DOI:
10.1039/D4FO04911J
(Paper)
Food Funct., 2025,
16, 147-167
Exploiting conjugated linoleic acid for health: a recent update
Received
8th October 2024
, Accepted 7th November 2024
First published on 6th December 2024
Abstract
Conjugated linoleic acid (CLA) is widely used as a dietary supplement due to its reported benefits in enhancing immunity, regulating inflammation, treating obesity, and preventing cancer. However, there is a lack of comprehensive studies on its mechanisms and dose-effect relationships. Moreover, there are insufficient in-depth studies on CLA's new functions, safety, side effects, and clinical utility. This review systematically examines the structure and sources of CLA, summarizes its role in improving human health, and critically reviews the potential mechanisms behind these benefits. It also analyzes the side effects of CLA and addresses issues related to dosing and oxidative decomposition in CLA research. Additionally, the potential of using CLA-producing probiotics to manage diseases is explored. This review can guide and promote further research on CLA's functions and support the development of CLA dietary supplements. It will accelerate the development of CLA nutritional and medical foods, contribute to the improvement of human health, and have important social meaning and economic value.
1. Introduction
The composition of fatty acids has attracted considerable attention from researchers due to its effect on human health.1 Conjugated linoleic acid (CLA) is a type of polyunsaturated fatty acid found in ruminant animal foods (dairy products, milk, and meat). It is a positional and geometric isomer of linoleic acid and contains conjugated double bond systems, in which 28 CLA isomers have been identified.2 The main isomers of CLA in dietary sources are trans10, cis12-CLA (t10, c12-CLA) and cis9, trans11-CLA (c9, t11-CLA). t10, c12-CLA accounts for 1–10% of the total CLA, while c9, t11-CLA accounts for 90% of the total CLA in dietary sources (Fig. 1).3 Furthermore, CLA's status as a class of naturally occurring essential fatty acids indicates that it is involved in a variety of biological regulatory functions in vivo. CLA can inhibit cancer, reduce blood fat, resist atherosclerosis, and improve immunity. Additionally, it can regulate blood sugar, blood pressure, and other important physiological functions.4 Thus, CLA has health benefits that merit further examination.
 |
| Fig. 1 Source and structure of CLA. CLA is a ruminant animal foods (dairy products, milk, and meat) found in a kind of polyunsaturated fatty acids. The main isomer of CLA in dietary sources is t10, c12-CLA and c9, t11-CLA, which have proven to have health benefits. | |
The US Food and Drug Administration (FDA) defines trans fats as “unsaturated fatty acids (non-conjugated) containing one or more independent double bonds in trans configuration”.5 According to the above definition, CLA with conjugated bonds is not a trans fatty acid. Moreover, t10, c12-CLA, c9, t11-CLA, and a mixture of the two (50
:
50) have been classified as “generally recognized as safe, GRAS” by the FDA.6 According to clinical studies, intakes of up to 3.4 g CLA per day (2 years) or 6 g CLA per day (1 year) are currently considered safe.7,8 The FDA currently states that “the use of CLA-rich oils in certain specific foods in general categories, such as fruit juices, dairy products, beverages, meal replacement drinks, and soy milk, is considered safe at levels of approximately 1.5 g of CLA”.6
The limited availability of CLA-fortified foods currently reflects the challenges of using CLA as a food ingredient. Although CLA has been widely studied for regulating health, the analysis of its mechanism is insufficient. In addition, the side effects of long-term use of CLA have not received enough attention. Some of the new features such as prevent alzheimer's disease and depression in CLA studies lack in-depth clinical and safety studies. As a result, this review summarizes the potential health benefits and mechanisms of CLA, lists the potential side effects of CLA, proposes a dose-effect relationship study of CLA, discusses the efficient delivery of CLA, and recommends the use of CLA-producing microorganisms in future CLA research to regulate human health. This review will guide and promote research on CLA function and the development of dietary products.
2. Regulation of CLA on inflammation
2.1. Regulation of inflammatory bowel disease
Inflammatory bowel disease (IBD) is a chronic recurrent disease, mainly including Crohn's disease and ulcerative colitis (UC).3 CLA can alleviate IBD in animal models. The mucus layer can against harmful substances and microorganisms: it comprises a firm inner layer (composed of polymerized MUC2) and a loose outer layer (formed by hydrolysis of MUC2).9 Dietary supplementation of 20 mg per day of CLA significantly increased the number of goblet cells, upregulated MUC2, and repaired the intestinal mucus layer of mice with UC.3 The tight junctions (TJs) and adhesion junctions between epithelial cells can join epithelial cells and prevent microbial invasion through paracellular pathways.10 CLA can regulate TJ proteins, thereby affecting the intestinal mechanical barrier. For example, when 1% of CLA was added to the diet of mice, it upregulated mRNA levels of Occludin, ZO-1, MUC2, and Claudin-3 in colitis mice.11 However, oral administration of CLA at 20 mg per day upregulated TJ and AJ proteins in colitis mice.3 In addition, dextran sulphate sodium (DSS) can induce apoptosis of epithelial cells, thereby disrupting the intestinal mechanical barrier. 20 mg per day of CLA can significantly downregulate the apoptosis of epithelial cells in mice with colitis,3 which plays an important role in maintaining the intestinal epithelial cell layer. Therefore, CLA can upregulate MUC2 to improve the mucus layer, regulate TJ proteins, inhibit apoptosis, and improve colitis (Fig. 2 and 3).
 |
| Fig. 2 Schematic of CLA in improving health. CLA can regulate IBD, colorectal cancer, atopic dermatitis, arthritis, II diabetes mellitus, obesity, atherosclerosis, breast cancer, and psychiatric disease by regulating gut microbiota, intestinal metabolites, cytokines associated with inflammation, oxidative stress, and intestinal mechanical barrier. Docosahexaenoic acid (DHA); brain derived neurotrophic factor (BDNF); synaptic functional proteins (SFP); vascular cell adhesion molecule 1 (VCAM-1); monocyte chemotactic protein 1 (MCP-1); macrophage-1 antigen (Mac-1); very late appearing antigen 4 (VLA-4); total cholesterol (TC); fasting blood glucose (FBG); low density lipoprotein cholesterol (LDL-C); fatty acid (FA); prostaglandin E2 (PGE2); matrix metalloproteinases (MMP); leukotriene B4 (LTB4). | |
 |
| Fig. 3 The potential mechanisms of CLA in relieving colitis. CLA can upregulate MUC2 to improve mucus layer, regulate TJ proteins, and inhibit apoptosis. CLA can also inhibit oxidative stress. CLA regulates NF-κB signaling pathway and intestinal inflammatory cytokines through macrophage-mediated PPAR-γ. CLA inhibits harmful bacteria, promotes the growth of beneficial bacteria, and improves the imbalance of gut microbiota. | |
Oxidative stress can facilitate the release of inflammatory mediators, which lead to the destruction of the intestinal mechanical barrier.12 The antioxidant effects of CLA have been reported. For example, feeding rats with CLA (19.54 mg g−1 fat) during adolescence increased the activity of superoxide dismutase (SOD) and catalase (CAT) in the blood and liver.13 Dietary supplementation with 0.83% CLA significantly increased liver total antioxidant capacity (T-AOC) and CAT activity of large yellow croaker.14 Moreover, CLA (20 or 40 mg per day) significantly increased the activities of CAT, glutathione peroxidase (GSH-Px), and SOD in colon tissue.3 Thus, inhibition of oxidative stress is an important way for CLA to relieve colitis (Fig. 2 and 3).
IBD may be associated with inflammation of gut mucosa. CLA (100 mg per kg per d) prevented colon shortening, significantly reduced NF-κB expression, and increased the expression of PPAR-γ and treleaf factor family 3 (TFF3).15 In addition, supplementation of 1% CLA in the diet of colitis mice reduced inflammatory mediator infiltration and mucosal damage, indicating a macrophage-mediated PPAR-γ dependent mechanism.16 Moreover, CLA intervention significantly downregulated IL-6, IL-1β, and TNF-α expression, while it upregulated IL-10 expression.11 CLA (40 or 20 mg per day) decreased IL-6 and TNF-α and increased IL-10 in the colon. This phenomenon may be associated with activation of PPAR-γ and inhibition of NF-κB, decreasing the secretion of proinflammatory factors.3 Therefore, CLA can improve colitis by activating PPAR-γ to inhibit the NF-κB signaling pathway and intestinal inflammatory cytokines (Fig. 2 and 3).
Gut microbiota are closely related to the pathogenesis of IBD.17 DSS treatment can affect the gut microbiota, while CLA has a regulatory effect on the gut microbiota. For example, CLA treatment increased Verrucomicrobia, which might be attributed to the high expression of MUC2.17 At genus level, CLA treatment increased the abundance of Odoribacter and Bifidobacterium but significantly decreased Bacteroides. Bifidobacterium, as a probiotic, has a certain relieving effect on colitis.3 In addition, studies have shown that DSS treatment reduced Odoribacter and increased host inflammation by reducing short-chain fatty acid production.18Odoribacter can produce butyric acid, which improves the intestinal barrier and relieves colitis.19 Research has shown that Bacteroides has a proinflammatory effect and can degrade the mucosal layer and destroy the intestinal barrier.20 Therefore, the regulatory effect of CLA on intestinal inflammation is partly attributable to the regulation of gut microbiota (Fig. 2 and 3).
2.2. Regulation of arthritis
Arthritis is an inflammatory disease that occurs in the joints. As a dietary supplement, CLA has been reported to have some regulatory effects on arthritis.21 The supplementation of 0.5% c9, t11-CLA and t10, c12-CLA can reduce arachidonic acid (ARA) and the incidence of arthritis in mice.22 In collagen-induced arthritis (CA) and rheumatoid arthritis (RA), prostaglandin E2 (PGE2) produced by ARA cascades promotes Th17 cell differentiation, expansion, and disease. In contrast, inhibition of ARA cascades and certain Th cytokines may inhibit the development of arthritis.23 Moreover, 0.25% t10, c12-CLA increased IgG to aggravate the severity of CA, while 0.5% c9, t11-CLA increased IL-4 and IL-10 to reduce the incidence of CA.24 Therefore, the effect of t10, c12-CLA on arthritis needs further study. Huebner et al. found that 0.5% c9, t11-CLA reduced plasma concentrations of IL-1β, IL-6, and TNF-α.25 In addition, t10, c12-CLA reduced the inflammatory response in the mouse CA model in a dose-dependent manner.26 In another study, 0.5% t10, c12-CLA or 0.5% c9, t11-CLA significantly reduced arthritis scores due to reduced IL-1β, IL-17, and IL-21.27 Therefore, CLA can reduce arthritis by regulating Th1, Th2, and Th17 cells, inhibiting proinflammatory cytokines, and upregulating anti-inflammatory cytokines (Fig. 2 and 4).
 |
| Fig. 4 The potential mechanisms of CLA in relieving arthritis. CLA can regulate Th1, Th2 and Th17 cells, as well as inhibit pro-inflammatory cytokines and upregulate anti-inflammatory cytokines. CLA can also inhibit COX-2 and its derivatives, such as prostacyclin and PGE2. Prostaglandin E2 (PGE2). | |
In RA mouse models, 0.5% c9, t11-CLA reduced the incidence of arthritis. As a result, c9, t11-CLA may be an effective adjuvant of cyclooxygenase-2 (COX-2) inhibition in the treatment of chronic inflammation.28 COX-2 inhibitors relieve pain and inflammation mainly by inhibiting the production of proinflammatory mediators. Additionally, inhibiting COX-2 also leads to the inhibition of PGE2 and prostacyclin, collectively promoting inflammatory diseases, such as RA.28 Inhibition of COX-2 can promote IL-10 secretion and produce anti-inflammatory properties.28 Therefore, inhibiting COX-2 and its derivatives, such as prostacyclin and PGE2, is an important strategy for using CLA to regulate arthritis (Fig. 2 and 4).
2.3. Regulation of atopic dermatitis
Atopic dermatitis is one of the most common recurrent inflammatory skin diseases in the world.29 It is characterized by clinical signs typical of Th2/Th1 imbalance, IgE hypersensitivity, chronic pruritus, and eczema. Th2 cytokines, including IL-13 and IL-4, predominate in the acute phase of atopic dermatitis. IL-4 can stimulate the synthesis of IgE by B lymphocytes. Th1 cytokines (IL-12 and IFN-γ) progress to chronic atopic dermatitis through the proliferation and differentiation of infiltrating macrophages.29 CLA (100 mg kg−1) reduced the number of CD4+ T cells in the skin and inhibited the production of IFN-γ and IL-4 in serum and atopic dermatitis-like lesion skin.30 In addition, elevated serum total IgE levels are one of the key features of atopic dermatitis.29 Oral administration of 100 mg c9, t11-CLA significantly inhibited allergic dermatitis in mice mainly by reducing the infiltration of inflammatory cells (neutrophils, eosinophils, monocytes, mast cells, and leukocytes) and IgE levels.31 Oral CLA mixture (200 μL per day) reduced the clinical score of allergic dermatitis, and this effect was attributed to decreased ear IL-4 and plasma IgE levels.32 Antigen production and IgE conversion stimulate mast cell activation, prompting mast cell release of itch-related substances, such as histamine, leukotrienes, prostaglandins, inflammatory cytokines, and other inflammatory mediators.33 PGE2 and leukotriene B4 (LTB4) can promote inflammation and pruritus. Therefore, reducing the number of mast cells and the production of pruritus mediators is beneficial to relieving pruritus.34 In addition, supplementation with CLA can reduce levels of PGE2 and LTB4 in the skin, which is consistent with previous studies.35 Thus, CLA can regulate atopic dermatitis by regulating IgE and inhibiting mast cell activity and mast cell release of itch-related substances, such as PGE2 and LTB4 (Fig. 2 and 5).
 |
| Fig. 5 The potential mechanisms of CLA in relieving atopic dermatitis. CLA can regulate IgE and inhibit mast cell activity and mast cell release of itch related substances. CLA can also downregulate COX-2 and 5-LOX and inhibit the production of ARA derivatives PGE2 and LTB4. Oral CLA supplementation significantly inhibit atopic dermatitis-like skin lesions and inflammation by attenuating the NF-κB inflammatory signaling pathway and reducing the inflammatory factor. Arachidonic acid (ARA); prostaglandin E2 (PGE2); leukotriene B4 (LTB4). | |
In addition, CLA can downregulate the expression of NF-κB, myeloid differentiation factor88 (MyD88), 5-lipoxygenase (5-LOX), COX-2, toll-like receptor 4 (TLR4), and TNF-α.30 For example, CLA dose-dependently inhibited proinflammatory cytokines, including inducible nitric oxide synthase (iNOS), COX-2, IL-1β, IL-6, and TNF-α, in the skin of inflamed mice.36 Increased expression of 5-LOX and COX-2 has been proposed as a mechanism leading to increased production of LTB4 and PGE2. 5-LOX and COX-2 are two important enzymes that catalyze the conversion of ARA to bioactive lipid derivatives LTB4 and PGE2.37 COX-2 and 5-LOX can also induce atopic dermatitis, implying that the downregulation of COX-2 or 5-LOX reduces the development of inflammatory processes. CLA can significantly inhibit the expression of 5-LOX/LTB4 or COX-2/PGE2 in mouse skin tissues,30 which is an important mechanism for CLA to regulate atopic dermatitis. Therefore, CLA can improve atopic dermatitis by downregulating COX-2 and 5-LOX, inhibiting the production of ARA derivatives PGE2 and LTB4 (Fig. 2 and 5).
The production of a large number of proinflammatory cytokines (IL-6, TNF-α, and IL-1β), leads to severe and persistent skin inflammation and itching sensation.30 TNF-α produced by macrophages plays a crucial role in the acute phase of atopic dermatitis, mediating its onset.38 In addition, the NF-κB pathway regulates the production of proinflammatory cytokines in skin allergic reactions. Consistent with these findings, the DNFB-induced atopic dermatitis model increased NF-κB production in mouse skin, whereas CLA inhibited NF-κB production.30 Thus, oral CLA supplementation significantly inhibited atopic dermatitis-like skin lesions and inflammation by attenuating the NF-κB inflammatory signaling pathway and reducing the inflammatory factor (Fig. 2 and 5).
3. Regulation of metabolic syndrome
3.1. Regulation of obesity
Metabolic syndrome refers to a pathological state in which carbohydrates, fats, proteins, and other substances in the human body undergo metabolic disorders, including dyslipidemia, hypertension, type 2 diabetes, and obesity, which impose a significant economic burden on patients. Browning of white adipose tissue is a mechanism of fat mobilization that promotes weight and fat loss. For example, CLA (1.5% mixed) promoted uncoupling protein 1 (UCP1) expression in white adipose tissue and browning of epididymal (visceral) white fat in OB/OB spontaneously obese mice.39 Moreover, low doses of t10, c12-CLA (0.03, 0.1, and 0.3%) upregulated the expression of UCP1 in epididymal (visceral) white adipose tissue and inguinal (subcutaneous),40 thus promoting browning of white adipose tissue. Supplementation of compound CLA can reduce adipocyte size, increase lipolysis, and reduce body weight and mesenteric fat.41 In addition, the weight loss achieved by gavage of t10, c12-CLA (0.4%) mainly involved the inhibition of adipogenesis and the prevention of adipocyte differentiation.42 Dietary supplementation of t10, c12-CLA (0.1%) decreased the mass of brown adipose tissue and white adipose tissue and increased thermogenesis and adipose oxidative decomposition.43 Thus, the main modes of action of CLA against obesity include (1) suppressing appetite and reducing energy intake, (2) inhibiting fatty acid metabolism and adipogenesis, (3) increasing lipolysis or defatting, (4) decreasing adipocyte size, and (5) increasing fat oxidation and energy expenditure in white liver, muscle, and adipose tissue (Fig. 2).44,45
Table 1 presents human trials of CLA using obesity as an endpoint. A significant reduction was observed in body fat, while no significant difference was observed in body mass index in the oral CLA group.46 When CLA supplementation was extended to a year, subjects lost both fat mass and body weight.47,48 Many studies have reported beneficial effects of CLA on body weight and obesity-related parameters, including reduced body mass index, fat mass, subcutaneous fat, and appetite and increased fat oxidation rate, satiety, and serum leptin. However, no significant side effects were observed (Table 1).49–57 Some studies have shown that mixed CLA supplements did not show any affect physical metrics.58–64 In addition to causing weight and fat loss, CLA may also cause harmful side effects. A daily intake of 4.2 g of CLA was effective in reducing fat mass in obese men,65 but promoted insulin resistance. In addition, a daily intake of 3.4 g of t10, c12-CLA significantly reduced body fat and body weight but increased glucose levels and plasma insulin and decreased insulin sensitivity.66–68 These studies showed that t10, c12-CLA-mediated weight loss may occur at the expense of metabolic health.
Table 1 CLA studies in humans with the endpoint of body weight/composition
Study participants |
Patient group |
CLA dose |
Treatment period |
Effects due to CLA |
Year |
Ref. |
Body mass index (BMI); type 2 diabetes mellitus (T2DM); conjugated linoleic acid triglyceride (CLA-TG); conjugated linoleic acid free fatty acid (CLA-FFA); high-density lipoproteins (HDL); very low-density lipoproteins (VLDL); C-reactive protein (CRP); interleukin (IL); low-density lipoproteins (LDL). |
Healthy overweight men and women |
Placebo (26) vs. mixed CLA (26) |
Mixed CLA (1.7, 3.4, 5.1, or 6.8 g per day) or placebo |
3 months |
↑ Lean mass |
2000 |
46
|
↓ Fat mass |
Healthy overweight men and women |
Placebo (59) vs. mixed CLA (61) |
Mixed CLA (3.4 g per day) or placebo |
12 months |
↓ Fat, BMI and body weight |
2004 |
47
|
Healthy, overweight men and women |
Placebo (20) vs. mixed CLA (20) |
Mixed CLA (4 g per day) or placebo (safflower oil). |
12 months |
↓ Fat mass, BMI, body weight |
2007 |
48
|
Overweight but weight stable men and women |
Placebo (9) vs. mixed CLA (10) |
Mixed CLA (4 g per day) or placebo (safflower oil) |
6 months |
↓ Body weight |
2007 |
49
|
↑ Fat oxidation |
Overweight men and women |
Placebo (18) vs. mixed CLA (18) vs. 9,11 CLA (18) |
Mixed CLA (either 1.8 or 3.6 g per day) or placebo (oleic acid) |
3.5 months |
↓ Appetite |
2003 |
50
|
↑ Satiety |
Postmenopausal women with T2DM |
Placebo (17) vs. mixed CLA (18) |
Mixed CLA (1.7 g per day) or placebo |
4 months with a 1 month washout |
↓ Trunk adiposity, BMI and body weight |
2009 |
51
|
Women with metabolic syndrome |
Placebo (7) vs. mixed CLA (7) |
Mixed CLA (3 g per day) or placebo |
3 months, in conjunction with a low-calorie diet. |
↓ Body fat mass |
2012 |
52
|
↓ Plasma insulin levels |
Healthy men and women of stable weight |
Placebo (40) vs. mixed CLA (40) |
Mixed CLA (1.7 g per day) or placebo |
3 months |
↓ Body fat, BMI, body weight and subcutaneous fat |
2012 |
53
|
Healthy men and women |
Placebo (17) vs. mixed CLA (20) vs. mixed CLA (18) |
Mixed CLA (3.2 g per day or 6.4 g per day) or placebo |
3 months |
↑ Lean body mass |
2007 |
54
|
↑ serum IL-6 |
Healthy overweight men and women |
Placebo (37) vs. mixed CLA (44) |
Mixed CLA (3.4 g per day) or placebo |
12–24 months |
↓ Serum cholesterol |
2005 |
55
|
↑ Serum leptin |
↓ Fat, BMI and body weight |
Healthy men and women |
Placebo (38) vs. mixed CLA (38) |
Mixed CLA (5 g per day) or placebo |
3.5 months |
↑ Muscle mass |
2006 |
56
|
↓ Fat mass |
6–10 years-old children with a BMI >85th percentile |
Placebo (38) vs. mixed CLA (38) |
Mixed CLA (5 g per day) or placebo |
6 months |
↓ Body fat, BMI |
2010 |
57
|
↑ Lean body mass |
↓ HDL cholesterol |
Healthy weight-stable men and women |
Placebo (32) vs. mixed CLA (32) |
Mixed CLA (3.9 g per day) or placebo |
3 months |
No effects on body weight |
2007 |
58
|
Obese women |
Placebo (12) vs. mixed CLA (12) |
Mixed CLA (3.2 g per day) or placebo (olive oil). |
2 months with concurrent exercise |
None |
2016 |
59
|
Moderately overweight men and women |
Placebo (5) vs. mixed CLA (5) vs. 9,11 CLA (5) |
Mixed CLA (1.3 g per day), 9,11 CLA (1.3 g per day) or placebo |
2 months, treatment period with a 1 months washout. |
No effects on body weight or plasma lipids |
2010 |
60
|
Moderately obese weight-stable men and women |
Placebo (61) vs. mixed CLA (61) |
Mixed CLA (3.4 g per day) or placebo (olive oil) |
13 months |
No effects on body weight |
2006 |
61
|
Healthy men and women |
Placebo (20) vs. mixed CLA (20) |
Mixed CLA (4.5 g per day), or placebo (olive oil). |
3 months |
No effect on body weight, BMI, or body fat |
2006 |
62
|
Overweight and obese women |
Placebo (37) vs. mixed CLA (37) |
Mixed CLA (3 g per day) or placebo (sunflower oil). |
3 months |
No effect on body weight, BMI |
2016 |
63
|
Sedentary males |
Placebo (9) vs. CLA (9) |
Mixed CLA (3 g per day) or placebo |
1 month, concurrently with exercise |
No difference in body weight, fat or BMI |
2013 |
64
|
Abdominally obese older men |
Placebo (12) vs. mixed CLA (13) |
Mixed CLA (4.2 g per day) or placebo |
1 month |
↑ Insulin resistance |
2001 |
65
|
↓ Sagittal abdominal diameter |
Men with signs of metabolic syndrome |
Placebo (12) vs. CLA (13) |
10,12 CLA (3.4 g per day) or placebo |
3 months |
↑ Insulin resistance and glycemia |
2002 |
66
|
↓ HDL cholesterol |
3.2. Regulation of type II diabetes mellitus
Adipocytes can produce a variety of proinflammatory molecules, or “adipocytokines”, including IL-6, adiponectin, resistin, leptin, monocyte chemotactic protein 1 (MCP-1), IL-1β, and TNF-α. Abnormal expression of these adipokines occurs during obesity and leads to insulin resistance.69 Inflammatory mediators released by macrophages and T lymphocytes disrupt important insulin pathways in adipocytes and lead to subsequent accumulation of macrophages into the stromal vascular portion, further enhancing insulin resistance.70 Diabetic mice fed with fermented milk containing 0.01% CLA had significantly lower low density lipoprotein cholesterol (LDL-C), triglyceride, body weight, serum total cholesterol (TC), leptin levels, serum insulin level, and fasting blood glucose (FBG).71 Dietary administration of c9, t11 CLA reduced triglyceride levels, insulin, and fasting glucose and upregulated the production of insulin signaling pathway molecules.72 In addition, dietary supplementation with CLA (0.5 or 1%) improved plasma creatinine levels and overall inflammatory response, iNOS, and C-reactive protein in diabetic patients (Fig. 2).73 These results showed that regulation of fatty acid composition through administration of CLA may reduce obesity-induced insulin resistance. However, the effects of CLA observed in Type 2 diabetes mellitus animal models have not been comprehensively validated in human studies.
4. Regulation of atherosclerosis
Atherosclerosis is a progressive dyslipidemia and inflammatory disease in which monocyte-derived macrophages play a key role.74 Atherosclerotic plaque is characterized by an accumulation of lipid and inflammatory cells in the artery wall that can gradually become unstable, leading to rupture and subsequent ischemic events.75 Endothelial dysfunction and monocyte aggregation are the key markers of atherosclerosis.74 The t10, c12-CLA and c9, t11-CLA isomers can reduce the expression of macrophage-1 antigen (Mac-1) and late-appearing antigen 4 (VLA-4) on monocytes,76 indicating that CLA can regulate the adhesion of monocytes to endothelial cells. The aggregation and migration of monocytes and macrophages are caused by increased MCP-1 levels. MCP-1-deficient mice have been reported to significantly reduce the incidence of atherosclerosis.77 CLA inhibited the migration of monocytes to MCP-1 by PPAR-γ-dependent mechanism in vitro.78 This result shows that CLA is an effective inhibitor of monocyte migration in vitro and may be promising in combating migrating pathogenic monocytes in atherosclerosis. In addition to monocytes, CLA affects macrophages in atherosclerotic animals. Mixed CLA supplements can reduce the size of atherosclerotic lesions and macrophage accumulation.75,79 Feeding the animals with 0.1% CLA also reduced plaque macrophage content.80 In addition, CLA inhibited atherosclerosis by inhibiting foam cell formation and regulating the expression of the RCT gene, thereby facilitating the removal of cholesterol from the circulation.81,82 For example, CLA inhibited foam cell formation in vitro through PPAR-γ and LXRα dependent mechanisms.83 Thus, CLA can improve atherosclerosis by inhibiting monocyte migration, improving endothelial cell dysfunction, inhibiting macrophage secretion, and inhibiting the formation of foam cells (Fig. 2 and 6).
 |
| Fig. 6 The potential mechanisms of CLA in relieving atherosclerosis. CLA can improve endothelial cell dysfunction. CLA can also upregulate PPAR-γ to inhibit macrophage secretion and monocyte migration. Moreover, CLA may inhibit inflammatory factor and the formation of foam cells. | |
In Ldlr and ApoE mice, the improvement effect of t10, c12-CLA was significantly greater than that of c9, t11-CLA,84 indicating that t10, c12-CLA were isomers responsible for the protection of atherosclerosis.80 These studies collectively showed that CLA (particularly likely t10, c12-CLA) had an antiatherosclerotic effect.75 However, other studies showed that CLA supplementation had no significant effect on atherosclerosis. In a study of apoE mice, feeding the mice with a diet mixed with CLA for 12 weeks did not improve atherosclerosis.85 Similarly, administration of 0.5% t10, c12-CLA did not affect atherosclerosis in apoE/Ldlr/double knockout mice.86 Therefore, future studies are needed to confirm the role of CLA in atherosclerosis.
5. Regulation of cancer
CLA is a good anticancer agent. Different animal models have been used to demonstrate the role of CLA in the development of colorectal and breast cancers (Table 2). A diet containing 1% CLA inhibited the incidence of colon tumors, decreased PGE2 and ARA, and increased the apoptosis-regulating protein Bcl-2/bax ratio and caspase-3 in colon cancer (CRC) rats.87,88 In Apcmin/+ mice, c9, t11-CLA reduced the number of polyps, while supplementation of t10, c12-CLA increased the diameter of polyps in intestinal tumors through the PPAR-γ signaling pathway, indicating that different CLA isomers had different effects on colon tumors.89 However, a mixture of t10, c12-CLA (0.5%) and c9, t11-CLA (0.5%) did not show anticancer effects in Apcmin/+ mice.90 Mice injected with cancer cells were fed with a diet containing t10, c12-CLA and c9, t11-CLA for 4 weeks. The results showed that the two CLA isomers were found to be effective against CRC cell metastasis in vivo.91 However, more research is needed to explain the mechanisms by which various CLA isomers inhibit lung metastasis and CRC. Kim et al. found that CLA supplementation decreased the levels of TXB2 and PGE2 in the colonic mucosa, increased apoptosis, and reduced tumor incidence.92 In addition, CLA supplementation inhibited TNF-α and COX-2, increased PPAR-γ, induced apoptosis, and inhibited cell proliferation.93,94 Therefore, CLA can inhibit CRC cell proliferation and metastasis, promote apoptosis of CRC cells, activate PPAR-γ, inhibit proinflammatory cytokines, and reduce harmful substances, thereby reducing CRC (Fig. 2 and 7).
 |
| Fig. 7 The potential mechanisms of CLA in relieving cancer. CLA can inhibit cancer cell proliferation and cell cycle growth. CLA up-regulates bax and caspase-3 and promotes apoptosis of cancer cells. CLA can also inhibit MMP2 and MMP4 and cancer cell metastasis. CLA may activate PPAR-γ to inhibit pro-inflammatory cytokines. CLA inhibits oxidative stress and inflammatory substance receptors. Matrix metalloproteinases (MMP). | |
Table 2 Effects of CLA in carcinogenesis (animal models)
Cancer type |
Inducer |
Animal |
CLA dose |
Treatment period |
Effects |
Year |
Ref. |
1,2-Dimethylhydrazin (DMD); azoxymethane (AOM); N-nitroso-N-methylurea (NMU); 1,2-dimethylhydrazine (DMH); 7,12-dimethylbenz[a]anthracene (DMBA); N-methyl-nitrosourea (MNU); 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP); N-nitrosobis-2-oxopropylamine (BOP). |
Colon |
DMD |
SD rats |
CLA mixture (1%) |
14 weeks |
↑ Apoptotic index |
2001 |
87
|
↓ Number of tumors |
↓ Prostaglandin E2, thromboxane B2 and arachidonic acid |
Colon |
AOM |
SD rats |
CLA mixture (1%) |
44 weeks |
↑ Apoptosis and Caspase-3 |
2010 |
88
|
↓ Wnt signaling and arachidonic acid |
Colon |
— |
Apc(min/+) mice |
c9, t11-CLA (1%) |
8 weeks |
↓ Polyp size (c9, t11-CLA, CLA mixture) |
2008 |
89
|
CLA mixture (1%) |
↑ Polyp size (t10, c12-CLA) |
t10, c12-CLA (1%) |
Colon |
— |
Apc(min/+) mice |
CLA mixture (1%) |
7 weeks |
No effect |
2000 |
90
|
Colon |
NMU |
BALB/c mice |
c9, t11-CLA (0.1%) |
4 weeks |
↓ Cancer cell migration, pulmonary nodules |
2007 |
91
|
t10, c12-CLA (0.1%) |
Colon |
DMH |
SD rats |
CLA mixture (1%) |
30 weeks |
↓ Tumor incidence, arachidonic acid, thromboxane B2 and DAG |
2003 |
92
|
↑ Apoptosis |
Colon |
AOM + DSS |
C57BL/6 mice |
CLA mixture (1%) |
12 weeks |
↓ Macrophage |
2010 |
93
|
↑ PPAR g |
↓TNF-α |
Colon |
AOM, colonic aberrant crypt foci |
Male F344 rats |
c9, t11-CLA (0.1%, or 1%) |
4 weeks |
Induced apoptosis and suppressed cell proliferation, ↑ PPAR g |
2007 |
94
|
↓ COX-2 |
Mammary |
DMBA |
SD rats |
CLA mixture (80 mg per kg BW) |
14 weeks |
Inhibit PUFA oxidation, MDA |
2019 |
95
|
Mammary |
DMBA |
BALB/c mice |
CLA mixture (0.1%, 0.5% or 1%) |
4 weeks |
Decresed tumor burden |
2000 |
96
|
Mammary |
MNU |
SD rats |
c9, t11-CLA (1%) or CLA mixture (1%) |
20 weeks |
Decressed tumor mass |
2003 |
97
|
Mammary |
DMBA |
SD rats |
c9, t11-CLA (80 mg per kg BW) |
20 weeks |
↓ Decreased tumor weight and Volume |
2020 |
98
|
↓ Ki-67 |
Mammary |
DMBA |
SD rats |
CLA mixture (0.5%, 1%, 1.5% or 2%) |
24 weeks |
No effect |
1997 |
99
|
CLA can downregulate the proliferation of breast cancer cells and also has certain effects on breast cancer animals. For example, mixture CLA intervention in rats reduced the incidence of breast cancer by inhibiting PUFA oxidation and regulating the expression of COX-2 and PGE2 receptors.95 Another study showed that CLA (0.1–1%) reduced lung tumor load in mice.96 Oral administration of c9, t11-CLA reduced tumor weight and volume, hormone receptor PR, and Ki-67 expression, demonstrating a protective effect on breast cancer.97,98 Therefore, CLA can inhibit oxidative stress, inhibit inflammatory substance receptors, and thus regulate breast cancer (Fig. 2 and 7). However, another study showed that CLA did not significantly affect breast cancer. For example, rats with breast cancer were fed with 0.5, 1, 1.5, and 2% CLA. The results showed that CLA did not affect breast cancer.99
6. Regulation of psychiatric disease
Alzheimer's disease (AD) is a progressive neurodegenerative disease. c9, t11-CLA diet significantly reduced amyloid beta and regulated the expression of anti-inflammatory cytokines IL-19 and IL-10 in AD model mice.100 Neuroinflammation caused by Aβ accumulation is thought to be the key to memory loss in AD.101 Thus, dietary supplementation of c9, t11-CLA can upregulate anti-inflammatory cytokines in the brain and may inhibit neuroinflammation by activating PPAR-γ. In addition, CLA supplementation can enhance cognitive function and reduce cerebral oxidative stress, inflammation, apoptosis, and acetylcholinesterase activity in AD mice.102 Therefore, CLA can inhibit amyloid beta and cell apoptosis and activate PPAR-γ to regulate inflammatory cytokines, thereby improving AD (Fig. 2 and 8).
 |
| Fig. 8 The potential mechanisms of CLA in relieving psychiatric disease. CLA can inhibit amyloid beta and cell apoptosis, and activate PPAR-γ to regulate inflammatory cytokines, leading to improvement of AD. Supplementation of CLA can significantly improve redox homeostasis, inhibit Nrf2 overactivation, and increase DHA, BDNF and SFP in depressed mice. Alzheimer's disease (AD); docosahexaenoic acid (DHA); brain derived neurotrophic factor (BDNF); synaptic functional proteins (SFP). | |
Depression is a global mental illness characterized by anhedonia, insomnia, loss of appetite, sadness, and even suicide.103 Supplementation of 30 mg mixed CLA significantly improved redox homeostasis and inhibited Nrf2 overactivation. Additionally, it increased synaptic functional proteins (SFP), brain-derived neurotrophic factor (BDNF), and docosahexaenoic acid (DHA) in depressed mice.103 Oxidative stress, one of the etiology of depression/neurodegenerative disease, is often accompanied by activation of the Nrf2 pathway. This pathway is a key antioxidant/detoxification mechanism and is considered a promising target for the treatment of psychiatric disorders and depression-like behaviors.104 The beneficial effects of CLA supplementation are clearly multifactorial. Nrf2 is involved in regulating multiple physiological pathways (lipid metabolism, inflammation, and redox homeostasis), this mechanism may at least partially explain the antidepressant effects of CLA (Fig. 2 and 8). Currently, there are a limited amount of studies on the effects of CLA on depression and AD. Therefore, more animal and even clinical studies are needed to expand the current results in the future.
