Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(1)2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38203294

ABSTRACT

Ketone bodies (KBs), such as acetoacetate and ß-hydroxybutyrate, serve as crucial alternative energy sources during glucose deficiency. KBs, generated through ketogenesis in the liver, are metabolized into acetyl-CoA in extrahepatic tissues, entering the tricarboxylic acid cycle and electron transport chain for ATP production. Reduced glucose metabolism and mitochondrial dysfunction correlate with increased neuronal death and brain damage during cerebral ischemia and neurodegeneration. Both KBs and the ketogenic diet (KD) demonstrate neuroprotective effects by orchestrating various cellular processes through metabolic and signaling functions. They enhance mitochondrial function, mitigate oxidative stress and apoptosis, and regulate epigenetic and post-translational modifications of histones and non-histone proteins. Additionally, KBs and KD contribute to reducing neuroinflammation and modulating autophagy, neurotransmission systems, and gut microbiome. This review aims to explore the current understanding of the molecular mechanisms underpinning the neuroprotective effects of KBs and KD against brain damage in cerebral ischemia and neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease.


Subject(s)
Brain Injuries , Diet, Ketogenic , Neurodegenerative Diseases , Neuroprotective Agents , Humans , Ketone Bodies , Neuroprotection , Neuroprotective Agents/therapeutic use , Cerebral Infarction
2.
Nutrients ; 14(22)2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36432618

ABSTRACT

The ketone bodies (KBs) ß-hydroxybutyrate and acetoacetate are important alternative energy sources for glucose during nutrient deprivation. KBs synthesized by hepatic ketogenesis are catabolized to acetyl-CoA through ketolysis in extrahepatic tissues, followed by the tricarboxylic acid cycle and electron transport chain for ATP production. Ketogenesis and ketolysis are regulated by the key rate-limiting enzymes, 3-hydroxy-3-methylglutaryl-CoA synthase 2 and succinyl-CoA:3-oxoacid-CoA transferase, respectively. KBs participate in various cellular processes as signaling molecules. KBs bind to G protein-coupled receptors. The most abundant KB, ß-hydroxybutyrate, regulates gene expression and other cellular functions by inducing post-translational modifications. KBs protect tissues by regulating inflammation and oxidative stress. Recently, interest in KBs has been increasing due to their potential for treatment of various diseases such as neurological and cardiovascular diseases and cancer. Cancer cells reprogram their metabolism to maintain rapid cell growth and proliferation. Dysregulation of KB metabolism also plays a role in tumorigenesis in various types of cancer. Targeting metabolic changes through dietary interventions, including fasting and ketogenic diets, has shown beneficial effects in cancer therapy. Here, we review current knowledge of the molecular mechanisms involved in the regulation of KB metabolism and cellular signaling functions, and the therapeutic potential of KBs and ketogenic diets in cancer.


Subject(s)
Diet, Ketogenic , Neoplasms , Humans , 3-Hydroxybutyric Acid , Ketone Bodies/metabolism , Signal Transduction , Neoplasms/drug therapy
4.
Int J Mol Sci ; 23(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36077072

ABSTRACT

Compound C (CompC), an inhibitor of AMP-activated protein kinase, reduces the viability of various renal carcinoma cells. The molecular mechanism underlying anti-proliferative effect was investigated by flow cytometry and western blot analysis in Renca cells. Its effect on the growth of Renca xenografts was also examined in a syngeneic BALB/c mouse model. Subsequent results demonstrated that CompC reduced platelet-derived growth factor receptor signaling pathways and increased ERK1/2 activation as well as reactive oxygen species (ROS) production. CompC also increased the level of active Wee1 tyrosine kinase (P-Ser642-Wee1) and the inactive form of Cdk1 (P-Tyr15-Cdk1) while reducing the level of active histone H3 (P-Ser10-H3). ROS-dependent ERK1/2 activation and sequential alterations in Wee1, Cdk1, and histone H3 might be responsible for the CompC-induced G2/M cell cycle arrest and cell viability reduction. In addition, CompC reduced the adhesion, migration, and invasion of Renca cells in the in vitro cell systems, and growth of Renca xenografts in the BALB/c mouse model. Taken together, the inhibition of in vivo tumor growth by CompC may be attributed to the blockage of cell cycle progression, adhesion, migration, and invasion of tumor cells. These findings suggest the therapeutic potential of CompC against tumor development and progression.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Carcinoma, Renal Cell/pathology , Cell Division , Disease Models, Animal , Histones , Humans , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism
6.
Aging (Albany NY) ; 14(1): 4-27, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35020602

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a key glycolytic enzyme that participates in various cellular events, such as DNA repair and apoptosis. The functional diversity of GAPDH depends on its intracellular localization. Because AMP-activated protein kinase (AMPK) regulates the nuclear translocation of GAPDH in young cells and AMPK activity significantly increases during aging, we investigated whether altered AMPK activity is involved in the nuclear localization of GAPDH in senescent cells. Age-dependent nuclear translocation of GAPDH was confirmed by confocal laser scanning microscopy in human diploid fibroblasts (HDFs) and by immunohistochemical analysis in aged rat skin cells. Senescence-induced nuclear localization was reversed by lysophosphatidic acid but not by platelet-derived growth factor. The extracellular matrix from young cells also induced the nuclear export of GAPDH in senescent HDFs. An activator of AMPK, 5-Aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR), increased the level of nuclear GAPDH, whereas an inhibitor of AMPK, Compound C, decreased the level of nuclear GAPDH in senescent HDFs. Transfection with AMPKα siRNA prevented nuclear translocation of GAPDH in senescent HDFs. The stimulatory effect of AICAR and serum depletion on GAPDH nuclear translocation was reduced in AMPKα1/α2-knockout mouse embryonic fibroblasts. Overall, increased AMPK activity may play a role in the senescence-associated nuclear translocation of GAPDH.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/physiology , Cellular Senescence/physiology , Fibroblasts/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Extracellular Matrix , Gene Expression Regulation/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lysophospholipids/pharmacology , Rats , Ribonucleotides/pharmacology
8.
Biology (Basel) ; 10(2)2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33671902

ABSTRACT

Proteasome inhibitors, such as bortezomib (BZ) and carfilzomib (CFZ), have been suggested as treatments for various cancers. To utilize BZ and/or CFZ as effective therapeutics for treating melanoma, we studied their molecular mechanisms using B16-F1 melanoma cells. Flow cytometry of Annexin V-fluorescein isothiocyanate-labeled cells indicated apoptosis induction by treatment with BZ and CFZ. Apoptosis was evidenced by the activation of various caspases, including caspase 3, 8, 9, and 12. Treatment with BZ and CFZ induced endoplasmic reticulum (ER) stress, as indicated by an increase in eIF2α phosphorylation and the expression of ER stress-associated proteins, including GRP78, ATF6α, ATF4, XBP1, and CCAAT/enhancer-binding protein homologous protein. The effects of CFZ on ER stress and apoptosis were lower than that of BZ. Nevertheless, CFZ and BZ synergistically induced ER stress and apoptosis in B16-F1 cells. Furthermore, the combinational pharmacological interactions of BZ and CFZ against the growth of B16-F1 melanoma cells were assessed by calculating the combination index and dose-reduction index with the CompuSyn software. We found that the combination of CFZ and BZ at submaximal concentrations could obtain dose reduction by exerting synergistic inhibitory effects on cell growth. Moreover, this drug combination reduced tumor growth in C57BL/6 syngeneic mice. Taken together, these results suggest that CFZ in combination with BZ may be a beneficial and potential strategy for melanoma treatment.

9.
Aging (Albany NY) ; 11(11): 3731-3749, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31199782

ABSTRACT

Nectandrin B (NecB) is a bioactive lignan compound isolated from Myristica fragrans (nutmeg), which functions as an activator of AMP-activated protein kinase (AMPK). Because we recently found that treatment with NecB increased the cell viability of old human diploid fibroblasts (HDFs), the underlying molecular mechanism was investigated. NecB treatment in old HDFs reduced the activity staining of senescence-associated ß-galactosidase and the levels of senescence markers, such as the Ser15 phosphorylated p53, caveolin-1, p21waf1, p16ink4a, p27kip1, and cyclin D1. NecB treatment increased that in S phase, indicating a enhancement of cell cycle entry. Interestingly, NecB treatment ameliorated age-dependent activation of AMPK in old HDFs. Moreover, NecB reversed the age-dependent expression and/or activity changes of certain sirtuins (SIRT1-5), and cell survival/death-related proteins. The transcriptional activity of Yin-Yang 1 and the expression of downstream proteins were elevated in NecB-treated old HDFs. In addition, NecB treatment exerted a radical scavenging effect in vitro, reduced cellular ROS levels, and increased antioxidant enzymes in old HDFs. Moreover, NecB-mediated activation of the AMPK pathway reduced intracellular ROS levels. These results suggest that NecB-induced protection against cellular senescence is mediated by ROS-scavenging through activation of AMPK. NecB might be useful in ameliorating age-related diseases and extending human lifespan.


Subject(s)
Adenylate Kinase/metabolism , Cellular Senescence/drug effects , Fibroblasts/drug effects , Lignans/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Diploidy , Fibroblasts/metabolism , Humans , Phosphorylation , Sirtuins/metabolism
10.
Cancers (Basel) ; 11(6)2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31200558

ABSTRACT

We recently observed that Compound C (CompC), a reversible inhibitor of AMP-activated protein kinase, reduced the cell viability of B16-F1 melanoma cells. To establish its molecular mechanism(s) of action, the cell cycle was examined by flow cytometry and the expression of cell cycle regulatory proteins and angiogenesis-related proteins were examined by western blot analysis. In addition, its effect on tumor growth was investigated using C57BL/6 syngeneic mice bearing B16-F1 xenografts. We found that CompC induced G2/M cell cycle arrest, which was associated with reduced levels of cell cycle regulatory proteins, such as phosphorylated pRB, cyclin-dependent protein kinases (Cdks), cyclins, and phosphorylated P-Ser10-histone H3, and increased levels of Cdk inhibitors, such as p21 and p53. We also found that CompC inhibits proliferation, migration, and tube formation of human umbilical vascular endothelial cells via the inhibition of vascular endothelial growth factor receptor-induced signaling pathways. As expected, CompC significantly reduced the tumor size of B16-F1 xenografts in the syngeneic mouse model. Inhibition of tumor growth may be attributed to reduced cell proliferation via cell cycle inhibition and in part to decreased angiogenesis in CompC-treated mice. These findings suggest the potential use of CompC against melanoma development and progression.

12.
Exp Mol Med ; 51(6): 1-15, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31221957

ABSTRACT

Eukaryotic cells require sufficient oxygen (O2) for biological activity and survival. When the oxygen demand exceeds its supply, the oxygen levels in local tissues or the whole body decrease (termed hypoxia), leading to a metabolic crisis, threatening physiological functions and viability. Therefore, eukaryotes have developed an efficient and rapid oxygen sensing system: hypoxia-inducible factors (HIFs). The hypoxic responses are controlled by HIFs, which induce the expression of several adaptive genes to increase the oxygen supply and support anaerobic ATP generation in eukaryotic cells. Hypoxia also contributes to a functional decline during the aging process. In this review, we focus on the molecular mechanisms regulating HIF-1α and aging-associated signaling proteins, such as sirtuins, AMP-activated protein kinase, mechanistic target of rapamycin complex 1, UNC-51-like kinase 1, and nuclear factor κB, and their roles in aging and aging-related diseases. In addition, the effects of prenatal hypoxia and obstructive sleep apnea (OSA)-induced intermittent hypoxia have been reviewed due to their involvement in the progression and severity of many diseases, including cancer and other aging-related diseases. The pathophysiological consequences and clinical manifestations of prenatal hypoxia and OSA-induced chronic intermittent hypoxia are discussed in detail.


Subject(s)
Aging , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Humans , Hypoxia/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction , Sirtuins/metabolism
14.
Sci Rep ; 9(1): 5039, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911132

ABSTRACT

Neuroblastoma is a solid malignant tumor of the sympathetic nervous system, which accounts for 8-10% of childhood cancers. Considering the overall high risk and poor prognosis associated with neuroblastoma, effective therapeutics should be developed to improve patient survival and quality of life. A recent study showed that a proteasome inhibitor, carfilzomib (CFZ), reduced cell viability of SK-N-BE(2)-M17 neuroblastoma cells. Therefore, we investigated the molecular mechanisms by which CFZ lower the cell viability of neuroblastoma cells. CFZ reduced cell viability via cell cycle arrest at G2/M and apoptosis, which involved caspase activation (caspases-8, 9, 4, and 3), endoplasmic reticulum stress, reactive oxygen species production, mitochondrial membrane potential loss, and autophagy in a dose- and time-dependent manner. The effect of CFZ was additive to that of cisplatin (Cis), a well-known chemotherapeutic drug, in terms of cell viability reduction, cell cycle arrest, and apoptosis. Importantly, the additive effect of CFZ was maintained in Cis-resistant neuroblastoma cells. These results suggest that CFZ can be used in combination therapy for patients with neuroblastoma to overcome the resistance and adverse side effects of Cis.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Neuroblastoma/drug therapy , Oligopeptides/pharmacology , Cell Line, Tumor , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology
15.
Neurotoxicology ; 71: 39-51, 2019 03.
Article in English | MEDLINE | ID: mdl-30508555

ABSTRACT

α-Naphthoflavone (αNF) is a prototype flavone, also known as a modulator of aryl hydrocarbon receptor (AhR). In the present study, we investigated the molecular mechanisms of αNF-induced cytotoxic effects in HT22 mouse hippocampal neuronal cells. αNF induced apoptotic cell death via activation of caspase-12 and -3 and increased expression of endoplasmic reticulum (ER) stress-associated proteins, including C/EBP homologous protein (CHOP). Inhibition of ER stress by treatment with the ER stress inhibitor, salubrinal, or by CHOP siRNA transfection reduced αNF-induced cell death. αNF activated mitogen-activated protein kinases (MAPKs), such as p38, JNK, and ERK, and inhibition of MAPKs reduced αNF-induced CHOP expression and cell death. αNF also induced accumulation of reactive oxygen species (ROS) and an antioxidant, N-acetylcysteine, reduced αNF-induced MAPK phosphorylation, CHOP expression, and cell death. Furthermore, αNF activated c-Src kinase, and inhibition of c-Src by a kinase inhibitor, SU6656, or siRNA transfection reduced αNF-induced ROS accumulation, MAPK activation, CHOP expression, and cell death. Inhibition of AhR by an AhR antagonist, CH223191, and siRNA transfection of AhR and AhR nuclear translocator reduced αNF-induced AhR-responsive luciferase activity, CHOP expression, and cell death. Finally, we found that inhibition of c-Src and MAPKs reduced αNF-induced transcriptional activity of AhR. Taken together, these findings suggest that αNF induces apoptosis through ER stress via c-Src-, ROS-, MAPKs-, and AhR-dependent pathways in HT22 cells.


Subject(s)
Apoptosis , Benzoflavones/metabolism , Endoplasmic Reticulum Stress , Hippocampus/metabolism , Neurons/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Cell Line , Mice , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Reactive Oxygen Species
16.
Apoptosis ; 24(1-2): 200-203, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30390185

ABSTRACT

The original version of this article contained mistakes in figures. The western blot data for pro-caspase-3 and cleaved caspase-3 (Fig. 1d), ß-actin (Fig. 1d), PLCγ1 (Fig. 5d), and eIF2α (Fig. 7d) are incorrect. The corrected Figs. 1d, 5d, and 7d are shown below. The corrections do not influence either the validity of the published data or the conclusion described in the article.

17.
Apoptosis ; 24(1-2): 198-199, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30390184

ABSTRACT

The original version of this article contained a mistake in the figure. The Ca2 + confocal image for the 2-APB/Apicidin-120 min in Fig. 5d is incorrect. The correction does not influence either the validity of the published data or the conclusion described in the article. The corrected Fig. 5d is given below.

20.
Toxicol In Vitro ; 48: 170-178, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29391262

ABSTRACT

Recent studies demonstrate that the autophagy-dependent turnover of mitochondria (mitophagy) mediates pulmonary epithelial cell death in response to cigarette smoke extract (CSE) exposure, and contributes to emphysema development in vivo during chronic cigarette smoke (CS)-exposure, although the underlying mechanisms remain unclear. Here, we investigated the role of mitophagy in regulating apoptosis in CSE-exposed human lung bronchial epithelial cells. Furthermore, we investigated the potential of the polymethoxylated flavone antioxidant quercetogetin (QUE) to inhibit CSE-induced mitophagy-dependent apoptosis. Our results demonstrate that CSE induces mitophagy in epithelial cells via mitochondrial dysfunction, and causes increased expression levels of the mitophagy-regulator protein PTEN-induced putative kinase-1 (PINK1) and the mitochondrial fission protein dynamin-1-like protein (DRP-1). CSE induced epithelial cell death and increased the expression of the apoptosis-related proteins cleaved caspase-3, -8 and -9. Caspase-3 activity was significantly increased in Beas-2B cells exposed to CSE, and decreased by siRNA-dependent knockdown of DRP-1. Treatment of epithelial cells with QUE inhibited CSE-induced mitochondrial dysfunction and mitophagy by inhibiting phospho (p)-DRP-1 and PINK1 expression. QUE suppressed mitophagy-dependent apoptosis by inhibiting the expression of cleaved caspase-3, -8 and -9 and downregulating caspase activity in human bronchial epithelial cells. These findings suggest that QUE may serve as a potential therapeutic in CS-induced pulmonary diseases.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Epithelial Cells/drug effects , Flavones/pharmacology , Mitophagy/drug effects , Nicotiana/toxicity , Protective Agents/pharmacology , Smoke/adverse effects , Bronchi/cytology , Bronchi/drug effects , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Dynamins , GTP Phosphohydrolases/biosynthesis , Humans , Microtubule-Associated Proteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Protein Kinases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL