Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 70
1.
Biomed Pharmacother ; 168: 115759, 2023 Dec.
Article En | MEDLINE | ID: mdl-37865993

Mesenchymal stem cell (MSC) transplantation offers significant potential for the treatment of diabetes mellitus (DM) and its complications. However, hyperglycemic conditions can induce senescence and dysfunction in both transplanted and resident MSCs, thereby limiting their therapeutic potential. Mitochondrial dysfunction and oxidative stress are key contributors to this process in MSCs exposed to hyperglycemia. As such, strategies aimed at mitigating mitochondrial dysfunction could enhance the therapeutic efficacy of MSC transplantation in DM. In this review, we provide an updated overview of how mitochondrial dysfunction mediates MSC senescence. We present experimental evidence for the molecular mechanisms behind high glucose-induced mitochondrial dysfunction in MSCs, which include impairment of mitochondrial biogenesis, mitochondrial calcium regulation, the mitochondrial antioxidant system, mitochondrial fusion-fission dynamics, mitophagy, and intercellular mitochondrial transfer. Furthermore, we propose potential pharmacological candidates that could improve the efficacy of MSC transplantation by enhancing mitochondrial function in patients with DM and related complications.


Diabetes Mellitus , Hyperglycemia , Mesenchymal Stem Cell Transplantation , Humans , Diabetes Mellitus/therapy , Diabetes Mellitus/metabolism , Mitochondria/metabolism , Hyperglycemia/metabolism , Oxidative Stress/physiology , Cellular Senescence
2.
Antioxidants (Basel) ; 12(9)2023 Sep 06.
Article En | MEDLINE | ID: mdl-37760030

Diabetic retinopathy (DR) is a leading cause of blindness in diabetic patients. Umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are emerging as a promising new drug for degenerative disease associated with diabetes. Recent studies have shown that high glucose-increased excessive calcium levels are a major risk factor for mitochondrial reactive oxygen species (mtROS) accumulation and apoptosis. This study aimed to investigate the role of high glucose-induced NFATC1 signaling in mitochondrial oxidative stress-stimulated apoptosis and the effect of tacrolimus on the therapeutic efficacy of subconjunctival transplantation of UCB-MSCs in a DR rat model. High glucose increased mtROS and cleaved caspase-9 expression in UCB-MSCs. High glucose conditions increased O-GlcNAcylated protein expression and nuclear translocation of NFATC1. Tacrolimus pretreatment recovered high glucose-induced mtROS levels and apoptosis. In the DR rat model, subconjunctival transplantation of tacrolimus-pretreated MSCs improved retinal vessel formation, retinal function, and uveitis. In high glucose conditions, tacrolimus pretreatment reduced protein and mRNA expression levels of DRP1 and inhibited mitochondrial fission. In conclusion, we demonstrated that high glucose-induced O-GlcNAcylation activates NFATC1 signaling, which is important for DRP1-mediated mitochondrial fission and mitochondrial apoptosis. Finally, we proposed NFATC1 suppression by tacrolimus as a promising therapeutic strategy to improve the therapeutic efficacy of UCB-MSC transplantation for DR treatment.

3.
Autophagy ; 19(10): 2752-2768, 2023 10.
Article En | MEDLINE | ID: mdl-37357416

ABBREVIATIONS: Aß: amyloid ß; AD: Alzheimer disease; AMPK: 5' adenosine monophosphate-activated protein kinase; CTSB: cathepsin B; CTSD: cathepsin D; DM: diabetes mellitus; ESCRT: endosomal sorting complex required for transport; FBXO27: F-box protein 27; iPSC-NDs: induced pluripotent stem cell-derived neuronal differentiated cells; LAMP1: lysosomal-associated membrane protein 1; LMP: lysosomal membrane permeabilization; LRSAM1: leucine rich repeat and sterile alpha motif containing 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTORC1: mechanistic target of rapamycin kinase complex 1; p-MAPT/tau: phosphorylated microtubule associated protein tau; ROS: reactive oxygen species; STZ: streptozotocin; TFE3: transcription factor E3; TFEB: transcription factor EB; TRIM16: tripartite motif containing 16; UBE2QL1: ubiquitin conjugating enzyme E2 Q family like 1; VCP: valosin containing protein.


Autophagy , Macroautophagy , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Glucose/metabolism , Lysosomes/metabolism
4.
Neurobiol Dis ; 177: 106009, 2023 02.
Article En | MEDLINE | ID: mdl-36689912

Heavy alcohol consumption causes neuronal cell death and cognitive impairment. Neuronal cell death induced by ethanol may result from increased production of the sphingolipid metabolite ceramide. However, the molecular mechanisms of neuronal cell death caused by ethanol-induced ceramide production have not been elucidated. Therefore, we investigated the mechanism through which ethanol-induced ceramide production causes neuronal cell apoptosis using human induced-pluripotent stem cell-derived neurons and SH-SY5Y cells and identified the effects of ceramide on memory deficits in C57BL/6 mice. First, we found that ethanol-induced ceramide production was decreased by inhibition of the de novo synthesis pathway, mediated by serine palmitoyltransferase (SPT). The associated alterations of the molecules related to the ceramide pathway suggest that the elevated level of ceramide activated protein phosphatase 1 (PP1), which inhibited the nuclear translocation of serine/arginine-rich splicing factor 1 (SRSF1). This led to aberrant splicing of myeloid cell leukemia 1 (MCL-1) pre-mRNA, which upregulated MCL-1S expression. Our results demonstrated that the interaction of MCL-1S with the inositol 1, 4, 5-trisphosphate receptor (IP3R) increases calcium release from the endoplasmic reticulum (ER) and then activated ER-bound inverted formin 2 (INF2). In addition, we discovered that F-actin polymerization through INF2 activation promoted ER-mitochondria contacts, which induced mitochondrial calcium influx and mitochondrial reactive oxygen species (mtROS) production. Markedly, MCL-1S silencing decreased mitochondria-associated ER membrane (MAM) formation and prevented mitochondrial calcium influx and mtROS accumulation, by inhibiting INF2-dependent actin polymerization interacting with mitochondria. Furthermore, the inhibition of ceramide production in ethanol-fed mice reduced MCL-1S expression, neuronal cell death, and cognitive impairment. In conclusion, we suggest that ethanol-induced ceramide production may lead to mitochondrial calcium overload through MCL-1S-mediated INF2 activation-dependent MAM formation, which promotes neuronal apoptosis.


Ceramides , Neuroblastoma , Humans , Mice , Animals , Ceramides/metabolism , Ethanol/pharmacology , Calcium/metabolism , Mice, Inbred C57BL , Neuroblastoma/metabolism , Apoptosis , Mitochondria/metabolism , Endoplasmic Reticulum/metabolism , Serine-Arginine Splicing Factors
5.
J Vet Sci ; 24(1): e3, 2023 Jan.
Article En | MEDLINE | ID: mdl-36560835

BACKGROUND: Zinc (Zn) is an essential cofactor for physiological homeostasis in the body. Zn oxide (ZnO), an inorganic compound that supplies Zn, exists in various sizes, and its bioavailability may vary depending on the size in vivo. However, comparative studies on the nutritional effects of micro-sized ZnO (M-ZnO) and nano-sized ZnO (N-ZnO) supplementation on Zn deficiency (ZnD) animal models have not been reported. OBJECTIVES: This study investigated the nutritional bioavailability of N-ZnO and M-ZnO particles in dietary-induced ZnD mice. METHODS: Animals were divided into six experimental groups: normal group, ZnD control group, and four ZnO treatment groups (Nano-Low, Nano-High, Micro-Low, and Micro-High). After ZnD induction, N-ZnO or M-ZnO was administered orally every day for 4 weeks. RESULTS: ZnD-associated clinical signs almost disappeared 7 days after N-ZnO or M-ZnO administration. Serum Zn concentrations were higher in the Nano-High group than in the ZnD and M-ZnO groups on day 7 of ZnO treatment. In the liver and testis, Nano-Low and Nano-High groups showed significantly higher Zn concentrations than the other groups after 14-day treatment. ZnO supplementation increased Mt-1 mRNA expression in the liver and testis and Mt-2 mRNA expression in the liver. Based on hematoxylin-and-eosin staining results, N-ZnO supplementation alleviated histological damage induced by ZnD in the testis and liver. CONCLUSIONS: This study suggested that N-ZnO can be utilized faster than M-ZnO for nutritional restoration at the early stage of ZnD condition and presented Mt-1 as an indicator of Zn status in the serum, liver, and testis.


Zinc Oxide , Animals , Male , Mice , RNA, Messenger , Zinc/therapeutic use , Zinc Oxide/pharmacology
6.
J Vet Sci ; 23(5): e74, 2022 Sep.
Article En | MEDLINE | ID: mdl-36174978

BACKGROUND: Previous studies have presented evidence to support the significant association between red meat intake and colon cancer, suggesting that heme iron plays a key role in colon carcinogenesis. Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, exhibits anti-oxidative and anti-cancer effects. However, the effect of EGCG on red meat-associated colon carcinogenesis is not well understood. OBJECTIVES: We aimed to investigate the regulatory effects of hemin and EGCG on colon carcinogenesis and the underlying mechanism of action. METHODS: Hemin and EGCG were treated in Caco2 cells to perform the water-soluble tetrazolium salt-1 assay, lactate dehydrogenase release assay, reactive oxygen species (ROS) detection assay, real-time quantitative polymerase chain reaction and western blot. We investigated the regulatory effects of hemin and EGCG on an azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced colon carcinogenesis mouse model. RESULTS: In Caco2 cells, hemin increased cell proliferation and the expression of cell cycle regulatory proteins, and ROS levels. EGCG suppressed hemin-induced cell proliferation and cell cycle regulatory protein expression as well as mitochondrial ROS accumulation. Hemin increased nuclear factor erythroid-2-related factor 2 (Nrf2) expression, but decreased Keap1 expression. EGCG enhanced hemin-induced Nrf2 and antioxidant gene expression. Nrf2 inhibitor reversed EGCG reduced cell proliferation and cell cycle regulatory protein expression. In AOM/DSS mice, hemin treatment induced hyperplastic changes in colon tissues, inhibited by EGCG supplementation. EGCG reduced the hemin-induced numbers of total aberrant crypts and malondialdehyde concentration in the AOM/DSS model. CONCLUSIONS: We demonstrated that EGCG reduced hemin-induced proliferation and colon carcinogenesis through Nrf2-inhibited mitochondrial ROS accumulation.


NF-E2-Related Factor 2 , Rodent Diseases , Animals , Antioxidants , Azoxymethane , Caco-2 Cells , Carcinogenesis , Catechin/analogs & derivatives , Cell Cycle Proteins , Colon , Dextrans , Hemin/pharmacology , Humans , Iron , Kelch-Like ECH-Associated Protein 1 , Lactate Dehydrogenases , Malondialdehyde , Mice , Reactive Oxygen Species , Tea , Tetrazolium Salts
7.
Free Radic Biol Med ; 192: 1-12, 2022 11 01.
Article En | MEDLINE | ID: mdl-36096355

Tumor necrosis factor-α (TNF-α) is an inflammatory cytokine involved in cell survival, apoptosis, and homeostasis. However, the regulatory effect of TNF-α on mesenchymal stem cell (MSC) redox regulation remains unknown. The process of delaying the senescence of MSCs and maintaining antioxidation mechanism is important in transplantation therapy to treat inflammatory diseases that result from restricted immunomodulatory effects of senescent MSCs. Thus, we examined the role of TNF-α-mediated signaling and its regulatory mechanisms on the senescence of umbilical cord blood-derived MSCs (UCB-MSCs) and identified its therapeutic efficacy in a collagen-induced arthritis (CIA) mouse model. We found that TNF-α increased fatty acid synthesis and lipid droplet (LD) formation through NF-κB/SREBP1-mediated FASN, SCD1, and DGAT2 expression, which protects UCB-MSCs from oxidative stress against accumulated toxic lipids. Additionally, DGAT2-mediated LD formation was regulated by TNF-α-activated TNF receptor (TNFR)1 signaling. We also found that storage of unsaturated FAs in LDs is regulated by SIRT5-dependent ß-oxidation of FAs, which reduces mitochondrial ROS (mtROS) accumulation. Particularly, mtROS homeostasis was maintained by superoxide dismutase 2 (SOD2) upregulation through TNFR2-mediated SIRT5/Nrf2 signaling. In a CIA mouse model, UCB-MSCs transfected with SIRT5 siRNA exhibited reduced therapeutic effects compared with UCB-MSCs transfected with NT siRNA. Overall, the results indicated that SIRT5 plays a central role in protecting TNF-α-induced UCB-MSC senescence through FA ß-oxidation and SOD2-mediated antioxidation.


NF-E2-Related Factor 2 , Sirtuins , Animals , Cytokines/metabolism , Disease Models, Animal , Fatty Acids , Gene Silencing , Lipids , Mice , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , Oxidation-Reduction , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Cell Mol Life Sci ; 79(6): 294, 2022 May 13.
Article En | MEDLINE | ID: mdl-35562616

Exposure to maternal stress irreversibly impairs neurogenesis of offspring by inducing life-long effects on interaction between neurons and glia under raging differentiation process, culminating in cognitive and neuropsychiatric abnormalities in adulthood. We identified that prenatal exposure to stress-responsive hormone glucocorticoid impaired neurogenesis and induced abnormal behaviors in ICR mice. Then, we used human induced pluripotent stem cell (iPSC)-derived neural stem cell (NSC) to investigate how neurogenesis deficits occur. Following glucocorticoid treatment, NSC-derived astrocytes were found to be A1-like neurotoxic astrocytes. Moreover, cortisol-treated astrocytic conditioned media (ACM) then specifically downregulated AMPA receptor-mediated glutamatergic synaptic formation and transmission in differentiating neurons, by inhibiting localization of ionotropic glutamate receptor (GluR)1/2 into synapses. We then revealed that downregulated astrocytic fibroblast growth factor 2 (FGF2) and nuclear fibroblast growth factor receptor 1 (FGFR1) of neurons are key pathogenic factors for reducing glutamatergic synaptogenesis. We further confirmed that cortisol-treated ACM specifically decreased the binding of neuronal FGFR1 to the synaptogenic NLGN1 promoter, but this was reversed by FGFR1 restoration. Upregulation of neuroligin 1, which is important in scaffolding GluR1/2 into the postsynaptic compartment, eventually normalized glutamatergic synaptogenesis and subsequent neurogenesis. Moreover, pretreatment of FGF2 elevated neuroligin 1 expression and trafficking of GluR1/2 into the postsynaptic compartment of mice exposed to prenatal corticosterone, improving spatial memory and depression/anxiety-like behaviors. In conclusion, we identified neuroligin 1 restoration by astrocytic FGF2 and its downstream neuronal nuclear FGFR1 as a critical target for preventing prenatal stress-induced dysfunction in glutamatergic synaptogenesis, which recovered both neurogenesis and hippocampal-related behaviors.


Astrocytes , Induced Pluripotent Stem Cells , Adult , Animals , Astrocytes/metabolism , Cell Adhesion Molecules, Neuronal , Female , Fibroblast Growth Factor 2/metabolism , Glucocorticoids/metabolism , Hippocampus/metabolism , Humans , Hydrocortisone/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred ICR , Neurogenesis , Neurons/metabolism , Pregnancy , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
9.
J Biomed Sci ; 29(1): 17, 2022 Mar 07.
Article En | MEDLINE | ID: mdl-35255899

BACKGROUND: Androgenetic alopecia (AGA) is a genetic disorder caused by dihydrotestosterone (DHT), accompanied by the senescence of androgen-sensitive dermal papilla cells (DPCs) located in the base of hair follicles. DHT causes DPC senescence in AGA through mitochondrial dysfunction. However, the mechanism of this pathogenesis remains unknown. In this study, we investigated the protective role of cyanidins on DHT-induced mitochondrial dysfunction and DPC senescence and the regulatory mechanism involved. METHODS: DPCs were used to investigate the effect of DHT on mitochondrial dysfunction with MitoSOX and Rhod-2 staining. Senescence-associated ß-galactosidase activity assay was performed to examine the involvement of membrane AR-mediated signaling in DHT-induced DPC senescence. AGA mice model was used to study the cyanidins on DHT-induced hair growth deceleration. RESULTS: Cyanidin 3-O-arabinoside (C3A) effectively decreased DHT-induced mtROS accumulation in DPCs, and C3A reversed the DHT-induced DPC senescence. Excessive mitochondrial calcium accumulation was blocked by C3A. C3A inhibited p38-mediated voltage-dependent anion channel 1 (VDAC1) expression that contributes to mitochondria-associated ER membrane (MAM) formation and transfer of calcium via VDAC1-IP3R1 interactions. DHT-induced MAM formation resulted in increase of DPC senescence. In AGA mice models, C3A restored DHT-induced hair growth deceleration, which activated hair follicle stem cell proliferation. CONCLUSIONS: C3A is a promising natural compound for AGA treatments against DHT-induced DPC senescence through reduction of MAM formation and mitochondrial dysfunction.


Dihydrotestosterone , Hair Follicle , Animals , Anthocyanins , Cellular Senescence , Dihydrotestosterone/metabolism , Dihydrotestosterone/pharmacology , Mice , Mitochondria
10.
Stem Cell Res Ther ; 12(1): 114, 2021 02 05.
Article En | MEDLINE | ID: mdl-33546749

BACKGROUND: Retarded wound healing in patients with obesity contributes to a risk of complications associated with vascular insufficiency and oxidative stress. The high cholesterol levels of patients with obesity are associated with apoptosis of engrafted umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs). Melatonin contributes to the prevention of cholesterol accumulation in patients with obesity via a mechanism that is poorly understood. We therefore investigated the regulatory mechanism of melatonin in cholesterol-induced apoptosis. METHODS: The protective effects of melatonin on cholesterol-induced apoptosis were investigated in UCB-MSCs. We used a mouse model of induced obesity to show that melatonin treatment restored the survival rate of transplanted UCB-MSCs and their wound-healing capacity. The mean values of the treatment groups were compared with those of the control group using Student's t test, and differences among three or more groups were analyzed using one-way analysis of variance with Dunnett's multiple comparison test. RESULTS: Melatonin treatment increased the expression of ATP-binding cassette subfamily A member 1 (ABCA1), which reduced cholesterol accumulation and cholesterol-induced apoptosis. The mouse skin wound healing model showed that melatonin treatment restored the survival rate of transplanted UCB-MSCs and the wound-healing capacity of obese mice. Melatonin inhibited the expression of binding immunoglobulin protein (BiP) through the regulation of MT2/Sp1-dependent microRNA-597-5p. Melatonin decreased the co-localization of BiP with nuclear factor erythroid 2-related factor 1 (NRF1), which resulted in increased ABCA1 expression. CONCLUSION: Melatonin induced the efflux of intracellular cholesterol through ABCA1 to decrease apoptosis of UCB-MSCs via an MT2-dependent BiP/NRF1 pathway.


Hypercholesterolemia , Melatonin , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , ATP Binding Cassette Transporter 1/genetics , Animals , Apoptosis , Carrier Proteins , Humans , Immunoglobulins , Melatonin/pharmacology , Mice
11.
Nat Commun ; 12(1): 487, 2021 01 20.
Article En | MEDLINE | ID: mdl-33473105

Stress-induced glucocorticoids disturb mitochondrial bioenergetics and dynamics; however, instead of being removed via mitophagy, the damaged mitochondria accumulate. Therefore, we investigate the role of glucocorticoids in mitophagy inhibition and subsequent synaptic defects in hippocampal neurons, SH-SY5Y cells, and ICR mice. First, we observe that glucocorticoids decrease both synaptic density and vesicle recycling due to suppressed mitophagy. Screening data reveal that glucocorticoids downregulate BNIP3-like (BNIP3L)/NIX, resulting in the reduced mitochondrial respiration function and synaptic density. Notably, we find that glucocorticoids direct the glucocorticoid receptor to bind directly to the PGC1α promoter, downregulating its expression and nuclear translocation. PGC1α downregulation selectively decreases NIX-dependent mitophagy. Consistent with these results, NIX enhancer pre-treatment of a corticosterone-exposed mouse elevates mitophagy and synaptic density in hippocampus, improving the outcome of a spatial memory task. In conclusion, glucocorticoids inhibit mitophagy via downregulating NIX and that NIX activation represents a potential target for restoring synapse function.


Glucocorticoids/adverse effects , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Protective Agents/metabolism , Synapses/metabolism , Animals , Cell Death , Corticosterone/pharmacology , Hydrocortisone/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitophagy/drug effects , Neuronal Plasticity/physiology , Neurons/metabolism , Protein Kinases/metabolism
12.
Dig Dis Sci ; 66(2): 605-611, 2021 02.
Article En | MEDLINE | ID: mdl-32222926

BACKGROUND: The prevalence and risk factors of low bone mineral density (BMD) in Asian patients newly diagnosed with inflammatory bowel disease (IBD) have not been fully suggested. AIMS: We aimed to examine the prevalence and risk factors of low BMD in young Korean patients newly diagnosed with IBD. METHODS: We prospectively enrolled 132 patients aged less than 50 years and newly diagnosed with IBD from six tertiary referral centers in Korea between November 2014 and April 2017. BMD was measured by dual-energy X-ray absorptiometry, and then the Z-score was determined. We defined low BMD as a Z-score ≤ - 1.0. RESULTS: Of 68 patients with ulcerative colitis (UC), 22 (32.4%) had low BMD. Also, of 64 patients with Crohn's disease (CD), 24 (37.5%) showed low BMD. Results from multivariate regression analysis identified the risk factors for low BMD as a high level of alkaline phosphatase (ALP) (≥ 140 U/L) (P = 0.010) in UC patients, and being underweight (body mass index ≤ 18.5 kg/m2) (P = 0.017) in CD patients. CONCLUSIONS: Our study showed that about one-third of newly diagnosed IBD Asian patients had low BMD. The clinical factors associated with low BMD were a high level of ALP in UC patients, and being underweight, in CD patients. Therefore, measurements of BMD in young patients should be considered at the diagnosis of IBD.


Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/epidemiology , Inflammatory Bowel Diseases/diagnostic imaging , Inflammatory Bowel Diseases/epidemiology , Absorptiometry, Photon/methods , Adult , Bone Density/physiology , Female , Humans , Male , Prospective Studies , Republic of Korea/epidemiology , Risk Factors , Young Adult
13.
Cell Death Differ ; 28(1): 184-202, 2021 01.
Article En | MEDLINE | ID: mdl-32704090

Hyperglycemia in diabetes mellitus (DM) patients is a causative factor for amyloidogenesis and induces neuropathological changes, such as impaired neuronal integrity, neurodegeneration, and cognitive impairment. Regulation of mitochondrial calcium influx from the endoplasmic reticulum (ER) is considered a promising strategy for the prevention of mitochondrial ROS (mtROS) accumulation that occurs in the Alzheimer's disease (AD)-associated pathogenesis in DM patients. Among the metabolites of ellagitannins that are produced in the gut microbiome, urolithin A has received an increasing amount of attention as a novel candidate with anti-oxidative and neuroprotective effects in AD. Here, we investigated the effect of urolithin A on high glucose-induced amyloidogenesis caused by mitochondrial calcium dysregulation and mtROS accumulation resulting in neuronal degeneration. We also identified the mechanism related to mitochondria-associated ER membrane (MAM) formation. We found that urolithin A-lowered mitochondrial calcium influx significantly alleviated high glucose-induced mtROS accumulation and expression of amyloid beta (Aß)-producing enzymes, such as amyloid precursor protein (APP) and ß-secretase-1 (BACE1), as well as Aß production. Urolithin A injections in a streptozotocin (STZ)-induced diabetic mouse model alleviated APP and BACE1 expressions, Tau phosphorylation, Aß deposition, and cognitive impairment. In addition, high glucose stimulated MAM formation and transglutaminase type 2 (TGM2) expression. We first discovered that urolithin A significantly reduced high glucose-induced TGM2 expression. In addition, disruption of the AIP-AhR complex was involved in urolithin A-mediated suppression of high glucose-induced TGM2 expression. Markedly, TGM2 silencing inhibited inositol 1, 4, 5-trisphosphate receptor type 1 (IP3R1)-voltage-dependent anion-selective channel protein 1 (VDAC1) interactions and prevented high glucose-induced mitochondrial calcium influx and mtROS accumulation. We also found that urolithin A or TGM2 silencing prevented Aß-induced mitochondrial calcium influx, mtROS accumulation, Tau phosphorylation, and cell death in neuronal cells. In conclusion, we suggest that urolithin A is a promising candidate for the development of therapies to prevent DM-associated AD pathogenesis by reducing TGM2-dependent MAM formation and maintaining mitochondrial calcium and ROS homeostasis.


Alzheimer Disease/prevention & control , Calcium/metabolism , Coumarins/pharmacology , Endoplasmic Reticulum/drug effects , Glucose/metabolism , Mitochondria/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Homeostasis , Humans , Male , Mice , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Voltage-Dependent Anion Channel 1/metabolism
14.
Cell Commun Signal ; 18(1): 123, 2020 08 12.
Article En | MEDLINE | ID: mdl-32787872

BACKGROUND: Neurodegeneration is a representative phenotype of patients with chronic alcoholism. Ethanol-induced calcium overload causes NOD-like receptor protein 3 (NLRP3) inflammasome formation and an imbalance in mitochondrial dynamics, closely associated with the pathogenesis of neurodegeneration. However, how calcium regulates this process in neuronal cells is poorly understood. Therefore, the present study investigated the detailed mechanism of calcium-regulated mitochondrial dynamics and NLRP3 inflammasome formation in neuronal cells by ethanol. METHODS: In this study, we used the SK-N-MC human neuroblastoma cell line. To confirm the expression level of the mRNA and protein, real time quantitative PCR and western blot were performed. Co-immunoprecipitation and Immunofluorescence staining were conducted to confirm the complex formation or interaction of the proteins. Flow cytometry was used to analyze intracellular calcium, mitochondrial dysfunction and neuronal apoptosis. RESULTS: Ethanol increased cleaved caspase-3 levels and mitochondrial reactive oxygen species (ROS) generation associated with neuronal apoptosis. In addition, ethanol increased protein kinase A (PKA) activation and cAMP-response-element-binding protein (CREB) phosphorylation, which increased N-methyl-D-aspartate receptor (NMDAR) expression. Ethanol-increased NMDAR induced intracellular calcium overload and calmodulin-dependent protein kinase II (CaMKII) activation leading to phosphorylation of dynamin-related protein 1 (Drp1) and c-Jun N-terminal protein kinase 1 (JNK1). Drp1 phosphorylation promoted Drp1 translocation to the mitochondria, resulting in excessive mitochondrial fission, mitochondrial ROS accumulation, and loss of mitochondrial membrane potential, which was recovered by Drp1 inhibitor pretreatment. Ethanol-induced JNK1 phosphorylation activated the NLRP3 inflammasome that induced caspase-1 dependent mitophagy inhibition, thereby exacerbating ROS accumulation and causing cell death. Suppressing caspase-1 induced mitophagy and reversed the ethanol-induced apoptosis in neuronal cells. CONCLUSIONS: Our results demonstrated that ethanol upregulated NMDAR-dependent CaMKII phosphorylation which is essential for Drp1-mediated excessive mitochondrial fission and the JNK1-induced NLRP3 inflammasome activation resulting in neuronal apoptosis. Video abstract.


Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dynamins/metabolism , Ethanol/pharmacology , Inflammasomes/metabolism , Mitochondrial Dynamics , Mitogen-Activated Protein Kinase 8/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurons/metabolism , Apoptosis/drug effects , Calcium/metabolism , Caspase 1/metabolism , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Humans , Intracellular Space/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Models, Biological , Neurons/drug effects , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism
15.
Cell Death Dis ; 11(6): 469, 2020 06 18.
Article En | MEDLINE | ID: mdl-32555166

The gut-brain axis is currently being studied as a therapeutic strategy for neurological diseases, especially Alzheimer's disease (AD). Obesity results in the gut microbiota dysbiosis, which includes butyrate-producing bacteria are reduced. Although sodium butyrate (NaB) has emerged as the potential therapeutic substance in AD, there is a lack of detailed results into what signaling pathways affect amyloidogenesis in AD induced by obesity. Thus, we investigated the regulatory role of NaB on amyloidogenesis in neuronal cells under high cholesterol. In our results, we verified that increased amyloid ß peptide (Aß) accumulation in the brain of obese mice and a reduction in butyrate-producing bacteria due to the gut microbiota dysbiosis induced by obesity. We showed that NaB decreased the expression levels of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and Aß accumulation induced by high cholesterol in SK-N-MC cells. We demonstrated that NaB was absorbed in cells through sodium-coupled monocarboxylate transporter 1 (SMCT1) and then inhibited high cholesterol-induced Aß accumulation. Subsequently, we also observed that reactive oxygen species (ROS) were overproduced because of increased NADPH oxidase 2 (NOX2) expression under high cholesterol. Meanwhile, NaB decreased NOX2 levels through a reduction of NF-κB activity, which ultimately inhibited Aß accumulation caused by high cholesterol. We demonstrated that NaB increased the expression levels of p21 under high cholesterol, contributing to p21/NRF2 (Nuclear factor erythroid 2-related factor 2) colocalization, which leads to NRF2 stabilization. NRF2 stabilization causes NF-κB inactivation, followed by NOX2 suppression and superoxide dismutase 1 (SOD1) upregulation. Thus, NaB with SOD1 silencing under high cholesterol did not eliminate excessive ROS, and eventually resulted in Aß accumulation. In conclusion, we demonstrated that NaB prevents excessive ROS through NOX2 suppression and SOD1 upregulation by p21/NRF2 pathway, which is critical for inhibiting BACE1-dependent amyloidogenesis in neuronal cells exposed to high cholesterol environment.


Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Butyric Acid/therapeutic use , NADPH Oxidase 2/metabolism , NF-E2-Related Factor 2/metabolism , Obesity/complications , Superoxide Dismutase-1/metabolism , Amyloid beta-Peptides/metabolism , Animals , Butyric Acid/pharmacology , Humans , Mice , Reactive Oxygen Species , Transfection
16.
Br J Pharmacol ; 177(16): 3828-3847, 2020 08.
Article En | MEDLINE | ID: mdl-32436237

BACKGROUND AND PURPOSE: Although diabetes mellitus (DM) is an important risk factor for Alzheimer's disease (AD), the detailed mechanism(s) by which DM regulates amyloid ß (Aß) processing is still unclear. The longer residence time of amyloid precursor protein (APP) in endosomes is critical for Aß production and DM is known to cause endosomal dysregulation. Here we have examined the effects of high glucose on APP-producing endosomes and related signaling pathways. EXPERIMENTAL APPROACH: To identify the underlying mechanisms, we investigated the effects of high glucose on abnormalities in early endosomes and related signalling pathways in human neuroblastoma cells. In vivo, diabetic mice treated with pharmacological inhibitors were used to examine endosomal dysfunction. KEY RESULTS: The hippocampus of diabetic animals presented endosomal abnormalities and Aß up-regulation. High glucose increased Aß production through early endosomal enlargement achieved by increased lipid raft-mediated APP endocytosis. High glucose induced ROS-stimulated Sp1 activation, up-regulating phosphatidylinositol binding clathrin assembly protein (PICALM), clathrin heavy chain, and adaptor-related protein complex 2 alpha 1. PICALM facilitated clathrin-mediated APP endocytosis resulting in early endosomal enlargement. Meanwhile, AMPK/mTORC1-mediated autophagy defect and ROS- and mTORC1-mediated lysosomal dysfunction aggravated early endosomal enlargement under high glucose. Moreover, the increased Aß production and cognitive deficits in diabetic mice were reversed by inhibition of early endosomal enlargement. CONCLUSION AND IMPLICATIONS: High glucose induces early endosomal abnormalities through PICALM-induced APP endocytosis and mTORC1-inhibited endosomal clearance, up-regulating Aß production. Thus, targeting PICALM and mTORC1 to prevent endosomal disorders is a promising strategy for managing diabetes-induced AD.


Alzheimer Disease , Diabetes Mellitus, Experimental , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Endosomes , Glucose , Mechanistic Target of Rapamycin Complex 1 , Mice
17.
J Biomed Sci ; 27(1): 21, 2020 Jan 06.
Article En | MEDLINE | ID: mdl-31906951

BACKGROUND: Melatonin (5-methoxy-N-acetyltryptamine), a hormone produced in the pineal gland, has a variety of biological functions as an antioxidant, but a functional role of melatonin in the regulation of intestinal mucin (Muc) production during bacterial infection has yet to be described in detail. In this study, we investigate the effects of melatonin during Muc2 repression elicited by the Gram-negative bacterium V. vulnificus. METHODS: Mucus-secreting human HT29-MTX cells were used to study the functional role of melatonin during Muc2 depletion induced by the recombinant protein (r) VvpM produced by V. vulnificus. The regulatory effects of melatonin coupling with melatonin receptor 2 (MT2) on the production of reactive oxygen species (ROS), the activation of PKCδ and ERK, and the hypermethylation of the Muc2 promoter as induced by rVvpM were examined. Experimental mouse models of V. vulnificus infection were used to study the role of melatonin and how it neutralizes the bacterial toxin activity related to Muc2 repression. RESULTS: Recombinant protein (r) VvpM significantly reduced the level of Muc2 in HT29-MTX cells. The repression of Muc2 induced by rVvpM was significantly restored upon a treatment with melatonin (1 µM), which had been inhibited by the knockdown of MT2 coupling with Gαq and the NADPH oxidase subunit p47 phox. Melatonin inhibited the ROS-mediated phosphorylation of PKCδ and ERK responsible for region-specific hypermethylation in the Muc2 promoter in rVvpM-treated HT29-MTX cells. In the mouse models of V. vulnificus infection, treatment with melatonin maintained the level of Muc2 expression in the intestine. In addition, the mutation of the VvpM gene from V. vulnificus exhibited an effect similar to that of melatonin. CONCLUSIONS: These results demonstrate that melatonin acting on MT2 inhibits the hypermethylation of the Muc2 promoter to restore the level of Muc2 production in intestinal epithelial cells infected with V. vulnificus.


Bacterial Toxins/metabolism , DNA Methylation , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Melatonin/pharmacology , Mucin-2/biosynthesis , Receptor, Melatonin, MT2/metabolism , Vibrio Infections/metabolism , Vibrio vulnificus/metabolism , Animals , Bacterial Toxins/pharmacology , HT29 Cells , Humans , Mice , Vibrio Infections/pathology
18.
Adv Exp Med Biol ; 1232: 271-276, 2020.
Article En | MEDLINE | ID: mdl-31893420

Adaptation to hypoxia is essential for regulating the survival and functions of hypoxic cells; it is mainly mediated by the hypoxia-inducible factor 1 (HIF1). The alpha subunit of HIF1 (HIF1α) is a well-known regulatory component of HIF1, which is tightly controlled by various types of HIF1α-regulating processes. Previous research has shown that microtubule-regulated HIF1α nuclear translocation is a key factor for HIF1 activation under hypoxia. In this review, we summarize experimental reports on the role of microtubule-associated factors, such as microtubule, dynein, and dynein adaptor protein, in nuclear translocation of HIF1α. Based upon scientific evidence, we propose a bicaudal D homolog (BICD) as a novel HIF1α translocation regulating factor. A deeper understanding of the mechanism of the action of regulatory factors in controlling HIF1α nuclear translocation will provide novel insights into cell biology under hypoxia.


Active Transport, Cell Nucleus , Hypoxia-Inducible Factor 1, alpha Subunit , Active Transport, Cell Nucleus/genetics , Cell Hypoxia/physiology , Cell Nucleus/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microtubules/metabolism , Protein Transport/genetics
19.
Gut Liver ; 14(2): 257-264, 2020 03 15.
Article En | MEDLINE | ID: mdl-31060119

Background/Aims: Few studies have addressed the relationship between the occurrence of adverse events (AEs) in endoscopic retrograde cholangiopancreatography (ERCP) and hospital case volume or endoscopist's experience with inconsistent results. The aim of our study was to investigate the impact of hospital case volume and endoscopist's experience on the AEs associated with ERCP and to analyze patient- and procedure-related risk factors for post-ERCP AEs. Methods: From January 2015 to December 2015, we prospectively enrolled patients with naïve papilla who underwent ERCP at six centers. Patient- and procedure-related variables were recorded on data collection sheets at the time of and after ERCP. Results: A total of 1,191 patients (median age, 71 years) were consecutively enrolled. The overall success rate of biliary cannulation was 96.6%. Overall, 244 patients (20.5%) experienced post-ERCP AEs, including pancreatitis (9.0%), bleeding (11.8%), perforation (0.4%), cholangitis (1.2%), and others (0.9%). While post-ERCP pancreatitis (PEP) was more common when the procedure was performed by less experienced endoscopists, bleeding was more common in high-volume centers and by less experienced endoscopists. Multivariate analysis showed that a less experience in ERCP was significantly associated with PEP (odds ratio [OR], 1.630; 95% confidence interval [CI], 1.050 to 2.531; p=0.030) and post-ERCP bleeding (OR, 1.439; 95% CI, 1.003 to 2.062; p=0.048). Conclusions: Our study demonstrated that overall AEs following ERCP were associated with the experience of the endoscopist. To minimize post-ERCP AEs, rigorous training with a sufficient case volume is required, and treatment strategies should be modified according to the endoscopist's expertise.


Cholangiopancreatography, Endoscopic Retrograde/statistics & numerical data , Clinical Competence/statistics & numerical data , Hospitals, High-Volume/statistics & numerical data , Hospitals, Low-Volume/statistics & numerical data , Surgeons/statistics & numerical data , Aged , Female , Humans , Male , Middle Aged , Odds Ratio , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Prospective Studies , Risk Factors , Treatment Outcome
20.
Cell Death Dis ; 10(8): 590, 2019 08 05.
Article En | MEDLINE | ID: mdl-31383843

O-cyclic phytosphingosine-1-phosphate (cP1P) is a novel chemically synthesized sphingosine metabolite derived from phytosphingosine-1-phosphate. Although structurally similar to sphingosine-1-phosphate (S1P), its biological properties in stem cells remain to be reported. We investigated the effect of cP1P on the therapeutic potential of mesenchymal stem cells (MSCs) and their regulatory mechanism. We found that, under hypoxia, cP1P suppressed MSC mitochondrial dysfunction and apoptosis. Metabolic data revealed that cP1P stimulated glycolysis via the upregulation of glycolysis-related genes. cP1P-induced hypoxia-inducible factor 1 alpha (HIF1α) plays a key role for MSC glycolytic reprogramming and transplantation efficacy. The intracellular calcium-dependent PKCα/mammalian target of the rapamycin (mTOR) signaling pathway triggered by cP1P regulated HIF1α translation via S6K1, which is critical for HIF1 activation. Furthermore, the cP1P-activated mTOR pathway induced bicaudal D homolog 1 expression, leading to HIF1α nuclear translocation. In conclusion, cP1P enhances the therapeutic potential of MSC through mTOR-dependent HIF1α translation and nuclear translocation.


Glycolysis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Sphingosine/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Hypoxia , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Fetal Blood/cytology , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lysophospholipids/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Transport , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sphingosine/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transfection , Up-Regulation/drug effects
...