Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Nitric Oxide ; 149: 18-31, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38823434

ABSTRACT

Redox-based protein posttranslational modifications, such as S-nitrosylation of critical, active site cysteine thiols have garnered significant clinical attention and research interest, reasoning for one of the crucial biological implications of reactive messenger molecule, nitric oxide in the cellular repertoire. The stringency of the S-(de)nitrosylation-based redox switch governs the activity and contribution of several susceptible enzymes in signal transduction processes and diverse pathophysiological settings, thus establishing it as a transient yet reasonable, and regulated mechanism of NO adduction and release. Notably, endogenous proteases like cytosolic and mitochondrial caspases with a molecular weight ranging from 33 to 55 kDa are susceptible to performing this biochemistry in the presence of major oxidoreductases, which further unveils the enormous redox-mediated regulational control of caspases in the etiology of diseases. In addition to advancing the progress of the current state of understanding of 'redox biochemistry' in the field of medicine and enriching the existing dynamic S-nitrosoproteome, this review stands as a testament to an unprecedented shift in the underpinnings for redundancy and redox relay between the major redoxin/antioxidant systems, fine-tuning of which can command the apoptotic control of caspases at the face of nitro-oxidative stress. These intricate functional overlaps and cellular backups, supported rationally by kinetically favorable reaction mechanisms suggest the physiological relevance of identifying and involving such cognate substrates for cellular S-denitrosylases that can shed light on the bigger picture of extensively proposing targeted therapies and redox-based drug designing to potentially alleviate the side effects of NOx/ROS in disease pathogenesis.


Subject(s)
Caspases , Oxidation-Reduction , Humans , Caspases/metabolism , Animals , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Cysteine/metabolism
2.
Biochim Biophys Acta Gen Subj ; 1867(11): 130452, 2023 11.
Article in English | MEDLINE | ID: mdl-37652366

ABSTRACT

The interdependent and finely tuned balance between the well-established redox-based modification, S-nitrosylation, and its counteractive mechanism of S-nitrosothiol degradation, i.e., S-denitrosylation of biological protein or non-protein thiols defines the cellular fate in the context of redox homeostasis. S-nitrosylation of cysteine residues by S-nitrosoglutathione, S-nitroso-L-cysteine-like physiological and S-nitroso-L-cysteine ethyl ester-like synthetic NO donors inactivate Caspase-3, 8, and 9, thereby hindering their apoptotic activity. However, spontaneous restoration of their activity upon S-denitrosylation of S-nitrosocaspases into their reduced, free thiol active states, aided by the members of the ubiquitous cellular redoxin (thioredoxin/ thioredoxin reductase/ NADPH) and low molecular weight dithiol (lipoic acid/ lipoamide dehydrogenase/ dihydrolipoic acid/ NADPH) systems imply a direct relevance to their proteolytic activities and further downstream signaling cascades. Additionally, our previous and current findings offer crucial insight into the concept of redundancy between thioredoxin and lipoic acid systems, and the redox-modulated control of the apoptotic and proteolytic activity of caspases, triggering their cyto- and neurotoxic effects in response to nitro-oxidative stress. Thus, this might lay the foundation for the exogenous introduction of precise and efficient NO or related donor drug delivery systems that can directly participate in catering to the S-(de)-nitrosylation-mediated functional outcomes of the cysteinyl proteases in pathophysiological settings.


Subject(s)
Nitric Oxide , Thioctic Acid , Humans , Nitric Oxide/metabolism , Caspase 9/metabolism , Hep G2 Cells , NADP/metabolism , Thioredoxins/metabolism , Thioredoxin-Disulfide Reductase
3.
Biochem Biophys Res Commun ; 653: 83-92, 2023 04 23.
Article in English | MEDLINE | ID: mdl-36863212

ABSTRACT

Proteins become S-glutathionylated as a result of the derivatization of their cysteine thiols with the thiolate anion derivative of glutathione; this process is frequently linked to diseases and protein misbehavior. Along with the other well-known oxidative modifications like S-nitrosylation, S-glutathionylation has quickly emerged as a major contributor to a number of diseases, with a focus on neurodegeneration. The immense clinical significance of S-glutathionylation in cell signaling and the genesis of diseases are progressively coming to light with advanced research, which is also creating new opportunities for prompt diagnostics that utilize this phenomenon. In-depth investigation in recent years has revealed other significant deglutathionylases in addition to glutaredoxin, necessitating the hunt for their specific substrates. The precise catalytic mechanisms of these enzymes must also be understood, along with how the intracellular environment affects their impact on protein conformation and function. These insights must then be extrapolated to the understanding of neurodegeneration and the introduction of novel and clever therapeutic approaches to clinics. Clarifying the importance of the functional overlap of glutaredoxin and other deglutathionylases and examining their complementary functions as defense systems in the face of stress are essential prerequisites for predicting and promoting cell survival under high oxidative/nitrosative stress.


Subject(s)
Glutaredoxins , Protein Processing, Post-Translational , Glutaredoxins/metabolism , Proteins/metabolism , Glutathione/metabolism , Cysteine/metabolism , Oxidation-Reduction , Oxidative Stress
4.
Acta Neuropathol Commun ; 11(1): 19, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36691076

ABSTRACT

We recently discovered that the expression of PRKN, a young-onset Parkinson disease-linked gene, confers redox homeostasis. To further examine the protective effects of parkin in an oxidative stress model, we first combined the loss of prkn with Sod2 haploinsufficiency in mice. Although adult prkn-/-//Sod2± animals did not develop dopamine cell loss in the S. nigra, they had more reactive oxidative species and a higher concentration of carbonylated proteins in the brain; bi-genic mice also showed a trend for more nitrotyrosinated proteins. Because these redox changes were seen in the cytosol rather than mitochondria, we next explored the thiol network in the context of PRKN expression. We detected a parkin deficiency-associated increase in the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG) in murine brain, PRKN-linked human cortex and several cell models. This shift resulted from enhanced recycling of GSSG back to GSH via upregulated glutathione reductase activity; it also correlated with altered activities of redox-sensitive enzymes in mitochondria isolated from mouse brain (e.g., aconitase-2; creatine kinase). Intriguingly, human parkin itself showed glutathione-recycling activity in vitro and in cells: For each GSSG dipeptide encountered, parkin regenerated one GSH molecule and was S-glutathionylated by the other (GSSG + P-SH [Formula: see text] GSH + P-S-SG), including at cysteines 59, 95 and 377. Moreover, parkin's S-glutathionylation was reversible by glutaredoxin activity. In summary, we found that PRKN gene expression contributes to the network of available thiols in the cell, including by parkin's participation in glutathione recycling, which involves a reversible, posttranslational modification at select cysteines. Further, parkin's impact on redox homeostasis in the cytosol can affect enzyme activities elsewhere, such as in mitochondria. We posit that antioxidant functions of parkin may explain many of its previously described, protective effects in vertebrates and invertebrates that are unrelated to E3 ligase activity.


Subject(s)
Glutathione , Proteins , Adult , Mice , Humans , Animals , Glutathione Disulfide/metabolism , Glutathione/metabolism , Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Ubiquitin-Protein Ligases/genetics , Antioxidants , Cysteine/metabolism , Brain/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Mammals/metabolism
5.
Antioxidants (Basel) ; 11(10)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36290644

ABSTRACT

S-nitrosylation of proteins occurs as a consequence of the derivatization of cysteine thiols with nitric oxide (NO) and is often associated with diseases and protein malfunction. Aberrant S-nitrosylation, in addition to other genetic and epigenetic factors, has gained rapid importance as a prime cause of various metabolic, respiratory, and cardiac disorders, with a major emphasis on cancer and neurodegeneration. The S-nitrosoproteome, a term used to collectively refer to the diverse and dynamic repertoire of S-nitrosylated proteins, is relatively less explored in the field of redox biochemistry, in contrast to other covalently modified versions of the same set of proteins. Advancing research is gradually unveiling the enormous clinical importance of S-nitrosylation in the etiology of diseases and is opening up new avenues of prompt diagnosis that harness this phenomenon. Ever since the discovery of the two robust and highly conserved S-nitrosoglutathione reductase and thioredoxin systems as candidate denitrosylases, years of rampant speculation centered around the identification of specific substrates and other candidate denitrosylases, subcellular localization of both substrates and denitrosylases, the position of susceptible thiols, mechanisms of S-denitrosylation under basal and stimulus-dependent conditions, impact on protein conformation and function, and extrapolating these findings towards the understanding of diseases, aging and the development of novel therapeutic strategies. However, newer insights in the ever-expanding field of redox biology reveal distinct gaps in exploring the crucial crosstalk between the redoxins/major denitrosylase systems. Clarifying the importance of the functional overlap of the glutaredoxin, glutathione, and thioredoxin systems and examining their complementary functions as denitrosylases and antioxidant enzymatic defense systems are essential prerequisites for devising a rationale that could aid in predicting the extent of cell survival under high oxidative/nitrosative stress while taking into account the existence of the alternative and compensatory regulatory mechanisms. This review thus attempts to highlight major gaps in our understanding of the robust cellular redox regulation system, which is upheld by the concerted efforts of various denitrosylases and antioxidants.

6.
Anal Biochem ; 655: 114872, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36027970

ABSTRACT

Tumor formation and progression have been much of a study over the last two centuries. Recent studies have seen different developments for the early diagnosis and treatment of the disease; some of which even promise survival of the patient. Cysteine proteases, mainly cathepsins have been unequivocally identified as putative worthy players of redox imbalance that contribute to the premonition and further progression of cancer by interfering in the normal extracellular and intracellular proteolysis and initiating a proteolytic cascade. The present review article focuses on the study of cancer so far, while establishing facts on how future studies focused on the cellular interrelation between nitric oxide (NO) and cancer, can direct their focus on cathepsins. For a tumor cell to thrive and synergize in a cancerous environment, different mutations in the proteolytic and signaling pathways and the proto-oncogenes, oncogenes, and the tumor suppressor genes are made possible through cellular biochemistry and some cancer-stimulating environmental factors. The accumulated findings show that S-nitrosylation of cathepsins under the influence of NO-donors can prevent the invasion of cancer and cause cancer cell death by blocking the activity of cathepsins as well as the major denitrosylase systems using a multi-way approach. Faced with a conundrum of how to fill the gap between the dodging of established cancer hallmarks with cathepsin activity and gaining appropriate research/clinical accreditation using our hypothesis, the scope of this review also explores the interplay and crosstalk between S-nitrosylation and S-(de)nitrosylation of this protease and highlights the utility of charging thioredoxin (Trx) reductase inhibitors, low-molecular-weight dithiols, and Trx mimetics using efficient drug delivery system to prevent the denitrosylation or regaining of cathepsin activity in vivo. In foresight, this raises the prospect that drugs or novel compounds that target cathepsins taking all these factors into consideration could be deployed as alternative or even better treatments for cancer, though further research is needed to ascertain the safety, efficiency and effectiveness of this approach.


Subject(s)
Cathepsins , Neoplasms , Cathepsins/metabolism , Cysteine/metabolism , Humans , Nitric Oxide/metabolism , Oxidation-Reduction
7.
Nitric Oxide ; 127: 26-43, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35850377

ABSTRACT

Ribonucleotide reductase (RNR) is a multi-subunit enzyme responsible for catalyzing the rate-limiting step in the production of deoxyribonucleotides essential for DNA synthesis and repair. The active RNR complex is composed of multimeric R1 and R2 subunits. The RNR catalysis involves the formation of tyrosyl radicals in R2 subunits and thiyl radicals in R1 subunits. Despite the quaternary structure and cofactor diversity, all the three classes of RNR have a conserved cysteine residue at the active site which is converted into a thiyl radical that initiates the substrate turnover, suggesting that the catalytic mechanism is somewhat similar for all three classes of the RNR enzyme. Increased RNR activity has been associated with malignant transformation, cancer cell growth, and tumorigenesis. Efforts concerning the understanding of RNR inhibition in designing potent RNR inhibitors/drugs as well as developing novel approaches for antibacterial, antiviral treatments, and cancer therapeutics with improved radiosensitization have been made in clinical research. This review highlights the precise and potent roles of NO in RNR inhibition by targeting both the subunits. Under nitrosative stress, the thiols of the R1 subunits have been found to be modified by S-nitrosylation and the tyrosyl radicals of the R2 subunits have been modified by nitration. In view of the recent advances and progresses in the field of nitrosative modifications and its fundamental role in signaling with implications in health and diseases, the present article focuses on the regulations of RNR activity by S-nitrosylation of thiols (R1 subunits) and nitration of tyrosyl residues (R2 subunits) which will further help in designing new drugs and therapies.


Subject(s)
Ribonucleotide Reductases , Catalysis , Catalytic Domain , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Sulfhydryl Compounds , Tyrosine
8.
Mol Biol Rep ; 49(8): 8179-8183, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35655055

ABSTRACT

BACKGROUND: The thioredoxin system (Trx), comprising of Trx, Thioredoxin reductase (TrxR) and NADPH aids in donating hydrogen group to support Ribonucleotide reductase (RNR) catalysis during de-novo DNA biosynthesis. However, it has been observed that inhibiting TrxR does not affect the viability of cancer cells that are susceptible to pharmacological glutathione (GSH) depletion. This prompted us to study the potential antioxidant redundancies that might prolong RNR activity. METHODS: To study the RNR activity assay, the RNR complex was reconstituted by mixing purified mouse recombinant RNR subunits and the conversion of [3 H] CDP into [3 H] dCDP was monitored. In the assay system, either purified Trx and GSH or Lipoamide system was supplemented as reducing agents to support RNR catalysis. RESULTS: Herein, we have found that GSH-dependent Trx reduction supports mammalian class I RNR catalysis in absence of TrxR in the system. Our data also presents the first report that the LAM system is capable of supporting in-vitro RNR activity in the complete absence of either Trx or Grx systems. CONCLUSIONS: We conclude that GSH-mediated Trx reduction and LAM systems support basal level RNR activity in vitro; in absence of TrxR and complete redoxin systems respectively and hypothesize that potential redundancy between the various antioxidant systems might synergize in sustaining RNR activity.


Subject(s)
Antioxidants , Ribonucleotide Reductases , Animals , Catalysis , Glutathione/metabolism , Mammals/metabolism , Mice , Oxidation-Reduction , Ribonucleotide Reductases/metabolism , Ribonucleotides , Thioctic Acid/analogs & derivatives , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism
9.
Nitric Oxide ; 118: 26-30, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34742907

ABSTRACT

The intracellular concentration of reduced glutathione (GSH) lies in the range of 1-10 mM, thereby indisputably making it the most abundant intracellular thiol. Such a copious amount of GSH makes it the most potent and robust cellular antioxidant that plays a crucial role in cellular defence against redox stress. The role of GSH as a denitrosylating agent is well established; in this study, we demonstrate GSH mediated denitrosylation of HepG2 cell-derived protein nitrosothiols (PSNOs), by a unique spin-trapping mechanism, using 5,5-dimethyl-1-pyrroline N-oxide (DMPO) as the spin trapping agent, followed by a western blot analysis. We also report our findings of two, hitherto unidentified substrates of GSH mediated S-denitrosylation, namely S-nitrosoglutaredoxin 1 (Grx1-SNO) and S-nitrosylated R1 subunit of ribonucleotide reductase (R1-SNO).


Subject(s)
Glutaredoxins/metabolism , Glutathione/metabolism , Ribonucleoside Diphosphate Reductase/metabolism , S-Nitrosothiols/metabolism , Cyclic N-Oxides/chemistry , Glutaredoxins/chemistry , Hep G2 Cells , Humans , Ribonucleoside Diphosphate Reductase/chemistry , S-Nitrosothiols/chemistry , Spin Labels , Spin Trapping , Thioredoxins/chemistry , Thioredoxins/metabolism
10.
Mol Biol Rep ; 48(11): 7621-7626, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34599703

ABSTRACT

Ribonucleotide reductases (RNR) catalyze the rate-limiting step in DNA synthesis during the S-phase of the cell cycle. Its constant activity in order to maintain dNTP homeostasis is a fascinating area of research and an attractive candidate for cancer research and antiviral drugs. Redox modification such as S-glutathionylation of the R1 subunit of mammalian RNR protein has been presumed to regulate the activity of RNR during catalytic cycles. Herein, we report S-glutathionylation of the R2 subunit. We have also shown Grx1 system can efficiently deglutathionylate the S-glutathionylated R2 subunit. Additionally, our data also showed for the very first time S-glutathionylation of mammalian p53R2 subunit that regulates DNA synthesis outside S-phase during DNA damage and repair. Taken together, these data will open new avenues for future research relating to exact physiological significance, target thiols, and/or overall RNR activity due to S-glutathionylation of R2 and p53R2 subunits and provide valuable insights for effective treatment regimes.


Subject(s)
Cell Cycle Proteins , DNA Replication , Glutathione , Protein Subunits , Ribonucleotide Reductases , S Phase , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Glutathione/chemistry , Glutathione/metabolism , Mice , Protein Subunits/chemistry , Protein Subunits/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism
11.
Curr Protoc ; 1(9): e262, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34570435

ABSTRACT

The dynamic and unstable nature of protein nitrosothiols (PSNOs) derived from complex biological matrices (like cell lysates) make them unsuitable for proteomic/biochemical analysis in vitro. In an attempt to increase the stability of cell-derived PSNOs, scientists have devised methods to derivatize thiols undergoing nitrosylation, with a suitable molecule, to yield a stable adduct that could easily be detected using appropriate antibodies. The Biotin Switch Assay (BTSA) is currently the most widely used method for tagging PSNOs; however, the error-prone and cumbersome nature of the BTSA protocol prompted the development of alternative mechanisms of tagging cell-derived PSNOs. One such method is the immuno-spin trapping method using 5,5-dimethyl-1-pyrroline N-oxide (DMPO), which effectively overcomes the shortcomings of the BTSA and proves to be a promising alternative. Here we describe the protocol for DMPO-based PSNO labeling and subsequent proteomic analysis by western blotting with an anti-DMPO antibody. © 2021 Wiley Periodicals LLC. Basic Protocol: Labeling of cell-derived PSNOs by DMPO-based immuno-spin trapping and their subsequent analysis by immunostaining.


Subject(s)
Proteins , Proteomics , Free Radicals , Spin Trapping , Sulfhydryl Compounds
12.
J Cell Biochem ; 122(11): 1579-1593, 2021 11.
Article in English | MEDLINE | ID: mdl-34472139

ABSTRACT

Nitric oxide is a well-known gasotransmitter molecule that covalently docks to sulfhydryl groups of proteins resulting in S-nitrosylation of proteins and nonprotein thiols that serve a variety of cellular processes including cGMP signaling, vasodilatation, neurotransmission, ion-channel modulation, and cardiac signaling. S-nitrosylation is an indispensable modification like phosphorylation that directly regulates the functionality of numerous proteins. However, recently there has been a controversy over the stability of S-nitrosylated proteins (PSNOs) within the cell. It has been argued that PSNOs formed within the cell is a transient intermediate step to more stable disulfide formation and disulfides are the predominant end effector modifications in NO-mediated signaling. The present article accumulates state-of-the-art evidence from numerous research that strongly supports the very existence of PSNOs within the cell and attempts to put an end to the controversy. This review illustrates critical points including comparative bond dissociation energies of S-NO bond, the half-life of S-nitrosothiols and PSNOs, cellular concentrations of PSNOs, X ray crystallographic studies on PSNOs, and stability of PSNOs at physiological concentration of antioxidants. These logical evidence cumulatively support the endogenous stability and inevitable existence of PSNOs/RSNOs within the cell that directly regulate the functionality of proteins and provide valuable insight into understanding stable S-nitrosylation mediated cell signaling.


Subject(s)
Proteins/chemistry , Proteins/metabolism , S-Nitrosothiols/metabolism , Animals , Disulfides/chemistry , Disulfides/metabolism , Glutathione/metabolism , Humans , Nitric Oxide/metabolism , Protein Stability , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , S-Nitrosothiols/chemistry , Signal Transduction
13.
Free Radic Biol Med ; 172: 604-621, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34245859

ABSTRACT

S-nitrosylation is a very fundamental post-translational modification of protein and non-protein thiols due the involvement of it in a variety of cellular processes including activation/inhibition of several ion channels such as ryanodine receptor in the cardiovascular system; blood vessel dilation; cGMP signaling and neurotransmission. S-nitrosothiol homeostasis in the cell is tightly regulated and perturbations in homeostasis result in an altered redox state leading to a plethora of disease conditions. However, the exact role of S-nitrosylated proteins and nitrosative stress metabolites in inflammation and in inflammation modulation is not well-reviewed. The cell utilizes its intricate defense mechanisms i.e. cellular denitrosylases such as Thioredoxin (Trx) and S-nitrosoglutathione reductase (GSNOR) systems to combat nitric oxide (NO) pathology which has also gained current attraction as novel anti-inflammatory molecules. This review attempts to provide state-of-the-art knowledge from past and present research on the mechanistic role of nitrosative stress intermediates (RNS, OONO-, PSNO) in pulmonary and autoimmune diseases and how cellular denitrosylases particularly GSNOR and Trx via imparting opposing effects can modulate and reduce inflammation in several health and disease conditions. This review would also bring into notice the existing gaps in current research where denitrosylases can be utilized for ameliorating inflammation that would leave avenues for future therapeutic interventions.


Subject(s)
Aldehyde Oxidoreductases , Nitrosative Stress , Aldehyde Oxidoreductases/metabolism , Humans , Inflammation , Nitric Oxide , Nitrosation
14.
Arch Biochem Biophys ; 704: 108869, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33819447

ABSTRACT

Parkinson's disease (PD) is one of the fastest-growing neurodegenerative disorders of increasing global prevalence. It represents the second most common movement disorder after tremor and the second most common neurodegenerative disorder after Alzheimer's disease. The incidence rate of idiopathic PD increases steadily with age, however, some variants of autosomal recessive inheritance are present with an early age-at-onset (ARPD). Approximately 50 percent of ARPD cases have been linked to bi-allelic mutations in genes encoding Parkin, DJ-1, and PINK1. Each protein has been implicated in maintaining proper mitochondrial function, which is particularly important for neuronal health. Aberrant post-translational modifications of these proteins may disrupt their cellular functions and thus contributing to the development of idiopathic PD. Some post-translational modifictions can be attributed to the dysregulation of potentially harmful reactive oxygen and nitrogen species inside the cell, which promote oxidative and nitrosative stress, respectively. Unlike oxidative modifications, the covalent modification by Nitric Oxide under nitrosative stress, leading to S-nitrosylation of Parkin, DJ-1; and PINK1, is less studied. Here, we review the available literature on S-nitrosylation of these three proteins, their implications in the pathogenesis of PD, and provide an overview of currently known, denitrosylating systems in eukaryotic cells.


Subject(s)
Nitric Oxide/metabolism , Nitrosative Stress , Parkinson Disease/metabolism , Protein Deglycase DJ-1/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational , Ubiquitin-Protein Ligases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Humans , Mutation , Nitric Oxide/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Deglycase DJ-1/genetics , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics
15.
Acta Neuropathol ; 141(5): 725-754, 2021 05.
Article in English | MEDLINE | ID: mdl-33694021

ABSTRACT

The mechanisms by which parkin protects the adult human brain from Parkinson disease remain incompletely understood. We hypothesized that parkin cysteines participate in redox reactions and that these are reflected in its posttranslational modifications. We found that in post mortem human brain, including in the Substantia nigra, parkin is largely insoluble after age 40 years; this transition is linked to its oxidation, such as at residues Cys95 and Cys253. In mice, oxidative stress induces posttranslational modifications of parkin cysteines that lower its solubility in vivo. Similarly, oxidation of recombinant parkin by hydrogen peroxide (H2O2) promotes its insolubility and aggregate formation, and in exchange leads to the reduction of H2O2. This thiol-based redox activity is diminished by parkin point mutants, e.g., p.C431F and p.G328E. In prkn-null mice, H2O2 levels are increased under oxidative stress conditions, such as acutely by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxin exposure or chronically due to a second, genetic hit; H2O2 levels are also significantly increased in parkin-deficient human brain. In dopamine toxicity studies, wild-type parkin, but not disease-linked mutants, protects human dopaminergic cells, in part through lowering H2O2. Parkin also neutralizes reactive, electrophilic dopamine metabolites via adduct formation, which occurs foremost at the primate-specific residue Cys95. Further, wild-type but not p.C95A-mutant parkin augments melanin formation in vitro. By probing sections of adult, human midbrain from control individuals with epitope-mapped, monoclonal antibodies, we found specific and robust parkin reactivity that co-localizes with neuromelanin pigment, frequently within LAMP-3/CD63+ lysosomes. We conclude that oxidative modifications of parkin cysteines are associated with protective outcomes, which include the reduction of H2O2, conjugation of reactive dopamine metabolites, sequestration of radicals within insoluble aggregates, and increased melanin formation. The loss of these complementary redox effects may augment oxidative stress during ageing in dopamine-producing cells of mutant PRKN allele carriers, thereby enhancing the risk of Parkinson's-linked neurodegeneration.


Subject(s)
Aging/metabolism , Dopamine/metabolism , Mesencephalon/metabolism , Nerve Degeneration/metabolism , Ubiquitin-Protein Ligases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aging/pathology , Animals , Child , Child, Preschool , Female , Humans , Male , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Degeneration/pathology , Oxidation-Reduction , Young Adult
16.
Int J Biochem Cell Biol ; 131: 105904, 2021 02.
Article in English | MEDLINE | ID: mdl-33359085

ABSTRACT

Nitric Oxide is a very well known gaseous second messenger molecule and vasorelaxant agent involved in a variety of signaling in the body such as neurotransmission, ion channel modulation, and inflammation modulation. However, it's reversible covalent attachment to thiol groups of cysteine residues under nitrosative stress leading to aberrant protein S-nitrosylation (PSNO) has been reported in several pathological conditions in the body stemming from neurodegenerative diseases, cancer, cardiovascular system, and immune system disorders. In the cell, PSNOs are partly unstable and transit to a more stable disulfide state serving as an intermediate step towards disulfide formation thus eliciting the biological response. Scientists have identified several cellular thiol-dependent disulfide reductases that have the intrinsic capability to reverse the modification by reducing the stable disulfides formed in PSNOs and thereby rescue S-nitrosylation-induced altered proteins. The physiological roles of these major cellular ubiquitous S-denitrosylases and their probable implementations have not been fully explored. Gaining knowledge from current research and development this review provides a deeper insight into understanding the interplay and role of the major ubiquitous S-denitrosylases in maintaining cellular redox homeostasis. This review umbrellas the mechanism of Thioredoxin, TRP14, and Glutaredoxin systems and highlights their substrates specificities at different cellular conditions, physiological roles, and importance in diseased conditions that would allow researchers to investigate effective therapeutic interventions for nitrosative stress-related diseases and disorders.


Subject(s)
Cardiovascular Diseases/enzymology , Glutaredoxins/metabolism , Immune System Diseases/enzymology , Neoplasms/enzymology , Neurodegenerative Diseases/enzymology , Thioredoxins/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cysteine/metabolism , Gene Expression Regulation , Glutaredoxins/genetics , Humans , Immune System Diseases/genetics , Immune System Diseases/pathology , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nitric Oxide/metabolism , Nitrosation , Nitrosative Stress/genetics , Oxidation-Reduction , S-Nitrosothiols/metabolism , Signal Transduction , Substrate Specificity , Thioredoxins/genetics
17.
J Biol Chem ; 294(34): 12708-12716, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31266802

ABSTRACT

Deoxyribonucleotides are DNA building blocks and are produced de novo by reduction of ribose to deoxyribose. This reduction is catalyzed by ribonucleotide reductase (RNR), a heterodimeric tetramer enzyme in mammalian cells, having one of two free radical-containing subunits called R2 and p53R2. R2 is S-phase specific and used for DNA replication, whereas p53R2 functions in DNA repair and mitochondrial DNA synthesis. The larger RNR subunit, R1, has catalytically active cysteine thiols in its buried active site and a C-terminal swinging arm, with a Cys-Leu-Met-Cys sequence suggested to act as a shuttle dithiol/disulfide for electron transport. After each catalytic cycle the active site contains a disulfide, which has to be reduced for turnover. Thioredoxin (Trx) and glutaredoxin (Grx) systems have been implicated as electron donors for the RNR disulfide reduction via the swinging arm. Using mouse R1-R2 and R1-p53R2 complexes, we found here that the catalytic efficiency of the GSH-Grx system is 4-6 times higher than that of the Trx1 system. For both complexes, the Vmax values for Grx are strongly depended on GSH concentrations. The GSH disulfide resulting from the Grx reaction was reduced by NADPH and GSH reductase and this enzyme was essential because reaction with GSH alone yielded only little activity. These results indicate that C-terminal shuttle dithiols of mammalian R1 have a crucial catalytic role and that the GSH-Grx system favors the R1-p53R2 enzyme for DNA replication in hypoxic conditions, mitochondrial DNA synthesis, and in DNA repair outside the S-phase.


Subject(s)
Electrons , Glutaredoxins/metabolism , Glutathione/metabolism , Ribonucleotide Reductases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Mice , Models, Molecular , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
18.
FEBS Lett ; 593(14): 1799-1806, 2019 07.
Article in English | MEDLINE | ID: mdl-31125428

ABSTRACT

Glutaredoxins (Grx) are involved in many reactions including defense against oxidative stress. However, the role of the Grx system under nitrosative stress has barely been investigated. In this study, we found that human Grxs denitrosylated both low and high molecular weight S-nitrosothiols. Some S-nitrosylated proteins, stable in the presence of a physiological concentration of glutathione (GSH), were denitrosylated by Grxs. Caspase 3 and cathepsin B were identified as substrates of Grx1-catalysed denitrosylation. In addition, mono-thiol Grxs, such as Grx5, exhibited denitrosylase activity coupled with GSH via a monothiol mechanism. Our study demonstrates the ability of Grxs to act as S-denitrosylases and pinpoint a new mechanism for denitrosylation.


Subject(s)
Glutaredoxins/metabolism , Oxygenases/metabolism , Caspase 3/metabolism , Cathepsin B/metabolism , HEK293 Cells , Homeostasis , Humans , Nitric Oxide/metabolism , Protein Isoforms/metabolism , Signal Transduction
19.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27226571

ABSTRACT

The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-ß was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-ß production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-ß production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.


Subject(s)
Hepatocytes/immunology , Hepatocytes/metabolism , Interferon-beta/biosynthesis , Nitric Oxide Synthase Type II/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/pharmacology , Toll-Like Receptor 3/metabolism , eIF-2 Kinase/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Hepatocytes/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Phosphorylation/drug effects , Poly I-C/pharmacology , RNA Interference , Signal Transduction/drug effects , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Tyrosine/chemistry , Up-Regulation/drug effects , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
20.
Proc Natl Acad Sci U S A ; 111(19): 6964-9, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778250

ABSTRACT

Thioredoxin-related protein of 14 kDa (TRP14, also called TXNDC17 for thioredoxin domain containing 17, or TXNL5 for thioredoxin-like 5) is an evolutionarily well-conserved member of the thioredoxin (Trx)-fold protein family that lacks activity with classical Trx1 substrates. However, we discovered here that human TRP14 has a high enzymatic activity in reduction of l-cystine, where the catalytic efficiency (2,217 min(-1)⋅µM(-1)) coupled to Trx reductase 1 (TrxR1) using NADPH was fivefold higher compared with Trx1 (418 min(-1)⋅µM(-1)). Moreover, the l-cystine reduction with TRP14 was in contrast to that of Trx1 fully maintained in the presence of a protein disulfide substrate of Trx1 such as insulin, suggesting that TRP14 is a more dedicated l-cystine reductase compared with Trx1. We also found that TRP14 is an efficient S-denitrosylase with similar efficiency as Trx1 in catalyzing TrxR1-dependent denitrosylation of S-nitrosylated glutathione or of HEK293 cell-derived S-nitrosoproteins. Consequently, nitrosylated and thereby inactivated caspase 3 or cathepsin B could be reactivated through either Trx1- or TRP14-catalyzed denitrosylation reactions. TRP14 was also, in contrast to Trx1, completely resistant to inactivation by high concentrations of hydrogen peroxide. The oxidoreductase activities of TRP14 thereby complement those of Trx1 and must therefore be considered for the full understanding of enzymatic control of cellular thiols and nitrosothiols.


Subject(s)
Cystine/metabolism , Oxidative Stress/physiology , Thioredoxin Reductase 1/metabolism , Thioredoxins/metabolism , Carcinoma, Squamous Cell , Cysteine/metabolism , Enzyme Activation/physiology , Glutathione/metabolism , HEK293 Cells , HT29 Cells , Humans , Hydrogen Peroxide/pharmacology , Lung Neoplasms , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADP/metabolism , Nitric Oxide/metabolism , Oxidants/pharmacology , Oxidation-Reduction , Substrate Specificity , Sulfur/metabolism , Thioredoxin Reductase 1/genetics , Thioredoxins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...