Nitrated fatty acids: from diet to disease

https://doi.org/10.1016/j.cophys.2019.04.013Get rights and content

Highlights

  • Formation of nitro-fatty acids.

  • Signaling of nitro-fatty acids.

  • Anti-fibrotic effects of nitro-fatty acids.

Fatty acids not only provide caloric energy in our diets and building blocks of lipids but are also precursors of potent signaling molecules. Fatty acids can undergo enzymatic and non-enzymatic transformations to form autocrine and paracrine signaling molecules that regulate energy balance and metabolic homeostasis. A new class of lipid signaling mediators known as nitro-fatty acids (NO2-FAs) have recently been identified. These NO2-FAs are generated endogenously through non-enzymatic reactions of secondary products of nitrite and nitric oxide and are readily detected in human plasma and urine. NO2-FAs are potent anti-inflammatory and antioxidant cell signaling mediators and exert protective effects in numerous pre-clinical animal models of disease including cardiovascular, pulmonary and renal fibrosis. Chronic unresolved inflammation is a common key feature underlying most fibrotic disorders. Two signaling pathways that converge on inflammation and oxidative stress are nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and nuclear factor kappa B (NF-κB). NO2-FAs are pleiotropic signaling modulators that target both of these pathways providing a therapeutic strategy directed toward an integrated decrease in inflammation. This review summarizes the latest findings and understanding of the formation, signaling and anti-fibrotic effects of NO2-FA.

Introduction

The need for fatty acids goes far beyond the obligatory role of building blocks for fat and dietary sources for fuel in our bodies. The discovery of leukotrienes and prostaglandins, as enzymatic oxygenation products of unsaturated fatty acids, advanced the field demonstrating that fatty acid mediators are potent autocrine and paracrine signaling molecules. The technical advances provided by mass spectrometry-based approaches led to renewed interest and discoveries in this area of research. It has become clear that this large group of newly discovered bioactive molecules derived from polyunsaturated fatty acids executes physiological responses that are central to tissue homeostasis. Among them, NO2-FAs are formed that participate in anti-inflammatory and tissue protective actions through activation of Nrf2-dependent-signaling and concomitant inhibition of NF-κB-derived inflammation. The latest advances are provided in areas of NO2-FA formation and role in fibrotic disorders with a focus on unresolved inflammation, which appears to be a common feature of fibrosis across all tissues, and examining the signaling actions of NO2-FAs as a novel therapeutic.

Section snippets

Formation of nitro-fatty acids

Dietary interventions have been shown to be effective in a broad range of pathologies including diabetes and metabolic syndrome [1]. In addition to providing the nutrients to support metabolism, certain dietary components modulate metabolic processes. Moreover, these can be further metabolized and processed by the microbiome, providing a new set of modified molecules with unique signaling properties that regulate metabolism. These microbial transformations are important [2,3], the microbiome

Stomach as a bioreactor

The acidic conditions of the stomach during digestion result in its protonation and the secondary generation of nitric oxide (NO) through reductive reactions, the nitrosating species dinitrogen trioxide (N2O3) and nitrating species nitrogen dioxide (radical dotNO2) [9]. Initial studies evaluating the intake effects of high doses of nitrite demonstrated the formation of these reactive species in the stomach to be responsible for the formation of tumorigenic nitrosamines [10], prompting stringent standards

Endogenous NO2-FA

While NO2-CLA detection is well established and its levels are modulated by diet and inflammation, the remaining classes of NO2-FAs still need to be explored [13••,14]. Nitro-oleic acid (NO2-OA), which has long been used as a surrogate to investigate the actions of endogenous NO2-FA [15], has been reported in the subnanomolar level in plasma [16] but is usually below the limit of detection and/or quantification. In contrast to NO2-CLA, NO2-OA and its metabolites are normally not detected in

Nitro-fatty acid reactivity

The interest in the physiology and pharmacology of NO2-FA is fueled by the investigation of their anti-inflammatory and protective actions reported in several preclinical animal models of disease by NO2-OA, which is used as a surrogate to interrogate the effects of endogenous NO2-FA [15,18]. NO2-FAs are electrophilic molecules that contain a nitroalkene group that reacts mainly with cysteines through Michael addition reactions [19]. While cysteine adducts are the most relevant to NO2-FA

Nitro-fatty acid signaling

The reversibility of the NO2-FA reaction is central to their pleiotropic signaling activity. While initially nuclear factor (erythroid-derived 2)-like 2 (Nrf2), heat shock response (HSR) activation and nuclear factor kappa B (NF-κB) inhibition were proposed as main drivers of their signaling mechanisms, emerging evidence reveals new pathways that are inhibited, specifically STING, epoxide hydrolase and angiotensin II receptor [23, 24, 25]. The current understanding of the signaling of NO2-FA

Complexities of fibrosis

Fibrosis is a complex and vital program to repair injured tissue. It occurs after repetitive insults to the epithelium and is defined by the accumulation of extracellular matrix (ECM) molecules such as collagen and fibronectin [30]. Under normal conditions that lead to wound healing following an injury, the fibrotic ECM is degraded, the epithelium is repaired and fibrosis is resolved. In a fibrotic state, however, the normal repair and resolution mechanisms are dysfunctional leading to scarring

The need for novel therapeutics: NO2-FA

Electrophilic drugs inhibit pro-inflammatory signaling mediators and have been shown to provide a valuable approach in several pre-clinical fibrosis models. NO2-OA and other electrophiles, such as dimethyl fumarate and 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO), protect against kidney and pulmonary animal models of fibrosis [32, 33, 34, 35]. In cardiovascular disease, NO2-OA reverses hypoxia-induced right ventricular (RV) pressure and fibrotic RV remodeling in a pulmonary arterial

NF-κB inhibition

Under basal conditions, NF-κB is inactive as it is complexed with inhibitory κB (IκB)-α subunit, which contains a strong nuclear export signal, in the cytoplasm. In the canonical pathway, most stress signals activate NF-κB by controlling the redox-sensitive serine-specific IkB kinase (IKK). The activation of the IKK complex facilitates IκB-α phosphorylation (Ser-32 and Ser-36) by IκB kinase β (IKK-β) leading to IκB-α ubiquitination and proteasomal degradation resulting in NF-κB transactivation [

Nrf2 activation

NO2-FA derivatives activate Nrf2-dependent gene transcription by alkylating two functionally significant cysteine residues (Cys-273 and Cys-288) of Kelch-like ECH-associated protein (Keap)-1 in the cytoplasm. This inhibits Keap1-dependent Nrf2 degradation and results in nuclear translocation of de novo synthesized Nrf2 protein, binding to antioxidant response element and transactivation of gene signaling [41,42]. Nrf2-regulated genes are critical in protecting against oxidative stress, by

Crosstalk between Nrf2 and NF-κB pathways

Not only does Nrf2 activate antioxidant gene expression but Nrf2 directly limits inflammation by binding to promoter regions of pro-inflammatory cytokines [46]. Additionally, Nrf2 inhibits RNA polymerase II recruitment to the transcription start site of IL-6 and IL-1β genes without altering the required recruitment of NF-κB [46]. In this case, Nrf2 and NF-κB signaling appear to be acting individually. However, there is considerable crosstalk between these two redox-regulated transcription

Summary and perspective

Unresolved inflammation is the backbone of many diseases, especially during the progression to fibrosis. Electrophilic NO2-FAs have been proven safe in Phase I clinical trials and provide new dietary and pharmacological approaches to target inflammatory and fibrotic disorders by reversibly regulating signaling pathways. The decrease of inflammation and oxidative stress through Nrf2 and NF-κB pathways provides the conditions to return to homeostasis. The use of electrophiles is not free of

Conflict of interest statement

F.J.S. has financial interest in Complexa Inc. N.K.H.K. has no conflicts of interest to declare.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We would like to thank Ms. Sonia Salvatore for input and graphic design. This work was supported by R01-GM125944 and R01-DK112854 (F.J.S), AHA17GRN33660955 (F.J.S), P01-HL103455 (N.K.H.K.) and University of Pittsburgh Medical Center Competitive Medical Research Fund Award (N.K.H.K.).

References (52)

  • C. Batthyany et al.

    Reversible post-translational modification of proteins by nitrated fatty acids in vivo

    J Biol Chem

    (2006)
  • A.L. Hansen et al.

    Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling

    Proc Natl Acad Sci U S A

    (2018)
  • R.L. Charles et al.

    Protection from hypertension in mice by the Mediterranean diet is mediated by nitro fatty acid inhibition of soluble epoxide hydrolase

    Proc Natl Acad Sci U S A

    (2014)
  • L. di Nuzzo et al.

    Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis

    Drug Des Devel Ther

    (2014)
  • S. Liu et al.

    Nitro-oleic acid protects against adriamycin-induced nephropathy in mice

    Am J Physiol Renal Physiol

    (2013)
  • L. Villacorta et al.

    Electrophilic nitro-fatty acids inhibit vascular inflammation by disrupting LPS-dependent TLR4 signalling in lipid rafts

    Cardiovasc Res

    (2013)
  • W. Gong et al.

    CKIP-1 affects the polyubiquitination of Nrf2 and Keap1 via mediating Smurf1 to resist HG-induced renal fibrosis in GMCs and diabetic mice kidneys

    Free Radic Biol Med

    (2018)
  • E.H. Kobayashi et al.

    Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription

    Nat Commun

    (2016)
  • A. Cuadrado et al.

    Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases

    Nat Rev Drug Discov

    (2019)
  • Joanna D. Wardyn et al.

    Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways

    Biochem Soc Trans

    (2015)
  • H.M. Roager et al.

    Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: a randomised cross-over trial

    Gut

    (2019)
  • Y. Heianza et al.

    Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: the POUNDS lost trial

    Gut

    (2019)
  • E.E. Canfora et al.

    Gut microbial metabolites in obesity, NAFLD and T2DM

    Nat Rev Endocrinol

    (2019)
  • B.S. Rocha et al.

    Role of nitrite, urate and pepsin in the gastroprotective effects of saliva

    Redox Biol

    (2016)
  • G. Fuke et al.

    Systematic evaluation on the effectiveness of conjugated linoleic acid in human health

    Crit Rev Food Sci Nutr

    (2017)
  • J.O. Lundberg et al.

    Metabolic effects of dietary nitrate in health and disease

    Cell Metab

    (2018)
  • Cited by (19)

    • Nitro-fatty acids as novel Virgin olive oil quality markers

      2023, Journal of Food Composition and Analysis
    View all citing articles on Scopus
    View full text