Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Dennis R. Petersen is active.

Publication


Featured researches published by Dennis R. Petersen.


Chemico-Biological Interactions | 2000

Role of aldehyde dehydrogenases in endogenous and xenobiotic metabolism

Vasilis Vasiliou; Aglaia Pappa; Dennis R. Petersen

Aldehydes are highly reactive molecules that are intermediates or products involved in a broad spectrum of physiologic, biologic and pharmacologic processes. Aldehydes are generated from chemically diverse endogenous and exogenous precursors and aldehyde-mediated effects vary from homeostatic and therapeutic to cytotoxic, and genotoxic. One of the most important pathways for aldehyde metabolism is their oxidation to carboxylic acids by aldehyde dehydrogenases (ALDHs). Oxidation of the carbonyl functional group is considered a general detoxification process in that polymorphisms of several human ALDHs are associated a disease phenotypes or pathophysiologies. However, a number of ALDH-mediated oxidation form products that are known to possess significant biologic, therapeutic and/or toxic activities. These include the retinoic acid, an important element for vertebrate development, gamma-aminobutyric acid (GABA), an important neurotransmitter, and trichloroacetic acid, a potential toxicant. This review summarizes the ALDHs with an emphasis on catalytic properties and xenobiotic substrates of these enzymes.


Pharmacological Reviews | 2012

Aldehyde Dehydrogenase Inhibitors: a Comprehensive Review of the Pharmacology, Mechanism of Action, Substrate Specificity, and Clinical Application

Vindhya Koppaka; David C. Thompson; Ying Chen; Manuel Ellermann; K. C. Nicolaou; Risto O. Juvonen; Dennis R. Petersen; Richard A. Deitrich; Thomas D. Hurley; Vasilis Vasiliou

Aldehyde dehydrogenases (ALDHs) belong to a superfamily of enzymes that play a key role in the metabolism of aldehydes of both endogenous and exogenous derivation. The human ALDH superfamily comprises 19 isozymes that possess important physiological and toxicological functions. The ALDH1A subfamily plays a pivotal role in embryogenesis and development by mediating retinoic acid signaling. ALDH2, as a key enzyme that oxidizes acetaldehyde, is crucial for alcohol metabolism. ALDH1A1 and ALDH3A1 are lens and corneal crystallins, which are essential elements of the cellular defense mechanism against ultraviolet radiation-induced damage in ocular tissues. Many ALDH isozymes are important in oxidizing reactive aldehydes derived from lipid peroxidation and thereby help maintain cellular homeostasis. Increased expression and activity of ALDH isozymes have been reported in various human cancers and are associated with cancer relapse. As a direct consequence of their significant physiological and toxicological roles, inhibitors of the ALDH enzymes have been developed to treat human diseases. This review summarizes known ALDH inhibitors, their mechanisms of action, isozyme selectivity, potency, and clinical uses. The purpose of this review is to 1) establish the current status of pharmacological inhibition of the ALDHs, 2) provide a rationale for the continued development of ALDH isozyme-selective inhibitors, and 3) identify the challenges and potential therapeutic rewards associated with the creation of such agents.


Chemical Research in Toxicology | 2009

Molecular Mechanisms of 4-Hydroxy-2-nonenal and Acrolein Toxicity: Nucleophilic Targets and Adduct Formation

Richard M. LoPachin; Terrence Gavin; Dennis R. Petersen; David S. Barber

Acrolein and 4-hydroxy-2-nonenal (HNE) are byproducts of lipid peroxidation and are thought to play central roles in various traumatic injuries and disease states that involve cellular oxidative stress, for example, spinal cord trauma, diabetes, and Alzheimers disease. In this review, we will discuss the chemical attributes of acrolein and HNE that determine their toxicities. Specifically, these aldehydes are classified as type 2 alkenes and are characterized by an alpha,beta-unsaturated carbonyl structure. This structure is a conjugated system that contains mobile pi-electrons. The carbonyl oxygen atom is electronegative and can promote the withdrawal of mobile electron density from the beta-carbon atom causing regional electron deficiency. On the basis of this type of electron polarizability, both acrolein and HNE are considered to be soft electrophiles that preferentially form 1,4-Michael type adducts with soft nucleophiles. Proteomic, quantum mechanical, and kinetic data will be presented, indicating that cysteine sulfhydryl groups are the primary soft nucleophilic targets of acrolein and HNE. This is in contrast to nitrogen groups on harder biological nucleophiles such as lysine or histidine residues. The toxicological outcome of adduct formation is not only dependent upon residue selectivity but also the importance of the targeted amino acid in protein function or structure. In attempting to discern the toxicological significance of a given adduct, we will consider the normal roles of cysteine, lysine, and histidine residues in proteins and the relative merits of corresponding adducts in the manifestations of diseases or toxic states. Understanding the molecular actions of acrolein and HNE could provide insight into many pathogenic conditions that involve initial cellular oxidative stress and could, thereby, offer new efficacious avenues of pharmacological defense.


Human Genomics | 2011

The human fatty acid-binding protein family: Evolutionary divergences and functions

Rebecca L. Smathers; Dennis R. Petersen

Fatty acid-binding proteins (FABPs) are members of the intracellular lipid-binding protein (iLBP) family and are involved in reversibly binding intracellular hydrophobic ligands and trafficking them throughout cellular compartments, including the peroxisomes, mitochondria, endoplasmic reticulum and nucleus. FABPs are small, structurally conserved cytosolic proteins consisting of a water-filled, interior-binding pocket surrounded by ten anti-parallel beta sheets, forming a beta barrel. At the superior surface, two alpha-helices cap the pocket and are thought to regulate binding. FABPs have broad specificity, including the ability to bind long-chain (C16-C20) fatty acids, eicosanoids, bile salts and peroxisome proliferators. FABPs demonstrate strong evolutionary conservation and are present in a spectrum of species including Drosophila melanogaster, Caenorhabditis elegans, mouse and human. The human genome consists of nine putatively functional protein-coding FABP genes. The most recently identified family member, FABP12, has been less studied.


Toxicology and Applied Pharmacology | 1987

The oxidation of α-β unsaturated aldehydic products of lipid peroxidation by rat liver aldehyde dehydrogenases☆

David Y. Mitchell; Dennis R. Petersen

Lipid peroxidation of microsomal membranes produces a large number of aldehydes, alcohols, and ketones, of which some are cytotoxic. trans-4-Hydroxy-2-nonenal (4HN) and trans-2-hexenal (HX) are two α-β unsaturated aldehydes which are major and minor lipid peroxidation products, respectively. The role of aldehyde dehydrogenase (ALDH) in the oxidation of 4HN and HX was examined using semipurified mitochondrial, cytosolic, and microsomal ALDH isozymes prepared from male Sprague-Dawley rat liver. High- and low-affinity mitochondrial and high-affinity cytosolic ALDH isozymes were able to oxidize 4HN. The affinities of the three isozymes for 4HN, reported as the VK values, are 0.258, 0.032 and 0.030 nmol NADH formed/min/mg protein/μmol 4HN/liter, respectively. The low-affinity cytosolic and microsomal forms of ALDH are unable to oxidize 4HN. The high-affinity mitochondrial, low-affinity cytosolic, and microsomal ALDH isozymes oxidized HX, displaying VK values of 0.600, 0.058, and 0.058 nmol NADH formed/min/mg protein/μmol HX/liter, respectively. Oxidation of HX by the low-affinity mitochondrial and high-affinity cytosolic isozyme was not detected. This study indicates that ALDH may participate in the in vivo metabolism of cytotoxic aldehydic products formed during lipid peroxidation.


Chemico-Biological Interactions | 2003

Covalent adduction of nucleophilic amino acids by 4-hydroxynonenal and 4-oxononenal

Jonathan A. Doorn; Dennis R. Petersen

Peroxidation of polyunsaturated fatty acids yields the lipid aldehydes 4-hydroxynonenal (4HNE) and 4-oxononenal (4ONE). Adduction of proteins by 4HNE is thought to be involved in the pathogenesis of several diseases. At the present time, the reactivity of 4ONE toward proteins is unknown. The purpose of this study was to identify amino acids that react with 4HNE and 4ONE, characterize the chemical structure of the adduct, and determine the preference for amino acid modification. Model peptides containing one or more nucleophilic residues (i.e. Arg, Cys, His, Met and Lys) were reacted with 4HNE and 4ONE at pH 7.4, 37 degrees C and analyzed using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-TOF-MS). Post-source decay (PSD) analysis was used to confirm peptide modification. The bimolecular rate constant for adduction of amino acids and peptides by 4HNE and 4ONE was measured. Results of this work indicate that Cys, His and Lys are modified by 4HNE. In contrast, 4ONE was found to react with Arg, Cys, His and Lys. The predominant adduct resulting from modification of peptides by 4HNE or 4ONE had a mass of 156 or 154 Da (respectively), indicating that both lipid aldehydes react primarily via Michael addition with peptide nucleophiles to yield a covalent adduct. Reactivity of amino acids toward 4HNE was found to have the following order of potency: Cys>>His>Lys. Preference for the reaction of amino acid nucleophiles with 4ONE was determined to have the following order: Cys>>His>Lys>Arg. The presence of an Arg on a Cys-containing peptide increased the reaction rate with 4HNE and 4ONE by a factor of approximately 5-6 compared to the Cys nucleophile alone. Rate constants for the modification of Cys by 4HNE and 4ONE were determined to be 1.21 and 186 M(-1) s(-1) (respectively), indicating a >150-fold difference in reactivity between the lipid aldehydes toward Cys. Spontaneous conjugation of glutathione (GSH) with the lipid aldehydes was found to occur with rate constants of 1.33 and 145 M(-1) s(-1) for 4HNE and 4ONE (respectively), demonstrating a 110-fold difference in the rate of GSH modification between the two compounds. Results of the present study indicate that both 4HNE and 4ONE react with amino acid nucleophiles via Michael addition with the following order of potency: Cys>>His>Lys. However, the reactivity of these lipid aldehydes toward amino acid nucleophiles differs qualitatively with Arg being a target for 4ONE but not 4HNE and quantitatively by a remarkable >100-fold difference in the rate of Cys modification between 4HNE and 4ONE.


Chemico-Biological Interactions | 2011

Overview of lipid peroxidation products and hepatic protein modification in alcoholic liver disease

Rebecca L. Smathers; James J. Galligan; Benjamin J. Stewart; Dennis R. Petersen

OBJECTIVES Oxidative stress is one component of alcoholic liver disease (ALD) that is manifested in the peroxidation of cellular lipids producing the electrophile, 4-hydroxynonenal (4-HNE). This electrophile is proposed to modify essential cellular proteins resulting in loss of protein function and cellular homeostasis. Studies were initiated to identify hepatic proteins that are targets of 4-HNE modification and determine their relationship with progression of the early stages of ALD. METHODS Rat and mouse models were developed using the Lieber-DeCarli diet to simulate early stages of ALD consisting of fatty liver (steatosis) and hepatocellular injury indicated by a 1.5-2-fold elevation of plasma ALT activity. Liver samples obtained from control and ethanol treated animals were subjected to two-dimensional electrophoresis and immunoblotting using polyclonal antibodies generated against 4-HNE epitopes for detection of proteins modified by 4-HNE. Following identification of 4-HNE adducted proteins, the respective recombinant proteins modified with physiologic concentrations of 4-HNE were evaluated to determine the functional consequences of 4-HNE modification. RESULTS One group of proteins identified included Hsp70, Hsp90 and protein disulfide isomerase (PDI), all of which are involved in protein folding or processing are targets of adduction. In vitro assays indicated significant impairment of the protein activities following modification with physiologically relevant concentrations of 4-HNE. Liver fatty acid binding protein, L-FABP, was also identified as a target and additional studies revealed that the levels of this protein were significantly decreased because of chronic ethanol ingestion. Erk1/2 was identified as a target for modification and subsequently determined to have impaired activity. CONCLUSIONS Inhibition of Hsp70, Hsp90 and PDI function could be involved in initiation of the early phases of ER stress contributing to stimulation and accumulation of hepatic lipids. Likewise, impairment of L-FABP activity could also disrupt lipid transport also contributing to steatosis. The modification and inhibition of Erk1/2 by 4-HNE may also contribute to the decreased hepatocellular proliferation associated with ALD. Collectively, these results provide new information concerning the mechanisms whereby the modification of hepatic proteins by 4-HNE contributes to ALD.


Journal of Proteome Research | 2012

Mitochondrial Acetylome Analysis in a Mouse Model of Alcohol-Induced Liver Injury Utilizing SIRT3 Knockout Mice

Kristofer S. Fritz; James J. Galligan; Matthew D. Hirschey; Eric Verdin; Dennis R. Petersen

Mitochondrial protein hyperacetylation is a known consequence of sustained ethanol consumption and has been proposed to play a role in the pathogenesis of alcoholic liver disease (ALD). The mechanisms underlying this altered acetylome, however, remain unknown. The mitochondrial deacetylase sirtuin 3 (SIRT3) is reported to be the major regulator of mitochondrial protein deacetylation and remains a central focus for studies on protein acetylation. To investigate the mechanisms underlying ethanol-induced mitochondrial acetylation, we employed a model for ALD in both wild-type (WT) and SIRT3 knockout (KO) mice using a proteomics and bioinformatics approach. Here, WT and SIRT3 KO groups were compared in a mouse model of chronic ethanol consumption, revealing pathways relevant to ALD, including lipid and fatty acid metabolism, antioxidant response, amino acid biosynthesis and the electron-transport chain, each displaying proteins with altered acetylation. Interestingly, protein hyperacetylation resulting from ethanol consumption and SIRT3 ablation suggests ethanol-induced hyperacetylation targets numerous biological processes within the mitochondria, the majority of which are known to be acetylated through SIRT3-dependent mechanisms. These findings reveal overall increases in 91 mitochondrial targets for protein acetylation, identifying numerous critical metabolic and antioxidant pathways associated with ALD, suggesting an important role for mitochondrial protein acetylation in the pathogenesis of ALD.


Chemical Research in Toxicology | 2011

4-Hydroxynonenal Inhibits SIRT3 via Thiol-Specific Modification

Kristofer S. Fritz; James J. Galligan; Rebecca L. Smathers; James R. Roede; Colin T. Shearn; Philip Reigan; Dennis R. Petersen

4-Hydroxynonenal (4-HNE) is an endogenous product of lipid peroxidation known to play a role in cellular signaling through protein modification and is a major precursor for protein carbonyl adducts found in alcoholic liver disease (ALD). In the present study, a greater than 2-fold increase in protein carbonylation of sirtuin 3 (SIRT3), a mitochondrial class III histone deacetylase, is reported in liver mitochondrial extracts of ethanol-consuming mice. The consequence of this in vivo carbonylation on SIRT3 deacetylase activity is unknown. Interestingly, mitochondrial protein hyperacetylation was observed in a time-dependent increase in a model of chronic ethanol consumption; however, the underlying mechanisms for this remain unknown. Tandem mass spectrometry was used to identify and characterize the in vitro covalent modification of rSIRT3 by 4-HNE at Cys(280), a critical zinc-binding residue, and the resulting inhibition of rSIRT3 activity via pathophysiologically relevant concentrations of 4-HNE. Computational-based molecular modeling simulations indicate that 4-HNE modification alters the conformation of the zinc-binding domain inducing minor changes within the active site, resulting in the allosteric inhibition of SIRT3 activity. These conformational data are supported by the calculated binding energies derived from molecular docking studies suggesting the substrate peptide of acetyl-CoA synthetase 2 (AceCS2-K(ac)) and display a greater affinity for native SIRT3 as compared with the 4-HNE adducted protein. The results of this study characterize altered mitochondrial protein acetylation in a mouse model of chronic ethanol ingestion and thiol-specific allosteric inhibition of rSIRT3 resulting from 4-HNE adduction.


Biochemical Pharmacology | 1991

Lipid aldehyde oxidation as a physiological role for class 3 aldehyde dehydrogenases

Ronald Lindahl; Dennis R. Petersen

A large number of different unsaturated, saturated and hydroxylated aliphatic aldehydes can be generated during the peroxidation of cellular lipids. This study examined the kinetic properties of purified Class 3 rat aldehyde dehydrogenase (ALDH) with respect to the oxidation of various lipid aldehyde substrates. It also compared the substrate preference of the prototypic Class 3 ALDH with that of the constitutive rat microsomal aldehyde dehydrogenase. The results suggest that (1) microsomal ALDH is a member of the Class 3 aldehyde dehydrogenase family, and (2) the physiological role of the Class 3 ALDHs, including the microsomal form, is the oxidation of medium (6 to 9 carbon) chain length saturated and unsaturated aldehydes generated by the peroxidation of cellular lipids. Short chain aliphatic aldehydes, such as a malondialdehyde and 4-hydroxyalkenals, are not substrates for the Class 3 aldehyde dehydrogenases.

Collaboration


Dive into the Dennis R. Petersen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James J. Galligan

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca L. Smathers

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Reigan

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Richard A. Deitrich

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Benjamin J. Stewart

Lawrence Livermore National Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge