Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.594
Filter
1.
PLoS One ; 19(6): e0304149, 2024.
Article in English | MEDLINE | ID: mdl-38848430

ABSTRACT

Glioblastoma, the most aggressive form of brain cancer, poses a significant global health challenge with a considerable mortality rate. With the predicted increase in glioblastoma incidence, there is an urgent need for more effective treatment strategies. In this study, we explore the potential of caerin 1.1 and 1.9, host defence peptides derived from an Australian tree frog, in inhibiting glioblastoma U87 and U118 cell growth. Our findings demonstrate the inhibitory impact of caerin 1.1 and 1.9 on cell growth through CCK8 assays. Additionally, these peptides effectively curtail the migration of glioblastoma cells in a cell scratch assay, exhibiting varying inhibitory effects among different cell lines. Notably, the peptides hinder the G0/S phase replication in both U87 and U118 cells, pointing to their impact on the cell cycle. Furthermore, caerin 1.1 and 1.9 show the ability to enter the cytoplasm of glioblastoma cells, influencing the morphology of mitochondria. Proteomics experiments reveal intriguing insights, with a decrease in CHI3L1 expression and an increase in PZP and JUNB expression after peptide treatment. These proteins play roles in cell energy metabolism and inflammatory response, suggesting a multifaceted impact on glioblastoma cells. In conclusion, our study underscores the substantial anticancer potential of caerin 1.1 and 1.9 against glioblastoma cells. These findings propose the peptides as promising candidates for further exploration in the realm of glioblastoma management, offering new avenues for developing effective treatment strategies.


Subject(s)
Cell Proliferation , Down-Regulation , Glioblastoma , Mitochondria , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Cell Proliferation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Cell Respiration/drug effects , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/metabolism , Cell Movement/drug effects
2.
PLoS One ; 19(5): e0303528, 2024.
Article in English | MEDLINE | ID: mdl-38753618

ABSTRACT

Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,ß; TRα,ß) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,ß, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRß and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.


Subject(s)
Arsenic , Hypothalamus , Mitochondria , PPAR gamma , RNA, Messenger , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Mitochondria/metabolism , Mitochondria/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Arsenic/toxicity , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/genetics , Male , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Cell Respiration/drug effects , Gene Expression Regulation/drug effects
3.
Biomolecules ; 14(4)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38672489

ABSTRACT

Colorectal cancer (CRC) is a leading cause of death worldwide. Conventional therapies are available with varying effectiveness. Acetate, a short-chain fatty acid produced by human intestinal bacteria, triggers mitochondria-mediated apoptosis preferentially in CRC but not in normal colonocytes, which has spurred an interest in its use for CRC prevention/therapy. We previously uncovered that acetate-induced mitochondrial-mediated apoptosis in CRC cells is significantly enhanced by the inhibition of the lysosomal protease cathepsin D (CatD), which indicates both mitochondria and the lysosome are involved in the regulation of acetate-induced apoptosis. Herein, we sought to determine whether mitochondrial function affects CatD apoptotic function. We found that enhancement of acetate-induced apoptosis by CatD inhibition depends on oligomycin A-sensitive respiration. Mechanistically, the potentiating effect is associated with an increase in cellular and mitochondrial superoxide anion accumulation and mitochondrial mass. Our results provide novel clues into the regulation of CatD function and the effect of tumor heterogeneity in the outcome of combined treatment using acetate and CatD inhibitors.


Subject(s)
Apoptosis , Cathepsin D , Colorectal Neoplasms , Mitochondria , Oligomycins , Humans , Acetates/pharmacology , Apoptosis/drug effects , Cathepsin D/metabolism , Cathepsin D/antagonists & inhibitors , Cell Line, Tumor , Cell Respiration/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Mitochondria/metabolism , Mitochondria/drug effects , Oligomycins/pharmacology
4.
Am J Physiol Cell Physiol ; 326(6): C1669-C1682, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38646781

ABSTRACT

We previously showed that the transaminase inhibitor, aminooxyacetic acid, reduced respiration energized at complex II (succinate dehydrogenase, SDH) in mitochondria isolated from mouse hindlimb muscle. The effect required a reduction in membrane potential with resultant accumulation of oxaloacetate (OAA), a potent inhibitor of SDH. To specifically assess the effect of the mitochondrial transaminase, glutamic oxaloacetic transaminase (GOT2) on complex II respiration, and to determine the effect in intact cells as well as isolated mitochondria, we performed respiratory and metabolic studies in wildtype (WT) and CRISPR-generated GOT2 knockdown (KD) C2C12 myocytes. Intact cell respiration by GOT2KD cells versus WT was reduced by adding carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP) to lower potential. In mitochondria of C2C12 KD cells, respiration at low potential generated by 1 µM FCCP and energized at complex II by 10 mM succinate + 0.5 mM glutamate (but not by complex I substrates) was reduced versus WT mitochondria. Although we could not detect OAA, metabolite data suggested that OAA inhibition of SDH may have contributed to the FCCP effect. C2C12 mitochondria differed from skeletal muscle mitochondria in that the effect of FCCP on complex II respiration was not evident with ADP addition. We also observed that C2C12 cells, unlike skeletal muscle, expressed glutamate dehydrogenase, which competes with GOT2 for glutamate metabolism. In summary, GOT2 KD reduced C2C12 respiration in intact cells at low potential. From differential substrate effects, this occurred largely at complex II. Moreover, C2C12 versus muscle mitochondria differ in complex II sensitivity to ADP and differ markedly in expression of glutamate dehydrogenase.NEW & NOTEWORTHY Impairment of the mitochondrial transaminase, GOT2, reduces complex II (succinate dehydrogenase, SDH)-energized respiration in C2C12 myocytes. This occurs only at low inner membrane potential and is consistent with inhibition of SDH. Incidentally, we observed that C2C12 mitochondria compared with muscle tissue mitochondria differ in sensitivity of complex II respiration to ADP and in the expression of glutamate dehydrogenase.


Subject(s)
Cell Respiration , Membrane Potential, Mitochondrial , Mitochondria, Muscle , Animals , Mice , Membrane Potential, Mitochondrial/drug effects , Cell Respiration/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/enzymology , Mitochondria, Muscle/drug effects , Cell Line , Aspartate Aminotransferase, Mitochondrial/metabolism , Aspartate Aminotransferase, Mitochondrial/genetics , Electron Transport Complex II/metabolism , Electron Transport Complex II/genetics , Cell Differentiation/drug effects , Succinate Dehydrogenase/metabolism , Succinate Dehydrogenase/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Oxygen Consumption/drug effects
5.
Biomed Pharmacother ; 175: 116664, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678966

ABSTRACT

Mitochondrial dysmorphology/dysfunction follow global cerebral ischemia-reperfusion (GCI/R) injury, leading to neuronal death. Our previous researches demonstrated that Levodopa (L-DOPA) improves learning and memory impairment in GCI/R rats by increasing synaptic plasticity of hippocampal neurons. This study investigates if L-DOPA, used in Parkinson's disease treatment, alleviates GCI/R-induced cell death by enhancing mitochondrial quality. Metabolomics and transcriptomic results showed that GCI/R damage affected the Tricarboxylic acid (TCA) cycle in the hippocampus. The results of this study show that L-DOPA stabilized mitochondrial membrane potential and ultrastructure in hippocampus of GCI/R rats, increased dopamine level in hippocampus, decreased succinic acid level, and stabilized Ca2+ level in CA1 subregion of hippocampus. As a precursor of dopamine, L-DOPA is presumed to improves mitochondrial function in hippocampus of GCI/R rats. However, dopamine cannot cross the blood-brain barrier, so L-DOPA is used in clinical therapy to supplement dopamine. In this investigation, OGD/R models were established in isolated mouse hippocampal neurons (HT22) and primary rat hippocampal neurons. Notably, dopamine exhibited a multifaceted impact, demonstrating inhibition of mitochondrial reactive oxygen species (mitoROS) production, stabilization of mitochondrial membrane potential and Ca2+ level, facilitation of TCA circulation, promotion of aerobic respiratory metabolism, and downregulation of succinic acid-related gene expression. Consistency between in vitro and in vivo results underscores dopamine's significant neuroprotective role in mitigating mitochondrial dysfunction following global cerebral hypoxia and ischemia injury. Supplement dopamine may represent a promising therapy to the cognitive impairment caused by GCI/R injury.


Subject(s)
Hippocampus , Levodopa , Membrane Potential, Mitochondrial , Mitochondria , Rats, Sprague-Dawley , Reperfusion Injury , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Levodopa/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Membrane Potential, Mitochondrial/drug effects , Male , Mice , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Rats , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Dopamine/metabolism , Reactive Oxygen Species/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell Respiration/drug effects , Citric Acid Cycle/drug effects , Calcium/metabolism , Neuroprotective Agents/pharmacology
6.
Nutrients ; 15(9)2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37432297

ABSTRACT

Diosgenin (DIO) is a dietary steroid sapogenin possessing multiple biological functions, such as the amelioration of diabetes. However, the remission effect of DIO on diabetic nephropathy (DN) underlying oxidative stress and cell apoptosis remains unclear. Here, the effect of DIO on ROS generation and its induced cell apoptosis was studied in vitro and in vivo. Renal proximal tubular epithelial (HK-2) cells were treated with DIO (1, 2, 4 µM) under high glucose (HG, 30 mM) conditions. DN rats were induced by a high-fat diet combined with streptozotocin, followed by administration of DIO for 8 weeks. Our data suggested that DIO relieved the decline of HK-2 cell viability and renal pathological damage in DN rats. DIO also relieved ROS (O2- and H2O2) production. Mechanistically, DIO inhibited the expression of NOX4 and restored mitochondrial respiratory chain (MRC) complex I-V expressions. Further, DIO inhibited mitochondrial apoptosis by ameliorating mitochondrial membrane potential (MtMP) and down-regulating the expressions of CytC, Apaf-1, caspase 3, and caspase 9, while up-regulating Bcl2 expression. Moreover, the ER stress and its associated cell apoptosis were inhibited through decreasing PERK, p-PERK, ATF4, IRE1, p-CHOP, and caspase 12 expressions. Collectively, DIO inhibited ROS production by modulating NOX4 and MRC complexes, which then suppressed apoptosis regulated by mitochondria and ER stress, thereby attenuating DN.


Subject(s)
Apoptosis , Diabetic Neuropathies , Humans , Cell Line , Apoptosis/drug effects , Diosgenin/pharmacology , Reactive Oxygen Species/metabolism , Cell Respiration/drug effects , Diabetic Neuropathies/metabolism , Animals , Rats , Endoplasmic Reticulum Stress , Rats, Sprague-Dawley , Male , Mitochondria/drug effects , Mitochondria/metabolism
7.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35298717

ABSTRACT

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Subject(s)
Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
8.
Biochem Biophys Res Commun ; 588: 140-146, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34954521

ABSTRACT

Smoking cessation increases body weight. The underlying mechanisms, however, have not been fully understood. We here report an establishment of a mouse model that exhibits an augmented body weight gain after nicotine withdrawal. High fat diet-fed mice were infused with nicotine for two weeks, and then with vehicle for another two weeks using osmotic minipumps. Body weight increased immediately after nicotine cessation and was significantly higher than that of mice continued on nicotine. Mice switched to vehicle consumed more food than nicotine-continued mice during the first week of cessation, while oxygen consumption was comparable. Elevated expression of orexigenic agouti-related peptide was observed in the hypothalamic appetite center. Pair-feeding experiment revealed that the accelerated weight gain after nicotine withdrawal is explained by enhanced energy intake. As a showcase of an efficacy of pharmacologic intervention, exendin-4 was administered and showed a potent suppression of energy intake and weight gain in mice withdrawn from nicotine. Our current model provides a unique platform for the investigation of the changes of energy regulation after smoking cessation.


Subject(s)
Nicotine/adverse effects , Substance Withdrawal Syndrome/pathology , Weight Gain , Agouti-Related Protein/metabolism , Animals , Calorimetry , Cell Respiration/drug effects , Disease Models, Animal , Energy Intake/drug effects , Exenatide/pharmacology , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substance Withdrawal Syndrome/genetics , Weight Gain/drug effects , Weight Gain/genetics
9.
PLoS Genet ; 17(12): e1009971, 2021 12.
Article in English | MEDLINE | ID: mdl-34965247

ABSTRACT

Rothmund-Thomson syndrome (RTS) is an autosomal recessive genetic disorder characterized by poikiloderma, small stature, skeletal anomalies, sparse brows/lashes, cataracts, and predisposition to cancer. Type 2 RTS patients with biallelic RECQL4 pathogenic variants have multiple skeletal anomalies and a significantly increased incidence of osteosarcoma. Here, we generated RTS patient-derived induced pluripotent stem cells (iPSCs) to dissect the pathological signaling leading to RTS patient-associated osteosarcoma. RTS iPSC-derived osteoblasts showed defective osteogenic differentiation and gain of in vitro tumorigenic ability. Transcriptome analysis of RTS osteoblasts validated decreased bone morphogenesis while revealing aberrantly upregulated mitochondrial respiratory complex I gene expression. RTS osteoblast metabolic assays demonstrated elevated mitochondrial respiratory complex I function, increased oxidative phosphorylation (OXPHOS), and increased ATP production. Inhibition of mitochondrial respiratory complex I activity by IACS-010759 selectively suppressed cellular respiration and cell proliferation of RTS osteoblasts. Furthermore, systems analysis of IACS-010759-induced changes in RTS osteoblasts revealed that chemical inhibition of mitochondrial respiratory complex I impaired cell proliferation, induced senescence, and decreased MAPK signaling and cell cycle associated genes, but increased H19 and ribosomal protein genes. In summary, our study suggests that mitochondrial respiratory complex I is a potential therapeutic target for RTS-associated osteosarcoma and provides future insights for clinical treatment strategies.


Subject(s)
Electron Transport Complex I/genetics , Osteosarcoma/genetics , RNA, Long Noncoding/genetics , RecQ Helicases/genetics , Rothmund-Thomson Syndrome/genetics , Adenosine Triphosphate/biosynthesis , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cellular Senescence/genetics , Electron Transport Complex I/antagonists & inhibitors , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation/genetics , Osteoblasts/drug effects , Osteogenesis/genetics , Osteosarcoma/complications , Osteosarcoma/pathology , Oxadiazoles/pharmacology , Oxidative Phosphorylation/drug effects , Piperidines/pharmacology , Rothmund-Thomson Syndrome/complications , Rothmund-Thomson Syndrome/pathology
10.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769217

ABSTRACT

Amiodarone is a potent antiarrhythmic drug and displays substantial liver toxicity in humans. It has previously been demonstrated that amiodarone and its metabolite (desethylamiodarone, DEA) can inhibit mitochondrial function, particularly complexes I (CI) and II (CII) of the electron transport system in various animal tissues and cell types. The present study, performed in human peripheral blood cells, and one liver-derived human cell line, is primarily aimed at assessing the concentration-dependent effects of these drugs on mitochondrial function (respiration and cellular ATP levels). Furthermore, we explore the efficacy of a novel cell-permeable succinate prodrug in alleviating the drug-induced acute mitochondrial dysfunction. Amiodarone and DEA elicit a concentration-dependent impairment of mitochondrial respiration in both intact and permeabilized platelets via the inhibition of both CI- and CII-supported respiration. The inhibitory effect seen in human platelets is also confirmed in mononuclear cells (PBMCs) and HepG2 cells. Additionally, amiodarone elicits a severe concentration-dependent ATP depletion in PBMCs, which cannot be explained solely by mitochondrial inhibition. The succinate prodrug NV118 alleviates the respiratory deficit in platelets and HepG2 cells acutely exposed to amiodarone. In conclusion, amiodarone severely inhibits metabolism in primary human mitochondria, which can be counteracted by increasing mitochondrial function using intracellular delivery of succinate.


Subject(s)
Amiodarone/toxicity , Anti-Arrhythmia Agents/toxicity , Mitochondria/drug effects , Protective Agents/pharmacology , Succinic Acid/pharmacology , Adenosine Triphosphate/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Respiration/drug effects , Hep G2 Cells , Humans , Mitochondria/metabolism , Prodrugs/pharmacology
11.
Cells ; 10(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34831040

ABSTRACT

Preeclampsia (PE) is characterized by new onset hypertension in association with placental ischemia, reduced fetal weight, elevated soluble fms-like tyrosine kinase-1 (sFlt-1), and placental mitochondrial (mt) dysfunction and oxidative stress (ROS). Progesterone induced blocking factor (PIBF) is a product of progesterone signaling that blocks inflammatory processes and we have previously shown PIBF to lower mean arterial blood pressure (MAP) and sFlt-1 in a rat model of PE. Infusion of sFlt-1 causes hypertension and many characteristics of PE in pregnant rodents, however, its role in causing mt dysfunction is unknown. Therefore, we hypothesize that PIBF will improve mt function and MAP in response to elevated sFlt-1 during pregnancy. We tested our hypothesis by infusing sFlt-1 via miniosmotic pumps in normal pregnant (NP) Sprague-Dawley rats (3.7 µg·kg-1·day-1) on gestation days (GD) 13-19 in the presence or absence of PIBF (2.0 µg/mL) injected intraperitoneally on GD 15 and examined mean arterial blood pressure (MAP) and placental mt ROS on GD 19. sFlt-1 increased MAP to 112 + 2 (n = 11) compared to NP rats (98 + 2 mmHg, n = 15, p < 0.05), which was lowered in the presence of sFlt-1 (100 + 1 mmHg, n = 5, p < 0.05). Placental mtATP was reduced in sFlt-1 infused rats versus NP controls, but was improved with PIBF. Placental mtROS was elevated with sFlt-1 compared to NP controls, but was reduced with PIBF. Sera from NP + sFlt-1 increased endothelial cell mtROS, which was attenuated with PIBF. These data demonstrate sFlt-1 induced HTN during pregnancy reduces placental mt function. Importantly, PIBF improved placental mt function and HTN, indicating the efficacy of improved progesterone signaling as potential therapeutics for PE.


Subject(s)
Antigens, Neoplasm/pharmacology , Hypertension/pathology , Mitochondria/pathology , Placenta/metabolism , Progesterone/pharmacology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Blood Pressure , Body Weight/drug effects , Cell Respiration/drug effects , Female , Fetus/metabolism , Hypertension/blood , Hypertension/physiopathology , Mitochondria/drug effects , Organ Size/drug effects , Pregnancy , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Solubility
12.
Cells ; 10(11)2021 11 05.
Article in English | MEDLINE | ID: mdl-34831251

ABSTRACT

Cardiorenal syndrome (CRS) is a pathological link between the kidneys and heart, in which an insult in a kidney or heart leads the other organ to incur damage. CRS is classified into five subtypes, and type 3 (CRS3) is characterized by acute kidney injury as a precursor to subsequent cardiovascular changes. Mitochondrial dysfunction and oxidative and nitrosative stress have been reported in the pathophysiology of CRS3. It is known that vitamin C, an antioxidant, has proven protective capacity for cardiac, renal, and vascular endothelial tissues. Therefore, the present study aimed to assess whether vitamin C provides protection to heart and the kidneys in an in vivo CRS3 model. The unilateral renal ischemia and reperfusion (IR) protocol was performed for 60 min in the left kidney of adult mice, with and without vitamin C treatment, immediately after IR or 15 days after IR. Kidneys and hearts were subsequently collected, and the following analyses were conducted: renal morphometric evaluation, serum urea and creatinine levels, high-resolution respirometry, amperometry technique for NO measurement, gene expression of mitochondrial dynamic markers, and NOS. The analyses showed that the left kidney weight was reduced, urea and creatinine levels were increased, mitochondrial oxygen consumption was reduced, NO levels were elevated, and Mfn2 expression was reduced after 15 days of IR compared to the sham group. Oxygen consumption and NO levels in the heart were also reduced. The treatment with vitamin C preserved the left kidney weight, restored renal function, reduced NO levels, decreased iNOS expression, elevated constitutive NOS isoforms, and improved oxygen consumption. In the heart, oxygen consumption and NO levels were improved after vitamin C treatment, whereas the three NOS isoforms were overexpressed. These data indicate that vitamin C provides protection to the kidneys and some beneficial effects to the heart after IR, indicating it may be a preventive approach against cardiorenal insults.


Subject(s)
Ascorbic Acid/pharmacology , Cardio-Renal Syndrome/pathology , Kidney/pathology , Mitochondria/pathology , Animals , Cell Respiration/drug effects , Isoenzymes/metabolism , Kidney/drug effects , Kidney/physiopathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Models, Biological , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
13.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681781

ABSTRACT

In this paper, we examined the effects of melittin, a bee venom membrane-active peptide, on mitochondrial respiration and cell viability of healthy human lymphocytes (HHL) and Jurkat cells, as well as on lymphoblasts from acute human T cell leukemia. The viability of melittin-treated cells was related to changes in O2 consumption and in the respiratory control index (RCI) of mitochondria isolated from melittin-pretreated cells as well as of mitochondria first isolated from cells and then directly treated with melittin. It was shown that melittin is three times more cytotoxic to Jurkat cells than to HHL, but O2 consumption and RCI values of mitochondria from both cell types were equally affected by melittin when melittin was directly added to mitochondria. To elucidate the molecular mechanism of melittin's cytotoxicity to healthy and cancer cells, the effects of melittin on lipid-packing and on the dynamics in model plasma membranes of healthy and cancer cells, as well as of the inner mitochondrial membrane, were studied by EPR spin probes. The affinity of melittin binding to phosphatidylcholine, phosphatidylserine, phosphatidic acid and cardiolipin, and binding sites of phospholipids on the surface of melittin were studied by 31P-NMR, native PAGE and AutoDock modeling. It is suggested that the melittin-induced decline of mitochondrial bioenergetics contributes primarily to cell death; the higher cytotoxicity of melittin to cancer cells is attributed to its increased permeability through the plasma membrane.


Subject(s)
Lymphocytes/drug effects , Melitten/pharmacology , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Bee Venoms/chemistry , Blood Cells/drug effects , Blood Cells/metabolism , Cell Respiration/drug effects , Cells, Cultured , Humans , Jurkat Cells , Lipid Bilayers/chemistry , Lymphocytes/metabolism , Melitten/isolation & purification , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Models, Biological , Permeability/drug effects
14.
Cell Death Dis ; 12(11): 953, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34657120

ABSTRACT

Many cell death pathways, including apoptosis, regulated necrosis, and ferroptosis, are relevant for neuronal cell death and share common mechanisms such as the formation of reactive oxygen species (ROS) and mitochondrial damage. Here, we present the role of the actin-regulating protein cofilin1 in regulating mitochondrial pathways in oxidative neuronal death. Cofilin1 deletion in neuronal HT22 cells exerted increased mitochondrial resilience, assessed by quantification of mitochondrial ROS production, mitochondrial membrane potential, and ATP levels. Further, cofilin1-deficient cells met their energy demand through enhanced glycolysis, whereas control cells were metabolically impaired when challenged by ferroptosis. Further, cofilin1 was confirmed as a key player in glutamate-mediated excitotoxicity and associated mitochondrial damage in primary cortical neurons. Using isolated mitochondria and recombinant cofilin1, we provide a further link to toxicity-related mitochondrial impairment mediated by oxidized cofilin1. Our data revealed that the detrimental impact of cofilin1 on mitochondria depends on the oxidation of cysteine residues at positions 139 and 147. Overall, our findings show that cofilin1 acts as a redox sensor in oxidative cell death pathways of ferroptosis, and also promotes glutamate excitotoxicity. Protective effects by cofilin1 inhibition are particularly attributed to preserved mitochondrial integrity and function. Thus, interfering with the oxidation and pathological activation of cofilin1 may offer an effective therapeutic strategy in neurodegenerative diseases.


Subject(s)
Cofilin 1/metabolism , Mitochondria/pathology , Neurons/pathology , Oxidative Stress , Animals , Cell Death/drug effects , Cell Line , Cell Respiration/drug effects , Cell Survival/drug effects , Cofilin 1/deficiency , Down-Regulation/drug effects , Energy Metabolism/drug effects , Glutamic Acid/toxicity , Glycolysis/drug effects , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Neurons/drug effects , Neurotoxins/toxicity , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Piperazines/toxicity , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology
15.
Cells ; 10(10)2021 10 19.
Article in English | MEDLINE | ID: mdl-34685775

ABSTRACT

IL-2 is a cytokine released from CD4+T cells with dual actions and can either potentiate the inflammatory response or quell a chronic inflammatory response depending on its circulating concentration. IL-2 is elevated in many chronic inflammatory conditions and is increased during preeclampsia (PE). PE is characterized by new-onset hypertension during pregnancy and organ dysfunction and increasing evidence indicates that proinflammatory cytokines cause hypertension and mitochondrial (mt) dysfunction during pregnancy. The reduced uterine perfusion pressure (RUPP) model of placental ischemia is a rat model of PE that we commonly use in our laboratory and we have previously shown that low doses of recombinant IL-2 can decrease blood pressure in RUPP rats. The objective of this study was to determine the effects of a low dose of recombinant IL-2 on multi-organ mt dysfunction in the RUPP rat model of PE. We tested our hypothesis by infusing recombinant IL-2 (0.05 ng/mL) into RUPP rats on GD14 and examined mean arterial pressure (MAP), renal, placental and endothelial cell mt function compared to control RUPP. MAP was elevated in RUPP rats (n = 6) compared to controls (n = 5) (122 ± 5 vs. 102 ± 3 mmHg, p < 0.05), but was reduced by administration of LD recombinant IL-2 (107 ± 1 vs. 122 ± 5 mmHg, n = 9, p < 0.05). Renal, placental and endothelial mt ROS were significantly increased in RUPP rats compared to RUPP+ IL-2 and controls. Placental and renal respiration rates were reduced in RUPP rats compared to control rats but were normalized with IL-2 administration to RUPPs. These data indicate that low-dose IL-2 normalized multi-organ mt function and hypertension in response to placental ischemia.


Subject(s)
Hypertension/complications , Interleukin-2/pharmacology , Ischemia/complications , Mitochondria/metabolism , Placenta/pathology , Animals , Blood Pressure/drug effects , Cell Respiration/drug effects , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/blood , Hypertension/physiopathology , Interleukin-2/blood , Ischemia/blood , Mitochondria/drug effects , Organ Size/drug effects , Organ Specificity/drug effects , Pre-Eclampsia/blood , Pre-Eclampsia/physiopathology , Pregnancy , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
16.
Oxid Med Cell Longev ; 2021: 5577541, 2021.
Article in English | MEDLINE | ID: mdl-34707777

ABSTRACT

Mitochondrial dysfunction and oxidative stress are extensively linked to Parkinson's disease (PD) pathogenesis. Melatonin is a pleiotropic molecule with antioxidant and neuroprotective effects. The aim of this study was to evaluate the effect of melatonin on oxidative stress markers, mitochondrial complex 1 activity, and mitochondrial respiratory control ratio in patients with PD. A double-blind, cross-over, placebo-controlled randomized clinical trial study was conducted in 26 patients who received either 25 mg of melatonin or placebo at noon and 30 min before bedtime for three months. At the end of the trial, in patients who received melatonin, we detected a significant diminution of lipoperoxides, nitric oxide metabolites, and carbonyl groups in plasma samples from PD patients compared with the placebo group. Conversely, catalase activity was increased significantly in comparison with the placebo group. Compared with the placebo group, the melatonin group showed significant increases of mitochondrial complex 1 activity and respiratory control ratio. The fluidity of the membranes was similar in the melatonin group and the placebo group at baseline and after three months of treatment. In conclusion, melatonin administration was effective in reducing the levels of oxidative stress markers and restoring the rate of complex I activity and respiratory control ratio without modifying membrane fluidity. This suggests that melatonin could play a role in the treatment of PD.


Subject(s)
Antioxidants/therapeutic use , Antiparkinson Agents/therapeutic use , Melatonin/therapeutic use , Mitochondria/drug effects , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Antioxidants/adverse effects , Antiparkinson Agents/adverse effects , Biomarkers/blood , Cell Respiration/drug effects , Cross-Over Studies , Double-Blind Method , Electron Transport Complex I/metabolism , Humans , Lipid Peroxidation/drug effects , Melatonin/adverse effects , Mexico , Mitochondria/metabolism , Parkinson Disease/blood , Parkinson Disease/diagnosis , Time Factors , Treatment Outcome
17.
PLoS One ; 16(10): e0258845, 2021.
Article in English | MEDLINE | ID: mdl-34699527

ABSTRACT

The ectoparasite Varroa destructor Anderson and Trueman is the most important parasites of the western honey bee, Apis mellifera L. The most widely currently used treatment uses formic acid (FA), but the understanding of its effects on V. destructor is limited. In order to understand the mechanism of action of FA, its effect on Varroa mites was investigated using proteomic analysis by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). V. destructor was collected from honey bee colonies with natural mite infestation before and 24 h after the initiation of FA treatment and subjected to proteome analysis. A total of 2637 proteins were identified. Quantitative analysis of differentially expressed candidate proteins (fold change ≥ 1.5; p ≤ 0.05) revealed 205 differentially expressed proteins: 91 were induced and 114 repressed in the FA-treated group compared to the untreated control group. Impaired protein synthesis accompanied by increased protein and amino acid degradation suggest an imbalance in proteostasis. Signs of oxidative stress included significant dysregulation of candidate proteins of mitochondrial cellular respiration, increased endocytosis, and induction of heat shock proteins. Furthermore, an increased concentration of several candidate proteins associated with detoxification was observed. These results suggest dysregulated cellular respiration triggered by FA treatment as well as an increase in cellular defense mechanisms, including induced heat shock proteins and detoxification enzymes.


Subject(s)
Arthropod Proteins/metabolism , Formates/pharmacology , Proteomics/methods , Varroidae/metabolism , Animals , Bees/parasitology , Cell Respiration/drug effects , Chromatography, Liquid , Gene Expression Regulation/drug effects , Oxidative Stress , Tandem Mass Spectrometry , Varroidae/drug effects
18.
Cell Death Dis ; 12(10): 893, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593753

ABSTRACT

Uncontrolled mitosis is one of the most important features of cancer, and mitotic kinases are thought to be ideal targets for anticancer therapeutics. However, despite numerous clinical attempts spanning decades, clinical trials for mitotic kinase-targeting agents have generally stalled in the late stages due to limited therapeutic effectiveness. Alisertib (MLN8237) is a promising oral mitotic aurora kinase A (AURKA, Aurora-A) selective inhibitor, which is currently under several clinical evaluations but has failed in its first Phase III trial due to inadequate efficacy. In this study, we performed genome-wide CRISPR/Cas9-based screening to identify vulnerable biological processes associated with alisertib in breast cancer MDA-MB-231 cells. The result indicated that alisertib treated cancer cells are more sensitive to the genetic perturbation of oxidative phosphorylation (OXPHOS). Mechanistic investigation indicated that alisertib treatment, as well as other mitotic kinase inhibitors, rapidly reduces the intracellular ATP level to generate a status that is highly addictive to OXPHOS. Furthermore, the combinational inhibition of mitotic kinase and OXPHOS by alisertib, and metformin respectively, generates severe energy exhaustion in mitotic cells that consequently triggers cell death. The combination regimen also enhanced tumor regression significantly in vivo. This suggests that targeting OXPHOS by metformin is a potential strategy for promoting the therapeutic effects of mitotic kinase inhibitors through the joint targeting of mitosis and cellular energy homeostasis.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Mitosis , Oxidative Phosphorylation , Adenosine Triphosphate/metabolism , Animals , Aurora Kinase A/metabolism , Azepines/pharmacology , Breast Neoplasms/pathology , CRISPR-Cas Systems/genetics , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cytosol/metabolism , Drug Synergism , Energy Metabolism/drug effects , Female , Homeostasis/drug effects , Humans , Metformin/pharmacology , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Mitosis/drug effects , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pyrimidines/pharmacology
19.
Plant Physiol ; 186(3): 1487-1506, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34624108

ABSTRACT

Because it is the precursor for various essential cellular components, the amino acid serine is indispensable for every living organism. In plants, serine is synthesized by two major pathways: photorespiration and the phosphorylated pathway of serine biosynthesis (PPSB). However, the importance of these pathways in providing serine for plant development is not fully understood. In this study, we examine the relative contributions of photorespiration and PPSB to providing serine for growth and metabolism in the C3 model plant Arabidopsis thaliana. Our analyses of cell proliferation and elongation reveal that PPSB-derived serine is indispensable for plant growth and its loss cannot be compensated by photorespiratory serine biosynthesis. Using isotope labeling, we show that PPSB-deficiency impairs the synthesis of proteins and purine nucleotides in plants. Furthermore, deficiency in PPSB-mediated serine biosynthesis leads to a strong accumulation of metabolites related to nitrogen metabolism. This result corroborates 15N-isotope labeling in which we observed an increased enrichment in labeled amino acids in PPSB-deficient plants. Expression studies indicate that elevated ammonium uptake and higher glutamine synthetase/glutamine oxoglutarate aminotransferase (GS/GOGAT) activity causes this phenotype. Metabolic analyses further show that elevated nitrogen assimilation and reduced amino acid turnover into proteins and nucleotides are the most likely driving forces for changes in respiratory metabolism and amino acid catabolism in PPSB-deficient plants. Accordingly, we conclude that even though photorespiration generates high amounts of serine in plants, PPSB-derived serine is more important for plant growth and its deficiency triggers the induction of nitrogen assimilation, most likely as an amino acid starvation response.


Subject(s)
Arabidopsis/growth & development , Cell Proliferation/drug effects , Cell Respiration/drug effects , Nitrogen/metabolism , Plant Development/physiology , Plant Growth Regulators/metabolism , Serine/biosynthesis , Biosynthetic Pathways , Phosphorylation
20.
Clin Sci (Lond) ; 135(19): 2243-2263, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34569605

ABSTRACT

The protein tyrosine kinase inhibitor imatinib is used in the treatment of various malignancies but may also promote beneficial effects in the treatment of diabetes. The aim of the present investigation was to characterize the mechanisms by which imatinib protects insulin producing cells. Treatment of non-obese diabetic (NOD) mice with imatinib resulted in increased beta-cell AMP-activated kinase (AMPK) phosphorylation. Imatinib activated AMPK also in vitro, resulting in decreased ribosomal protein S6 phosphorylation and protection against islet amyloid polypeptide (IAPP)-aggregation, thioredoxin interacting protein (TXNIP) up-regulation and beta-cell death. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) mimicked and compound C counteracted the effect of imatinib on beta-cell survival. Imatinib-induced AMPK activation was preceded by reduced glucose/pyruvate-dependent respiration, increased glycolysis rates, and a lowered ATP/AMP ratio. Imatinib augmented the fractional oxidation of fatty acids/malate, possibly via a direct interaction with the beta-oxidation enzyme enoyl coenzyme A hydratase, short chain, 1, mitochondrial (ECHS1). In non-beta cells, imatinib reduced respiratory chain complex I and II-mediated respiration and acyl-CoA carboxylase (ACC) phosphorylation, suggesting that mitochondrial effects of imatinib are not beta-cell specific. In conclusion, tyrosine kinase inhibitors modestly inhibit mitochondrial respiration, leading to AMPK activation and TXNIP down-regulation, which in turn protects against beta-cell death.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/drug therapy , Energy Metabolism/drug effects , Hypoglycemic Agents/pharmacology , Imatinib Mesylate/pharmacology , Insulin-Secreting Cells/drug effects , Mitochondria/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Line , Cell Respiration/drug effects , Diabetes Mellitus/enzymology , Diabetes Mellitus/pathology , Disease Models, Animal , Enoyl-CoA Hydratase/metabolism , Enzyme Activation , Humans , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Islet Amyloid Polypeptide/metabolism , Male , Mice, Inbred NOD , Mitochondria/enzymology , Mitochondria/pathology , Phosphorylation , Rats, Sprague-Dawley , Ribosomal Protein S6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...