How Seed Oils Destroy Your Mitochondria and Lead To Chronic Disease, with Tucker Goodrich

Tucker Goodrich is a technology executive in the financial industry who designs, runs, and debugs complex systems in high-risk environments. Areas of expertise include risk management, systems management, and cyber-security. 

 

After experiencing some personal health crises and realizing that the ‘solutions’ offered by medical professionals weren’t working or addressing causation he started applying the same approach in research and evaluation of data to his own health issues to determine root causes. 

 

His interests have focused on dietary and environmental drivers of chronic disease, including carbohydrate, wheat, and various classes of fats. Specifically, he's attempting to understand and popularize understanding of the mechanisms driving the diet-derived explosion in so-called chronic diseases (or diseases of civilization). He is active on twitter (@tuckergoodrich, has a blog called Yelling Stop, is an Expert Advisor for the nutrition start-up Nutrita, and has been a guest on numerous podcasts.





Time Stamps:

Podcast Youtube

0:09:38   0:03:21   Podcast Begins

0:11:12   0:04:55   What is Linoleic Acid?

0:16:44   0:10:27   How Linoleic acid caused disease

0:21:06   0:14:49   Understanding cardiolipin and the electron transport chain

0:23:28   0:17:11   Linoleic acid drives mitochondrial dysfunction, causes chronic illness

0:26:41   0:20:24   Maintenance of Cardiolipin and Crista Structure Requires Cooperative Functions of Mitochondrial Dynamics and Phospholipid Transport   https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7026740/

0:32:56   0:26:39   Linoleic acid is uniquely damaging

0:35:30   0:29:13   Myths surrounding linoleic acid

0:38:19   0:32:02   The supremacy of animal fats

0:41:05   0:34:48   Brief episode of STZ-induced hyperglycemia produces cardiac abnormalities in rats fed a diet rich in n-6 PUFA   https://journals.physiology.org/doi/full/10.1152/ajpheart.00480.2004?rfr_dat=cr_pub++0pubmed&url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org

0:42:25   0:36:08   How seed oil consumption leads to necrosis and CVD

0:49:38   0:43:21   Mainstream understanding of PUFAs is built on faulty epidemiology

0:56:21   0:50:04   Circulating levels of linoleic acid and HDL-cholesterol are major determinants of 4-hydroxynonenal protein adducts in patients with heart failurehttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC3909262/

0:58:49   0:52:32   All about OxLAMs

1:03:32   0:57:15   Oxidized omega-6s are essential for atherosclerosis 

1:13:33   1:07:16   Lowering dietary linoleic acid reduces bioactive oxidized linoleic acid metabolites in humans   https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3467319/

1:14:30   1:08:13   Strong increase in hydroxy fatty acids derived from linoleic acid in human low density lipoproteins of atherosclerotic patients   https://www.sciencedirect.com/science/article/abs/pii/S0009308497000959?via%3Dihub

1:15:38   1:09:21   The root cause of oxidative stress is linoleic acid

1:18:17   1:12:00   Metabolites of arachidonic acid and linoleic acid in early stages of non-alcoholic fatty liver disease—A pilot study   https://www.sciencedirect.com/science/article/abs/pii/S1098882315300101

1:19:56   1:13:39   How Linoleic acid affects immunity

1:44:03   1:37:46   The linoleic acid-chronic illness hypothesis is air-tight

1:48:14   1:41:57   Calories In, Calories Out is the wrong paradigm

1:51:01   1:44:44   Role of Physiological Levels of 4-Hydroxynonenal on Adipocyte Biology: Implications for Obesity and Metabolic Syndrome   https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4038367/

1:51:53   1:45:36   Eating seed oils is about as smart as eating poison ivy

1:54:54   1:48:37   Tucker Goodrich radical paleo evangelism

1:58:19   1:52:02   Where to find Tucker Goodrich

 

Tucker's Blog Post:

Linoleic acid and its metabolites, a primer

Big topic, steadily getting bigger
https://pubmed.ncbi.nlm.nih.gov/?term=(linoleic)%20NOT%20(conjugated)&sort=pubdate&timeline=expanded

Most studied metabolite is HNE (aka 4-HNE)
https://pubmed.ncbi.nlm.nih.gov/?term=(HNE)%20OR%20(4-HNE)%20NOT%20(human%20neutrophil%20elastase)&sort=pubdate&timeline=expanded

Many others, including 13-HODE, MDA, leukotoxin, ONA, leukotoxin, 2-AG, ad nauseum. Full number not known.
But crucial to a much larger topic, Oxidative Stress:
https://pubmed.ncbi.nlm.nih.gov/?term=(oxidative%20stress)&sort=pubdate&timeline=expanded

But first, a little context and a caveat. Cardiolipin and Essential Fatty Acids

Cardiolipin

Cardiolipin is a molecule that is found in mitochondria in the human body, and in bacteria and chloroplasts.
“Cardiolipin is a phospholipid located exclusively in energy transducing membranes and it was identified in mitochondria, bacteria, hydrogenosomes and chloroplasts. In eukaryotes, cardiolipin is the only lipid that is synthesized in the mitochondria.” (Rosa et al., 2008)

I very much enjoyed the podcast with Peter. His is one of two blogs where I have gone back to the first post and read everything that he has written. Peter and I have different, but complementary, focuses though. He is interested in what is happening in the ETC, I am interested in what happens around that. So I’m just going to posit that everything he says is correct, and talk about what’s going on around the ETC and the functionality he’s discussed.

Cardiolipin is comprised of four fatty acids (unlike a triglyceride, which is made from three). This structure is key to its function, as is demonstrated by Barth’s Syndrome, in which cardiolipin cannot be constructed properly, due to a genetic defect. Peter’s thread you discussed is titled Protons. Cardiolipins are what conducts protons and electrons along the ETC, and, as you discussed the various complexes that make up the ETC, those complexes are bound into functional supercomplexes comprised of cardiolipin. (Hoch, 1992)
The very shape of the mitochondria is determined by cardiolipin.:
“Energy production, a central role of mitochondria, demands highly folded structures of the mitochondrial inner membrane (MIM) called cristae and a dimeric phospholipid (PL) cardiolipin (CL).”

(Kojima et al., 2019)

Cardiolipin fatty acid composition is determined by diet and by cell-type-specific DNA. This is important since cardiolipin composition determines how susceptible the molecule is to oxidative damage
Quick summation of three blog posts: (Goodrich, 2016a, 2016b, 2016c)
Dietary linoleic acid controls cardiolipin composition, linoleic-acid-containing cardiolipin are uniquely susceptible to oxidative damage. Cardiolipin are in contact with cytochrome c, which is an iron-containing molecule. Iron in cytochrome causes adjacent LA molecules in CL to auto oxidize, this can become a self-sustaining reaction, in vitro will continue until all CL is gone. Oxidized CL releases oxylipins like those mentioned above. (Liu et al., 2011) Oxidized CL then becomes a trigger for mitosis and apoptosis.

This paper shows exactly what this process looks like in vivo, in mice. (Ghosh et al., 2004) In my blog post discussing it (Goodrich, 2018) I show the following two images:

The first image shows a mitochondrion that has physically collapsed in the N-6+Hyperglycemia group, and the next shows the near inability of these mice to burn glucose. Apparently Complex I has largely failed, leading to massive necrosis in the heart. This follows from a major loss of cardiolipin after n-6 feeding commences, which was similar in both N-6 and N-6+Hyperglycemia groups.
QED for those posts on cardiolipin above.
Mitochondria are essential to life. Cardiolipin, essential to mitochondria, is also essential to life. N-6 feeding seems to cause cardiolipin to become very fragle…

Essential Fatty Acids

When you read all these papers, you will continuously come across the claim that linoleic acid is an EFA. This is based on studies in rodents, dating back to 1930. (Burr & Burr, 1930)
More careful work recently has determined that LA is not an EFA, in rodents (Carlson et al., 2019) or in humans. (Gura et al., 2005)
So when you are told that you should eat seed oils because they are “essential”, you can snort in derision. The amount of LA in Gura 2005 was tiny, about ½%. Eating a diet based on real food an you will get that much, it’s only possible to become EFA “deficient” under the care of a physician.

Notable metabolites

Oxidized Cardiolipin

Anti-phospholipid syndrome is an auto-immune condition in which the body attacks its own phospholipids, specifically oxidized cardiolipin. (Tuominen Anu et al., 2006) This is an antigen in lupus, atherosclerosis, chronic fatigue syndrome, (Hokama et al., 2008) and fibromyalgia (Gräfe et al., 1999). It’s unclear what the role of oxCL is in these diseases, although as discussed above LA appears to be required for CL to oxidize in large quantities, and it induces it.
Several drugs have been developed to protect cardiolipin from oxidation, and they seem to show benefit in a variety of age-related and chronic diseases. (Chavez et al., 2020; Díaz-Quintana et al., 2020; Skulachev et al., 2010)

Oxidized LDL

OxLDL was demonstrated to be essential to the progression of atherosclerosis in the late 1980s, shortly after the LDL receptor was discovered and it was shown that non-oxidized LDL would not induce macrophages to become foam cells, and that dietary LA induced LDL to be more susceptible to oxidation, while fats such as oleic acid were protective (similar to what has been shown with cardiolipin). (Palinski W et al., 1990; Parthasarathy et al., 1990; Witztum & Steinberg, 1991) OxLDL is a normal part of immune function (Kaplan et al., 2017), but in an industrial diet context it seems to become pathogenic, playing a role in CVD, cancer, T2DM, and the metabolic syndrome. 
OxLDL is an auto-antigen, antibodies for oxLDL are cross-reactive to LPS and Staph.
Treatment of obese rhesus monkeys with an oxLDL antibody reduces insulin resistance and inflammation.
(Crisby et al., 2009; Deleanu et al., 2016; González-Chavarría et al., 2018; Kruit et al., 2010; Marin et al., 2015)

Fig. 5: "Free 4-HNE and total MDA in native low density lipoproteins (nLDL), oxidized low density lipoproteins (oxLDL) and glycated low density lipoproteins (gLDL)." (Deleanu et al., 2016)

(Li et al., 2013)

Leukotoxin (EpOME, (±)9(10)-epoxy-12Z- and (±)12(13)-epoxy-9Z-octadecenoic acid [9(10)- and 12(13)]-EpOME)

Leukotoxin is produced in leukocytes as part of the respiratory burst used as an anti-pathogen strategy. It is derived from linoleic acid, and is responsible for the effects of ARDS and diseases that induce ARDS, like COVID-19 in severe cases. Covered at length in this post (Goodrich, 2020) or (Hildreth et al., 2020). It’s also involved in brown adipose tissue regulation.

ONA (9-ONA, 9-oxononanoic acid)

ONA induces arterial calcification in mice, and appears to also do so in humans. (Riad et al., 2017). “These results indicated that 9-ONA is the primary inducer of PLA2 activity and TxA2 production, and is probably followed by the development of diseases such as thrombus formation.” It also appears to induce platelet aggregation. (Ren et al., 2013)

2-AG (2-arachidonoylglycerol)

An endocannabinoid derived from arachidonic acid (AA) which is derived from dietary LA. Induces over-consumption of carbohydrates and obesity in rodents and humans. (Alvheim et al., 2012; Silvestri & Di Marzo, 2013)
Rimonabant, which was a human-approved anti-obesity drug for a brief time, treated this pathway in humans. “Large randomized trials with rimonabant have demonstrated efficacy in treatment of overweight and obese individuals with weight loss significantly greater than a reduced calorie diet alone. In addition, multiple other cardiometabolic parameters were improved in the treatment groups including increased levels of high density lipoprotein cholesterol, reduced triglycerides, reduced waist circumference, improved insulin sensitivity, decreased insulin levels, and in diabetic patients improvement in glycosylated hemoglobin percentage.” (Bronander & Bloch, 2007)
This phenomenon is the largest issue I have with Peter’s Protons hypothesis, as it seems odd that the endocannabinoid system might counteract the effect he describes, yet it does.

MDA (Malondialdehyde)

“Indeed, oxidation products such as oxidized phosphatidylcholine, MDA, 4-HNE and others have been documented in virtually all inflammatory diseases including atherosclerosis, pulmonary, renal, and liver diseases, as well as diseases affecting the central nervous system like multiple sclerosis and Alzheimer's disease [8–14].” (Weismann & Binder, 2012)
I frankly haven’t looked too closely at MDA for the simple reason that it can be made from n-6 or n-3 fats. Although in practice, it’s from n-6 fats.
MDA is the most-common marker of oxidative stress (OxStr), which is the process of n-6 fats breaking down into toxins, via the rather inaccurate TBARS test. (Specialties, n.d.). It’s also the substance used for oxLDL, via the E06 test. (Yeang et al., 2016)

HNE (4-HNE, 4-Hydroxynonenal, or 4-hydroxy-2-nonenal)

HNE is the most-studied linoleic acid metabolite, since it’s rediscovery by Esterbauer. (Esterbauer et al., 1991). HNE is a major toxic component of oxLDL (see that section) along with MDA. Unlike MDA, HNE is derived exclusively from n-6 fats, linoleic and arachidonic acid, hence is a good tracker of their effects in the body.
HNE is used as a mitochondrial regulator, along with ROS (your discussion w/ Peter didn’t mention that point) (Speijer, 2016), so this is a fundamental part of the body with regular and pathological functions.
If you’ve heard that glutathione (GSH) is an important antioxidant, it’s in part because it protects the body from HNE. Depressed levels of GSH indicate excessive production of HNE, typically from LA. Aldehyde dehydrogenase (ALDH) is also involved in detoxifying HNE, HNE has the unique ability to damage both GSH and ALDH, thus breaking its own regulatory system.
HNE can be produced in the mitochondria from the oxidation of LA-containing cardiolipin (Liu et al., 2011).

Protein damage

HNE damages a significant subset of proteins in the cell (~27%) (Codreanu et al., 2009)
HNE is associated with the major type of DNA damage (Okamoto et al., 1994), which is induced by LA oxylipins (Kanazawa et al., 2016).

DNA damage

HNE induces the major mutation seen in cancer, it damages the TP53 cancer-protection gene:
“P53 is often mutated in solid tumors, in fact, somatic changes involving the gene encoding for p53 (TP53) have been discovered in more than 50% of human malignancies and several data confirmed that p53 mutations represent an early event in cancerogenesis.” (Perri et al., 2016)
“The major lipid peroxidation product, trans-4-hydroxy-2-nonenal, preferentially forms DNA adducts at codon 249 of human p53 gene, a unique mutational hotspot in hepatocellular carcinoma” (Hu et al., 2002)

Lipid damage

"These reactive oxygen species readily attack the polyunsaturated fatty acids of the fatty acid membrane, initiating a self-propagating chain reaction." (Mylonas & Kouretas, 1999)

Alzheimer’s Disease

HNE induces beta-amyloid:
“The present study demonstrates a direct cause-and-effect correlation between oxidative stress and altered amyloid-β production, and provides a molecular mechanism by which naturally occurring product of lipid peroxidation may trigger generation of toxic amyloid-β42 species.” (Arimon et al., 2015)

It breaks pyruvate dehydrogenase. (Hardas et al., 2013; Humphries & Szweda, 1998)

It breaks ATP synthase. (Terni et al., 2010)

8-OHdG (8-oxo-dG , 8-Oxo-2'-deoxyguanosine)

“The biomarker 8-OHdG or 8-oxodG has been a pivotal marker for measuring the effect of endogenous oxidative damage to DNA and as a factor of initiation and promotion of carcinogenesis.” (Valavanidis et al., 2009)

“Linoleic acid hydroperoxides (LOOH) formed 8-oxo-dG at a higher level than H2O2 in guanosine or double-stranded DNA.” (Kanazawa et al., 2016)

13-HODE (

Asthma: “13-S-HODE causes severe airway dysfunction, airway neutrophilia, mitochondrial dysfunction and epithelial injury in naïve mouse…” (Mabalirajan et al., 2013; Panda et al., 2017)

Bibliography

Alvheim, A. R., Malde, M. K., Osei‐Hyiaman, D., Hong, Y. H., Pawlosky, R. J., Madsen, L., Kristiansen, K., Frøyland, L., & Hibbeln, J. R. (2012). Dietary Linoleic Acid Elevates Endogenous 2-AG and Anandamide and Induces Obesity. Obesity, 20(10), 1984–1994. https://doi.org/10.1038/oby.2012.38

Arimon, M., Takeda, S., Post, K. L., Svirsky, S., Hyman, B. T., & Berezovska, O. (2015). Oxidative stress and lipid peroxidation are upstream of amyloid pathology. Neurobiology of Disease, 84, 109–119. https://doi.org/10.1016/j.nbd.2015.06.013

Bronander, K. A., & Bloch, M. J. (2007). Potential role of the endocannabinoid receptor antagonist rimonabant in the management of cardiometabolic risk: A narrative review of available data. Vascular Health and Risk Management, 3(2), 181–190. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1994026/

Burr, G. O., & Burr, M. M. (1930). On the Nature and Rôle of the Fatty Acids Essential in Nutrition. Journal of Biological Chemistry, 86(2), 587–621. http://www.jbc.org/content/86/2/587

Carlson, S. J., O’Loughlin, A. A., Anez-Bustillos, L., Baker, M. A., Andrews, N. A., Gunner, G., Dao, D. T., Pan, A., Nandivada, P., Chang, M., Cowan, E., Mitchell, P. D., Gura, K. M., Fagiolini, M., & Puder, M. (2019). A Diet With Docosahexaenoic and Arachidonic Acids as the Sole Source of Polyunsaturated Fatty Acids Is Sufficient to Support Visual, Cognitive, Motor, and Social Development in Mice. Frontiers in Neuroscience, 13, 72. https://doi.org/10.3389/fnins.2019.00072

Chavez, J. D., Tang, X., Campbell, M. D., Reyes, G., Kramer, P. A., Stuppard, R., Keller, A., Zhang, H., Rabinovitch, P. S., Marcinek, D. J., & Bruce, J. E. (2020). Mitochondrial protein interaction landscape of SS-31. Proceedings of the National Academy of Sciences, 117(26), 15363–15373. https://doi.org/10.1073/pnas.2002250117

Codreanu, S. G., Zhang, B., Sobecki, S. M., Billheimer, D. D., & Liebler, D. C. (2009). Global Analysis of Protein Damage by the Lipid Electrophile 4-Hydroxy-2-nonenal. Molecular & Cellular Proteomics, 8(4), 670–680. https://doi.org/10.1074/mcp.M800070-MCP200

Crisby, M., Kublickiene, K., Henareh, L., & Agewall, S. (2009). Circulating levels of autoantibodies to oxidized low-density lipoprotein and C-reactive protein levels correlate with endothelial function in resistance arteries in men with coronary heart disease. Heart and Vessels, 24(2), 90–95. https://doi.org/10.1007/s00380-008-1089-y

Deleanu, M., Sanda, G. M., Stancu, C. S., Popa, M. E., & Sima, A. V. (2016). Profiles of Fatty Acids and the Main Lipid Peroxidation Products of Human Atherogenic Low Density Lipoproteins. Revista De Chimie, 67(1), 8–12. http://www.revistadechimie.ro/article_eng.asp?ID=4799

Díaz-Quintana, A., Pérez-Mejías, G., Guerra-Castellano, A., De la Rosa, M. A., & Díaz-Moreno, I. (2020). Wheel and Deal in the Mitochondrial Inner Membranes: The Tale of Cytochrome c and Cardiolipin. Oxidative Medicine and Cellular Longevity, 2020, e6813405. https://doi.org/10.1155/2020/6813405

Esterbauer, H., Schaur, R. J., & Zollner, H. (1991). Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radical Biology and Medicine, 11(1), 81–128. https://doi.org/10.1016/0891-5849(91)90192-6

Ghosh, S., Qi, D., An, D., Pulinilkunnil, T., Abrahani, A., Kuo, K.-H., Wambolt, R. B., Allard, M., Innis, S. M., & Rodrigues, B. (2004). Brief episode of STZ-induced hyperglycemia produces cardiac abnormalities in rats fed a diet rich in n-6 PUFA. American Journal of Physiology. Heart and Circulatory Physiology, 287(6), H2518-2527. https://doi.org/10.1152/ajpheart.00480.2004

González-Chavarría, I., Fernandez, E., Gutierrez, N., González-Horta, E. E., Sandoval, F., Cifuentes, P., Castillo, C., Cerro, R., Sanchez, O., & Toledo, J. R. (2018). LOX-1 activation by oxLDL triggers an epithelial mesenchymal transition and promotes tumorigenic potential in prostate cancer cells. Cancer Letters, 414, 34–43. https://doi.org/10.1016/j.canlet.2017.10.035

Goodrich, T. (2016a, February 5). The Cause of Metabolic Syndrome: Excess Omega-6 Fats (Linoleic Acid) in Your Mitochondria. Yelling Stop. http://yelling-stop.blogspot.com/2016/02/the-cause-of-metabolic-syndrome-excess.html

Goodrich, T. (2016b, February 23). How To Prevent Oxidative Damage In Your Mitochondria. Yelling Stop. http://yelling-stop.blogspot.com/2016/02/how-to-prevent-oxidative-damage-in-your.html

Goodrich, T. (2016c, February 24). What Effect Does Linoleic Acid Have On Mitochondria? Yelling Stop. http://yelling-stop.blogspot.com/2016/02/what-effect-does-linoleic-acid-have-on.html

Goodrich, T. (2018, June 28). What’s Worse—Carbs or Seed Oils? Understanding a High-PUFA Diet. Yelling Stop. http://yelling-stop.blogspot.com/2018/06/whats-worsecarbs-or-seed-oils.html

Goodrich, T. (2020, June 2). Does Consumption of Omega-6 Seed Oils Worsen ARDS and COVID-19? [Blog]. Yelling Stop. http://yelling-stop.blogspot.com/2020/06/does-consumption-of-omega-6-seed-oils.html

Gräfe, A., Wollina, U., Tebbe, B., Sprott, H., Uhlemann, C., & Hein, G. (1999). Fibromyalgia in lupus erythematosus. Acta Dermato-Venereologica, 79(1), 62–64. https://doi.org/10.1080/000155599750011732

Gura, K. M., Parsons, S. K., Bechard, L. J., Henderson, T., Dorsey, M., Phipatanakul, W., Duggan, C., Puder, M., & Lenders, C. (2005). Use of a fish oil-based lipid emulsion to treat essential fatty acid deficiency in a soy allergic patient receiving parenteral nutrition. Clinical Nutrition, 24(5), 839–847. https://doi.org/10.1016/j.clnu.2005.05.020

Hardas, S. S., Sultana, R., Clark, A. M., Beckett, T. L., Szweda, L. I., Murphy, M. P., & Butterfield, D. A. (2013). Oxidative modification of lipoic acid by HNE in Alzheimer disease brain. Redox Biology, 1(1), 80–85. https://doi.org/10.1016/j.redox.2013.01.002

Hildreth, K., Kodani, S. D., Hammock, B. D., & Zhao, L. (2020). Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: A review of recent studies. The Journal of Nutritional Biochemistry, 86, 108484. https://doi.org/10.1016/j.jnutbio.2020.108484

Hoch, F. L. (1992). Cardiolipins and biomembrane function. Biochimica et Biophysica Acta (BBA) - Reviews on Biomembranes, 1113(1), 71–133. https://doi.org/10.1016/0304-4157(92)90035-9

Hokama, Y., Empey‐Campora, C., Hara, C., Higa, N., Siu, N., Lau, R., Kuribayashi, T., & Yabusaki, K. (2008). Acute phase phospholipids related to the cardiolipin of mitochondria in the sera of patients with chronic fatigue syndrome (CFS), chronic ciguatera fish poisoning (CCFP), and other diseases attributed to chemicals, Gulf War, and marine toxins. Journal of Clinical Laboratory Analysis, 22(2), 99–105. https://doi.org/10.1002/jcla.20217

Hu, W., Feng, Z., Eveleigh, J., Iyer, G., Pan, J., Amin, S., Chung, F.-L., & Tang, M. (2002). The major lipid peroxidation product, trans- 4-hydroxy-2-nonenal, preferentially forms DNA adducts at codon 249 of human p53 gene, a unique mutational hotspot in hepatocellular carcinoma. Carcinogenesis, 23(11), 1781–1789. https://doi.org/10.1093/carcin/23.11.1781

Humphries, K. M., & Szweda, L. I. (1998). Selective Inactivation of α-Ketoglutarate Dehydrogenase and Pyruvate Dehydrogenase: Reaction of Lipoic Acid with 4-Hydroxy-2-nonenal. Biochemistry, 37(45), 15835–15841. https://doi.org/10.1021/bi981512h

Kanazawa, K., Sakamoto, M., Kanazawa, K., Ishigaki, Y., Aihara, Y., Hashimoto, T., & Mizuno, M. (2016). Lipid peroxides as endogenous oxidants forming 8-oxo-guanosine and lipid-soluble antioxidants as suppressing agents. Journal of Clinical Biochemistry and Nutrition, 59(1), 16–24. https://doi.org/10.3164/jcbn.15-122

Kaplan, H., Thompson, R. C., Trumble, B. C., Wann, L. S., Allam, A. H., Beheim, B., Frohlich, B., Sutherland, M. L., Sutherland, J. D., Stieglitz, J., Rodriguez, D. E., Michalik, D. E., Rowan, C. J., Lombardi, G. P., Bedi, R., Garcia, A. R., Min, J. K., Narula, J., Finch, C. E., … Thomas, G. S. (2017). Coronary atherosclerosis in indigenous South American Tsimane: A cross-sectional cohort study. The Lancet, 389(10080), 1730–1739. https://doi.org/10.1016/S0140-6736(17)30752-3

Kojima, R., Kakimoto, Y., Furuta, S., Itoh, K., Sesaki, H., Endo, T., & Tamura, Y. (2019). Maintenance of Cardiolipin and Crista Structure Requires Cooperative Functions of Mitochondrial Dynamics and Phospholipid Transport. Cell Reports, 26(3), 518-528.e6. https://doi.org/10.1016/j.celrep.2018.12.070

Kruit, J. K., Brunham, L. R., Verchere, C. B., & Hayden, M. R. (2010). HDL and LDL cholesterol significantly influence β-cell function in type 2 diabetes mellitus. Current Opinion in Lipidology, 21(3), 178. https://doi.org/10.1097/MOL.0b013e328339387b

Li, S., Kievit, P., Robertson, A.-K., Kolumam, G., Li, X., von Wachenfeldt, K., Valfridsson, C., Bullens, S., Messaoudi, I., Bader, L., Cowan, K. J., Kamath, A., van Bruggen, N., Bunting, S., Frendéus, B., & Grove, K. L. (2013). Targeting oxidized LDL improves insulin sensitivity and immune cell function in obese Rhesus macaques. Molecular Metabolism, 2(3), 256–269. https://doi.org/10.1016/j.molmet.2013.06.001

Liu, W., Porter, N. A., Schneider, C., Brash, A. R., & Yin, H. (2011). Formation Of 4-Hydroxynonenal From Cardiolipin Oxidation: Intramolecular Peroxyl Radical Addition And Decomposition. Free Radical Biology & Medicine, 50(1), 166–178. https://doi.org/10.1016/j.freeradbiomed.2010.10.709

Mabalirajan, U., Rehman, R., Ahmad, T., Kumar, S., Singh, S., Leishangthem, G. D., Aich, J., Kumar, M., Khanna, K., Singh, V. P., Dinda, A. K., Biswal, S., Agrawal, A., & Ghosh, B. (2013). Linoleic acid metabolite drives severe asthma by causing airway epithelial injury. Scientific Reports, 3(1), 1349. https://doi.org/10.1038/srep01349

Marin, M. T., Dasari, P. S., Tryggestad, J. B., Aston, C. E., Teague, A. M., & Short, K. R. (2015). Oxidized HDL and LDL in adolescents with type 2 diabetes compared to normal weight and obese peers. Journal of Diabetes and Its Complications, 29(5), 679–685. https://doi.org/10.1016/j.jdiacomp.2015.03.015

Mylonas, C., & Kouretas, D. (1999). Lipid peroxidation and tissue damage. In Vivo (Athens, Greece), 13(3), 295–309. http://europepmc.org/article/med/10459507

Okamoto, K., Toyokuni, S., Uchida, K., Ogawa, O., Takenewa, J., Kakehi, Y., Kinoshita, H., Hattori-Nakakuki, Y., Hiai, H., & Yoshida, O. (1994). Formation of 8-hydroxy-2’-deoxyguanosine and 4-hydroxy-2-nonenal-modified proteins in human renal-cell carcinoma. International Journal of Cancer, 58(6), 825–829. https://doi.org/10.1002/ijc.2910580613

Palinski W, Ylä-Herttuala S, Rosenfeld M E, Butler S W, Socher S A, Parthasarathy S, Curtiss L K, & Witztum J L. (1990). Antisera and monoclonal antibodies specific for epitopes generated during oxidative modification of low density lipoprotein. Arteriosclerosis: An Official Journal of the American Heart Association, Inc., 10(3), 325–335. https://doi.org/10.1161/01.ATV.10.3.325

Panda, L., Gheware, A., Rehman, R., Yadav, M. K., Jayaraj, B. S., Madhunapantula, S. V., Mahesh, P. A., Ghosh, B., Agrawal, A., & Mabalirajan, U. (2017). Linoleic acid metabolite leads to steroid resistant asthma features partially through NF-κB. Scientific Reports, 7. https://doi.org/10.1038/s41598-017-09869-9

Parthasarathy, S., Khoo, J. C., Miller, E., Barnett, J., Witztum, J. L., & Steinberg, D. (1990). Low density lipoprotein rich in oleic acid is protected against oxidative modification: Implications for dietary prevention of atherosclerosis. Proceedings of the National Academy of Sciences, 87(10), 3894–3898. https://doi.org/10.1073/pnas.87.10.3894

Perri, F., Pisconti, S., & Della Vittoria Scarpati, G. (2016). P53 mutations and cancer: A tight linkage. Annals of Translational Medicine, 4(24). https://doi.org/10.21037/atm.2016.12.40

Ren, R., Hashimoto, T., Mizuno, M., Takigawa, H., Yoshida, M., Azuma, T., & Kanazawa, K. (2013). A lipid peroxidation product 9-oxononanoic acid induces phospholipase A2 activity and thromboxane A2 production in human blood. Journal of Clinical Biochemistry and Nutrition, 52(3), 228–233. https://doi.org/10.3164/jcbn.12-110

Riad, A., Narasimhulu, C. A., Deme, P., & Parthasarathy, S. (2017). A Novel Mechanism for Atherosclerotic Calcification: Potential Resolution of the Oxidation Paradox. Antioxidants & Redox Signaling, 29(5), 471–483. https://doi.org/10.1089/ars.2017.7362

Rosa, I. de A., Einicker-Lamas, M., Bernardo, R. R., & Benchimol, M. (2008). Cardiolipin, a lipid found in mitochondria, hydrogenosomes and bacteria was not detected in Giardia lamblia. Experimental Parasitology, 120(3), 215–220. https://doi.org/10.1016/j.exppara.2008.07.009

Silvestri, C., & Di Marzo, V. (2013). The Endocannabinoid System in Energy Homeostasis and the Etiopathology of Metabolic Disorders. Cell Metabolism, 17(4), 475–490. https://doi.org/10.1016/j.cmet.2013.03.001

Skulachev, V. P., Antonenko, Y. N., Cherepanov, D. A., Chernyak, B. V., Izyumov, D. S., Khailova, L. S., Klishin, S. S., Korshunova, G. A., Lyamzaev, K. G., Pletjushkina, O. Yu., Roginsky, V. A., Rokitskaya, T. I., Severin, F. F., Severina, I. I., Simonyan, R. A., Skulachev, M. V., Sumbatyan, N. V., Sukhanova, E. I., Tashlitsky, V. N., … Zvyagilskaya, R. A. (2010). Prevention of cardiolipin oxidation and fatty acid cycling as two antioxidant mechanisms of cationic derivatives of plastoquinone (SkQs). Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1797(6), 878–889. https://doi.org/10.1016/j.bbabio.2010.03.015

Specialties, N. (n.d.). Trouble With TBARS [Store]. The Trouble with TBARS. Retrieved October 23, 2020, from https://www.nwlifescience.com/information/trouble-with-tbars

Speijer, D. (2016). Being right on Q: Shaping eukaryotic evolution. The Biochemical Journal, 473(22), 4103–4127. https://doi.org/10.1042/BCJ20160647

Terni, B., Boada, J., Portero‐Otin, M., Pamplona, R., & Ferrer, I. (2010). Mitochondrial ATP-Synthase in the Entorhinal Cortex Is a Target of Oxidative Stress at Stages I/II of Alzheimer’s Disease Pathology. Brain Pathology, 20(1), 222–233. https://doi.org/10.1111/j.1750-3639.2009.00266.x

Tuominen Anu, Miller Yury I., Hansen Lotte F., Kesäniemi Y. Antero, Witztum Joseph L., & Hörkkö Sohvi. (2006). A Natural Antibody to Oxidized Cardiolipin Binds to Oxidized Low-Density Lipoprotein, Apoptotic Cells, and Atherosclerotic Lesions. Arteriosclerosis, Thrombosis, and Vascular Biology, 26(9), 2096–2102. https://doi.org/10.1161/01.ATV.0000233333.07991.4a

Valavanidis, A., Vlachogianni, T., & Fiotakis, C. (2009). 8-hydroxy-2’ -deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. Journal of Environmental Science and Health. Part C, Environmental Carcinogenesis & Ecotoxicology Reviews, 27(2), 120–139. https://doi.org/10.1080/10590500902885684

Weismann, D., & Binder, C. J. (2012). The innate immune response to products of phospholipid peroxidation. Biochimica et Biophysica Acta (BBA) - Biomembranes, 1818(10), 2465–2475. https://doi.org/10.1016/j.bbamem.2012.01.018

Witztum, J. L., & Steinberg, D. (1991). Role of oxidized low density lipoprotein in atherogenesis. Journal of Clinical Investigation, 88(6), 1785–1792. https://doi.org/10.1172/JCI115499

Yeang, C., Hung, M. Y., Pattison, J., Bowden, K., Dalton, N., Peterson, K. L., Witztum, J. L., Tsimikas, S., & Que, X. (2016). Expression of E06, a natural monoclonal antibody targeted to oxidized phospholipids (OXPL), attenuates the progression of aortic sclerosis in aged hyperlipidemic mice. Atherosclerosis, 252, e229. https://doi.org/10.1016/j.atherosclerosis.2016.07.212