7. Limitations, challenges, and prospects in functional studies of CLA
7.1. Effects of isomer, dose, animal model, and test time on results of CLA functional studies
Commercial CLAs usually contain equal proportions of t10, c12-CLA and c9, t11-CLA. Both isomers are currently used in animal and clinical studies. Most of the beneficial properties of CLA are attributed to the two main isomers. In addition, different CLA isomers may have different effects, and the effects of different concentrations of isomers in CLA mixtures are difficult to distinguish. For example, t10, c12-CLA is involved in lipid oxidative catabolism, while c9, t11-CLA can act as active anabolic agents with anti-inflammatory effects.105 Both isomers have anticancer properties, although they have different effects on apoptosis regulation, oncogene regulation, and lipid metabolism.106 Therefore, the function of each CLA isomers should be studied to make the research results more explicit and clearer.
Functional studies of CLA involve different animal models, including monkeys, rabbits, rats, mice, and pigs. Differences in animal genetic background and internal structure can lead to varying effects. Using different animals may result in different, and sometimes opposite, outcomes. Therefore, in the functional study of CLA, animal models should be clarified, and in the clinical study of CLA, age, gender, and physical status of subjects should also be clarified to facilitate reference and inspection by other researchers. In addition, CLA supplement time can affect the experimental results. As a result, it is necessary to clarify the CLA supplement time in the study. In conclusion, in the study of CLA function, clarifying the animal model, determining the composition, dose, and duration of CLA, and controlling these factors are necessary to standardize the study of CLA function and obtain valuable results.
7.2. Limitations, challenges, and prospects in dose-effect studies of CLA
At present, there are limited dose-effect studies on the function of CLA. Beef and dairy products are major sources of CLA in foods. Many people supplement these foods due to the difficulty of quantifying dietary CLA intake. Different countries have published recommended intakes for CLA: 52–137 mg per day in the United States, 600–800 mg per day in the United Kingdom, and 1500 mg per day in Australia.107 The health benefits of CLA can only be achieved if sufficient doses are taken.108 Studies have shown that a daily intake of 3–6 g of CLA can produce significant health benefits.109,110 In human studies, a mixture of t10, c12-CLA and c9, t11-CLA has been used. Therefore, in the study of the function of CLA, more attention should be paid to the dosage. Animal models can be used to study the influence of different doses of CLA on a certain disease to determine the dose-effect relationship and find the optimal dose of CLA. Further study on the dose effect of CLA on humans based on the results of animal experiments would be beneficial for the development of CLA products (Fig. 10).
7.3. Limitations, challenges, and prospects in the studies of side effects of CLA
Although many of the effects of CLA are well known, the potential health problems of CLA are rarely reported. Most studies have not reported the relationship between glucose or insulin levels and CLA supplementation.66,111 Some studies have shown that CLA supplementation has no significant effect on glucose homeostasis,52,53,112,113 whereas some showed that CLA supplementation can cause impaired glucose tolerance and disrupt glucose homeostasis.64,67 Studies have reported that potential adverse effects of CLA on glucose homeostasis were related to the isomer t10, c12-CLA, while c9, t11-CLA showed no relationship with glucose homeostasis.67 When the two isomers of CLA are jointly used, c9, t11-CLA may antagonize t10, c12-CLA.67,68 In addition, the effects of CLA on glucose homeostasis are transient. Moreover, in relatively long-term human studies, the use of CLA has no effect on glucose homeostasis.55,114 Therefore, the effects of CLA on glucose homeostasis need to be evaluated.
In addition, CLA supplementation can cause fatty liver (hepatic steatosis) (Fig. 9). The c9, t11-CLA can decrease G6PC and PCK1 to reduce hepatic gluconeogenesis, while t10, c12-CLA increases hepatic gluconeogenesis by increasing G6PC expression.115 Regardless of species, CLA induces hepatic steatosis by influencing insulin secretion, regulating hepatic fatty acid (FA) composition, and promoting the secretion of inflammatory response and adipokine.116 CLA can increase liver PKCε, diacylglycerol, and TG to induce insulin resistance, hepatic steatosis, and liver enlargement, which is attributed to the up-regulation of hepatic gluconeogenic gene expression (Fig. 9).86,117–119 Moreover, high doses of CLA led to fat atrophy, steatosis and inflammation in mice.40 Long-term feeding of t10, c12-CLA (0.3%) diets activates lipogenesis, induces FA oxidation and inhibits LDL secretion, which leads to increase liver mass and lipid and TG accumulation.120 Therefore, caution should be exercised when using t10, c12-CLA, and attention should be paid to the adverse reactions caused by long-term supplementation.121
 |
| Fig. 9 Possible pathways by which CLA induces hepatic steatosis. CLA in adipose tissue causes a reduction in fatty acid oxidation, lipogenesis and uptake of lipid and glucose, leading to higher circulatory levels of free fatty acids and glucose. Pancreas-induced systemic insulin resistance. Alterations in hepatic lipid metabolism and increased hepatic lipid accumulation and liver mass, leading to hepatic steatosis. | |
In addition, studies have shown that CLA supplementation inhibits milk fat synthesis in goats, dairy cows, and mice.122,123 This problem raises questions about the use of CLA in humans, particularly concerning infant health. However, there are significant differences in milk fat sources between humans and ruminants, with milk fat in ruminants being more sensitive to CLA.124 Dietary supplementation of t10, c12-CLA can decrease milk fat production.125 In addition to animals, several studies reported the effects of CLA on milk fat content in humans. For example, compared with placebo, a daily intake of 1.5 g of CLA for 5 days significantly reduced milk fat, although milk fat levels remained within normal ranges.126 Another study showed no significant change in milk fat content after 5 days of CLA supplementation (750 mg CLA isomer per day or 2–4 g per day).127 The effect of CLA on the fat content in human milk needs to be confirmed by more clinical trials.
Thus, the effects of CLA on milk fat suppression, hepatic steatosis, and glucose homeostasis have shown different results in animal and clinical studies. More long-term clinical trials are needed to confirm these results in the future (Fig. 10).
 |
| Fig. 10 The limitations, challenges, and prospects in functional studies of CLA. In future basic research on CLA in improving health, attention should be paid to the dose-effect relationship (the function and dose of each CLA isomers should be studied), CLA embedding (influence of different embedding methods on CLA function), side effects (the effects of CLA on milk fat suppression, hepatic steatosis, and glucose homeostasis), effects of CLA-producing LAB on health (CLA-producing microorganisms can be used to improve inflammation-related diseases). | |
7.4. Limitations, challenges, and prospects of CLA embedding and encapsulation
CLA has many functions, but it needs to be taken orally into the intestine. CLA is a polyunsaturated FA that is lipophilic (or hydrophobic) and is easily degraded and oxidized during storage and digestion.128 The low solubility of CLA also significantly reduced its bioacceptability and bioavailability. CLA is extremely unstable and even more prone to autooxidation than oleic acid, linolenic acid, or ARA. To overcome these challenges, some carriers have been developed to encapsulate CLA. We summarize the current carrier of CLA packaging and its characteristics and prospects. Chang developed isopass linear dextrin encapsulated CLA, which improved the solubility of the complex and enabled targeted delivery of CLA to the small intestine.129 Moreover, the degradation rate of CLA was reduced by encapsulating CLA in whey protein concentrate or a mixture of whey protein concentrate and maltodextrin by spray drying.130 However, CLA is rapidly decomposed into furan FAs when heated in air.131 Therefore, spray drying and other heating methods have limitations and are ineffective for CLA packaging (Fig. 10).
Increasing the bioavailability of CLA at the nanoscale is a promising approach. This approach delays the degradation of CLA through oxidation and other chemical reactions, thus preventing the development of undesirable odors and loss of metabolic value.132 Soybean lecithin is a good emulsifying surfactant owing to its high affinity with cell membranes and its ability to increase the absorption of bioactive compounds. For example, in a study of obese mice, nanoemulsified water-soluble-conjugated linoleic acid increased CLA availability, reduced triglyceride and cholesterol volumes in liver tissue, and significantly increased CLA anti-obesity activity.133 Moreover, results of human intestinal Caco-2 cells showed that nano-emulsification enhanced the absorption rate of CLA.134 These results provide important information for the development of nanoemulsion carriers, which may improve the thermal stability and bioavailability of CLA (Fig. 10). Although emulsion technology is particularly suitable for the design and manufacture of delivery systems that encapsulate bioactive lipids such as CLA, these lipids are highly sensitive to oxidation due to their high interfacial area.135 Therefore, future research needs to overcome this limitation. Soybean pro-lipoprotein (LP) is characterized by high loading capacity, antioxidant activity, and sustained release in vitro. Therefore, soybean proto-lipoprotein (LPP) nanoparticles were prepared as a new CLA carrier, which encapsulated CLA to prevent oxidation and achieve continuous release.136 Further experiments on animals and humans are needed in the future to determine the effect.
7.5. Limitations, challenges, and prospects in the studies of health effect of CLA-producing lactic acid bacteria
Although CLA has many functions, long-term use of CLA has certain side effects. Side effects of CLA have been reported in animal studies, including altered glucose/insulin metabolism and liver dysfunction. CLA supplementation in improving health or preventing disease remains a controversial topic. In addition, CLA supplementation can affect liver transglutaminase and liver fatty acid synthesis and oxidation. A large number of CLA-producing bacteria such as Butyrivibrio, Lachnospiraceae, Bulleidia, and Coriobacteriaceae naturally exist in the rumen of animals that eat food containing high linoleic acid content.137 In addition, many non-ruminant bacteria, including strains naturally present in the human gastrointestinal tract, such as Lactococcus, Pediococcus, Propionibacterium, Roseburia, Bifidobacterium and Lactobacillus, can use LA to synthesize CLA.138 These bacteria similarly express linoleic isomerase to ruminant bacteria in order to convert linoleic acid to c9, t11-CLA. Only a few studies have reported the production of t10, c12-CLA by gastrointestinal bacteria. Some researchers have considered using CLA-producing strains to directly produce CLA in the colon, thereby eliminating the side effects of taking large doses of CLA directly. For example, for colitis, Yang et al. screened several strains, including Lactobacillus plantarum ZS2058, Bifidobacterium breve CCFM683, B. pseudocatenulatum MY40C, B. pseudocatenulatum CCFM680, and B. longum CCFM681. The effect of these bacteria on UC remission was investigated in a series of studies.11,139–142 The results showed that these CLA-producing probiotics inhibited NF-κB and inflammatory cytokines. Additionally, they regulated the immune response and played an anti-inflammatory role by activating PPAR-γ. Moreover, they could promote the secretion of MUC2 through goblet cells to protect the intestinal mucus layer and reduce the apoptosis of colon epithelial cells. In addition, they could regulate the diversity and composition of gut microbiota (Fig. 10). These features indicate that colitis can be regulated by the use of CLA-producing microorganisms. Therefore, CLA-producing microorganisms can be used to improve inflammation-related diseases, such as arthritis, dermatitis, and colon cancer, in future studies.
8. Conclusion
CLA has been shown to have some regulatory effects on health in animal experiments, but human studies are lacking. In addition, the side effects of CLA need to be further explored. Based on the summary, analysis, and reflection of numerous studies, the following recommendations are made: (1) identify and control animal models, CLA composition, dose, and duration; (2) control the optimal intake of CLA through dose-effect studies; (3) encapsulate CLA using spray drying, microcapsule, emulsion technology, and embedding to reduce oxidative decomposition and improve bioavailability; and (4) use CLA-producing microbes to improve inflammatory diseases. This review will accelerate the development of nutritional and medical foods containing CLA, ultimately helping to improve human health.
Abbreviation
FDA | The US Food and Drug Administration |
IBD | Inflammatory bowel disease |
UC | Ulcerative colitis |
TJs | Tight junctions |
SOD | Superoxide dismutase |
CAT | Catalase |
T-AOC | Total antioxidant capacity |
GSH-Px | Glutathione peroxidase |
DSS | Dextran sulphate sodium |
COX-2 | Cyclooxygenase-2 |
iNOS | Inducible nitric oxide synthase |
TFF3 | Treleaf factor family 3 |
CA | Collagen-induced arthritis |
RA | Rheumatoid arthritis |
PGE2 | Prostaglandin E2 |
ARA | Arachidonic acid |
DHA | Docosahexaenoic acid |
BDNF | Brain derived neurotrophic factor |
SFP | Synaptic functional proteins |
VCAM-1 | Vascular cell adhesion molecule 1 |
MCP-1 | Monocyte chemotactic protein 1 |
Mac-1 | Macrophage-1 antigen |
VLA-4 | Very late appearing antigen 4 |
TC | Total cholesterol |
FBG | Fasting blood glucose |
LDL-C | Low density lipoprotein cholesterol |
FA | Fatty acid |
MMP | Matrix metalloproteinases |
LTB4 | Leukotriene B4 |
AD | Alzheimer's disease |
5-LOX | 5-Lipoxygenase |
TLR4 | Toll-like receptor 4 |
MyD88 | Myeloid differentiation factor88 |
UCP1 | Uncoupling protein 1 |
CRC | Colon cancer |
TXB2 | Thromboxane 2 |
LPP | Lipoprotein |
Consent for publication
All authors have read and agreed with the manuscript.
Data availability
No primary research results, software or code have been included and no new data were generated or analysed as part of this review.
Conflicts of interest
The authors declare that they have no financial conflicts of interest.
Acknowledgements
This research was supported by the National Natural Science Foundation of China (No. 3227161449).
References
- J. K. Wang, L. X. Han, D. Y. Wang, P. P. Li and F. Shahidi, Conjugated fatty acids in muscle food products and their potential health benefits: A review, J. Agric. Food Chem., 2020, 68, 13530–13540 CrossRef CAS PubMed.
- C. K. Virsangbhai, A. Goyal, B. Tanwar and M. K. Sihag, Potential health benefits of conjugated linoleic acid: An important functional dairy ingredient, Eur. J. Nutr. Food. Saf., 2020, 11, 200–213 CrossRef.
- Y. Chen, B. Yang, R. P. Ross, Y. Jin, C. Stanton, J. Zhao, H. Zhang and W. Chen, Orally administered CLA ameliorates DSS-induced colitis in mice via intestinal barrier improvement, oxidative stress reduction, and inflammatory cytokine and gut microbiota modulation, J. Agric. Food Chem., 2019, 67, 13282–13298 CrossRef CAS PubMed.
- A. K. Fleck, S. Hucke, F. Teipel, M. Eschborn, C. Janoschka, M. Liebmann, H. Wami, L. Korn, G. Pickert, M. Hartwig, T. Wirth, M. Herold, K. Koch, M. Falk-Paulsen, U. Dobrindt, S. Kovac, C. C. Gross, P. Rosenstiel, M. Trautmann, H. Wiendl, D. Schuppan, T. Kuhlmann and L. Klotz, Dietary conjugated linoleic acid links reduced intestinal inflammateon to amelioration of CNS autoimmunity, Brain, 2021, 144, 1152–1166 CrossRef.
- M. Temkov and V. Muresan, Tailoring the structure of lipids, oleogels and fat replacers by different approaches for solving the trans-fat issue-A review, Foods, 2021, 10, 1376 CrossRef CAS PubMed.
- A. K. Mauro, D. M. Berdahl, N. Khurshid, L. Clemente, A. C. Ampey, D. M. Shah, I. M. Bird and D. S. Boeldt, Conjugated linoleic acid improves endothelial Ca2+signaling by blocking growth factor and cytokine-mediated Cx43 phosphorylation, Mol. Cell. Endocrinol., 2020, 510, 110814 CrossRef CAS PubMed.
- I. J. Onakpoya, P. P. Posadzki, L. K. Watson, L. A. Davies and E. Ernst, The efficacy of long-term conjugated linoleic acid (CLA) supplementation on body composition in overweight and obese individuals: a systematic review and meta-analysis of randomized clinical trials, Eur. J. Nutr., 2012, 51, 127–134 CrossRef CAS PubMed.
- Y. Park, Conjugated linoleic acid in human health effects on weight control, J Nutr. Prev. Treat. Abdom. Obes., 2014, 1, 429–446 Search PubMed.
- K. Bergstrom, X. Shan, D. Casero, A. Batushansky, V. Lagishetty, J. P. Jacobs, C. Hoover, Y. Kondo, B. J. Shao, L. Gao, W. Zandberg, B. Noyovitz, J. M. McDaniel, D. L. Gibson, S. Pakpour, N. Kazemian, S. McGee, C. W. Houchen, C. V. Rao, T. M. Griffin, J. L. Sonnenburg, R. P. McEver, J. Braun and L. J. Xia, Proximal colon-derived O-glycosylated mucus encapsulates and modulates the microbiota, Science, 2020, 370, 467–472 CrossRef CAS PubMed.
- T. Otani and M. Furuse, Tight junction structure and function revisited, Trends Cell Biol., 2020, 30, 805–817 CrossRef CAS PubMed.
- J. Wang, H. Chen, B. Yang, Z. Gu, H. Zhang, W. Chen and Y. Q. Chen,
Lactobacillus plantarum ZS2058 produces CLA to ameliorate DSS-induced acute colitis in mice, RSC Adv., 2016, 6, 14457–14464 RSC.
- L. Shi, Y. Dai, B. Y. Jia, Y. F. Han, Y. Guo, T. H. Xie, J. L. Liu, X. Tan, P. H. Ding and J. X. Li, The inhibitory effects of Qingchang Wenzhong granule on the interactive network of inflammation, oxidative stress, and apoptosis in rats with dextran sulfate sodium-induced colitis, J. Cell. Biochem., 2019, 120, 9979–9991 CrossRef CAS PubMed.
- K. Chinnadurai, H. K. Kanwal, A. K. Tyagi, C. Stanton and P. Ross, High conjugated linoleic acid enriched ghee (clarified butter) increases the antioxidant and antiatherogenic potency in female Wistar rats, Lipids Health Dis., 2013, 12, 1–9 CrossRef PubMed.
- R. T. Zuo, Q. H. Ai, K. S. Mai and W. Xu, Effects of conjugated linoleic acid on growth, non-specific immunity, antioxidant capacity, lipid deposition and related gene expression in juvenile large yellow croaker Larmichthys crocea fed soyabean oil-based diets, Br. J. Nutr., 2013, 110, 1220–1232 CrossRef CAS PubMed.
- S. Borniquel, C. Jädert and J. O. Lundberg, Dietary conjugated linoleic acid activates PPARγ and the intestinal trefoil factor in SW480 cells and mice with dextran sulfate sodium-induced colitis, J. Nutr., 2012, 142, 2135–2140 CrossRef CAS PubMed.
- J. Bassaganya-Riera, M. Viladomiu, M. Pedragosa, C. De Simone, A. Carbo, R. Shaykhutdinov, C. Jobin, J. C. Arthur, B. A. Corl, H. Vogel, M. Storr and R. Hontecillas, Probiotic bacteria produce conjugated linoleic acid locally in the gut that targets macrophage PPAR γ to suppress colitis, PLoS One, 2012, 7, e31238 CrossRef CAS.
- S. Fujio-Vejar, Y. Vasquez, P. Morales, F. Magne, P. Vera-Wolf, J. A. Ugalde, P. Navarrete and M. Gotteland, The gut microbiota of healthy chilean subjects reveals a high abundance of the phylum Verrucomicrobia, Front. Microbiol., 2017, 8, 1221 CrossRef PubMed.
- P. J. Torres, M. Siakowska, B. Banaszewska, L. Pawelczyk, A. J. Duleba, S. T. Kelley and V. G. Thackray, Gut microbial diversity in women with polycystic ovary syndrome correlates with hyperandrogenism, J. Clin. Endocrinol. Metab., 2018, 103, 1502–1511 CrossRef PubMed.
- L. Chen, M. M. Sun, W. Wu, W. J. Yang, X. S. Huang, Y. Xiao, C. Y. Ma, L. Q. Xu, S. X. Yao, Z. J. Liu and Y. Z. Cong, Microbiota metabolite butyrate differentially regulates Th1 and Th17 cells’ differentiation and function in induction of colitis, Inflamm. Bowel Dis., 2019, 25, 1450–1461 CrossRef PubMed.
- F. J. Ryan, A. M. Ahern, R. S. Fitzgerald, E. J. Laserna-Mendieta, E. M. Power, A. G. Clooney, K. W. O'Donoghue, P. J. McMurdie, S. Iwai, A. Crits-Christoph, D. Sheehan, C. Moran, B. Flemer, A. L. Zomer, A. Fanning, J. O'Callaghan, J. Walton, A. Temko, W. Stack, L. Jackson, S. A. Joyce, S. Melgar, T. Z. DeSantis, J. T. Bell, F. Shanahan and M. J. Claesson, Colonic microbiota is associated with inflammation and host epigenomic alterations in inflammatory bowel disease, Nat. Commun., 2020, 11, 1512 CrossRef CAS.
- S. J. Hur and Y. Park, Effect of conjugated linoleic acid on bone formation and rheumatoid arthritis, Eur. J. Pharmacol., 2007, 568, 16–24 CrossRef CAS PubMed.
- J. A. Muhlenbeck, J. M. Olson, A. B. Hughes and M. E. Cook, Conjugated linoleic acid isomers Trans-10, Cis-12 and Cis-9, Trans-11 prevent collagen-induced arthritis in a direct comparison, Lipids, 2018, 53, 689–698 CrossRef CAS PubMed.
- L. Ye, B. Jiang, J. Deng, J. Du, W. Xiong, Y. F. Guan, Z. Y. Wen, K. Z. Huang and Z. Huang, IL-37 alleviates rheumatoid arthritis by suppressing IL-17 and IL-17-triggering cytokine production and limiting Th17 cell proliferation, J. Immunol., 2015, 194, 5110–5119 CrossRef CAS PubMed.
- J. A. Muhlenbeck, D. E. Butz, J. M. Olson, D. Uribe-Cano and M. E. Cook, Dietary conjugated linoleic acid-c9t11 prevents collagen-induced arthritis, whereas conjugated linoleic acid-t10c12 increases arthritic severity, Lipids, 2017, 52, 303–314 CrossRef CAS PubMed.
- S. M. Huebner, J. M. Olson, J. P. Campbell, J. W. Bishop, P. M. Crump and M. E. Cook, Low dietary c9t11-conjugated linoleic acid intake from dairy fat or supplements reduces inflammation in collagen-induced arthritis, Lipids, 2016, 51, 807–819 CrossRef CAS.
- S. M. Huebner, J. M. Olson, J. P. Campbell, J. W. Bishop, P. M. Crump and M. E. Cook, Dietary trans-10, cis-12 CLA reduces murine collagen-induced arthritis in a dose-dependent manner, J. Nutr., 2014, 144, 177–184 CrossRef CAS.
- S. M. Huebner, J. P. Campbell, D. E. Butz, T. G. Fulmer, A. Gendron-Fitzpatrick and M. E. Cook, Individual isomers of conjugated linoleic acid reduce inflammation associated with established collagen-induced arthritis in DBA/1 mice, J. Nutr., 2010, 140, 1454–1461 CrossRef CAS.
- J. M. Olson, A. W. Haas, J. Lor, H. S. McKee and M. E. Cook, A comparison of the anti-inflammatory effects of Cis-9, Trans-11 conjugated linoleic acid to celecoxib in the collagen-induced arthritis model, Lipids, 2017, 52, 151–159 CrossRef CAS PubMed.
- Z. F. Fang, L. Z. Li, H. Zhang, J. X. Zhao, W. W. Lu and W. Chen, Gut microbiota, probiotics, and their interactions in prevention and treatment of atopic dermatitis: A review, Front. Immunol., 2021, 12, 720393 CrossRef CAS PubMed.
- L. Tang, X. L. Li, Z. X. Deng, Y. Xiao, Y. H. Cheng, J. Li and H. Ding, Conjugated linoleic acid attenuates 2,4-dinitrofluorobenzene-induced atopic dermatitis in mice through dual inhibition of COX-2/5-LOX and TLR4/NF-κB signaling, J. Nutr. Biochem., 2020, 81, 108379 CrossRef CAS PubMed.
- X. Sun, J. Zhang, A. K. H. MacGibbon, P. Black and G. W. Krissansen, Bovine milk fat enriched in conjugated linoleic and vaccenic acids attenuates allergic dermatitis in mice, Clin. Exp. Allergy, 2011, 41, 729–738 CrossRef CAS.
- T. Nakanishi, Y. Tokunaga, M. Yamasaki, L. Erickson and S. Kawahara, Orally administered conjugated linoleic acid ameliorates allergic dermatitis induced by repeated applications of oxazolone in mice, Anim. Sci. J., 2016, 87, 1554–1561 CrossRef CAS.
- Z. Paštar, J. Lipozenčić and S. Ljubojević, Etiopathogenesis of atopic dermatitis-an overview, Acta Dermatovenerol. Croat., 2005, 13, 54–62 Search PubMed.
- C. D. Sadik, T. Sezin and N. D. Kim, Leukotrienes orchestrating allergic skin inflammation, Exp. Dermatol., 2013, 22, 705–709 CrossRef CAS PubMed.
- L. D. Whigham, E. B. Cook, J. L. Stahl, R. Saban, D. E. Bjorling, M. W. Pariza and M. E. Cook, CLA reduces antigen-induced histamine and PGE(2) release from sensitized guinea pig tracheae, Am. J. Physiol. Regul. Integr. Comp. Physiol., 2001, 280, R908–R912 CrossRef CAS PubMed.
- L. Tang, X. Q. Cao, X. L. Li and H. Ding, Topical application with conjugated linoleic acid ameliorates 2, 4-dinitrofluorobenzene-induced atopic dermatitis-like lesions in BALB/c mice, Exp. Dermatol., 2021, 30, 237–248 CrossRef CAS PubMed.
- D. A. Yanes and J. L. Mosser-Goldfarb, Emerging therapies for atopic dermatitis: The prostaglandin/leukotriene pathway, J. Am. Acad. Dermatol., 2018, 78, S71–S75 CrossRef CAS PubMed.
- C. G. Kim, M. Kang, Y. H. Lee, W. G. Min, Y. H. Kim, S. J. Kang, C. H. Song, S. J. Park, J. H. Park, C. H. Han, Y. J. Lee and S. K. Ku, Bathing Effects of various seawaters on allergic (atopic) dermatitis-like skin lesions induced by 2,4-Dinitrochlorobenzene in hairless mice, Evidence-Based Complementary Altern. Med., 2015, 2015, 179185 Search PubMed.
- A. A. Wendel, A. Purushotham, L. F. Liu and M. A. Belury, Conjugated linoleic acid induces uncoupling protein 1 in white adipose tissue of ob/ob mice, Lipids, 2009, 44, 975–982 CrossRef CAS PubMed.
- W. Shen, C. C. Chuang, K. Martinez, T. Reid, J. M. Brown, L. Xi, L. Hixson, R. Hopkins, J. Starnes and M. McIntosh, Conjugated linoleic acid reduces adiposity and increases markers of browning and inflammation in white adipose tissue of mice, J. Lipid Res., 2013, 54, 909–922 CrossRef CAS PubMed.
- D. K. Dahiya-Renuka and A. K. Puniya, Conjugated linoleic acid enriched skim milk prepared with Lactobacillus fermentum DDHI27 endorsed antiobesity in mice, Future Microbiol., 2018, 13, 1007–1020 CrossRef PubMed.
- A. Yeganeh, P. Zahradka and C. G. Taylor, Trans-10, cis-12 conjugated linoleic acid (t10-c12 CLA) treatment and caloric restriction differentially affect adipocyte cell turnover in obese and lean mice, J. Nutr. Biochem., 2017, 49, 123–132 CrossRef CAS.
- W. Shen, J. Baldwin, B. Collins, L. Hixson, K. T. Lee, T. Herberg, J. Stames, P. Cooney, C. C. Chuang, R. Hopkins, T. Reid, S. Gupta and M. McIntosh, Low level of trans-10, cis-12 conjugated linoleic acid decreases adiposity and increases browning independent of inflammatory signaling in overweight Sv129 mice, J. Nutr. Biochem., 2015, 26, 616–625 CrossRef CAS PubMed.
- A. Kennedy, K. Martinez, S. Schmidt, S. Mandrup, K. LaPoint and M. McIntosh, Antiobesity mechanisms of action of conjugated linoleic acid, J. Nutr. Biochem., 2010, 21, 171–179 CrossRef CAS PubMed.
- C. P. B. Furlan, R. da Silva Marineli and M. R. M. Júnior, Conjugated linoleic acid and phytosterols counteract obesity induced by high-fat diet, Food Res. Int., 2013, 51, 429–435 CrossRef CAS.
- O. Gudmundsen, H. Blankson, J. A. Stakkestad, H. Fagertun, E. Thom and J. Wadstein, Conjugated linoleic acid reduces body fat mass in overweight and obese humans, J. Nutr., 2000, 130, 2943–2948 CrossRef PubMed.
- J. M. Gaullier, J. Halse, K. Høye, K. Kristiansen, H. Fagertun, H. Vik and O. Gudmundsen, Conjugated linoleic acid supplementation for 1 y reduces body fat mass in healthy overweight humans, Am. J. Clin. Nutr., 2004, 79, 1118–1125 CrossRef CAS PubMed.
- A. C. Watras, A. C. Buchholz, R. N. Close, Z. Zhang and D. A. Schoeller, The role of conjugated linoleic acid in reducing body fat and preventing holiday weight gain, Int. J. Obes., 2007, 31, 481–487 CrossRef CAS PubMed.
- R. N. Close, D. A. Schoeller, A. C. Watras and E. H. Nora, Conjugated linoleic acid supplementation alters the 6-mo change in fat oxidation during sleep, Am. J. Clin. Nutr., 2007, 86, 797–804 CrossRef CAS.
- M. M. J. W. Kamphuis, M. P. G. M. Lejeune, W. H. M. Saris and M. S. Westerterp-Plantenga, The effect of conjugated linoleic acid supplementation after weight loss on body weight regain, body composition, and resting metabolic rate in overweight subjects, Int. J. Obes., 2003, 27, 840–847 CrossRef CAS PubMed.
- L. E. Norris, A. L. Collene, M. L. Asp, J. C. Hsu, L. F. Liu, J. R. Richardson, D. Li, D. Bell, K. Osei and R. D. Jackson, Comparison of dietary conjugated linoleic acid with safflower oil on body composition in obese postmenopausal women with type 2 diabetes mellitus, Am. J. Clin. Nutr., 2009, 90, 468–476 CrossRef CAS PubMed.
- R. F. Carvalho, S. K. Uehara and G. Rosa, Microencapsulated conjugated linoleic acid associated with hypocaloric diet reduces body fat in sedentary women with metabolic syndrome, Vasc. Health Risk Manage., 2012, 2012, 661–667 CrossRef.
- S. C. Chen, Y. H. Lin, H. P. Huang, W. L. Hsu, J. Y. Houng and C. K. Huang, Effect of conjugated linoleic acid supplementation on weight loss and body fat composition in a Chinese population, Nutrition, 2012, 28, 559–565 CrossRef CAS.
- S. E. Steck, A. M. Chalecki, P. Miller, J. Conway, G. L. Austin, J. W. Hardin, C. D. Albright and T. Philippe, Conjugated linoleic acid supplementation for twelve weeks increases lean body mass in obese humans, J. Nutr., 2007, 137, 1188–1193 CrossRef CAS PubMed.
- J. M. Gaullier, J. Halse, K. Høye, K. Kristiansen, H. Fagertun, H. Vik and O. Gudmundsen, Supplementation with conjugated linoleic acid for 24 months is well tolerated by and reduces body fat mass in healthy, overweight humans, J. Nutr., 2005, 135, 778–784 CrossRef CAS.
- C. Pinkoski, P. D. Chilibeck, D. G. Candow, D. Esliger, J. B. Ewaschuk, M. Facci, J. P. Farthing and G. A. Zello, The effects of conjugated linoleic acid supplementation during resistance training, Med. Sci. Sports Exercise, 2006, 38, 339–348 CrossRef CAS PubMed.
- N. M. Racine, A. C. Watras, A. L. Carrel, D. B. Allen, J. J. McVean, R. R. Clark, A. R. O'Brien, M. O'Shea, C. E. Scott and D. A. Schoeller, Effect of conjugated linoleic acid on body fat accretion in overweight or obese children, Am. J. Clin. Nutr., 2010, 91, 1157–1164 CrossRef CAS PubMed.
- E. V. Lambert, J. H. Goedecke, K. Bluett, K. Heggie, A. Claassen, D. E. Rae, S. West, J. Dugas, L. Dugas and S. Meltzer, Conjugated linoleic acid versus high-oleic acid sunflower oil: effects on energy metabolism, glucose tolerance, blood lipids, appetite and body composition in regularly exercising individuals, Br. J. Nutr., 2007, 97, 1001–1011 CrossRef CAS PubMed.
- A. S. Ribeiro, F. L. C. Pina, S. R. Dodero, D. R. P. Silva, B. J. Schoenfeld, P. Sugihara, R. R. Fernandes, D. S. Barbosa, E. S. Cyrino and J. Tirapegui, Effect of conjugated linoleic acid associated with aerobic exercise on body fat and lipid profile in obese women: a randomized, double-blinded, and placebo-controlled trial, Int. J. Sport Nutr. Exercise Metab., 2016, 26, 135–144 CAS.
- S. Venkatramanan, S. V. Joseph, P. Y. Chouinard, H. Jacques, E. R. Farnworth and P. J. H. Jones, Milk enriched with conjugated linoleic acid fails to alter blood lipids or nody composition in moderately overweight, borderline hyperlipidemic individuals, J. Am. Coll. Nutr., 2010, 29, 152–159 CrossRef CAS.
- T. M. Larsen, S. Toubro, O. Gudmundsen and A. Astrup, Conjugated linoleic acid supplementation for 1 y does not prevent weight or body fat regain, Am. J. Clin. Nutr., 2006, 83, 606–612 CrossRef CAS PubMed.
- J. S. W. Taylor, S. R. P. Williams, R. Rhys, P. James and M. P. Frenneaux, Conjugated linoleic acid impairs endothelial function, Arterioscler., Thromb., Vasc. Biol., 2005, 26, 307–312 CrossRef.
- E. Mądry, I. Chudzicka-Strugała, K. Grabańska-Martyńska, K. Malikowska, P. Grebowiec, A. Lisowska and J. Walkowiak, Twelve weeks CLA supplementation decreases the hip circumference in overweight and obese women. A double-blind, randomized, placebo-controlled trial, Acta Sci. Pol., Technol. Aliment., 2016, 15, 107–113 CrossRef.
- S. Bulut, E. Bodur, R. Colak and H. Turnagol, Effects of conjugated linoleic acid supplementation and exercise on post-heparin lipoprotein lipase, butyrylcholinesterase, blood lipid profile and glucose metabolism in young men, Chem.-Biol. Interact., 2013, 203, 323–329 CrossRef CAS.
- U. Riserus, L. Berglund and B. Vessby, Conjugated linoleic acid (CLA) reduced abdominal adipose tissue in obese middle-aged men with signs of the metabolic syndrome: a randomised controlled trial, Int. J. Obes. Relat. Metab. Disord., 2001, 25, 1129–1135 CrossRef CAS.
- U. L. F. Riserus, P. Arner, K. Brismar and B. Vessby, Treatment with dietary trans10cis12 conjugated linoleic acid causes isomer-specific insulin resistance in obese men with the metabolic syndrome, Diabetes Care, 2002, 25, 1516–1521 CrossRef CAS PubMed.
- U. Riseérus, S. Basu, S. Jovinge, G. N. Fredrikson, J. Ärnlöv and B. Vessby, Supplementation with conjugated linoleic acid causes isomer-dependent oxidative stress and elevated C-reactive protein: a potential link to fatty acid-induced insulin resistance, Circulation, 2002, 106, 1925–1929 CrossRef.
- U. Risérus, B. Vessby, P. Arner and B. Zethelius, Supplementation with trans 10 cis 12-conjugated linoleic acid induces hyperproinsulinaemia in obese men: close association with impaired insulin sensitivity, Diabetologia, 2004, 47, 1016–1019 CrossRef PubMed.
- P. Dandona, A. Aljada, A. Chaudhuri, P. Mohanty and R. Garg, Metabolic syndrome a comprehensive perspective based on interactions between obesity, diabetes, and inflammation, Circulation, 2005, 111, 1448–1454 CrossRef PubMed.
- J. M. Bruun, A. S. Lihn, S. B. Pedersen and B. Richelsen, Monocyte chemoattractant protein-1 release is higher in visceral than subcutaneous human adipose tissue (AT): implication of macrophages resident in the AT, J. Clin. Endocrinol. Metab., 2005, 90, 2282–2289 CrossRef CAS PubMed.
- K. Song, I. B. Song, H. J. Gu, J. Y. Na, S. Kim, H. S. Yang, S. C. Lee, C. K. Huh and J. Kwon, Anti-diabetic Effect of fermented milk containing conjugated linoleic acid on type II diabetes mellitus, Korean J. Food Sci. Anim. Resour., 2016, 36, 170–177 CrossRef.
- F. Moloney, S. Toomey, E. Noone, A. Nugent, B. Allan, C. E. Loscher and H. M. Roche, Antidiabetic effects of cis-9, trans-11-conjugated linoleic acid may be mediated via anti-inflammatory effects in white adipose tissue, Diabetes, 2007, 56, 574–582 CrossRef CAS.
- N. Y. Park, H. Shin and Y. Lim, Effect of dietary CLA supplementation on renal inflammation in diabetic mice, Food Sci. Biotechnol., 2014, 23, 1623–1628 CrossRef CAS.
- X. L. Li, J. J. Cui, W. S. Zheng, J. L. Zhang, R. Li, X. L. Ma, M. Lin, H. H. Guo, C. Li and X. Y. Yu, Bicyclol alleviates atherosclerosis by manipulating gut microbiota, Small, 2022, 18, 2105021 CrossRef CAS.
- S. Toomey, B. Harhen, H. M. Roche, D. Fitzgerald and O. Belton, Profound resolution of early atherosclerosis with conjugated linoleic acid, Atherosclerosis, 2006, 187, 40–49 CrossRef CAS PubMed.
- E. Stachowska, A. Siennicka, M. Baskiewcz-Halasa, J. Bober, B. Machalinski and D. Chlubek, Conjugated linoleic acid isomers may diminish human macrophages adhesion to endothelial surface, Int. J. Food Sci. Nutr., 2012, 63, 30–35 CrossRef CAS.
- J. Gosling, S. Slaymaker, L. Gu, S. Tseng, C. H. Zlot, S. G. Young, B. J. Rollins and I. F. Charo, MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B, J. Clin. Invest., 1999, 103, 773–778 CrossRef CAS PubMed.
- S. McClelland, C. Cox, R. O'Connor, M. de Gaetano, C. McCarthy, L. Cryan and O. Belton, Conjugated linoleic acid suppresses the migratory and inflammatory phenotype of the monocyte/macrophage cell, Atherosclerosis, 2010, 211, 96–102 CrossRef CAS PubMed.
- S. Toomey, H. Roche, D. Fitzgerald and O. Belton, Regression of pre-established atherosclerosis in the apoE−/− mouse by conjugated linoleic acid, Biochem. Soc. Trans., 2003, 31, 1075–1079 CrossRef CAS PubMed.
- M. Franczyk-Zarów, R. B. Kostogrys, B. Szymczyk, J. Jawień, M. Gajda, T. Cichocki, L. Wojnar, S. Chlopicki and P. M. Pisulewski, Functional effects of eggs, naturally enriched with conjugated linoleic acid, on the blood lipid profile, development of atherosclerosis and composition of atherosclerotic plaque in apolipoprotein E and low-density lipoprotein receptor double-knockou mice (apoE/LDLR−/−), Br. J. Nutr., 2008, 99, 49–58 CrossRef PubMed.
- C. Mccarthy, N. T. Lieggi, D. Barry, D. Mooney, M. D. Gaetano, W. G. James, S. Mcclelland, M. C. Barry, L. Escoubet-Lozach and A. C. Li, Macrophage PPAR gamma Co-activator-1 alpha participates in repressing foam cell formation and atherosclerosis in response to conjugated linoleic acid, EMBO Mol. Med., 2013, 5, 1443–1457 CrossRef CAS.
- J. Ecker, T. Langmann, G. Liebisch and G. Schmitz, Isomer specific effects of conjugated linoleic acid on macrophage ABCG1 transcription by a SREBP-1c dependent mechanism, Biochem. Biophys. Res. Commun., 2007, 352, 805–811 CrossRef CAS PubMed.
- M. De Gaetano, K. Alghamdi, S. Marcone and O. Belton, Conjugated linoleic acid induces an atheroprotective macrophage MΦ2 phenotype and limits foam cell formation, J. Inflammation, 2015, 12, 1–14 CrossRef PubMed.
- P. L. Mitchell, T. K. Karakach, D. L. Currie and R. S. Mcleod, t-10, c-12 CLA dietary supplementation inhibits atherosclerotic lesion development despite adverse cardiovascular and hepatic metabolic marker profiles, PLoS One, 2012, 7, e52634 CrossRef CAS PubMed.
- M. H. Cooper, J. R. Miller, P. L. Mitchell, D. L. Currie and R. S. McLeod, Conjugated linoleic acid isomers have no effect on atherosclerosis and adverse effects on lipoprotein and liver lipid metabolism in apoE−/− mice fed a high-cholesterol diet, Atherosclerosis, 2008, 200, 294–302 CrossRef CAS.
- R. B. Kostogrys, M. Franczyk-Żarów, E. Maslak, M. Gajda and S. Chłopicki, Effects of margarine supplemented with T10C12 and C9T11 CLA on atherosclerosis and steatosis in apoE/LDLR−/− mice, J. Nutr. Health, 2012, 16, 482–490 CAS.
- H. S. Park, J. H. Ryu, Y. L. Ha and J. H. Y. Park, Dietary conjugated linoleic acid (CLA) induces apoptosis of colonic mucosa in 1,2-dimethylhydrazine-treated rats: a possible mechanism of the anticarcinogenic effect by CLA, Br. J. Nutr., 2001, 86, 549–555 CrossRef CAS PubMed.
- R. Shiraishi, R. Iwakiri, T. Fujise, T. Kuroki, T. Kakimoto, T. Takashima, Y. Sakata, S. Tsunada, Y. Nakashima, T. Yanagita and K. Fujimoto, Conjugated linoleic acid suppresses colon carcinogenesis in azoxymethane-pretreated rats with long-term feeding of diet containing beef tallow, J. Gastroenterol., 2010, 45, 625–635 CrossRef CAS PubMed.
- N. Mandir and R. A. Goodlad, Conjugated linoleic acids differentially alter polyp number and diameter in the Apc(min/+) mouse model of intestinal cancer, Cell Proliferation, 2008, 41, 279–291 CrossRef CAS.
- M. B. H. Petrik, B. T. Johnson, J. Whelan, M. F. McEntee and M. G. Obukowicz, Highly unsaturated (n-3) fatty acids, but not α-linolenic,conjugated linoleic or γ-linolenic acids, reduce tumorigenesis in ApcMin/+ mice, J. Nutr., 2000, 130, 2434–2443 CrossRef CAS PubMed.
- S. M. Soel, O. S. Choi, M. H. Bang, J. H. Y. Park and W. K. Kim, Influence of conjugated linoleic acid isomers on the metastasis of colon cancer cells in vitro and in vivo, J. Nutr. Biochem., 2007, 18, 650–657 CrossRef CAS PubMed.
- K. H. Kim and H. S. Park, Dietary supplementation of conjugated linoleic acid reduces colon tumor incidence in DMH-treated rats by increasing apoptosis with modulation of biomarkers, Nutrition, 2003, 19, 772–777 CrossRef CAS PubMed.
- N. P. Evans, S. A. Misyak, E. M. Schmelz, A. J. Guri, R. Hontecillas and J. Bassaganya-Riera, Conjugated linoleic acid ameliorates inflammation-induced colorectal cancer in mice through activation of PPARgamma, J. Nutr., 2010, 140, 515–521 CrossRef CAS.
- Y. Yasui, R. Suzuki, H. Kohno, S. Miyamoto, F. Beppu, M. Hosokawa, K. Miyashita and T. Tanaka, 9trans, 11trans conjugated linoleic acid inhibits the development of azoxymethane-induced colonic aberrant crypt foci in rats, Nutr. Cancer, 2007, 59, 82–91 CrossRef CAS.
- M. Biaek, A. Biaek and M. Czauderna, Conjugated linoleic acid isomers affect profile of lipid compounds and intensity of their oxidation in heart of rats with chemically-induced mammary tumors-preliminary study, Nutrients, 2019, 11, 2032 CrossRef.
- N. E. Hubbard, D. Lim, L. Summers and K. L. Erickson, Reduction of murine mammary tumor metastasis by conjugated linoleic acid, Cancer Lett., 2000, 150, 93–100 CrossRef CAS.
- F. Lavillonniere, V. Chajes, J. C. Martin, J. L. Sebedio, C. Lhuillery and P. Bougnoux, Dietary purified cis-9,trans-11 conjugated linoleic acid isomer has anticarcinogenic properties in chemically induced mammary tumors in rats, Nutrition, 2003, 45, 190–194 CAS.
- Y. Zeng, P. Liu, X. Yang, H. Li, H. Li, Y. Guo, X. Meng and X. Liu, The dietary c9,t11-conjugated linoleic acid enriched from butter reduces breast cancer progression in vivo, J. Food Biochem., 2020, 44, e13163 CrossRef PubMed.
- C. Ip and J. A. Scimeca, Conjugated linoleic acid and linoleic acid are distinctive modulators of mammary carcinogenesis, Nutr. Cancer, 1997, 27, 131–135 CrossRef CAS PubMed.
- Y. Fujita, K. Kano, S. Kishino, T. Nagao, X. Shen, C. Sato, H. Hatakeyama, Y. Ota, S. Niibori and A. Nomura, Dietary cis-9, trans-11-conjugated linoleic acid reduces amyloid β-protein accumulation and upregulates anti-inflammatory cytokines in an Alzheimer's disease mouse model, Sci. Rep., 2021, 11, 9749 CrossRef CAS PubMed.
- F. L. Heppner, R. M. Ransohoff and B. Becher, Immune attack: the role of inflammation in Alzheimer disease, Nat. Rev. Neurosci., 2015, 16, 358–372 CrossRef CAS PubMed.
- M. M. Agwa, D. A. Abdelmonsif, S. N. Khattab and S. Sabra, Self- assembled lactoferrin-conjugated linoleic acid micelles as an orally active targeted nanoplatform for Alzheimer's disease, Int. J. Biol. Macromol., 2020, 162, 246–261 CrossRef CAS PubMed.
- L. Cigliano, M. S. Spagnuolo, F. Boscaino, I. Ferrandino, A. Monaco, T. Capriello, E. Cocca and P. Bergamo, Dietary supplementation with fish oil or conjugated linoleic acid relieves depression markers in mice by modulation of the Nrf2 pathway, Mol. Nutr. Food Res., 2019, 63, 1900243 CrossRef CAS.
- K. Hashimoto, Essential role of Keap1-Nrf2 signaling in mood disorders: overview and future perspective, Front. Pharmacol., 2018, 9, 1182 CrossRef CAS.
- T. Wang and H. G. Lee, Advances in research on cis-9, trans-11 conjugated linoleic acid: A major functional conjugated linoleic acid isomer, Crit. Rev. Food Sci. Nutr., 2014, 55, 720–731 CrossRef.
- N. S. Kelley, N. E. Hubbard and K. L. Erickson, Conjugated linoleic acid isomers and cancer, J. Nutr., 2007, 137, 2599–2607 CrossRef CAS PubMed.
- M. W. Pariza, Y. Park and M. E. Cook, Mechanisms of action of conjugated linoleic acid: evidence and speculation, Proc. Soc. Exp. Biol. Med., 2000, 223, 8–13 CAS.
- J. M. Gaullier, G. Berven, H. Blankson and O. Gudmundsen, Clinical trial results support a preference for using CLA preparations enriched with two isomers rather than four isomers in human studies, Lipids, 2002, 37, 1019–1025 CrossRef CAS.
- C. G. Candela, El papel del CLA o ácido linoleico conjugado sobre la masa grasa corporal, Nutr. Clin. Diet. Hosp., 2004, 24, 55–60 Search PubMed.
- K. W. J. Wahle, S. D. Heys and D. Rotondo, Conjugated linoleic acids: are they beneficial or detrimental to health?, Prog. Lipid Res., 2004, 43, 553–587 CrossRef CAS PubMed.
- A. Dilzer and Y. Park, Implication of conjugated linoleic acid (CLA) in human health, Crit. Rev. Food Sci. Nutr., 2012, 52, 488–513 CrossRef CAS PubMed.
- N. D. M. Jenkins, S. L. Buckner, R. B. Baker, H. C. Bergstrom, K. C. Cochrane, J. P. Weir, T. J. Housh and J. T. Cramer, Effects of 6 weeks of aerobic exercise combined with conjugated linoleic acid on the physical working capacity at fatigue threshold, J Strength Cond Res, 2014, 28, 2127–2135 CrossRef PubMed.
- B. López-Plaza, L. M. Bermejo, T. K. Weber, P. Parra, F. Serra, M. Hernández, S. P. Milla and C. Gómez-Candela, Effects of milk supplementation with conjugated linoleic acid on weight control and body composition in healthy overweight people, Nutr. Hosp., 2013, 28, 2090–2098 Search PubMed.
- L. D. Whigham, M. O'Shea, I. C. M. Mohede, H. P. Walaski and R. L. Atkinson, Safety profile of conjugated linoleic acid in a 12-month trial
in obese humans, Food Chem. Toxicol., 2004, 42, 1701–1709 CrossRef CAS PubMed.
- B. K. Chai, M. Al-Shagga, Y. Pan, S. M. Then, K. N. Ting, H. S. Loh and S. K. Mohankumar, Cis-9, Trans-11 conjugated linoleic acid reduces phosphoenolpyruvate carboxykinase expression and hepatic glucose production in HepG2 cells, Lipids, 2019, 54, 369–379 CrossRef CAS PubMed.
- D. Vyas, A. K. G. Kadegowda and R. A. Erdman, Dietary conjugated linoleic acid and hepatic steatosis: species-specific effects on liver and adipose lipid metabolism and gene expression, J. Nutr. Metab., 2012, 2012, 932928 Search PubMed.
- D. M. Fedor, Y. Adkins, B. E. Mackey and D. S. Kelley, Docosahexaenoic acid prevents trans-10, cis-12-conjugated linoleic acid-induced nonalcoholic fatty liver disease in mice by altering expression of hepatic genes regulating fatty acid synthesis and oxidation, Metab. Syndr. Relat. Disord., 2012, 10, 175–180 CrossRef CAS PubMed.
- M. B. Stout, L. F. Liu and M. A. Belury, Hepatic steatosis by dietary-conjugated linoleic acid is accompanied by accumulation of diacylglycerol and increased membrane-associated protein kinase C & in mice, Mol. Nutr. Food Res., 2011, 55, 1010–1017 CrossRef CAS PubMed.
- R. B. Kostogrys, M. Franczyk-Żarów, E. Maślak, M. Gajda, Ł. Mateuszuk and S. Chłopicki, Conjugated linoleic acid has no effects on atherosclerosis but induces liver steatosis in apoE/LDLR mice fed a fructose-rich diet, J. Pre-Clin. Clin. Res., 2010, 4, 118–121 Search PubMed.
- J. Li, S. Viswanadha and J. J. Loor, Hepatic metabolic, inflammatory, and stress-related gene expression in growing mice consuming a low dose of Trans-10, cis-12-conjugated linoleic acid, J. Lipids, 2012, 2012, 571281 Search PubMed.
- L. Della-Casa, E. Rossi, C. Romanelli, L. Gibellini and A. Iannone, Effect of diets supplemented with different conjugated linoleic acid (CLA) isomers on protein expression in C57/BL6 mice, Genes Nutr., 2016, 11, 1–14 CrossRef CAS PubMed.
- M. Baldin, M. A. S. Gama, R. Dresch, K. J. Harvatine and D. E. Oliveira, A rumen unprotected conjugated linoleic acid supplement inhibits milk fat synthesis and improves energy balance in lactating goats, J. Anim. Sci., 2013, 91, 3305–3314 CrossRef CAS PubMed.
- K. J. Harvatine, M. M. Robblee, S. R. Thorn, Y. R. Boisclair and D. E. Bauman, Trans-10, cis-12 CLA dose-dependently inhibits milk fat synthesis without disruption of lactation in C57BL/6J mice, J. Nutr., 2014, 144, 1928–1934 CrossRef CAS PubMed.
- L. Bernard, C. Leroux and Y. Chilliard, Expression and nutritional regulation of lipogenic genes in the ruminant lactating mammary gland, Bioact. Compon. Milk, 2008, 606, 67–108 CAS.
- L. D. Granados-Rivera, O. Hernández-Mend, S. S. González-Muñoz, J. A. Burgueño-Ferreira, G. D. Mendoza-Martínez and C. M. Arriaga-Jordá, Effect of palmitic acid on the mitigation of milk fat depression syndrome caused by trans-10, cis-12-conjugated linoleic acid in grazing dairy cows, Arch. Anim. Nutr., 2017, 71, 428–440 CrossRef CAS PubMed.
- N. Masters, M. A. McGuire, K. A. Beerman, N. Dasgupta and M. K. McGuire, Maternal supplementation with CLA decreases milk fat in humans, Lipids, 2002, 37, 133–138 CrossRef CAS PubMed.
- S. A. Mosley, A. M. Shahin, J. Williams, M. A. Mcguire and M. K. Mcguire, Supplemental conjugated linoleic acid consumption does not influence milk macronutrient contents in all healthy lactating women, Lipids, 2007, 42, 723–729 CrossRef CAS PubMed.
- N. B. Leonard, Stability testing of nutraceuticals and functional foods, Handb. Nutraceuticals Funct. Foods, 2000, 13, 523–538 Search PubMed.
- Q. Chang, X. Zhou, C. Wu, X. Xu and Z. Jin, Preparation, characterization, water solubility, and targeted delivery of linear dextrin-conjugated linoleic acid inclusion complex, Starch/Staerke, 2015, 67, 521–527 CrossRef CAS.
- M. Jimenez, H. García and C. I. Beristain, Spray-dried encapsulation of Conjugated Linoleic Acid (CLA) with polymeric matrices, J. Sci. Food Agric., 2006, 86, 2431–2437 CrossRef CAS.
- M. P. Yurawecz, J. K. Hood, M. M. Mossoba, J. A. G. Roach and Y. Ku, Furan fatty acids determined as oxidation products of conjugated octadecadienoic acid, Lipids, 1995, 30, 595–598 CrossRef CAS PubMed.
- P. Shah, D. Bhalodia and P. Shelat, Nanoemulsion: a pharmaceutical review, Syst. Rev. Pharm., 2010, 1, 24–32 CrossRef CAS.
- D. Kim, J. H. Park, D. J. Kweon and G. D. Han, Bioavailability of nanoemulsified conjugated linoleic acid for an antiobesity effect, Int. J. Nanomed., 2013, 8, 451–459 Search PubMed.
- W. Heo, J. H. Kim, J. H. Pan and Y. J. Kim, Lecithin-based nano-emulsification improves the bioavailability of conjugated linoleic acid, J. Agric. Food Chem., 2016, 64, 1355–1360 CrossRef CAS PubMed.
- D. J. McClements, E. A. Decker and J. Weiss, Emulsion-based delivery systems for lipophilic bioactive components, J. Food Sci., 2007, 72, R109–R124 CrossRef CAS PubMed.
- Z. M. Gao, L. P. Zhu, X. Q. Yang, X. T. He, J. M. Wang, J. Guo, J. R. Qi, L. J. Wang and S. W. Yin, Soy lipophilic protein nanoparticles as a novel delivery vehicle for conjugated linoleic acid, Food Funct., 2014, 5, 1286–1293 RSC.
- D. W. Pitta, N. Indugu, B. Vecchiarelli, D. E. Rico and K. J. Harvatine, Alterations in ruminal bacterial populations at induction and recovery from diet-induced milk fat depression in dairy cows, J. Dairy Sci., 2017, 101, 295–309 CrossRef PubMed.
- B. Yang, H. Gao, C. Stanton, R. P. Ross, H. Zhang, Y. Q. Chen, H. Chen and W. Chen, Bacterial conjugated linoleic acid production and their applications, Prog. Lipid Res., 2017, 68, 26–36 CrossRef CAS PubMed.
- B. Yang, H. Chen, H. Gao, J. Wang, C. Stanton, R. P. Ross, H. Zhang and W. Chen,
Bifidobacterium breve CCFM683 could ameliorate DSS-induced colitis in mice primarily via conjugated linoleic acid production and gut microbiota modulation, J. Funct. Foods, 2018, 49, 61–72 CrossRef CAS.
- Y. Chen, Y. Jin, C. Stanton, R. P. Ross, J. Zhao, H. Zhan, B. Yang and W. Chen, Alleviation effects of Bifidobacterium breve on DSS-induced colitis depends on intestinal tract barrier maintenance and gut microbiota modulation, Eur. J. Nutr., 2021, 60, 369–387 CrossRef CAS PubMed.
- Y. Chen, B. Yang, C. Stanton, R. P. Ross, J. Zhao, H. Zhang and W. Chen,
Bifidobacterium pseudocatenulatum ameliorates DSS-Induced colitis by maintaining intestinal mechanical barrier, blocking proinflammatory cytokines, inhibiting TLR4/NF-κB signaling, and altering gut microbiota, J. Agric. Food Chem., 2021, 69, 1496–1512 CrossRef CAS PubMed.
- Y. Chen, H. Chen, J. Ding, C. Stanton, R. P. Ross, J. Zhao, H. Zhang, B. Yang and W. Chen,
Bifidobacterium longum ameliorates dextran sulfate sodium-induced colitis by producing conjugated linoleic acid, protecting intestinal mechanical barrier, restoring unbalanced gut microbiota, and regulating the toll-like receptor-4/nuclear factor-κB signaling pathway, J. Agric. Food Chem., 2021, 69, 14593–14608 CrossRef CAS PubMed.
|
This journal is © The Royal Society of Chemistry 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.