Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Med Sci ; 21(8): 1491-1499, 2024.
Article in English | MEDLINE | ID: mdl-38903928

ABSTRACT

Age-related structural and functional changes in the kidney can eventually lead to development of chronic kidney disease, which is one of the leading causes of mortality among elderly people. For effective management of age-related kidney complications, it is important to identify new therapeutic interventions with minimal side-effects. The present study was designed to evaluate the synergistic effect of a traditional Chinese herb, Alpinate Oxyphyllae Fructus (AOF), and adipose-derived mesenchymal stem cells (ADMSCs) in ameliorating D-galactose (D-gal)-induced renal aging phenotypes in WKY rats. The study findings showed that D-gal-induced alteration in the kidney morphology was partly recovered by the AOF and ADMSC co-treatment. Moreover, the AOF and ADMSC co-treatment reduced the expression of proinflammatory mediators (NFkB, IL-6, and Cox2) and increased the expression of redox regulators (Nrf2 and HO-1) in the kidney, which were otherwise augmented by the D-gal treatment. Regarding kidney cell death, the AOF and ADMSC co-treatment was found to abolish the proapoptotic effects of D-gal by downregulating Bax and Bad expressions and inhibiting caspase 3 activation. Taken together, the study findings indicate that the AOF and ADMSC co-treatment protect the kidney from D-gal-induced aging by reducing cellular inflammation and oxidative stress and inhibiting renal cell death. This study can open up a new path toward developing novel therapeutic interventions using both AOF and ADMSC to effectively manage age-related renal deterioration.


Subject(s)
Drugs, Chinese Herbal , Galactose , Kidney , Mesenchymal Stem Cells , Animals , Galactose/adverse effects , Rats , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Kidney/drug effects , Kidney/pathology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Oxidative Stress/drug effects , Male , Apoptosis/drug effects , Mesenchymal Stem Cell Transplantation/methods , Humans , Renal Insufficiency, Chronic/therapy , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/drug therapy
2.
Aging (Albany NY) ; 15(5): 1358-1370, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36863713

ABSTRACT

Parkinson's disease (PD) is a common disorder attributed to the loss of midbrain dopamine (mDA) neurons and reduced dopamine secretion. Currently, the treatment regimes for PD comprise deep brain stimulations, however, it attenuates the PD progression marginally and does not improve neuronal cell death. We investigated the function of Ginkgolide A (GA) to reinforce Wharton's Jelly-derived mesenchymal stem cells (WJMSCs) for treating the in vitro model of PD. GA enhanced the self-renewal, proliferation, and cell homing function of WJMSCs as assessed by MTT and transwell co-culture assay with a neuroblastoma cell line. GA pre-treated WJMSCs can restore 6-hydroxydopamine (6-OHDA)-induced cell death in a co-culture assay. Furthermore, exosomes isolated from GA pre-treated WJMSCs significantly rescued 6-OHDA-induced cell death as determined by MTT assay, flow cytometry, and TUNEL assay. Western blotting showed that apoptosis-related proteins were decreased following GA-WJMSCs exosomal treatment which further improved mitochondrial dysfunction. We further demonstrated that exosomes isolated from GA-WJMSCs could restore autophagy using immunofluorescence staining and immunoblotting assay. Finally, we used the alpha-synuclein recombinant protein and found that exosomes derived from GA-WJMSCs led to the reduced aggregation of alpha-synuclein compared to that in control. Our results suggested that GA could be a potential candidate for strengthening stem cell and exosome therapy for PD.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Neuroprotective Agents , Parkinson Disease , Humans , Oxidopamine/toxicity , alpha-Synuclein/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Dopamine/metabolism , Mesenchymal Stem Cells/metabolism
3.
Probiotics Antimicrob Proteins ; 15(5): 1287-1297, 2023 10.
Article in English | MEDLINE | ID: mdl-36044175

ABSTRACT

Diabetes-related brain complications have been reported in clinical patients and experimental models. The objective of the present study was to investigate the neuroprotective mechanisms of Lactobacillus reuteri GMNL-263 in streptozotocin (STZ)-induced diabetic rats. In this study, three different groups, namely control group, STZ-induced (55 mg/kg streptozotocin intraperitoneally) diabetic rats (DM), and DM rats treated with Lactobacillus reuteri GMNL-263 (1 × 109 CFU/rat/day), were utilized to study the protective effect of GMNL-263 in the hippocampus of STZ-induced diabetic rats. The results demonstrated that GMNL-263 attenuated diabetes-induced hippocampal damage by enhancing the cell survival pathways and repressing both inflammatory and apoptotic pathways. Histopathological analysis revealed that GMNL-263 prevented structural changes in the hippocampus in the DM group and decreased the level of inflammation and apoptosis in the hippocampus of DM rats. The IGF1R cell survival signaling pathway also improved after GMNL-263 treatment. These results indicate that probiotic GMNL-263 exerts beneficial effects in the brain of diabetic rats and has potential ability for clinical application.


Subject(s)
Diabetes Mellitus, Experimental , Limosilactobacillus reuteri , Neuroprotective Agents , Probiotics , Rats , Animals , Neuroprotective Agents/pharmacology , Streptozocin/adverse effects , Streptozocin/metabolism , Hippocampus
4.
Drug Chem Toxicol ; 46(5): 1044-1050, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36216784

ABSTRACT

To date, few studies have investigated the toxicological effects of the combined use of amphetamine and heroin in the heart. Hence, the aim of this study was to identify indicators for clinical evaluation and prevention of cardiac injury induced by the combined use of amphetamine and heroin. Four different groups were analyzed: (1) normal group (n=25;average age=35 ± 6.8); (2) heart disease group (n=25;average age=58 ± 17.2); (3) drug abusers (n = 27; average age = 37 ± 7.7); (4) drug abstainers (previous amphetamine-heroin users who had been drug-free for more than two weeks; n = 22; average age = 35 ± 5.6). The activity of MMPs, and levels of TNF-α, IL-6, GH, IGF-I, and several serum biomarkers were examined to evaluate the impact of drug abuse on the heart. The selected plasma biomarkers and classic cardiac biomarkers were significantly increased compared to the normal group. The zymography data showed the changes in cardiac-remodeling enzymes MMP-9 and MMP-2 among combined users of amphetamine and heroin. The levels of TNF-α and IL-6 only increased in the heart disease group. Growth hormone was increased; however, IGF-I level decreased with drug abuse and the level was not restored by abstinence. We speculated that the amphetamine-heroin users might pose risk to initiate heart disease even though the users abstained for more than two weeks. The activity change of MMP-9 and MMP-2 can be a direct reason affecting heart function. The indirect reason may be related to liver damage by drug abuse reduce IGF-1 production to protect heart function.


Subject(s)
Heart Diseases , Heart Injuries , Heroin Dependence , Humans , Adult , Middle Aged , Aged , Insulin-Like Growth Factor I , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Heroin , Heroin Dependence/complications , Interleukin-6 , Tumor Necrosis Factor-alpha , Amphetamine , Biomarkers
5.
J Food Biochem ; 46(5): e14041, 2022 05.
Article in English | MEDLINE | ID: mdl-35064587

ABSTRACT

Growing evidences indicate that high glucose toxicity-associated fibrotic effects play a pivotal role in diabetic nephropathy (DN). Tubular epithelial-myofibroblast transdifferentiation is a major hallmark of renal fibrosis event under diabetic stress. Roots of Glycyrrhiza uralensis (Radix glycyrrhizae) used as a sweetener and traditional Chinese medicine possess high potential for renal protection. In this study, a cell model for high glucose (HG) injury with HK-2 renal proximal tubular epithelial cell line and a type-II-diabetes model with Apoeem1/Narl /Narl mice was established and the beneficial effects of aqueous R. glycyrrhizae extract (RGE) was investigated. RGE-induced regulation on the high glucose-induced excessive production of TGF-ß1 and the Smad/Stat3 mechanisms of renal fibrosis were determined. HK-2 cells were challenged with 45 mM of high glucose for 48 hr. Following high glucose challenge, the cells were treated with 0.5, 1, and 1.5 mg/ml concentrations of RGE. The effect of RGE on DN was determined using high fructose diet-induced type-II-diabetes in Apoeem1/Narl /Narl mice models. Our results showed that RGE suppressed the expression of HG-induced TGFß signaling and associated fibrosis mechanism better than the pharmacological drug acarbose. These data suggest that RGE as a potential herbal supplement in attenuating fibrosis-associated diabetic nephropathy and a potential agent in diabetes treatments.


Subject(s)
Diabetic Nephropathies , Drugs, Chinese Herbal , Glycyrrhiza uralensis , Animals , Cell Line , Cell Transdifferentiation , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Drugs, Chinese Herbal/pharmacology , Fibrosis , Glucose , Glycyrrhiza uralensis/chemistry , Humans , Mice , Myofibroblasts/metabolism , Plant Extracts/pharmacology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
Mol Cell Biochem ; 477(1): 143-152, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34586566

ABSTRACT

The heart is a very dynamic pumping organ working perpetually to maintain a constant blood supply to the whole body to transport oxygen and nutrients. Unfortunately, it is also subjected to various stresses based on physiological or pathological conditions, particularly more vulnerable to damages caused by oxidative stress. In this study, we investigate the molecular mechanism and contribution of IGF-IIRα in endoplasmic reticulum stress induction in the heart under doxorubicin-induced cardiotoxicity. Using in vitro H9c2 cells, in vivo transgenic rat cardiac tissues, siRNAs against CHOP, chemical ER chaperone PBA, and western blot experiments, we found that IGF-IIRα overexpression enhanced ER stress markers ATF4, ATF6, IRE1α, and PERK which were further aggravated by DOX treatment. This was accompanied by a significant perturbation in stress-associated MAPKs such as p38 and JNK. Interestingly, PARKIN, a stress responsive cellular protective mediator was significantly downregulated by IGF-IIRα concomitant with decreased expression of ER chaperone GRP78. Furthermore, ER stress-associated pro-apoptotic factor CHOP was increased considerably in a dose-dependent manner followed by elevated c-caspase-12 and c-caspase-3 activities. Conversely, treatment of H9c2 cells with chemical ER chaperone PBA or siRNA against CHOP abolished the IGF-IIRα-induced ER stress responses. Altogether, these findings suggested that IGF-IIRα contributes to ER stress induction and inhibits cellular stress coping proteins while increasing pro-apoptotic factors feeding into a cardio myocyte damage program that eventually paves the way to heart failure.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Myocardium/metabolism , Receptor, IGF Type 2/metabolism , Animals , Cell Line , Cytotoxins/adverse effects , Cytotoxins/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Endoplasmic Reticulum/genetics , Rats , Rats, Transgenic , Receptor, IGF Type 2/genetics
7.
Aging (Albany NY) ; 13(13): 17536-17547, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34233296

ABSTRACT

Pathological manifestations in either heart or kidney impact the function of the other and form the basis for the development of cardiorenal syndrome. However, the mechanism or factors involved in such scenario are not completely elucidated. In our study, to find the correlation between late fetal gene expression in diabetic hearts and their influence on diabetic nephropathy, we created a rat model with cardiac specific overexpression of IGF-IIRα, which is an alternative splicing variant of IGFIIR, expressed in pathological hearts. In this study, transgenic rats over expressing cardiac specific IGF-IIRα and non-transgenic animal models established in SD rats were administered with single dose of streptozotocin (STZ, 55 mg/Kg) to induce Type I diabetes. The correlation between IGF-IIRα and kidney damages were further determined based on their intensity of damage in the kidneys. The results show that cardiac specific overexpression of IGF-IIRα elevates the diabetes associated inflammation and morphological changes in the kidneys. The diabetic transgenic rats showed advancement in the pathological features such a renal tubular damage, collagen accumulation and enhancement in STAT3 associated mechanism of renal fibrosis. The results therefore show that that IGF-IIRα expression in the heart during pathological condition may worsen symptoms of diabetic nephropathy in rats.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Receptor, IGF Type 2/genetics , Animals , Apoptosis/genetics , Collagen/metabolism , Fibrosis , Gene Expression Regulation , Kidney/pathology , Kidney Tubules/pathology , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , STAT3 Transcription Factor/genetics
9.
Int J Med Sci ; 17(13): 1916-1926, 2020.
Article in English | MEDLINE | ID: mdl-32788870

ABSTRACT

Aging is the most important current issue and is usually accompanied by complications, such as cardiovascular disorders and neurodegenerative diseases, which are the leading causes of death worldwide and the second major cause of death in Taiwan. In this study, we have investigated the protective effect of adipose-derived mesenchymal stem cells (ADSCs) and the role of epigallocatechin gallate (EGCG) in enhancing this effect in aging cerebral cortex of rats. Further, we attempted to elucidate the molecular mechanism through which EGCG influences the protective effects of ADSC. ADSCs, co-cultured with EGCG, were injected into 20-month-old Wistar rats. Hematoxylin and eosin staining of the cerebral cortex revealed noticeable neurogenic activity and visible improvements in the integrity of the pre-frontal cortex tissue, compared to that in rats treated with ADSCs alone. Western blot analysis confirmed that ADSC, co-cultured with EGCG, enhanced cell survival via the p-Akt pathway and improved mitochondrial biogenesis via the SIRT-1 pathway. Moreover, it increased the available brain-derived neurotrophic factor to a higher degree than that in the ADSC group. Furthermore, western blotting showed that EGCG improved the antioxidant activity of the ADSCs in the cortex tissues via the Nrf-2 and HO-1 pathway. Based on these findings, we propose that this variation in stem cell treatment may facilitate functional recovery and enhanced neuroprotection in aged brains.


Subject(s)
Aging/drug effects , Catechin/analogs & derivatives , Cerebral Cortex/diagnostic imaging , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Aging/pathology , Animals , Catechin/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Coculture Techniques , Humans , Mesenchymal Stem Cells/metabolism , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Rats , Signal Transduction/drug effects , Taiwan
10.
Aging (Albany NY) ; 12(8): 6852-6864, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32320382

ABSTRACT

Hippocampus is one of the most vulnerable brain regions in terms of age-related pathological change. Exercise is presumed to delay the aging process and promote health because it seems to improve the function of most of the aging mechanisms. The purpose of this study is to evaluate the effects of swimming exercise training on brain inflammation, apoptotic and survival pathways in the hippocampus of D-galactose-induced aging in SD rats. The rats were allocated to the following groups: (1) control; (2) swimming exercise; (3) induced-aging by injecting D-galactose; (4) induced-aging rats with swimming exercise. The longevity-related AMPK/SIRT1/PGC-1α signaling pathway and brain IGF1/PI3K/Akt survival pathway were significantly reduced in D-galactose-induced aging group compared to non-aging control group and increased after exercise training. The inflammation pathway markers were over-expressed in induced-aging hippocampus, exercise significantly inhibited the inflammatory signaling activity. Fas-dependent and mitochondrial-dependent apoptotic pathways were significantly increased in the induced-aging group relative to the control group whereas they were decreased in the aging-exercise group. This study demonstrated that swimming exercise not only reduced aging-induced brain apoptosis and inflammatory signaling activity, but also enhanced the survival pathways in the hippocampus, which provides one of the new beneficial effects for exercise training in aging brain.


Subject(s)
Aging/physiology , Hippocampus/physiology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Signal Transduction , Swimming/physiology , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Aging/pathology , Animals , Apoptosis , Caspases/metabolism , Cyclooxygenase 2/metabolism , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Hippocampus/pathology , Inflammation/metabolism , Male , NF-kappa B/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, IGF Type 1/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL