serum Troponin-I, oxidative stress and IL-1. Amiodarone pretreatment prevented the anticipated cardiac injury induced during epilepsy. Amiodarone possessed an anticonvulsive potential, protected the cardiac muscle and preserved its histological architecture. Therefore, amiodarone could be recommended as a protective therapy against cardiac dysfunction during epileptic seizures with favourable effect on seizure activity.


Asunto(s)
Amiodarona/uso terapéutico , Antiarrítmicos/uso terapéutico , Epilepsia/complicaciones , Cardiopatías/tratamiento farmacológico , Cardiopatías/etiología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/toxicidad , Animales , Biomarcadores/sangre , Epilepsia/inducido químicamente , Glutatión/sangre , Interleucina-1/metabolismo , Cloruro de Litio/administración & dosificación , Cloruro de Litio/toxicidad , Masculino , Malondialdehído/sangre , Agonistas Muscarínicos/administración & dosificación , Agonistas Muscarínicos/toxicidad , Contracción Miocárdica/efectos de los fármacos , Pilocarpina/administración & dosificación , Pilocarpina/toxicidad , Ratas , Ratas Wistar , Superóxido Dismutasa/sangre , Troponina I/sangre
4.
Clin Exp Pharmacol Physiol ; 49(4): 501-514, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35090059

RESUMEN

The recently defined necroptosis process participates in the pathophysiology of several tissue injuries. Targeting the necroptosis mediator receptor-interacting protein kinase (RIPK1) by necrostatin-1 in different phases of ischaemia-reperfusion injury (IRI) may provide new insight into the protection against renal IRI. The rat groups included (n = 8 in each group): 1) Sham; 2) Renal IRI; 3) Necrostatin-1 treatment 20 min before ischaemia induction in a dose of 1.65 mg/kg/intravenous; 4) Necrostatin-1 injection just before reperfusion; 5) Necrostatin-1 injection 20 min after reperfusion establishment; and 6) drug injection at both the pre-ischaemia and at reperfusion time in the same dose. Timing dependent, necrostatin-1 diminished RIPK1 (p < 0.001), and aborted the necroptosis-induced renal cell injury. Necrostatin-1 decreased the renal chemokine (CXCL1), interleukin-6, intercellular adhesion molecule (ICAM-1), myeloperoxidase, and the nuclear factor (NFκB), concomitant with reduced inducible nitric oxide synthase (iNOS), inflammatory cell infiltration, and diminished cell death represented by apoptotic cell count and the BAX/Bcl2 protein ratio. In group 6, the cell injury was minimal and the renal functions (creatinine, BUN and creatinine clearance) were almost normalised. The inflammatory markers were diminished (p < 0.001) compared to the IRI group. The results were confirmed by histopathological examination. In conclusion, RIPK1 inhibition ameliorates the inflammatory immune response induced by renal IRI. The use of two doses was more beneficial as the pathophysiology of cell injury is characterised.


Asunto(s)
Proteínas Quinasas , Daño por Reperfusión , Animales , Apoptosis/fisiología , Creatinina , Imidazoles , Inmunidad , Indoles , Isquemia , Ratas , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control
5.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G461-G476, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431405

RESUMEN

Multiple theories have been proposed describing the pathogenic mechanisms of Helicobacter pylori (H. pylori)-associated gastric motility disorders. We assessed ex vivo pyloric activity in H. pylori-infected rats, and tried to explore the associated ghrelin hormone alteration and pyloric fibrogenesis. In addition, miR-1 was assessed in pyloric tissue samples, being recently accused of having a role in smooth muscle dysfunction. Ninety adult male Wistar albino rats were assigned into nine groups: 1) control group, 2) sterile broth (vehicle group), 3) amoxicillin control, 4) omeperazole control, 5) clarithromycin control, 6) triple therapy control, 7) H. pylori- group, 8) H. pylori-clarithromycin group, and 9) H. pylori-triple therapy group. Urease enzyme activity was applied as an indicator of H. pylori infection. Ex vivo pyloric contractility was evaluated. Serum ghrelin was assessed, and histological tissue evaluation was performed. Besides, pyloric muscle miR-1 expression was measured. The immunological epithelial to mesenchymal transition (EMT) markers; transforming growth factor ß (TGFß), α-smooth muscle actin (α-SMA), and E-cadherin-3 were also evaluated. By H. pylori infection, a significant (P < 0.001) reduced pyloric contractility index was recorded. The miR-1 expression was decreased (P < 0.001) in the H. pylori-infected group, associated with reduced serum ghrelin, elevated TGFß, and α-SMA levels and reduced E-cadherin levels. Decreased miR-1 and disturbed molecular pattern were improved by treatment. In conclusion, H. pylori infection was associated with reduced miR-1, epithelial to mesenchymal transition, and pyloric hypomotility. The miR-1 may be a target for further studies to assess its possible involvement in H. pylori-associated pyloric dysfunction, which might help in the management of human H. pylori manifestations and complications.NEW & NOTEWORTHY This work is investigating functional, histopathological, and molecular changes underlying Helicobacter pylori hypomotility and is correlating these with miR-1, whose disturbance is supposed to be involved in smooth muscle dysfunction and cell proliferation according to literature. Epithelial to mesenchymal transition and reduced ghrelin hormone may contribute to H. pylori infection-associated hypomotility. H. pylori infection was associated with reduced pyloric miR-1 expression. Targeting miR-1 could be valuable in the clinical management of pyloric hypofunction.


Asunto(s)
Transición Epitelial-Mesenquimal , Motilidad Gastrointestinal , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Músculo Liso/microbiología , Píloro/microbiología , Gastropatías/microbiología , Actinas/metabolismo , Animales , Antibacterianos/farmacología , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Transición Epitelial-Mesenquimal/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Ghrelina/sangre , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/fisiopatología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Inhibidores de la Bomba de Protones/farmacología , Píloro/efectos de los fármacos , Píloro/metabolismo , Píloro/fisiopatología , Ratas Wistar , Gastropatías/tratamiento farmacológico , Gastropatías/metabolismo , Gastropatías/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 320(4): H1290-H1302, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33513084

RESUMEN

Bone marrow-derived mesenchymal stem cells (BM-MSCs) have demonstrated potential in treating diabetic cardiomyopathy. However, patients with diabetes are on multiple drugs and there is a lack of understanding of how transplanted stem cells would respond in presence of such drugs. Metformin is an AMP kinase (AMPK) activator, the widest used antidiabetic drug. In this study, we investigated the effect of metformin on the efficacy of stem cell therapy in a diabetic cardiomyopathy animal model using streptozotocin (STZ) in male Wistar rats. To comprehend the effect of metformin on the efficacy of BM-MSCs, we transplanted BM-MSCs (1 million cells/rat) with or without metformin. Our data demonstrate that transplantation of BM-MSCs prevented cardiac fibrosis and promoted angiogenesis in diabetic hearts. However, metformin supplementation downregulated BM-MSC-mediated cardioprotection. Interestingly, both BM-MSCs and metformin treatment individually improved cardiac function with no synergistic effect of metformin supplementation along with BM-MSCs. Investigating the mechanisms of loss of efficacy of BM-MSCs in the presence of metformin, we found that metformin treatment impairs homing of implanted BM-MSCs in the heart and leads to poor survival of transplanted cells. Furthermore, our data demonstrate that metformin-mediated activation of AMPK is responsible for poor homing and survival of BM-MSCs in the diabetic heart. Hence, the current study confirms that a conflict arises between metformin and BM-MSCs for treating diabetic cardiomyopathy. Approximately 10% of the world population is diabetic to which metformin is prescribed very commonly. Hence, future cell replacement therapies in combination with AMPK inhibitors may be more effective for patients with diabetes.NEW & NOTEWORTHY Metformin treatment reduces the efficacy of mesenchymal stem cell therapy for cardiac repair during diabetic cardiomyopathy. Stem cell therapy in diabetics may be more effective in combination with AMPK inhibitors.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Cardiomiopatías Diabéticas/cirugía , Hipoglucemiantes/toxicidad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Metformina/toxicidad , Miocardio/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/sangre , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Fibrosis , Hemoglobina Glucada/metabolismo , Insulina/sangre , Masculino , Células Madre Mesenquimatosas/metabolismo , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ratas Wistar , Recuperación de la Función , Estreptozocina
7.
Exp Physiol ; 106(11): 2262-2275, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34633737

RESUMEN

NEW FINDINGS: What is the central question of this study? What is the role of circulating exosomal miR-687 in remote hepatic injury following renal ischaemia-reperfusion injury (IRI) and does thymoquinone have a modulatory impact? What is the main finding and its importance? Exosomal miR-687 was expressed in renal IRI, entered the circulation and was deposited in the liver. Liver exosomal miR-687 was correlated with liver inflammation and apoptosis. Thymoquinone aborted the renal production of exosomal miR-687 and its further circulation to the liver. ABSTRACT: The pathophysiology of remote hepatic injury following acute renal ischaemia-reperfusion injury (IRI) is of particular clinical interest. Secreted small non-coding microRNA (miRs) are thought to exist in exosome-encapsulated form. Thymoquinone (TQ) is the main bioactive ingredient of Nigella sativa and has several renoprotective actions. We expected exosomal miR-687 to be relevant as it could act as a humoral mediator, with possible modulation by TQ. Thirty adult male Wister albino rats were assigned to three groups (n = 10); (1) sham-operated, (2) renal ischaemia-reperfusion injury (IRI), and (3) renal IRI pre-treated with TQ 10 mg/kg/day i.v. (TQ-IRI) for 10 days in addition to a dose administered at reperfusion onset. Following 24 h of reperfusion, the IRI group showed renal tissue hypoxia-inducible factor upregulation (P < 0.001). Electron microscopy images of exosomes and analysis of miR-687 revealed elevated levels, which appeared in the circulation. Large amounts of exosomal miR-687 were transmitted to the liver tissue. In the IRI group, liver transaminases (alanine aminotransferase, aspartate aminotransferase) were markedly (P < 0.001) elevated. The hepatic tissue inflammatory markers (vascular cell adhesion molecule-1, myeloperoxidase, monocyte chemotactic protein-1 and nuclear factor-κB) were upregulated (P < 0.001) accompanied with elevated caspase-3. TQ suppressed (P < 0.001) the renal expression and release of exosomal miR-687 into the circulation and its further deposition in the liver tissue; consequently, TQ diminished (P < 0.001) liver tissue inflammation and cellular apoptosis. The results were confirmed by histological tissue assessment. In conclusion, exosomal miR-687 liberated from injured renal tissues into the circulation may be an important factor in inducing remote hepatic injury. Exosomal miR-687 inhibition by TQ protected both renal and hepatic tissues from injury.


Asunto(s)
Lesión Renal Aguda , MicroARNs , Daño por Reperfusión , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Animales , Benzoquinonas , Isquemia/metabolismo , Hígado/metabolismo , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Reperfusión , Daño por Reperfusión/metabolismo
8.
Can J Physiol Pharmacol ; 99(4): 411-417, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32822562

RESUMEN

Despite that cyclosporine-A (CsA) is a widely used immunosuppressive drug, its nephrotoxic effect limits its long-term administration. Herein we tried to investigate its renal effect on endothelial dysfunction targeting the hypoxia-inducible factor (HIF-1α) / vascular endothelial growth factor (VEGF) / endothelial nitric oxide synthase (eNOS) pathway and the possible modulation by nicorandil. Eight groups of adult male Wistar rats were included: (1) control; (2) vehicle group (received oil); (3) glibenclamide 5 mg·kg-1·day-1 administered orally; (4) nicorandil 10 mg·kg-1·day-1 administered orally; (5) CsA 25 mg·kg-1·day-1 administered orally; (6) combined administration of CsA and nicorandil; (7) glibenclamide was added to CsA; and (8) both CsA and nicorandil were combined with glibenclamide. The treatment continued for six weeks. Combined nicorandil with CsA improved renal function deterioration initiated by CsA. CsA decreased the renal expression levels (P < 0.001) of HIF-1α, eNOS, and VEGF, inducing endothelial dysfunction and triggering inflammation, and upregulated the profibrotic marker transforming growth factor (TGF-ß). Nicorandil fixed the disturbed HIF-1α/VEGF/eNOS signaling. Nicorandil corrected the renal functions, confirmed by the improved histological glomerular tuft retraction that was obvious in the CsA group, without significant influence by glibenclamide. Proper protection from CsA-induced nephrotoxicity was achieved by nicorandil. Nicorandil reversed the disturbed HIF-1α/VEGF/eNOS pathway created by CsA.


Asunto(s)
Ciclosporina/efectos adversos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/efectos de los fármacos , Nicorandil/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Riñón/citología , Riñón/metabolismo , Masculino , Ratas
9.
Clin Exp Pharmacol Physiol ; 47(11): 1791-1797, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32881062

RESUMEN

At present, there is yet no specific antiviral treatment or immunization against the newly identified human severe acute respiratory syndrome virus (SARS-CoV2) that results in a rapidly progressive pandemic coronavirus disease 2019 (COVID-19). We believe in a crucial need for a clinical strategy to counteract this viral pandemic based on the known pathogenesis throughout the disease course. Evidence suggests that exaggerated patient's inflammatory response and oxidative stress are likely to aggravate the disease pathology. The resulting endothelial dysfunction further induces fibrosis and coagulopathy. These disturbances can generate severe acute respiratory distress syndrome (ARDS) that can progress into respiratory and circulatory failure. Nicorandil is an anti-anginal vasodilator drug acts by increasing nitric oxide bioavailability and opening of the KATP channel. Recently, nicorandil has been recognized to possess multiple protective effects against tissue injury. Here, we address a possible modulatory role of nicorandil against COVID-19 pathogenesis. We hypothesise nicorandil would be an effective form of adjuvant therapy against COVID-19.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Nicorandil/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Vasodilatadores/uso terapéutico , Antiinflamatorios/uso terapéutico , Anticoagulantes/uso terapéutico , Antioxidantes/uso terapéutico , COVID-19 , Infecciones por Coronavirus/fisiopatología , Fibrosis/prevención & control , Humanos , Pandemias , Neumonía Viral/fisiopatología , Tratamiento Farmacológico de COVID-19
10.
J Cell Physiol ; 234(7): 10942-10963, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30537190

RESUMEN

Wnt/ß-catenin signaling pathway plays a crucial role in diabetic cardiomyopathy (DCM), thus we aimed at investigating the effect of one therapeutic approach with resveratrol (RSV) given systemically and combined treatment of RSV with mesenchymal stem cells (MSCs) that was either RSV-preconditioned or not on Wnt/ß-catenin signaling pathway in streptozotocin-induced DCM, and to evaluate effects of RSV preconditioning on MSCs therapeutic potential. The rats were divided into control (C, n = 8), diabetic (DM, n = 8), diabetic treated with systemic RSV (DM-RSV, n = 8), diabetic treated with RSV and nonconditioned MSCs (DM-RSV-MSCs, n = 8), diabetic treated with RSV and RSV-incubated with MSCs (DM-RSV-MSCc, n = 8) and diabetic treated with RSV-conditioned MSCs (DM-MSCc, n = 8). Echocardiography (Echo) showed significant improvement of cardiac functions in all groups treated with RSV either systemic or added in culture media. Data of ejection fraction (EF%) of DM-RSV-MSCc (81.50; interquartile range [IQR], 80.00-83.00) was comparable to both DM-RSV-MSCs (77.50; IQR, 71.50-79.00), and DM-MSCc (71.50; IQR, 70.00-74.50). Histological examination of the left ventricles was performed for all groups. DM group revealed significant myocardial hypertrophy, apoptosis, interstitial fibrosis, and microvascular affection. All treated groups were associated, in variable degrees, with attenuation of cardiac hypertrophy and fibrosis, decreased area% for cardiac immunostaining of secreted frizzled-related protein (sFRP2) and Wnt/ß-catenin and improvement of the microvasculature. In conclusion, MSCs pretreated with resveratrol for 7 days showed increased 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and combined use of RSV (systemically and in culture media) significantly could improve cardiac remodeling capacity of MSCs via attenuation of sFRP2-mediated fibrosis and the downstream Wnt/ß-catenin pathway.


Asunto(s)
Antioxidantes/uso terapéutico , Cardiomiopatías Diabéticas/terapia , Fibrosis/terapia , Células Madre Mesenquimatosas/efectos de los fármacos , Resveratrol/uso terapéutico , Animales , Desviación Ósea , Diabetes Mellitus Experimental/complicaciones , Masculino , Trasplante de Células Madre Mesenquimatosas , Distribución Aleatoria , Ratas , Ratas Wistar
11.
Mod Rheumatol ; 29(4): 602-611, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30074417

RESUMEN

Objectives: The study aimed to evaluate the efficacy of combined gemfibrozil with prednisolone in the management of adjuvant-induced arthritis (AIA) rat model. Methods: Seventy two adult male Wistar albino rats were divided equally into 1-control group, three diseased groups: 2- Adjuvant induced arthritis (AIA), 3- high fat diet (HF), and 4- combined AIA-HF, and treated groups: 5- gemfibrozil 30 mg/kg treated AIA group (AIA-G) and the combined AIA-HF treated groups: 6- prednisolone equivalent to human 10 mg (AIA-HF-P10), 7- prednisolone equivalent to human 5 mg (AIA-HF-P5) 8- gemfibrozil (HF-AIA-G) and 9- combined treatment (HF-AIA-G-P5) Results: HF diet represents a precipitating factor for knee arthritis. Gemfibrozil improved the inflammatory findings in both AIA and AIA-HF groups. Combined administration of gemfibrozil with reduced steroid dose gave a similar therapeutic effect to the full steroid dose. Fortunately, we reported more reduction in the nuclear factor kappa-B (NF-κB) and high mobility group box 1 (HMGB1) in the HF-AIA-G-P5 compared with the HF-AIA-P10 group. The improvement was proved by the histological findings. Conclusion: Combined reduced prednisolone dose with gemfibrozil potentiates its anti-inflammatory activity. This could be a target in the management of rheumatoid arthritis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Gemfibrozilo/uso terapéutico , Prednisolona/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Artritis Experimental/metabolismo , Quimioterapia Combinada , Gemfibrozilo/administración & dosificación , Humanos , Masculino , FN-kappa B/metabolismo , PPAR alfa/agonistas , Prednisolona/administración & dosificación , Ratas , Ratas Wistar , Transducción de Señal
12.
Biochem Cell Biol ; 96(5): 564-571, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29425466

RESUMEN

The aim of this study was to investigate the effect of a regenerative therapy comprising mesenchymal stem cells (MSCs) pretreated with melatonin (MT) as a new therapy for underlying diabetic nephropathy (DN) pathogenesis in a rat model, and its possible effect on autophagy protein Beclin-1. Forty adult male albino Wistar rats were distributed among 4 groups: (i) control, (ii) DN, (iii) MSC-treated, and (iv) treated with MSCs that were pre-incubated in-vitro with MT (5 µmol·L-1 for 24 h; MSCs + MT). MSCs treatment significantly improved the renal functions and ameliorated the measured underlying DN pathogenesis and elevation of Beclin-1 protein levels compared with the DN group. In-vitro pretreatment of MSCs with MT enhanced proliferation and efficiency, and thus improved the kidney functions by increasing superoxide dismutase (SOD-1) and Beclin-1, and decreasing transforming growth factor (TGF-ß) markers in the kidney tissue, compared with the MSC group (P < 0.05). In conclusion: MSCs represent a promising target in DN management, and their effect can be intensified by pretreatment with MT. The elevated levels of Beclin-1 could be a mediator.


Asunto(s)
Nefropatías Diabéticas/terapia , Riñón/metabolismo , Melatonina/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Aloinjertos , Animales , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Riñón/patología , Masculino , Células Madre Mesenquimatosas/patología , Ratas , Ratas Wistar
13.
Clin Exp Nephrol ; 19(3): 419-26, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25002019

RESUMEN

BACKGROUND: Among the several disorders induced by sepsis, acute kidney injury (AKI) represents the most important economic burden problem that is associated with high mortality and morbidity rates. The aim of this study was to investigate the anti-inflammatory effects of ghrelin in sepsis-induced AKI and the possible role of vagus nerve. METHODS: Five groups were included: sham, cecal ligation and puncture (CLP), CLP-ghrelin, CLP-vagotomy and CLP-vagotomy-ghrelin group. RESULTS: Ghrelin treatment immediately after induction of CLP, significantly improved renal Glomerular filtration rate (GFR), serum creatinine, BUN and renal necrosis score as compared to the unprotected CLP group. In addition, ghrelin significantly decreased renal TNF alpha (111.5 ± 10.35 vs. 291.8 ± 15.8 pg/mg ptn), VCAM1 (6.28 ± 1.7 vs. 12.9 ± 1.2 µ/g ptn) and MPO (0.95 ± 0.13 vs. 2.5 ± 0.4 µ/g ptn) without significant increase in renal IL-10. Those effects were abolished by vagotomy. CONCLUSION: We concluded that ghrelin could represent new therapeutic window in early treatment of sepsis-induced AKI and this could be mainly due to its anti-inflammatory effects.


Asunto(s)
Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Ghrelina/uso terapéutico , Riñón/patología , Sepsis/complicaciones , Vagotomía , Lesión Renal Aguda/etiología , Animales , Presión Arterial/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Ciego/lesiones , Ciego/cirugía , Creatina/sangre , Creatina/orina , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Interleucina-10/metabolismo , Masculino , Necrosis/etiología , Peroxidasa/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/metabolismo , Nervio Vago , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Arch Physiol Biochem ; 129(1): 268-281, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36264662

RESUMEN

BACKGROUND: We sought to investigate thymoquinone (TQ)/quercetin combination in preventing hepatic steatosis (HS). MATERIALS AND METHODS: The included rat groups; (1) Control, (2) HS model, (3) HS treated with TQ 10 mg.kg-1.d-1, (4) HS treated with quercetin 50 mg.kg-1.d-1, and (5) HS treated with both compounds for 4 weeks. RESULTS: TQ/quercetin co-treatment augmented the anti-steatosis potential of each ingredient. The results revealed more (p < 0.001) sirtuin (SIRT1)/AMP-activated protein kinase (p-AMPK) upregulation compared to each treatment in line with autophagy protein Atg7 enhancement, and suppressed pro-inflammatory and oxidation markers. They diminished the hepatic lipogenic enzymes and perilipin-2 and activated the cytosolic lipases adipose triglyceride lipase (ATGL). Histological and Biochemical analysis revealed diminished lipid deposition and improved liver enzymes (alanine aminotransferase [ALT] and aspartate aminotransferase [AST]) compared to the data of separate treatments. CONCLUSION: TQ and quercitin effectively upregulated SIRT1/p-AMPK and regulated hepatic perilipin-2/ATGL, inflammation and oxidative stress, preserved liver structure and function. TQ/quercetin combination additively prevents HS.


Asunto(s)
Hígado Graso , Sirtuinas , Ratas , Animales , Quercetina/farmacología , Quercetina/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Perilipina-2/genética , Perilipina-2/metabolismo , Metabolismo de los Lípidos , Hígado Graso/prevención & control , Hígado/metabolismo , Lipasa , Autofagia
15.
Front Physiol ; 14: 1116705, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37415906

RESUMEN

Introduction: The high levels of angiotensin II (Ang II) can modify the vascular tone, enhance vascular smooth muscle cells (VSMCs) proliferation and hypertrophy and increase the inflammatory cellular infiltration into the vessel wall. The old herbal nonpharmacological agent, Hibiscus (HS) sabdariffa L has multiple cardioprotective impacts; thus, we investigated the role of HS extract in amelioration of renovascular hypertension (RVH)-induced aortic remodeling. Materials and methods: Thirty-five rats (7/group) were randomly allocated into 5 groups; group: I: Control-sham group, and RVH groups; II, III, IV, and V. The rats in RVH groups were subjected to the modified Goldblatt two-kidneys, one clip (2K1C) for induction of hypertension. In group: II, the rats were left untreated whereas in group III, IV, and V: RVH-rats were treated for 6 weeks with low dose hibiscus (LDH), medium dose hibiscus (MDH), and high dose hibiscus (HDH) respectively. Results: We found that the augmented pro-contractile response of the aortic rings was ameliorated secondary to the in-vivo treatment with HS dose dependently. The cyclophilin A (CyPA) protein levels positively correlated with the vascular adhesion molecule-1 (VCAM-1) and ERK1/2, which, in turn, contribute to the reactive oxygen species (ROS) production. Daily HS intake modified aortic renovation by enhancing the antioxidant capacity, restraining hypertrophy and fibrosis, downregulation of the metastasis associated lung adenocarcinoma transcript (MALAT1), and cyclophilin A (CyPA)/ERK1/2 levels. Discussion: Adding to the multiple beneficial effects, HS aqueous extract was able to inhibit vascular smooth muscle cell proliferation induced by 2K1C model. Thus, adding more privilege for the utilization of the traditional herbal extracts to attenuate RVH-induced aortopathy.

16.
Physiol Int ; 2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-36001412

RESUMEN

Scientific efforts have been made for a better understanding of the pathogenesis of hepatocellular carcinoma (HCC). We investigated the possible role of miR-192/nuclear factor-κB (NF-κB)/transforming growth factor-ß (TGF-ß)/E-cadherin in hepatic tumorigenesis. We expected a modulatory impact of thymoquinone. Thirty adult male rats were assigned into 3 groups (n = 10); (1) Control group. Group (2): Experimental HCC induced by intraperitoneal injection of diethylnitrosamine (DENA) followed by carbon tetrachloride (CCl4). Group (3): Thymoquinone 20 mg kg-1/oral supplementation starting from the model induction to the end of the 8th week. The HCC (DENA-CCL4) model was confirmed by elevated serum levels of alpha-fetoprotein and transaminases (ALT, AST) and by histopathological examination which denoted marked cellular atypia and features of neoplasia. Suppressed hepatic miR-192 and E-cadherin expression were detected in the HCC (DENA-CCL4) group accompanied by elevated tumor necrosis factor (TNF-α), interleukin (IL6)/NF-κB & TGF-ß1. Thymoquinone treatment protected the rat livers from hepatic tumorigenesis. Thymoquinone diminished (P < 0.001) alpha-fetoprotein and improved ALT, AST. It preserved hepatic miR-192 and normal E-cadherin expression. Thymoquinone-treated rats showed abrogated TNF-α, IL6/NF-κB/TGF-ß. Thymoquinone increased cell apoptosis markers Bax/Bcl2 and diminished cellular atypia. Pearson's correlations revealed positive association between miR-192 expression and E-cadherin and Bax/Bcl2 as well, and it was negatively correlated to alpha-fetoprotein, NF-κB and TGF-ß and the cellular atypia score. In conclusion, thymoquinone protected the liver tissues through preserving miR-192 and E-cadherin and aborting NF-κB & TGF-ß signaling. The current results highlight a new role for thymoquinone in preventing hepatic tumorigenesis.

17.
Diagnostics (Basel) ; 12(7)2022 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-35885665

RESUMEN

Liver fibrosis is a hallmark of thioacetamide (TAA) intoxications. MicroRNAs (miRs), such as miR-155, have been implied in the pathogenesis of liver disease, and regulated by the antioxidant and anti-inflammatory compound resveratrol (RES). The link between reactive oxygen species (ROS), tumour suppressor p53 (p53), and liver fibrosis-during the pathogenesis of TAA-induced liver injury-associated with miR-155 dysregulation with and without RES incorporation has not been previously studied. Therefore, one group of rats received TAA injections of 200 mg/kg; twice a week at the beginning of week 3 for 8 weeks (TAA group; or model group), whereas the protective group was pretreated daily with RES suspension (20 mg/kg; orally) for the first two weeks and subsequently sustained on receiving both RES and TAA until being sacrificed at the 10th week. Liver injuries developed in the model group were confirmed by a significant (p < 0.0001) elevation of hepatic tissue levels of miR-155, ROS, p53, and the profibrogenic biomarkers: tissue inhibitor of metalloproteinases-1 and α-smooth muscle actin, as well as collagen deposition (fibrosis). All these parameters were significantly (p ≤ 0.0234) protected by resveratrol (RES + TAA). In addition, we observed a significant (p < 0.0001) correlation between ROS/p53 axis mediated liver fibrosis and miR-155. Thus, TAA intoxication induced miR-155 imbalance and ROS/p53-mediated liver fibrosis, with resveratrol, conversely displaying beneficial hepatic pleiotropic effects for a period of 10 weeks.

18.
Egypt J Med Hum Genet ; 23(1): 97, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37521836

RESUMEN

Background: The angiotensin-converting enzyme-2 (ACE2) is recognized to be the fundamental receptor of severe acute respiratory syndrome coronavirus-2 (SARS-CoV2), responsible for the worldwide Coronavirus Disease-2019 (COVID-19) epidemic. However, genetic differences between people besides racial considerations and their relation to disease susceptibility are still not fully elucidated. Main body: To uncover the role of ACE2 in COVID-19 infection, we reviewed the published studies that explore the association of COVID-19 with the functional characteristics of ACE2 and its genetic variations. Notably, emerging studies tried to determine whether the ACE2 variants and/or expression could be associated with SARS-CoV/SARS-CoV2 have conflicting results. Some researchers investigated the potential of "population-specific" ACE2 genetic variations to impact the SARS-CoV2 vulnerability and suggested no ethnicity enrichment for ACE2 polymorphisms that could influence SARS-CoV2 S-protein binding. At the same time, some studies use data mining to predict several ACE2 variants that could enhance or decline susceptibility to SARS-CoV. On the other hand, fewer studies revealed an association of ACE2 expression with COVID-19 outcome reporting higher expression levels of ACE2 in East Asians. Conclusions: ACE2 gene variants and expression may modify the deleterious consequences of SARS-CoV2 to the host cells. It is worth noting that apart from the differences in gene expression and the genetic variations of ACE2, many other environmental and/or genetic factors could modify the disease outcome, including the genes for the innate and the adaptive immune response.

19.
Front Physiol ; 12: 744548, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899377

RESUMEN

Synchronized uterine receptivity with the time of implantation is crucial for pregnancy continuity. Vitamin D (VD) deficiency has been linked to the failure of implantation. Therefore, we tested the link between the Homeobox transcription factor-10/immunophilin FK506-binding protein 52 (HOXA-10/FKBP52) axis and the uterine receptivity in VD-deficient rats. The effect of VD supplementation at different doses was also investigated. Forty-eight pregnant rats were divided into six groups (eight/group); normal control rats fed with standard chow (control), control rats supplemented with VD (equivalent dose of 400 IU/day) (control-D400). VD-deficient group (DEF) and the three VD deficiency groups with VD supplementation were equivalent to 400, 4,000, and 10,000 IU/day (DEF-D400, DEF-D4000, and DEF-D10000, respectively). The expression levels of HOXA-10/FKBP52, progesterone level, and histological evaluation of decidualization using osteopontin (OSN) and progesterone receptor (PGR) were estimated. An assessment of the uterine contractility was conducted for all rats. This study showed the downregulation of HOXA-10/FKBP52 together with increased amplitude and frequency of the uterine contractility in the DEF group compared to control. VD dose-dependent supplementation restored progesterone/receptor competency, upregulated the expressional response of HOXA-10 and its downstream FKBP52, and improved uterine receptivity and endometrial decidualization at the time of implantation that was documented by increased area% of OSN and the number of implantation beads.

20.
Arch Physiol Biochem ; 124(5): 461-467, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29308676

RESUMEN

OBJECTIVE: We hypothesised that vitamin D has a beneficial renal protective effect from diabetic nephropathy (DN). METHODS: Four rat groups were included: normal control (control), type 2 diabetes for eight weeks (DM), treated group with angiotensin receptor blocker losartan (DM + L), and vitamin D-treated group started from the onset of diabetes (DM + Vit D). RESULTS: In the both treated groups, we found a significant (p < .05) reduction in the renal pro-inflammatory and profibrotic markers induced by diabetes. Vitamin D caused more reduction in monocyte chemoattractant protein-1 (MCP-1), transforming growth factor (TGFß-1), and renin-angiotensin levels that gave better kidney function compared to the DM + L group. CONCLUSION: Vitamin D may have a valuable role in the renal protective effect from DN, this may occur via expression of its VDR, Klotho and blocking renin-angiotensin activation, so vitamin D should be considered as a target in renal prophylactic measures against DN.


Asunto(s)
Nefropatías Diabéticas/prevención & control , Suplementos Dietéticos , Regulación de la Expresión Génica , Glucuronidasa/metabolismo , Riñón/metabolismo , Insuficiencia Renal/prevención & control , Vitamina D/uso terapéutico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Biomarcadores/sangre , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/terapia , Nefropatías Diabéticas/inmunología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Regulación hacia Abajo , Glucuronidasa/química , Glucuronidasa/genética , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/fisiopatología , Proteínas Klotho , Losartán/uso terapéutico , Masculino , Distribución Aleatoria , Ratas Wistar , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Insuficiencia Renal/complicaciones , Insuficiencia Renal/inmunología , Insuficiencia Renal/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA