Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Diabetes Res Clin Pract ; 203: 110877, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37579994

ABSTRACT

AIMS: Mitochondrial damage is implicated in diabetes pathogenesis and complications. Mitochondrial DNA copy number (mtDNA-cn) in human Type 1 diabetes (T1D) studies are lacking. We related mtDNA-cn in T1D and non-diabetic adults (CON) with diabetes complications and risk factors. METHODS: Cross-sectional study: 178 T1D, 132 non-diabetic controls. Associations of whole blood mtDNA-cn (qPCR) with complications, inflammation, and C-peptide. RESULTS: mtDNA-cn (median (LQ, UQ)) was lower in: T1D vs. CON (271 (189, 348) vs. 320 (264, 410); p < 0.0001); T1D with vs. without kidney disease (238 (180, 309) vs. 294 (198, 364); p = 0.02); and insulin injection vs. pump-users (251 (180, 340) vs. 322 (263, 406); p = 0.008). Significant univariate correlates of mtDNA-cn: T1D: (positive) HDL-C; (negative) fasting glucose, white cell count (WCC), sVCAM-1, sICAM-1; CON: (negative) WHR (waist-hip-ratio). Detectable C-peptide in T1D increased with lowest-highest mtDNA-cn tertiles (54%, 69%, 79%, p = 0.02). Independent determinants of mtDNA-cn: T1D: (positive) HDL-C; (negative) age, sICAM-1; AUROC 0.71; CON: WCC (negative), never smoking, (positive) female, pulse pressure; AUROC 0.74. CONCLUSIONS: mtDNA-cn is lower in T1D vs. CON, and in T1D kidney disease. In T1D, mtDNA-cn correlates inversely with age and inflammation, and positively with HDL-C, detectable C-peptide and pump use. Further clinical and basic science studies are merited.

2.
Redox Biol ; 46: 102038, 2021 10.
Article in English | MEDLINE | ID: mdl-34416478

ABSTRACT

Due to the high redox activity of the mitochondrion, this organelle can suffer oxidative stress. To manage energy demands while minimizing redox stress, mitochondrial homeostasis is maintained by the dynamic processes of mitochondrial biogenesis, mitochondrial network dynamics (fusion/fission), and mitochondrial clearance by mitophagy. Friedreich's ataxia (FA) is a mitochondrial disease resulting in a fatal hypertrophic cardiomyopathy due to the deficiency of the mitochondrial protein, frataxin. Our previous studies identified defective mitochondrial iron metabolism and oxidative stress potentiating cardiac pathology in FA. However, how these factors alter mitochondrial homeostasis remains uncharacterized in FA cardiomyopathy. This investigation examined the muscle creatine kinase conditional frataxin knockout mouse, which closely mimics FA cardiomyopathy, to dissect the mechanisms of dysfunctional mitochondrial homeostasis. Dysfunction of key mitochondrial homeostatic mechanisms were elucidated in the knockout hearts relative to wild-type littermates, namely: (1) mitochondrial proliferation with condensed cristae; (2) impaired NAD+ metabolism due to perturbations in Sirt1 activity and NAD+ salvage; (3) increased mitochondrial biogenesis, fusion and fission; and (4) mitochondrial accumulation of Pink1/Parkin with increased autophagic/mitophagic flux. Immunohistochemistry of FA patients' heart confirmed significantly enhanced expression of markers of mitochondrial biogenesis, fusion/fission and autophagy. These novel findings demonstrate cardiac frataxin-deficiency results in significant changes to metabolic mechanisms critical for mitochondrial homeostasis. This mechanistic dissection provides critical insight, offering the potential for maintaining mitochondrial homeostasis in FA and potentially other cardio-degenerative diseases by implementing innovative treatments targeting mitochondrial homeostasis and NAD+ metabolism.


Subject(s)
Cardiomyopathies , Friedreich Ataxia , Mitochondrial Diseases , Animals , Cardiomyopathies/metabolism , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Homeostasis , Humans , Iron/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , NAD/metabolism , Oxidation-Reduction
3.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165844, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32480040

ABSTRACT

The iron-containing protein, acireductone dioxygenase 1 (ADI1), is a dioxygenase important for polyamine synthesis and proliferation. Using differential proteomics, the studies herein demonstrated that ADI1 was significantly down-regulated by cellular iron depletion. This is important, since ADI1 contains a non-heme, iron-binding site critical for its activity. Examination of multiple human cell-types demonstrated a significant decrease in ADI1 mRNA and protein after incubation with iron chelators. The decrease in ADI1 after iron depletion was reversible upon incubation of cells with the iron salt, ferric ammonium citrate (FAC). A significant decrease in ADI1 mRNA levels was observed after 14 h of iron depletion. In contrast, the chelator-mediated reduction in ADI1 protein occurred earlier after 10 h of iron depletion, suggesting additional post-transcriptional regulation. The proteasome inhibitor, MG-132, prevented the iron chelator-mediated decrease in ADI1 expression, while the lysosomotropic agent, chloroquine, had no effect. These results suggest an iron-dependent, proteasome-mediated, degradation mechanism. Poly r(C)-binding protein (PCBPs) 1 and 2 act as iron delivery chaperones to other iron-containing dioxygenases and were shown herein for the first time to be regulated by iron levels. Silencing of PCBP1, but not PCBP2, led to loss of ADI1 expression. Confocal microscopy co-localization studies and proximity ligation assays both demonstrated decreased interaction of ADI1 with PCBP1 and PCBP2 under conditions of iron depletion using DFO. These data indicate PCBP1 and PCBP2 interact with ADI1, but only PCBP1 plays a role in ADI1 expression. In fact, PCBP2 appeared to play an accessory role, being involved as a potential co-chaperone.


Subject(s)
DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Iron/metabolism , Molecular Chaperones/metabolism , RNA-Binding Proteins/metabolism , Binding Sites , Cell Line , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Leupeptins , Membrane Potential, Mitochondrial , Molecular Chaperones/drug effects , Proteasome Inhibitors/pharmacology , RNA-Binding Proteins/genetics , Reactive Oxygen Species/metabolism , Zinc/metabolism
4.
Pharmacol Res ; 155: 104680, 2020 05.
Article in English | MEDLINE | ID: mdl-32032665

ABSTRACT

Friedreich's ataxia (FA) is due to deficiency of the mitochondrial protein, frataxin, which results in multiple pathologies including a deadly, hypertrophic cardiomyopathy. Frataxin loss leads to deleterious accumulations of redox-active, mitochondrial iron, and suppressed mitochondrial bioenergetics. Hence, there is an urgent need to develop innovative pharmaceuticals. Herein, the activity of the novel compound, 6-methoxy-2-salicylaldehyde nicotinoyl hydrazone (SNH6), was assessed in vivo using the well-characterized muscle creatine kinase (MCK) conditional frataxin knockout (KO) mouse model of FA. The design of SNH6 incorporated a dual-mechanism mediating: (1) NAD+-supplementation to restore cardiac bioenergetics; and (2) iron chelation to remove toxic mitochondrial iron. In these studies, MCK wild-type (WT) and KO mice were treated for 4-weeks from the asymptomatic age of 4.5-weeks to 8.5-weeks of age, where the mouse displays an overt cardiomyopathy. SNH6-treatment significantly elevated NAD+ and markedly increased NAD+ consumption in WT and KO hearts. In SNH6-treated KO mice, nuclear Sirt1 activity was also significantly increased together with the NAD+-metabolic product, nicotinamide (NAM). Therefore, NAD+-supplementation by SNH6 aided mitochondrial function and cardiac bioenergetics. SNH6 also chelated iron in cultured cardiac cells and also removed iron-loading in vivo from the MCK KO heart. Despite its dual beneficial properties of supplementing NAD+ and chelating iron, SNH6 did not mitigate cardiomyopathy development in the MCK KO mouse. Collectively, SNH6 is an innovative therapeutic with marked pharmacological efficacy, which successfully enhanced cardiac NAD+ and nuclear Sirt1 activity and reduced cardiac iron-loading in MCK KO mice. No other pharmaceutical yet designed exhibits both these effective pharmacological properties.


Subject(s)
Aldehydes/therapeutic use , Cardiomyopathies/drug therapy , Friedreich Ataxia/drug therapy , Hydrazones/therapeutic use , Iron Chelating Agents/therapeutic use , NAD/metabolism , Adenosine Triphosphate/metabolism , Aldehydes/pharmacology , Animals , Cardiomyopathies/metabolism , Cell Line , Creatine Kinase, MM Form/genetics , Disease Models, Animal , Friedreich Ataxia/metabolism , Hydrazones/pharmacology , Iron/metabolism , Iron Chelating Agents/pharmacology , Iron-Binding Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Rats , Frataxin
5.
Br J Pharmacol ; 177(9): 1967-1987, 2020 05.
Article in English | MEDLINE | ID: mdl-31895471

ABSTRACT

BACKGROUND AND PURPOSE: Alzheimer's disease (AD) is a multifactorial condition leading to cognitive decline and represents a major global health challenge in ageing populations. The lack of effective AD therapeutics led us to develop multifunctional nicotinoyl hydrazones to target several pathological characteristics of AD. EXPERIMENTAL APPROACH: We synthesised 20 novel multifunctional agents based on the nicotinoyl hydrazone scaffold, which acts as a metal chelator and a lipophilic delivery vehicle, donating a NAD+ precursor to cells, to target metal dyshomeostasis, oxidative stress, ß-amyloid (Aß) aggregation, and a decrease in the NAD+ /NADH ratio. KEY RESULTS: The most promising compound, 6-methoxysalicylaldehyde nicotinoyl hydrazone (SNH6), demonstrated low cytotoxicity, potent iron (Fe)-chelation efficacy, significant inhibition of copper-mediated Aß aggregation, oxidative stress alleviation, effective donation of NAD+ to NAD-dependent metabolic processes (PARP and sirtuin activity) and enhanced cellular NAD+ /NADH ratios, as well as significantly increased median Caenorhabditis elegans lifespan (to 1.46-fold of the control); partly decreased BACE1 expression, resulting in significantly lower soluble amyloid precursor protein-ß (sAPPß) and Aß1-40 levels; and favourable blood-brain barrier-permeation properties. Structure-activity relationships demonstrated that the ability of these nicotinoyl hydrazones to increase NAD+ was dependent on the electron-withdrawing or electron-donating substituents on the aldehyde- or ketone-derived moiety. Aldehyde-derived hydrazones containing the ONO donor set and electron-donating groups were required for NAD+ donation and low cytotoxicity. CONCLUSIONS AND IMPLICATIONS: The nicotinoyl hydrazones, particularly SNH6, have the potential to act as multifunctional therapeutic agents and delivery vehicles for NAD+ precursors for AD treatment.


Subject(s)
Alzheimer Disease , Hydrazones , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Animals , Aspartic Acid Endopeptidases , Caenorhabditis elegans , Humans , Hydrazones/pharmacology , Iron Chelating Agents , NAD
6.
Oxid Med Cell Longev ; 2019: 6392763, 2019.
Article in English | MEDLINE | ID: mdl-31057691

ABSTRACT

The mitochondrion is an essential organelle important for the generation of ATP for cellular function. This is especially critical for cells with high energy demands, such as neurons for signal transmission and cardiomyocytes for the continuous mechanical work of the heart. However, deleterious reactive oxygen species are generated as a result of mitochondrial electron transport, requiring a rigorous activation of antioxidative defense in order to maintain homeostatic mitochondrial function. Indeed, recent studies have demonstrated that the dysregulation of antioxidant response leads to mitochondrial dysfunction in human degenerative diseases affecting the nervous system and the heart. In this review, we outline and discuss the mitochondrial and oxidative stress factors causing degenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and Friedreich's ataxia. In particular, the pathological involvement of mitochondrial dysfunction in relation to oxidative stress, energy metabolism, mitochondrial dynamics, and cell death will be explored. Understanding the pathology and the development of these diseases has highlighted novel regulators in the homeostatic maintenance of mitochondria. Importantly, this offers potential therapeutic targets in the development of future treatments for these degenerative diseases.


Subject(s)
Antioxidants/metabolism , Apoptosis , Autophagy , Energy Metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Mitochondria/metabolism , Oxidative Stress , Heredodegenerative Disorders, Nervous System/pathology , Humans , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Reactive Oxygen Species/metabolism
7.
Curr Med Chem ; 26(2): 302-322, 2019.
Article in English | MEDLINE | ID: mdl-28685681

ABSTRACT

Metals are vital cellular elements necessary for multiple indispensable biological processes of living organisms, including energy transduction and cell proliferation. Interestingly, alterations in metal levels and also changes in the expression of proteins involved in metal metabolism have been demonstrated in a variety of cancers. Considering this and the important role of metals for cell growth, the development of drugs that sequester metals has become an attractive target for the development of novel anti-cancer agents. Interest in this field has surged with the design and development of new generations of chelators of the thiosemicarbazone class. These ligands have shown potent anticancer and anti-metastatic activity in vitro and in vivo. Due to their efficacy and safe toxicological assessment, some of these agents have recently entered multi-center clinical trials as therapeutics for advanced and resistant tumors. This review highlights the role and changes in homeostasis of metals in cancer and emphasizes the pre-clinical development and clinical assessment of metal ion-binding agents, namely, thiosemicarbazones, as antitumor agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Chelating Agents/therapeutic use , Metals, Heavy/metabolism , Neoplasms/drug therapy , Thiosemicarbazones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chelating Agents/chemistry , Chelating Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Ligands , Metals, Heavy/chemistry , Neoplasm Metastasis/prevention & control , Neoplasms/metabolism , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology
8.
Pharmacol Res ; 139: 298-313, 2019 01.
Article in English | MEDLINE | ID: mdl-30453033

ABSTRACT

Mitochondria play vital roles in various cellular processes, ranging from cellular metabolism to signal transduction and cell death regulation. As these properties are critical for cancer growth, the mitochondrion has recently become an attractive target for anti-cancer therapies. In addition, it has come to light that mitochondria are crucially involved in the regulation of stem cell identity, differentiation and fate. A similar role for mitochondria has been also demonstrated in malignant stem-like cells termed cancer stem cells (CSCs), which are implicated in progression and resistance of many tumors. In this review, we summarize different mitochondrial functions reported to promote acquisition and maintenance of CSC phenotype and discuss the rationale for their therapeutic targeting. Particular emphasis is given to therapeutics that act directly through modulation of these mitochondrial functions and have recently emerged as promising anti-CSC drugs in pre-clinical studies. This review highlights the intriguing aspects of mitochondrial biology that may have a crucial role in cancer initiation, progression, and resistance and which might facilitate pharmacological targeting. Indeed, understanding of mitochondrial function in the regulation of CSCs will promote the development of novel CSC-targeted therapeutic strategies, which could significantly improve the long-term survival of cancer patients.


Subject(s)
Mitochondria/physiology , Neoplastic Stem Cells/physiology , Animals , Antineoplastic Agents/pharmacology , Humans , Mitochondria/drug effects
9.
Carcinogenesis ; 40(6): 805-818, 2019 07 06.
Article in English | MEDLINE | ID: mdl-30561520

ABSTRACT

The metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), plays multifaceted roles in inhibiting oncogenic signaling and can suppress the epithelial mesenchymal transition (EMT), a key step in metastasis. In this investigation, NDRG1 inhibited the oncogenic effects of transforming growth factor-ß (TGF-ß) in PANC-1 pancreatic cancer cells, promoting expression and co-localization of E-cadherin and ß-catenin at the cell membrane. A similar effect of NDRG1 at supporting E-cadherin and ß-catenin co-localization at the cell membrane was also demonstrated for HT-29 colon and CFPAC-1 pancreatic cancer cells. The increase in E-cadherin in PANC-1 cells in response to NDRG1 was mediated by the reduction of three transcriptional repressors of E-cadherin, namely SNAIL, SLUG and ZEB1. To dissect the mechanisms how NDRG1 inhibits nuclear SNAIL, SLUG and ZEB1, we assessed involvement of the nuclear factor-κB (NF-κB) pathway, as its aberrant activation contributes to the EMT. Interestingly, NDRG1 comprehensively inhibited oncogenic NF-κB signaling at multiple sites in this pathway, suppressing NEMO, Iĸĸα and IĸBα expression, as well as reducing the activating phosphorylation of Iĸĸα/ß and IĸBα. NDRG1 also reduced the levels, nuclear co-localization and DNA-binding activity of NF-κB p65. Further, Iĸĸα, which integrates NF-κB and TGF-ß signaling to upregulate ZEB1, SNAIL and SLUG, was identified as an NDRG1 target. Considering this, therapies targeting NDRG1 could be a new strategy to inhibit metastasis, and as such, we examined novel anticancer agents, namely di-2-pyridylketone thiosemicarbazones, which upregulate NDRG1. These agents downregulated SNAIL, SLUG and ZEB1 in vitro and in vivo using a PANC-1 tumor xenograft model, demonstrating their marked potential.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Cycle Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/metabolism , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Thiosemicarbazones/pharmacology
10.
J Biol Chem ; 293(10): 3562-3587, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29305422

ABSTRACT

Multidrug resistance (MDR) is a major obstacle in cancer treatment due to the ability of tumor cells to efflux chemotherapeutics via drug transporters (e.g. P-glycoprotein (Pgp; ABCB1)). Although the mechanism of Pgp-mediated drug efflux is known at the plasma membrane, the functional role of intracellular Pgp is unclear. Moreover, there has been intense focus on the tumor micro-environment as a target for cancer treatment. This investigation aimed to dissect the effects of tumor micro-environmental stress on subcellular Pgp expression, localization, and its role in MDR. These studies demonstrated that tumor micro-environment stressors (i.e. nutrient starvation, low glucose levels, reactive oxygen species, and hypoxia) induce Pgp-mediated drug resistance. This occurred by two mechanisms, where stressors induced 1) rapid Pgp internalization and redistribution via intracellular trafficking (within 1 h) and 2) hypoxia-inducible factor-1α expression after longer incubations (4-24 h), which up-regulated Pgp and was accompanied by lysosomal biogenesis. These two mechanisms increased lysosomal Pgp and facilitated lysosomal accumulation of the Pgp substrate, doxorubicin, resulting in resistance. This was consistent with lysosomal Pgp being capable of transporting substrates into lysosomes. Hence, tumor micro-environmental stressors result in: 1) Pgp redistribution to lysosomes; 2) increased Pgp expression; 3) lysosomal biogenesis; and 4) potentiation of Pgp substrate transport into lysosomes. In contrast to doxorubicin, when stress stimuli increased lysosomal accumulation of the cytotoxic Pgp substrate, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), this resulted in the agent overcoming resistance. Overall, this investigation describes a novel approach to overcoming resistance in the stressful tumor micro-environment.


Subject(s)
Antineoplastic Agents/pharmacology , Lysosomes/drug effects , Models, Biological , Neoplasms/drug therapy , Thiosemicarbazones/pharmacology , Tumor Microenvironment/drug effects , ATP Binding Cassette Transporter, Subfamily B/agonists , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Acridines/pharmacology , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Biological Transport/drug effects , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysosomes/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Organelle Biogenesis , Protein Transport/drug effects , RNA Interference , Tetrahydroisoquinolines/pharmacology
11.
Neurochem Int ; 117: 35-48, 2018 07.
Article in English | MEDLINE | ID: mdl-28782591

ABSTRACT

Mitochondrial homeostasis is essential for maintaining healthy cellular function and survival. The detrimental involvement of mitochondrial dysfunction in neuro-degenerative diseases has recently been highlighted in human conditions, such as Parkinson's, Alzheimer's and Huntington's disease. Friedreich's ataxia (FA) is another neuro-degenerative, but also cardio-degenerative condition, where mitochondrial dysfunction plays a crucial role in disease progression. Deficient expression of the mitochondrial protein, frataxin, is the primary cause of FA, which leads to adverse alterations in whole cell and mitochondrial iron metabolism. Dys-regulation of iron metabolism in these compartments, results in the accumulation of inorganic iron deposits in the mitochondrial matrix that is thought to potentiate oxidative damage observed in FA. Therefore, the maintenance of mitochondrial homeostasis is crucial in the progression of neuro-degenerative conditions, particularly in FA. In this review, vital mitochondrial homeostatic processes and their roles in FA pathogenesis will be discussed. These include mitochondrial iron processing, mitochondrial dynamics (fusion and fission processes), mitophagy, mitochondrial biogenesis, mitochondrial energy production and calcium metabolism.


Subject(s)
Cardiovascular Diseases/metabolism , Friedreich Ataxia/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Homeostasis/physiology , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mitochondria/genetics , Mitochondria/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oxidative Stress/physiology , Frataxin
12.
Am J Pathol ; 187(12): 2858-2875, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28935570

ABSTRACT

Nuclear factor-erythroid 2-related factor-2 (Nrf2) is a master regulator of the antioxidant response. However, studies in models of Friedreich ataxia, a neurodegenerative and cardiodegenerative disease associated with oxidative stress, reported decreased Nrf2 expression attributable to unknown mechanisms. Using a mouse conditional frataxin knockout (KO) model in the heart and skeletal muscle, we examined the Nrf2 pathway in these tissues. Frataxin KO results in fatal cardiomyopathy, whereas skeletal muscle was asymptomatic. In the KO heart, protein oxidation and a decreased glutathione/oxidized glutathione ratio were observed, but the opposite was found in skeletal muscle. Decreased total and nuclear Nrf2 and increased levels of its inhibitor, Kelch-like ECH-associated protein 1, were evident in the KO heart, but not in skeletal muscle. Moreover, a mechanism involving activation of the nuclear Nrf2 export/degradation machinery via glycogen synthase kinase-3ß (Gsk3ß) signaling was demonstrated in the KO heart. This process involved the following: i) increased Gsk3ß activation, ii) ß-transducin repeat containing E3 ubiquitin protein ligase nuclear accumulation, and iii) Fyn phosphorylation. A corresponding decrease in Nrf2-DNA-binding activity and a general decrease in Nrf2-target mRNA were observed in KO hearts. Paradoxically, protein levels of some Nrf2 antioxidant targets were significantly increased in KO mice. Collectively, cardiac frataxin deficiency reduces Nrf2 levels via two potential mechanisms: increased levels of cytosolic Kelch-like ECH-associated protein 1 and activation of Gsk3ß signaling, which decreases nuclear Nrf2. These findings are in contrast to the frataxin-deficient skeletal muscle, where Nrf2 was not decreased.


Subject(s)
Friedreich Ataxia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Disease Models, Animal , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/physiology , Up-Regulation , Frataxin
13.
Mol Pharmacol ; 91(5): 499-517, 2017 05.
Article in English | MEDLINE | ID: mdl-28275050

ABSTRACT

Tumor necrosis factor α (TNFα) plays a vital role in cancer progression as it is associated with inflammation and promotion of cancer angiogenesis and metastasis. The effects of TNFα are mediated by its downstream target, the oncogene lysine-rich CEACAM1 coisolated protein (LYRIC, also known as metadherin or astrocyte elevated gene-1). LYRIC plays an important role in activating the nuclear factor-ĸB (NF-κB) signaling pathway, which controls multiple cellular processes, including proliferation, apoptosis, migration, etc. In contrast, the metastasis suppressor N-myc downstream regulated gene 1 (NDRG1) has the opposite effect on the NF-κB pathway, being able to inhibit NF-κB activation and reduce angiogenesis, proliferation, migration, and cancer cell invasion. These potent anticancer properties make NDRG1 an ideal therapeutic target. Indeed, a novel class of thiosemicarbazone anticancer agents that target this molecule has been developed; the lead agent, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone, has recently entered clinical trials for advanced and resistant cancers. To further elucidate the interaction between NDRG1 and oncogenic signaling, this study for the first time assessed the effects of NDRG1 on the tumorigenic properties of TNFα and its downstream target, LYRIC. We have demonstrated that NDRG1 inhibits the TNFα-mediated epithelial-to-mesenchymal transition. Further, NDRG1 also potently inhibited LYRIC expression, with a negative feedback loop existing between these two molecules. Examining the mechanism involved, we demonstrated that NDRG1 inhibited phosphatidylinositol 3-kinase/AKT signaling, leading to reduced levels of the LYRIC transcriptional activator, c-Myc. Finally, we demonstrated that novel thiosemicarbazones that upregulate NDRG1 also inhibit LYRIC expression, further highlighting their marked potential for cancer treatment.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Epithelial-Mesenchymal Transition/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Thiosemicarbazones/pharmacology , Up-Regulation/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Deferoxamine/pharmacology , Gene Silencing/drug effects , Humans , Membrane Proteins , Models, Biological , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , RNA-Binding Proteins , Thiosemicarbazones/chemistry , Tumor Necrosis Factor-alpha/pharmacology , Vimentin/metabolism
15.
Metallomics ; 8(9): 874-86, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27334916

ABSTRACT

Copper is an essential trace metal required by organisms to perform a number of important biological processes. Copper readily cycles between its reduced Cu(i) and oxidised Cu(ii) states, which makes it redox active in biological systems. This redox-cycling propensity is vital for copper to act as a catalytic co-factor in enzymes. While copper is essential for normal physiology, enhanced copper levels in tumours leads to cancer progression. In particular, the stimulatory effect of copper on angiogenesis has been established in the last several decades. Additionally, it has been demonstrated that copper affects tumour growth and promotes metastasis. Based on the effects of copper on cancer progression, chelators that bind copper have been developed as anti-cancer agents. In fact, a novel class of thiosemicarbazone compounds, namely the di-2-pyridylketone thiosemicarbazones that bind copper, have shown great promise in terms of their anti-cancer activity. These agents have a unique mechanism of action, in which they form redox-active complexes with copper in the lysosomes of cancer cells. Furthermore, these agents are able to overcome P-glycoprotein (P-gp) mediated multi-drug resistance (MDR) and act as potent anti-oncogenic agents through their ability to up-regulate the metastasis suppressor protein, N-myc downstream regulated gene-1 (NDRG1). This review provides an overview of the metabolism and regulation of copper in normal physiology, followed by a discussion of the dysregulation of copper homeostasis in cancer and the effects of copper on cancer progression. Finally, recent advances in our understanding of the mechanisms of action of anti-cancer agents targeting copper are discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/chemistry , Neoplasms/drug therapy , Thiosemicarbazones/pharmacology , Animals , Antineoplastic Agents/chemistry , Copper/metabolism , Humans , Thiosemicarbazones/chemistry
16.
Clin Sci (Lond) ; 130(11): 853-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27129098

ABSTRACT

The mitochondrion is a major site for the metabolism of the transition metal, iron, which is necessary for metabolic processes critical for cell vitality. The enigmatic mitochondrial protein, frataxin, is known to play a significant role in both cellular and mitochondrial iron metabolism due to its iron-binding properties and its involvement in iron-sulfur cluster (ISC) and heme synthesis. The inherited neuro- and cardio-degenerative disease, Friedreich's ataxia (FA), is caused by the deficient expression of frataxin that leads to deleterious alterations in iron metabolism. These changes lead to the accumulation of inorganic iron aggregates in the mitochondrial matrix that are presumed to play a key role in the oxidative damage and subsequent degenerative features of this disease. Furthermore, the concurrent dys-regulation of cellular antioxidant defense, which coincides with frataxin deficiency, exacerbates oxidative stress. Hence, the pathogenesis of FA underscores the importance of the integrated homeostasis of cellular iron metabolism and the cytoplasmic and mitochondrial redox environments. This review focuses on describing the pathogenesis of the disease, the molecular mechanisms involved in mitochondrial iron-loading and the dys-regulation of cellular antioxidant defense due to frataxin deficiency. In turn, current and emerging therapeutic strategies are also discussed.


Subject(s)
Friedreich Ataxia/drug therapy , Homeostasis/drug effects , Iron-Binding Proteins/pharmacology , Iron/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Animals , Friedreich Ataxia/metabolism , Humans , Mitochondria/metabolism , Frataxin
17.
Biochim Biophys Acta ; 1863(4): 727-48, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26844773

ABSTRACT

Essential metals, such as iron and copper, play a critical role in a plethora of cellular processes including cell growth and proliferation. However, concomitantly, excess of these metal ions in the body can have deleterious effects due to their ability to generate cytotoxic reactive oxygen species (ROS). Thus, the human body has evolved a very well-orchestrated metabolic system that keeps tight control on the levels of these metal ions. Considering their very high proliferation rate, cancer cells require a high abundance of these metals compared to their normal counterparts. Interestingly, new anti-cancer agents that take advantage of the sensitivity of cancer cells to metal sequestration and their susceptibility to ROS have been developed. These ligands can avidly bind metal ions to form redox active metal complexes, which lead to generation of cytotoxic ROS. Furthermore, these agents also act as potent metastasis suppressors due to their ability to up-regulate the metastasis suppressor gene, N-myc downstream regulated gene 1. This review discusses the importance of iron and copper in the metabolism and progression of cancer, how they can be exploited to target tumors and the clinical translation of novel anti-cancer chemotherapeutics.


Subject(s)
Antineoplastic Agents , Chelating Agents , Copper/metabolism , Drug Discovery , Iron/metabolism , Metals/metabolism , Animals , Antineoplastic Agents/therapeutic use , Chelating Agents/therapeutic use , Drug Discovery/methods , Drug Discovery/trends , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism
18.
Biochim Biophys Acta ; 1863(4): 770-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26844774

ABSTRACT

Melanoma has markedly increased worldwide during the past several decades in the Caucasian population and is responsible for 80% of skin cancer deaths. Considering that metastatic melanoma is almost completely resistant to most current therapies and is linked with a poor patient prognosis, it is crucial to further investigate potential molecular targets. Major cell-autonomous drivers in the pathogenesis of this disease include the classical MAPK (i.e., RAS-RAF-MEK-ERK), WNT, and PI3K signaling pathways. These pathways play a major role in defining the progression of melanoma, and some have been the subject of recent pharmacological strategies to treat this belligerent disease. This review describes the latest advances in the understanding of melanoma progression and the major molecular pathways involved. In addition, we discuss the roles of emerging molecular players that are involved in melanoma pathogenesis, including the functional role of the melanoma tumor antigen, p97/MFI2 (melanotransferrin).


Subject(s)
Melanoma/genetics , Melanoma/pathology , Oncogenes/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Disease Progression , Genes, ras/physiology , Humans , MAP Kinase Signaling System/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Wnt Signaling Pathway/physiology , raf Kinases/physiology
19.
Mol Pharmacol ; 89(5): 521-40, 2016 May.
Article in English | MEDLINE | ID: mdl-26895766

ABSTRACT

Metastasis is a complex process that is regulated by multiple signaling pathways, with the focal adhesion kinase (FAK)/paxillin pathway playing a major role in the formation of focal adhesions and cell motility. N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor in many solid tumor types, including prostate and colon cancer. Considering the antimetastatic effect of NDRG1 and the crucial involvement of the FAK/paxillin pathway in cellular migration and cell-matrix adhesion, we assessed the effects of NDRG1 on this important oncogenic pathway. In the present study, NDRG1 overexpression and silencing models of HT29 colon cancer and DU145 prostate cancer cells were used to examine the activation of FAK/paxillin signaling and the formation of focal adhesions. The expression of NDRG1 resulted in a marked and significant decrease in the activating phosphorylation of FAK and paxillin, whereas silencing of NDRG1 resulted in an opposite effect. The expression of NDRG1 also inhibited the formation of focal adhesions as well as cell migration and cell-collagen adhesion. Incubation of cells with novel thiosemicarbazones, namely di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone and di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone, that upregulate NDRG1 also resulted in decreased phosphorylation of FAK and paxillin. The ability of these thiosemicarbazones to inhibit cell migration and metastasis could be mediated, at least in part, through the FAK/paxillin pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Colonic Neoplasms/metabolism , Focal Adhesion Kinase 1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Paxillin/metabolism , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Signal Transduction , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Collagen/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Enzyme Activation/drug effects , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/genetics , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Focal Adhesions/pathology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Paxillin/agonists , Paxillin/antagonists & inhibitors , Phosphorylation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Thiosemicarbazones/pharmacology
20.
Oncotarget ; 6(34): 35522-41, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26431493

ABSTRACT

A major problem for cancer patients is the metastasis of cancer cells from the primary tumor. This involves: (1) migration through the basement membrane; (2) dissemination via the circulatory system; and (3) invasion into a secondary site. Metastasis suppressors, by definition, inhibit metastasis at any step of the metastatic cascade. Notably, Src is a non-receptor, cytoplasmic, tyrosine kinase, which becomes aberrantly activated in many cancer-types following stimulation of plasma membrane receptors (e.g., receptor tyrosine kinases and integrins). There is evidence of a prominent role of Src in tumor progression-related events such as the epithelial-mesenchymal transition (EMT) and the development of metastasis. However, the precise molecular interactions of Src with metastasis suppressors remain unclear. Herein, we review known metastasis suppressors and summarize recent advances in understanding the mechanisms of how these proteins inhibit metastasis through modulation of Src. Particular emphasis is bestowed on the potent metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1) and its interactions with the Src signaling cascade. Recent studies demonstrated a novel mechanism through which NDRG1 plays a significant role in regulating cancer cell migration by inhibiting Src activity. Moreover, we discuss the rationale for targeting metastasis suppressor genes as a sound therapeutic modality, and we review several examples from the literature where such strategies show promise. Collectively, this review summarizes the essential interactions of metastasis suppressors with Src and their effects on progression of cancer metastasis. Moreover, interesting unresolved issues regarding these proteins as well as their potential as therapeutic targets are also discussed.


Subject(s)
Genes, src , src-Family Kinases/genetics , src-Family Kinases/metabolism , Carcinogenesis , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...