Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int Immunopharmacol ; 129: 111662, 2024 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-38340421

RESUMEN

This study aimed to examine the effects of the secretome released by human umbilical cord-mesenchymal stem cells (MSC) as a result of preconditioning with deferoxamine (DFX), a hypoxia mimetic agent, on type 1 diabetes (T1D), by comparing it with the secretome produced by untreated MSCs. Initially, the levels of total protein, IL4, IL10, IL17, and IFNγ in the conditioned medium (CM) obtained from MSCs subjected to preconditioning with 150 µM DFX (DFX-CM) were analyzed in comparison to CM derived from untreated MSCs (N-CM). Subsequently, the CMs were administered to rats with T1D within a specific treatment plan. Following the sacrification, immunomodulation was evaluated by measuring serum cytokine levels and assessing the regulatory T cell (Treg) ratio in spleen mononuclear cells. Additionally, ß-cell mass was determined in the islets by immunohistochemical labeling of NK6 Homeobox 1 (Nkx6.1), Pancreatic duodenal homeobox-1 (Pdx1), and insulin antibodies in pancreatic sections. In vitro findings indicated that the secretome levels of MSCs were enhanced by preconditioning with DFX. In vivo, the use of DFX-CM significantly increased the Treg population, and accordingly, the level of inflammatory cytokines decreased. In ß-cell marker labeling, D + DFX-CM showed significantly increased PDX1 and insulin immunoreactivity. In conclusion, while the factors released by MSCs without external stimulation had limited therapeutic effects, substantial improvements in immunomodulation and ß-cell regeneration were seen with DFX-preconditioned cell-derived CM.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratas , Humanos , Animales , Deferoxamina/farmacología , Deferoxamina/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Secretoma , Citocinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Inmunomodulación , Cordón Umbilical , Regeneración
2.
Microsc Res Tech ; 87(1): 159-171, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37728208

RESUMEN

Mesenchymal stem/stromal cells (MSCs) hold significant therapeutic value due to their regeneration abilities, migration capacity, and immunosuppressive and immunomodulatory properties. These cells secrete soluble and insoluble factors, and this complex secretome contributes to their therapeutic effect. Furthermore, stimulation of cells by various external stimuli lead to secretome modifications that can increase the therapeutic efficacy. So, this study examined the effect of dimethyloxalylglycine (DMOG), a hypoxia-mimetic agent, on secretome profiles and exosome secretions of MSCs by evaluating conditioned medium (CM) and ultrastructural morphologies of the cells in comparison with unpreconditioned MSCs. The appropriate dose and duration of the use of DMOG were determined as 1000 µM and 24 h by evaluating the HIF-1α expression. DMOG-CM and N-CM were collected from MSCs incubated in serum-free medium with/without DMOG for 24 h, respectively. The content analysis of conditioned mediums (CMs) revealed that VEGF, NGF, and IL-4 levels were increased in DMOG-CM. Subsequently, exosomes were isolated from the CMs and were shown by transmission electron microscopy and Western blot analysis in both groups. The effects of CMs on proliferation and migration were determined by in vitro wound healing tests; both CMs increased the fibroblast's migratory and proliferative capacities. According to the ultrastructural evaluation, autophagosome, autolysosome, myelin figure, and microvesicular body structures were abundant in DMOG-preconditioned MSCs. Consistent with the high number of autophagic vacuoles, Beclin-1 expression was increased in those cells. These findings suggested that DMOG could alter MSCs' secretion profile, modify their ultrastructural morphology accordingly, and make the CM a more potent therapeutic tool. RESEARCH HIGHLIGHTS: Preconditioning mesenchymal stem/stromal cells with dimethyloxalylglycine, a hypoxia-mimetic agent, could modify cellular metabolism. Hypoxic mechanisms lead to alterations in the ultrastructural characteristics of mesenchymal stromal/stem cells. Preconditioning with dimethyloxalylglycine leads to ultrastructural and metabolic changes of mesenchymal stromal/stem cells along with modifications in their secretome profiles. Preconditioning of mesenchymal stromal/stem cells could render them a more potent therapeutic tool.


Asunto(s)
Células Madre Mesenquimatosas , Humanos , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Hipoxia/metabolismo , Cordón Umbilical/metabolismo
3.
Stem Cell Res Ther ; 13(1): 438, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36056427

RESUMEN

BACKGROUND: The therapeutic potential of mesenchymal stem cells (MSCs)-derived conditioned media (CM) can be increased after preconditioning with various chemical agents. The aim of this study is comparative evaluation of effects of N-CM and DFS-CM which are collected from normal (N) and deferoxamine (DFS) preconditioned umbilical cord-derived MSCs on rat diabetic nephropathy (DN) model. METHODS: After incubation of the MSCs in serum-free medium with/without 150 µM DFS for 48 h, the contents of N-CM and DFS-CM were analyzed by enzyme-linked immunosorbent assay. Diabetes (D) was induced by single dose of 55 mg/kg streptozotocin. Therapeutic effects of CMs were evaluated by biochemical, physical, histopathological and immunohistochemical analysis. RESULTS: The concentrations of vascular endothelial growth factor alpha, nerve growth factor and glial-derived neurotrophic factor in DFS-CM increased, while one of brain-derived neurotrophic factor decreased in comparison with N-CM. The creatinine clearance rate increased significantly in both treatment groups, while the improvement in albumin/creatinine ratio and renal mass index values were only significant for D + DFS-CM group. Light and electron microscopic deteriorations and loss of podocytes-specific nephrin and Wilms tumor-1 (WT-1) expressions were significantly restored in both treatment groups. Tubular beclin-1 expression was significantly increased for DN group, but it decreased in both treatment groups. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive apoptotic cell death increased in the tubules of D group, while it was only significantly decreased for D + DFS-CM group. CONCLUSIONS: DFS-CM can be more effective in the treatment of DN by reducing podocyte damage and tubular apoptotic cell death and regulating autophagic activity with its more concentrated secretome content than N-CM.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Células Madre Mesenquimatosas , Animales , Creatinina/metabolismo , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Deferoxamina , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratas , Cordón Umbilical/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
J Physiol Biochem ; 78(4): 869-883, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35907121

RESUMEN

The main goal of this study was to investigate the molecular changes in pancreatic progenitor cells subject to high glucose, aspartame, and metformin in vitro. This scope of work glucose, aspartame, and metformin were exposed to pancreatic islet derived progenitor cells (PID-PCs) for 10 days. GLUT1's role in beta-cell differentiation was examined by using GLUT1 inhibitor WZB117. Insulin+ cell ratio was measured by flow cytometry; the expression of beta-cell differentiation related genes was shown by RT-PCR; mitochondrial mass, mitochondrial ROS level, cytoplasmic Ca2+, glucose uptake, and metabolite analysis were made fluorometrically and spectrophotometrically; and proteins involved in related molecular pathways were determined by western blotting. Findings showed that glucose or aspartame exposed cells had similar metabolic and gene expression profile to control PID-PCs. Furthermore, relatively few insulin+ cells in aspartame treated cells were determined. Aspartame signal is transmitted through PLCß2, CAMKK2 and LKB1 in PID-PCs. The most obvious finding of this study is that metformin significantly increased beta-cell differentiation. The mechanism involves suppression of the sweet taste signal's molecules T1R3, PLCß2, cytoplasmic Ca+2, and AKT in addition to the direct effect of metformin on mitochondria and AMPK, and the energy metabolism of PID-PCs is remodelled in the direction of oxidative phosphorylation. These findings are very important in terms of determining that metformin stimulates the mitochondrial remodeling and the differentiation of PID-PCs to beta-cells and thus it may contribute to the compensation step, which is the first stage of diabetes development.


Asunto(s)
Metformina , Metformina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Fosfolipasa C beta/metabolismo , Fosfolipasa C beta/farmacología , Aspartame/metabolismo , Aspartame/farmacología , Mitocondrias/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Insulina/metabolismo , Diferenciación Celular , Células Madre/metabolismo
5.
Transl Res ; 246: 87-101, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35385790

RESUMEN

The small intestine, which is the area where sugars are absorbed, should be considered in the approaches developed for the treatment of diabetes. However, studies on small intestine damage in diabetic individuals, and the effects of current treatments on the small intestine are very limited. This is the first study to investigate the effects of exendin-4, a GLP-1 receptor agonist, on small intestine injury in diabetic mice. BALB/c male mice were divided into 4 groups for this study. The first group was given citrate buffer, the second group was given exendin-4, the third group was given streptozotocin (STZ), and the fourth group was given both exendin-4, and STZ. As the results, we determined a decrease in the edema and deterioration in the integrity of the villi, disruption in continuity of the brush border, fibrosis and enterocyte apoptosis, while the TNFα level and crypt cell proliferation were increased in the small intestinal tissue of exendin-4 treated STZ diabetic mice. Furthermore, the levels of duodenal tissue glucose, SGLT1, and GLUT2 were decreased, whereas there was an increase in GIP level in diabetic mice administered with exendin-4. Moreover, we determined that the sweet taste receptors T1R2/T1R3, downstream molecules PLCß2, α-gustducin and associated secondary messengers IP3, cAMP, which were increased in the duodenal tissue of STZ-diabetic mice, decreased with exendin-4 administration. These findings were evaluated as that exendin-4 reduces glucose absorption by suppressing the T1R2/T1R3 sweet taste signal perception pathway in duodenum of STZ diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental , Exenatida , Receptores Acoplados a Proteínas G , Gusto , Animales , Diabetes Mellitus Experimental/metabolismo , Exenatida/farmacología , Glucosa/metabolismo , Intestino Delgado/metabolismo , Masculino , Ratones , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Estreptozocina
6.
Food Chem Toxicol ; 153: 112264, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33992720

RESUMEN

This study aimed to investigate the molecular effects of the common natural sugar glucose and artificial sweetener aspartame on cancer stem cell (CSC) population and cancer aggressiveness of PANC-1 human pancreas adenocarcinoma cells. According to our findings while aspartame exposure significantly increased the CSC population, high glucose had no effect on it. The epithelial-mesenchymal transition marker N-cadherin increased only in the aspartame group. The findings indicate that a high level of glucose exposure does not effect the invasion and migration of PANC-1 cells, while aspartame increases both of these aggressiveness criteria. The findings also suggest that a high concentration of glucose maintains CSC population through induction of nuclear Oct3/4 and differentiation to parental cells via increasing cytoplasmic c-myc. Aspartame exposure to PANC-1 cells activated AKT and deactivated GSK3ß by increasing levels of ROS and cytoplasmic Ca+2, respectively, through T1R2/T1R3 stimulation. Then p-GSK3ß(Ser9) boosted the CSC population by increasing pluripotency factors Oct3/4 and c-myc via NICD, GLI1 and p21. In the aspartame group, T1R1 silencing further increased the CSC population but decreased cell viability and suppressed the p21, NICD and GLI activation. The presence and amount of T1R subunits in the membrane fraction of PANC-1 cells are demonstrated for the first time in this study, as is the regulatory effect of T1R1's on CSC population. In conclusion, the present study demonstrated that long-term aspartame exposure increases CSC population and tumor cell aggressiveness through p21, NICD, GLI1. Moreover, while aspartame had no tumorigenic effect, it could potentially advance an existing tumor.


Asunto(s)
Adenocarcinoma/metabolismo , Aspartame/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glucosa/farmacología , Humanos , Células Madre Neoplásicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
7.
Biol Trace Elem Res ; 199(4): 1515-1523, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32648196

RESUMEN

Vanadium compounds are being investigated as potential therapeutic agents in the treatment of many health problems, primarily diabetes. We aimed to provide the effect of N(1)-4-hydroxysalicylidene-N(4)-salicylidene-S-methyl-isothiosemicarbazidato-oxovanadium(IV) (VOL) on small intestinal injury in experimental male diabetic rats. Four groups were created of 3.0-3.5-month-old rats. The rats were made diabetic by a single dose of streptozotocin (STZ) at 65 mg/kg and grouped as follows: control animals, VOL-given control animals, STZ-induced diabetic animals and STZ-induced diabetic animals given VOL. A daily dose of 0.2 mM/kg vanadium complex was administered orally for 12 days after the inducement of diabetes. On the 12th day, small intestine tissue samples were taken. According to the data obtained from the biochemical analysis, reduced glutathione (GSH) level, catalase (CAT), glutathione peroxidase (GPx), glutathione-S-transferase (GST), superoxide dismutase (SOD), Na+/K+-ATPase and paraoxanase (PON) activities were increased, whereas sialic acid (SA), xanthine oxidase (XO) and disaccharidases (maltase and saccharidase) activities were decreased in the small intestine tissue of VOL-treated diabetic rats. Microscopic examinations revealed a remarkable decrease in the mucosal necrotic areas, discontinuity in the brush border, deterioration of the villi integrity and oedema inside the villi, but with a mild decrease in the inflammatory cells, deterioration and loss of integrity of the gland in the small intestine of VOL-treated diabetic rats. Moreover, VOL treatment markedly decreased the proliferation of villus cells and especially inflammatory cells in the small intestine of diabetic rats. According to the obtained data, the administration of VOL is a potentially convenient strategy to reducing small intestine injury in diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental , Tiosemicarbazonas , Animales , Glucemia , Catalasa/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Glutatión/metabolismo , Intestino Delgado/metabolismo , Masculino , Estrés Oxidativo , Ratas , Estreptozocina , Superóxido Dismutasa/metabolismo , Tiosemicarbazonas/farmacología
8.
Biochim Biophys Acta Mol Cell Res ; 1865(12): 1934-1944, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30290242

RESUMEN

The aim of this study was to investigate the molecular mechanism of pancreatic islet-derived mesenchymal stem cell (PID-MSC) differentiation into beta-cells in the presence of insulin and leptin resistance stimulators. We determined that beta-cell differentiation was stimulated by glucose, insulin, and leptin. Co-administration of insulin and leptin resulted in greater, at a further stage of differentiation but non-functional beta-cell formation. The levels of p-AKT(Ser473) did not change; SOCS3, PTP1B, p-IRS1(Ser307), PTEN levels increased and p-IRS1(Try) levels decreased due to insulin and leptin co-administration. These findings suggest that co-administration of insulin and leptin to PID-MSCs results in the development of both insulin and leptin resistance together. We showed that this differentiation signaling is mainly mediated by AKT/GSK-3ß/ß-catenin and Tub. Moreover, ß-catenin and Tub were linked to each other in the nucleus under this condition. Furthermore, we found that Tub and ß-catenin contributes to insulin production by increasing the expression of transcription factors by binding to the promoter regions of ins1, ins2, and pdx1 genes. In addition, Tub is also bound to the promoter region of the MafA gene. These findings demonstrate that when insulin and leptin resistance develop together in rat PID-MSCs beta-cell differentiation increases markedly via ß-catenin and Tub. New therapeutic agents that inhibit AKT/GSK-3ß/ß-catenin and in particular Tub may help prevent the development or retard the progression of type 2 diabetes.


Asunto(s)
Resistencia a la Insulina , Células Secretoras de Insulina/citología , Leptina/farmacología , Células Madre Mesenquimatosas/citología , Proteínas/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Insulina/farmacología , Células Secretoras de Insulina/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo
9.
Int Immunopharmacol ; 64: 52-59, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30145470

RESUMEN

Nerve growth factor (NGF) has been shown to protect the viability of kidney cells in acute phase of renal damage. However, since the half-life of NGF is very short, it is too large to pass the blood-brain barrier and rapidly transported to the liver for catabolizing its use in therapy is limited. 4-Methylcatechol (4MC) is a substance that increases NGF synthesis in many tissues. This study aimed to investigate the protective effects of 4MC against acute renal injury induced by streptozotocin (STZ). We have investigated the profibrotic, proinflammatory, oxidative changes in STZ-induced acute renal damage and the possible role of the NGF/TrkA system and Akt/GSK3ß/ß-catenin pathway in this mechanism. Experiment was designed as to be started with injection of 4MC for 10 days as a single dose (10 µg/kg) per day and to be terminated after 4 h of a single dose (75 mg/kg) STZ injection. As the result, 4MC pre-treatment decreased kidney damage, ROS production, the renal levels of TGFß1, CD68, tumor necrosis factor-α and interleukin 1ß. Moreover, 4MC pre-treatment increased levels of NGF and its receptor TrkA, p-Akt (Thr308), p-GSK3ß (Ser9) and nuclear ß-catenin. These data suggest that 4MC prevents the development of STZ-induced renal damage by suppressing ROS production and inflammation via Akt/GSK3ß/ß-catenin pathway which may be stimulated by NGF/TrkA signaling. Therefore, 4MC can be suggested as a potential agent for the prevention of acute renal injury.


Asunto(s)
Lesión Renal Aguda/prevención & control , Catecoles/farmacología , Glucógeno Sintasa Quinasa 3 beta/fisiología , Factor de Crecimiento Nervioso/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptor trkA/fisiología , Estreptozocina/toxicidad , beta Catenina/fisiología , Lesión Renal Aguda/inducido químicamente , Animales , Catecoles/uso terapéutico , Factor de Crecimiento Nervioso/análisis , Ratas , Ratas Wistar , Receptor trkA/análisis , Transducción de Señal/fisiología
10.
Peptides ; 99: 99-107, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29225158

RESUMEN

Glucagon-like peptide-1 (GLP-1) stimulates insulin secretion, - plays anti-inflammatory role in atherosclerosis, and has surfactant-releasing effects in lungs. GLP-1 analogues are used in diabetes therapy. This is the first study to investigate the effects of exendin-4, a GLP-1 receptor agonist, on lung injury in diabetic mice. BALB/c male mice were divided into four groups. The first group was given only citrate buffer, the second group was given only exendin-4, the third group was given only streptozotocin (STZ), and the fourth group was given both exendin-4 and STZ. Exendin-4 (3µg/kg) was administered daily by subcutaneous injection for 30days after mice were rendered diabetic with a single dose of STZ (200mg/kg). Structural alterations, oxidative stress, apoptosis, insulin signaling and expressions of prosurfactant-C, alpha-smooth muscle actin, collagen-I and fibronectin were evaluated in lung tissue. Diabetic mice lungs were characterized by induced oxidative stress, apoptosis, edema, and cell proliferation. They had honeycomb-like alveoli, thicker alveolar walls, and hypertrophic pneumocytes. Although exendin-4 treatment improved pulmonary edema, apoptosis, oxidative stress, and lung injury, it led to the disrupted insulin signaling and interstitial collagen accumulation in the lungs of diabetic mice. Exendin-4 ameliorates hyperglycemia-mediated lung damage by reducing glucose, -oxidative stress and stimulating cell proliferation. However, exendin-4 led to increased lung injury partly by reducing insulin signaling - and collagen accumulation around pulmonary vasculature in diabetic mice.


Asunto(s)
Complicaciones de la Diabetes/tratamiento farmacológico , Diabetes Mellitus Experimental/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Lesión Pulmonar/tratamiento farmacológico , Péptidos/farmacología , Ponzoñas/farmacología , Animales , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Exenatida , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Hiperglucemia/patología , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Mucosa Respiratoria/lesiones , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología
11.
J Cell Physiol ; 233(5): 4235-4244, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29058819

RESUMEN

Beta cell mass regulation represents a critical issue for understanding and treatment of diabetes. The most important process in the development of diabetes is beta cell death, generally induced by glucotoxicity or glucolipotoxicity, and the regeneration mechanism of new beta cells that will replace dead beta cells is still not fully understood. The aim of this study was to investigate the generation mechanism of new beta cells by considering the compensation phase of type 2 diabetes mellitus. In this study, pancreatic islet derived mesenchymal stem cells (PI-MSCs) were isolated from adult rats and characterized. Then, beta cells isolated from rats were co-cultured with PI-MSCs and they were exposed to glucotoxicity, lipotoxicity and glucolipotoxicity conditions for 72 hr. As the results apoptotic and necrotic cell death were increased in both PI-MSCs and beta cells especially by the exposure of glucotoxic and glucolipotoxic conditions to the co-culture systems. Glucotoxicity induced-differentiated beta cells were functional due to their capability of insulin secretion in response to rising glucose concentrations. Moreover, beta cell proliferation was induced in the glucotoxicity-treated co-culture system whereas suppressed in lipotoxicity or glucolipotoxicity-treated co-culture systems. In addition, 11 novel proteins, that may release from dead beta cells and have the ability to stimulate PI-MSCs in the direction of differentiation, were determined in media of glucotoxicity or glucolipotoxicity-treated co-culture systems. In conclusion, these molecules were considered as important for understanding cellular mechanism of beta cell differentiation and diabetes. Thus, they may be potential targets for diagnosis and cellular or therapeutic treatment of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Células Secretoras de Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Técnicas de Cocultivo , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica/genética , Glucosa/metabolismo , Humanos , Insulina/biosíntesis , Secreción de Insulina/genética , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Células Madre Mesenquimatosas/citología , Páncreas/metabolismo , Páncreas/patología , Ratas
12.
Int Immunopharmacol ; 36: 142-150, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27135545

RESUMEN

Gastrointestinal complications are frequent in renal transplant recipients. In this regard, renal ischemia/reperfusion injury (IRI)-induced gastric damage seems to be important and there is no data available on the mechanism of this pathology. Because of its anti-inflammatory and anti-oxidant properties, it can be suggested that prostaglandin-E1 (PGE1) protects cells from renal IRI-induced gastric damage. The aim of this study was to investigate the molecular mechanisms of gastric damage induced by renal IRI and the effect of PGE1 on these mechanisms. We set an experiment with four different animal groups: physiological saline-injected and sham-operated rats, PGE1 (20µg/kg)-administered and sham operated rats, renal IRI subjected rats, and PGE1-administered and renal IRI subjected rats. The protective effect of PGE1 on renal IRI-induced gastric damage was determined based on reduced histological damage and lactate dehydrogenase activity. Moreover, we demonstrated that PGE1 shows its protective effect through reducing the production of reactive oxygen species and malondialdehyde levels. During histological examination, we observed the presence of common mononuclear cell infiltration. Therefore, pro-inflammatory cytokines tumor necrosis factor-α and interleukin-1ß levels were measured and it has been shown that PGE1 suppressed both cytokines. Furthermore, it was found that PGE1 reduced the number of NF-κB(+) and caspase-3(+) inflammatory cells, and also NF-κB DNA-binding activity, while increasing proliferating cell nuclear antigen(+) epithelial cells in the stomach tissue of rats subjected to renal IR. Our data showed that PGE1 has a protective effect on renal IRI-induced oxidative stress and inflammation mediated gastric damage in rats.


Asunto(s)
Alprostadil/uso terapéutico , Antiinflamatorios/uso terapéutico , Gastritis/prevención & control , Trasplante de Riñón , Complicaciones Posoperatorias/prevención & control , Daño por Reperfusión/prevención & control , Estómago/efectos de los fármacos , Animales , Gastritis/etiología , Humanos , Interleucina-1beta/metabolismo , Riñón/patología , L-Lactato Deshidrogenasa/metabolismo , Masculino , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/etiología , Estómago/patología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Protoplasma ; 253(1): 127-35, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25802006

RESUMEN

The aim of present study was to investigate the effect of vitamin U (vit U, S-methylmethionine) on oxidative stress, inflammation, and fibrosis within the context of valproic acid (VPA)-induced renal damage. In this study, female Sprague Dawley rats were randomly divided into four groups: Group I consisted of intact animals, group II was given vit U (50 mg/kg/day, by gavage), group III was given VPA (500 mg/kg/day, intraperitonally), and group IV was given VPA + vit U. The animals were treated by vit U 1 h prior to treatment with VPA every day for 15 days. The following results were obtained in vit U + VPA-treated rats: (i) the protective effect of vit U on renal damage was shown by a significant decrease in histopathological changes and an increase in Na(+)/K(+)-ATPase activity; (ii) anti-oxidant property of vit U was demonstrated by a decrease in malondialdehyde levels and xanthine oxidase activity and an increase in glutathione levels, catalase and superoxide dismutase activities; (iii) anti-inflammatory property of vit U was demonstrated by a decrease in tumor necrosis factor-α, interleukin-1ß, monocyte chemoattractant protein-1 levels, and adenosine deaminase activity; (iv) anti-fibrotic effect of vit U was shown by a decrease in transforming growth factor-ß, collagen-1 levels, and arginase activity. Collectively, these data show that VPA is a promoter of inflammation, oxidative stress, and fibrosis which resulted in renal damage. Vit U can be proposed as a potential candidate for preventing renal damage which arose during the therapeutic usage of VPA.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Riñón/patología , Ácido Valproico/efectos adversos , Vitamina U/farmacología , Animales , Western Blotting , Catalasa/metabolismo , Colágeno Tipo I/metabolismo , Creatinina/sangre , Femenino , Fibrosis , Glutatión/metabolismo , Glutatión Transferasa/metabolismo , Immunoblotting , Inflamación/patología , Riñón/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Urea/sangre
14.
Pancreas ; 44(2): 243-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25401376

RESUMEN

OBJECTIVE: The aim of this study was to investigate the effects of nerve growth factor (NGF) neutralization on synthesis and secretion of activin A (Act-A) and betacellulin (BTC) from primary ß cells and the importance of these relations for ß-cell proliferation. METHODS: ß Cells were isolated from euglycemic and streptozotocin-induced (75 mg/kg) hyperglycemic rats and treated with NGF neutralization antibody. The gene expression levels of Act-A and BTC in the primary ß cells were evaluated using quantitative real-time polymerase chain reaction. The cellular and secreted levels of Act-A and BTC proteins were estimated using Western blot analysis. RESULTS: Nerve growth factor neutralization (1) reduced ß-cell proliferation, (2) decreased Act-A at gene expression and protein levels while increasing its secretion from ß cells, and (3) increased BTC at gene expression level while mildly decreasing its cellular protein level and secretion from ß cells. Nerve growth factor neutralization specifically affected ß cells of hyperglycemic rats. CONCLUSIONS: These findings indicate that NGF is an important regulator for the synthesis and secretion of Act-A and BTC from the ß cells. Moreover, the results suggested that ß-cell proliferation decreased through NGF neutralization is possibly related to decreased BTC and increased Act-A secretion from ß cells of hyperglycemic rats.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Betacelulina/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Subunidades beta de Inhibinas/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Betacelulina/genética , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica , Subunidades beta de Inhibinas/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Factor de Crecimiento Nervioso/inmunología , Factor de Crecimiento Nervioso/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Transducción de Señal/efectos de los fármacos
15.
Growth Factors ; 33(5-6): 419-29, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26728502

RESUMEN

In this study, we aimed to research the restorative effects of exendin-4, a GLP-1 analog, on renal tubular injury in streptozotocin-induced diabetes model. BALB/c male mice were divided into four groups: non-diabetic, non-diabetic + exendin-4 (3 µg/kg), diabetic and diabetic + exendin-4. In our diabetic model, we observed renal injury mainly in tubular area rather than glomeruli and exendin-4 decreased tubular injury with its glucose lowering effect. Besides, PCNA positive tubular cells, activities of LDH and Na(+)-K(+)-ATPase were also significantly declined by the administration of exendin-4. Furthermore, exendin-4 attenuated the levels of ROS, MDA, 8-OHdG, proinflammatory cytokines (TNF-α, IL-1ß), chemokine MCP-1, ICAM-1, and fibrosis-related molecules (transforming growth factor ß1 and fibronectin). In consistent with reducing tubular injury, macrophage infiltration and both MCP-1 and ICAM-1 production in tubular cells were decreased. These results indicate that exendin-4 may decrease renal tubular injury seen in the beginning of diabetic nephropathy by decreasing ROS production and inflammation.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Túbulos Renales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Péptidos/uso terapéutico , Ponzoñas/uso terapéutico , Animales , Quimiocina CCL2/biosíntesis , Diabetes Mellitus Experimental , Exenatida , Molécula 1 de Adhesión Intercelular/biosíntesis , Túbulos Renales/lesiones , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Antígeno Nuclear de Célula en Proliferación/metabolismo , Especies Reactivas de Oxígeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Estreptozocina , Factor de Necrosis Tumoral alfa/metabolismo
16.
Acta Histochem ; 116(1): 32-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23746671

RESUMEN

Chard is a plant used as an alternative hypoglycemic agent by diabetic people in Turkey. The aim of this study was to examine the molecular mechanism of hypoglycemic effects of chard extract. Male Sprague-Dawley rats (6-7 months old) were divided into five groups for this investigation: (1) control, (2) hyperglycemic, (3) hyperglycemic+chard, (4) hyperglycemic+insulin, (5) hyperglycemic+chard+insulin. Fourteen days after animals were rendered hyperglycemic by intraperitoneal injection of 60 mg/kg streptozotocin, the chard water extract (2 g/kg/day) or/and insulin (6 U/kg/day) was administered for 45 days. Hypoglycemic effect of chard extract was demonstrated by a significant reduction in the fasting blood glucose and increased glycogen levels in liver of chard extract-treated hyperglycemic rats. Moreover, activity of adenosine deaminase, which is suggested as an important enzyme for modulating the bioactivity of insulin, was decreased by chard treatment. Immunostaining analysis showed increased nuclear translocation of Akt2 and synthesis of GLUT2 in the hepatocytes of chard or/and insulin-treated hyperglycemic rats. The oxidative stress was decreased and antioxidant defense was increased by chard extract or/and insulin treatment to hyperglycemic rats according to the decreased malondialdehyde formation, the activities of catalase, superoxide dismutase, myeloperoxidase and increased glutathione levels. These findings suggest that chard extract might improve glucose response by increasing GLUT2 through Akt2 and antioxidant defense in the liver.


Asunto(s)
Beta vulgaris/química , Transportador de Glucosa de Tipo 2/metabolismo , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/farmacología , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transporte Activo de Núcleo Celular , Animales , Antioxidantes/metabolismo , Catalasa/metabolismo , Núcleo Celular/enzimología , Diabetes Mellitus Experimental/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Hipoglucemiantes/uso terapéutico , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Estrés Oxidativo , Peroxidasa/metabolismo , Extractos Vegetales/uso terapéutico , Ratas Sprague-Dawley , Estreptozocina , Superóxido Dismutasa/metabolismo
17.
Apoptosis ; 17(1): 14-24, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21938476

RESUMEN

Recent evidence suggested that tumor necrosis factor-alpha (TNF-α) and nerve growth factor (NGF) withdrawal activated a common apoptotic pathway. Here, we aimed to investigate the possible role of apoptotic Ras effectors RASSF1 and NORE1 in NGF reduction and TNF-α-related ß cell apoptosis in streptozotocin (STZ)-induced hyperglycemic rats. Rats were divided into four groups: the first group was given saline and citrate buffer, the second group was injected 4-methylcatechol (4-MC), an inducer of NGF synthesis, the third group received STZ, and the fourth group was given both 4-MC and STZ. 4-MC (10 µg/kg) was administered by daily intraperitoneal injection for 10 days before the animals were rendered hyperglycemic by administration of single dose STZ (75 mg/kg). With 4-MC pretreatment to hyperglycemic rats the following results were noted: (i) Decrease in pancreatic NGF level was blocked, (ii) Increase in pancreatic TNF-α level and the number of TNF-α(+) beta cell in the islets were prevented, (iii) Increase in the number of ß cell synthhesized apoptotic Ras effectors that RASSF1 and NORE1 was blocked, (iv) While pancreatic lipid peroxidation level decreased, antioxidant molecule glutathione and antioxidant enzymes glutathione peroxidase, catalase and superoxide dismutase activities increased, (v) Pancreatic caspase-3 activity and the number of cleaved caspase-3(+) ß cells were decreased. These results strengthen the idea that TNF-α and reduction in NGF can activate a common apoptotic pathway. Moreover, these data display that new apoptotic Ras effector molecules RASSF1 and NORE1 play important role with oxidative stress in NGF reduction and TNF-α-related pancreatic ß cell apoptosis in hyperglycemic rats. Furthermore, these findings suggest that 4-MC can prevent ß cell apoptosis possibly through increasing NGF synthesis in hyperglycemic rats.


Asunto(s)
Apoptosis , Hiperglucemia/metabolismo , Células Secretoras de Insulina/citología , Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Humanos , Hiperglucemia/genética , Hiperglucemia/fisiopatología , Células Secretoras de Insulina/metabolismo , Masculino , Factor de Crecimiento Nervioso/genética , Estrés Oxidativo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/genética , Proteínas Supresoras de Tumor/genética , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Neuropeptides ; 45(2): 143-50, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21295348

RESUMEN

In this study, it was aimed to investigate whether 4-methylcatechol (4-MC) could serve as an autocrine antiapoptotic agent by increasing nerve growth factor (NGF) in ß cells of hyperglycemic rats. Rats were divided into four groups: the first group was given citrate buffer and saline, the second group was administered 4-MC, the third group received streptozotocin (STZ), and the fourth group was given both 4-MC and STZ. 4-MC (10 µg/kg) was administered by daily intraperitoneal injection for 10 days before the animals were rendered hyperglycemic by administration of STZ (75 mg/kg). With 4-MC pretreatment on hyperglycemic rats the following results were noted: (i) Increase in plasma glucose, ß cell apoptosis and caspase-8 activation was prevented. (ii) Reduction of NGF+ and tyrosine receptor kinase A (TrkA)+ ß cell number was blocked. (iii) p75 neurotrophin receptor (p75(NTR))+ ß cell number was increased. These data suggest that 4-MC might exert its antiapoptotic actions through NGF/TrkA system which may block NGF/p75(NTR) activation in pancreatic ß cells of hyperglycemic rats.


Asunto(s)
Apoptosis/efectos de los fármacos , Catecoles/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Glucemia , Caspasa 8/metabolismo , Células Secretoras de Insulina/citología , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Estreptozocina/farmacología
19.
Peptides ; 32(2): 223-31, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21055431

RESUMEN

The hepatocytes express nerve growth factor (NGF) and its high affinity receptor tyrosine kinase A (TrkA). However, the link between NGF/TrkA system and hepatocyte proliferation in diabetic animals and the effects of exendin-4, a glucagon like peptide-1 (GLP-1) receptor agonist, on this system are not known. BALB/c male mice were divided into four groups. The first group was given citrate buffer only, the second group was administered exendin-4 alone, the third group received streptozotocin (STZ), and the fourth group was given both STZ and exendin-4. Exendin-4 (3µg/kg) was administered by subcutaneous injection daily for 30 days after the animals were rendered diabetic by administration of STZ (200mg/kg). With treatment of exendin-4 to the diabetic mice the following results were noted (i) NGF, TrkA and proliferating cell nuclear antigen positive hepatocytes were decreased; (ii) p75 neurotrophin receptor and caspase-3 positive hepatocyte could not be detected; (iii) liver alanine transaminase and aspartate transaminase activities, lipid peroxidation, protein carbonyl and myeloperoxidase levels were decreased; (iv) liver catalase, superoxide dismutase, glutathione peroxidase activities and glutathione levels were increased. These data suggest that exendin-4 might exerts its anti-proliferative action through blocking NGF/TrkA system and stimulating oxidative defense system in liver of diabetic mice.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Hepatocitos/patología , Hepatopatías/tratamiento farmacológico , Factor de Crecimiento Nervioso/metabolismo , Péptidos/farmacología , Receptor trkA/metabolismo , Ponzoñas/farmacología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Caspasa 3/metabolismo , Catalasa/metabolismo , Exenatida , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Peroxidación de Lípido/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Péptidos/uso terapéutico , Peroxidasa/metabolismo , Carbonilación Proteica/efectos de los fármacos , Receptor de Factor de Crecimiento Nervioso/metabolismo , Estreptozocina/farmacología , Superóxido Dismutasa/metabolismo , Ponzoñas/uso terapéutico
20.
Cell Biol Int ; 34(5): 543-52, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20128771

RESUMEN

This study was designed to evaluate the effect of Z-FA.FMK (benzyloxycarbonyl-l-phenylalanyl-alanine-fluoromethylketone), a pharmacological inhibitor of cathepsin B, on the proliferation of duodenal mucosal epithelial cells and the cellular system that controls this mechanism in these cells in vivo. For this investigation, BALB/c male mice were divided into four groups. The first group received physiological saline, the second group was administered Z-FA.FMK, the third group received D-GalN (D-galactosamine) and TNF-alpha (tumour necrosis factor-alpha) and the fourth group was given both D-GalN/TNF-alpha and Z-FA.FMK. When D-GalN/TNF-alpha was administered alone, we observed an increase in IL-1beta-positive and active NF-kappaB-positive duodenal epithelial cells, a decrease in PCNA (proliferative cell nuclear antigen)-positive duodenal epithelial cells and an increase in degenerative changes in duodenum. On the other hand, Z-FA.FMK pretreatment inhibited all of these changes. Furthermore, lipid peroxidation, protein carbonyl and collagen levels were increased, glutathione level and superoxide dismutase activity were decreased, while there was no change in catalase activity by D-GalN/TNF-alpha injection. On the contrary, the Z-FA.FMK pretreatment before D-GalN/TNF-alpha blocked these effects. Based on these findings, we suggest that Z-FA.FMK might act as a proliferative mediator which is controlled by IL-1beta through NF-kappaB and oxidative stress in duodenal epithelial cells of D-GalN/TNF-alpha-administered mice.


Asunto(s)
Dipéptidos/farmacología , Duodeno , Células Epiteliales , Galactosamina/farmacología , Interleucina-1beta/metabolismo , Cetonas/farmacología , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Duodeno/citología , Duodeno/efectos de los fármacos , Duodeno/patología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Fallo Hepático/inducido químicamente , Masculino , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA