Skip to main content

Research progress in lipid metabolic regulation of bioactive peptides

Abstract

Hyperlipidemia poses a serious threat to human health and evaluating the ability of natural active substances to regulate disorders of lipid metabolism is the focus of food functionality research in recent years. Bioactive peptides are distinguished by their broad range of sources, high nutritional content, ease of absorption and use by the body, and ease of determining their sequences. Bioactive peptides have a wide range of potential applications in the area of medicines and food. The regulation of lipid metabolism disorder caused by bioactive peptides from different sources provides a reference for the development and research of bioactive peptides for lipid reduction.

Graphical Abstract

Introduction

Hyperlipidemia occurs when one or more lipid metabolic pathways in the blood and lipid content are deemed abnormal. It is a common cardiovascular disease that poses a serious threat to human health (Ross 2017). Increased levels of total cholesterol (TC), total triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and lower levels of high-density lipoprotein cholesterol (HDL-C) are known indicators of a lipid metabolic disorder (Ajeigbe et al. 2022). Abnormal changes in these indicators related to lipid levels may increase the organism’s risk of atherosclerosis, hypertension, cardiovascular diseases, diabetes, and other related metabolic disorders, and therefore, is a key for the prevention of chronic diseases like metabolic syndrome (Oluwajuyitan et al. 2021; Huang et al. 2022). Dietary modifications and an appropriate increase in exercise can be used to effectively treat early abnormalities of lipid metabolism. However, due to unhealthy diets and lifestyles, including high sugar, high-fat, and high-calorie diet and a sedentary lifestyle with a lack of exercise, it is difficult to improve lipid metabolism disorder only through dietary intervention and exercise, and these methods requiring high-self control show less compliance (Li et al. 2021).

Common clinical lipid drugs include statins (like lovastatin, simvastatin, rosuvastatin, fluvastatin, fluvastatin, and atorvastatin), fibrates (like clobetl, bento, and fenofibrate), fibrate drugs (like clobebetl and gefbetil), and bile acid chelators (i.e., ketamine, test lidine, and cholilumine), which reduce the lipid level in vivo but are ineffective against oxidative stress and fundamental effect of lipid metabolism. These anti-lowering lipid drugs usually exert side effects unrelated to the purpose of treatment, like gastrointestinal irritation, myolytic symptoms, and liver and kidney function damage (Durkar et al. 2014; Wa et al. 2019). As a result, several researchers are evaluating ways to improve or treat lipid metabolic problems using natural active ingredients.

Bioactive peptides are composed of 2–50 amino acid residues, which form structures ranging from dipeptides to complex linear and annular configurations with different combinations and arrangements (Stoye et al. 2017). The biological activities of proteins are realized through their encoded peptide sequences, which usually express unique physiological functions in food processing (from room to high temperature), enzymatic reaction, microbial fermentation, and other processes (Chakrabarti et al. 2018; Chalamaiah et al. 2018; Xu et al. 2022). The sources of bioactive peptides mainly include natural bioactive peptides found in organisms, the active peptides obtained by the enzymatic digestion of the proteins, and synthetic bioactive peptides. The activity of the bioactive peptides typically depends on the electrostatic charge, amino acid composition and sequences, peptide chain length, and hydrophobicity of the molecular surface (Chalamaiah et al. 2018). To date, various physiological functions of bioactive peptides have received extensive research attention. The discovery of biological activities like lipid-lowering, blood pressure-lowering, and antithrombotic properties provides theoretical support for related studies of bioactive peptides (Cian et al. 2015; Wu et al. 2015; Zheng et al. 2020). This review collates the roles and mechanisms of peptides obtained from beans, cereals and tubers, nuts, aquatic animals, seaweed, eggs and milk, livestock organs or tissues, and other substances in the regulation of lipid metabolism (Table 1), for further development of bioactive peptides as potential lipid-lowering functional food or medicine.

Table 1 Bioactive peptides from different sources regulate lipid metabolism

Lipid metabolism pathways

The homeostasis in lipid transport and metabolism is regulated by multiple signaling pathways, including the adenylate-activated protein kinase (AMPK), phosphatidylinositol-3-kinase (PI3K), protein kinase B (Akt), mitogen-activated protein kinase (MAPK), peroxisome proliferator-activated receptor (PPAR), and insulin signaling cascades. These pathways maintain the body’s lipid metabolic balance by exerting complex regulatory effects on each other. Among them, the PPAR and AMPK/SREBPs pathways with PPAR and AMPK as the core, respectively, play important regulatory roles in stabilizing the body’s lipid levels and lipid metabolism (Fig. 1) (Fogarty & Hardie 2010). The regulation of the PPAR signaling system is implicated in atherosclerosis, obesity, insulin resistance, and cancer, and is known to affect lipid and energy metabolism. The SREBP-1 protein is an important transcription factor in liver cells, controlling several genes involved in cholesterol and lipid metabolism. Additionally, microRNAs have been increasingly demonstrated to play a significant role in aberrant lipid metabolism and associated disorders by controlling the expression of genes. Many microRNAs can directly or indirectly regulate the expression of SREBPs and PPAR, thereby affecting lipid metabolism (Cagen et al. 2005; Fogarty & Hardie 2010; Fleischmann & Iynedjian 2000).

Fig. 1
figure 1

Diagram showing the lipid metabolism pathways with AMPK/SREBPs as the core

Cholesterol metabolism

The liver and the small intestine are the two most critical organs involved in the entire process of cholesterol metabolism (Ahmadi et al. 2018). The function of the liver includes the synthesis of cholesterol, conversion into bile acids, and lipid reabsorption and metabolism in the body. Cholesterol balance in the liver is collectively regulated by SREBPs and liver X receptors (LXRs), whereby the former regulates cholesterol absorption by the liver, while the latter regulates cholesterol excretion from the liver. The genes regulated downstream of SREBPs mainly include enzymes related to cholesterol synthesis, like hydroxymethylglutaryl coenzyme A synthase (HMGCS), hydroxy methylglutaryl CoA reductase (HMGCR), squalene synthase (SQS), farnesyl diphosphate synthase (FDPs), and low-density lipoprotein receptor (LDLR) (Fig. 2). The downstream regulatory genes of LXRs typically include ATP-binding cassette protein A1 (ABCA1), responsible for cholesterol excretion from the liver through the cholesterol 7 α-hydroxylase (CYP7A1) axis, which plays a key rate-limiting role in the conversion of cholesterol to bile acids, following which bile is discharged from the liver and enters the small intestine. Most of the bile acid returns to the liver through the circulation between the liver and intestines and is metabolized.

Fig. 2
figure 2

Cholesterol metabolism. HDL, high-density lipoprotein; LDL, low-density lipoprotein; VLDL, very low density lipoprotein; ACAT, acyl coenzyme A-cholesterol acyltransferases; LIPA, lysosomal acid lipase A; MTP, microsomal triglyceride transfer protein

The small intestine is an important organ for cholesterol absorption. When dietary and bile cholesterol excreted from the liver enter the intestinal cavity of the small intestine, Niemann pick C1 like 1 (NPC1L1) transports free cholesterol (FC) through vesicles for absorption from the intestinal cavity to the small intestine cells. After cholesterol is absorbed into the small intestinal cells through NPC1L1, some of it is transported back to the small intestinal cavity by the homologous protein of ATP-binding cassette transporter G5/8 (ABCG5/8), and subsequently converted into fecal cholesterol by intestinal bacteria and discharged from the body. The remaining cholesterol is converted into cholesteryl ester (CE) under the action of acetyl coenzyme A acetyltransferase 2 (ACAT2) and is incorporated into chylomicrons (CM) through the microsomal triglyceride transfer protein (MTP).

CM’s function is the movement of cholesterol absorbed by the small intestine to the somatic cells to fuel the body’s energy needs and to the liver cells for recirculation and metabolism (Liu et al. 2017). After the CM enters the blood circulation through the lymphatic vessels, apo C-II from HDL can hydrolyze the fat in the CM by lipoprotein lipase (LPL) on the capillary wall into CM residues which are reabsorbed by hepatocytes, and the cholesterol contained therein reenters the liver. The liver and small intestine interact and cooperate, and the liver-intestinal circulation promotes the metabolism and circulation of cholesterol.

TG metabolism

Two ways for the biosynthesis of long-chain fatty acids are known—one is catalyzing the conversion of acetyl-Coenzyme A (AcCoA) and malondialdehyde-Coenzyme A in the palmitate pathway by fatty acid synthetase (FAS), and the other is catalyzing the conversion of AcCoA in the malonyl-CoA pathway by the acetyl-CoA carboxylase (ACC) for the generation of TGs (van de Beek et al. 2017). Thus, reducing the synthesis of TGs in the plasma can reduce their levels at the source by inhibiting both the ACC and FAS activities. TG synthesis is positively regulated by carbohydrate reaction element-binding proteins (ChREBP) and SREBP-1c, and reduction in TG levels in liver tissues can also be achieved by inhibiting SREBP-1c expression, concomitantly reducing adipogenesis (Ge et al. 2011). TGs are subjected to adipose triglyceride lipase (ATGL), hormone-sensitive triglyceride lipase (HSL), and monoacylglycerol lipase (MGL) actions, hydrolyzed into glycerol followed by removal of trimolecular fatty acids. They are then transported via fatty acid transferase (FAT) and alkaline phosphatase (ALP) to enterocytes and re-synthesized TGs are absorbed and used by the body (Fig. 3). Thus, TG synthesis can be slowed down by inhibiting the activities of FAT and ALP. During TG metabolism, fat absorption and production can be slowed down not only by inhibiting the activity of related enzymes or stimulating related transcription factors but also by promoting fatty acid oxidation to a certain extent, reducing the internal TG levels.

Fig. 3
figure 3

Triglyceride metabolism. PAK, protein kinase A; ATGL, adipose triglyceride lipase; HSL, hormone-sensitive lipase; MGL, monoacylglycerol lipase

Lipid metabolism and intestinal microflora

Several microorganisms in the human gastrointestinal tract play a vital role in the health of the host (Catalkaya et al. 2020). Intestinal microbes are important mediators of diet, the external environment, and various metabolic processes of the body. Given the continuous deepening of research on gut microbes, these have been implicated in the regulation of several metabolic diseases (including obesity, type 2 diabetes, tumors, non-alcoholic fatty liver diseases, cardiovascular diseases, etc.) by affecting the immune system and key metabolic pathways (Shuai et al. 2022; Chen et al. 2022). The four main phyla residing in the human gastrointestinal tract are Firmicutes (49–76%) and Bacteroidetes (16–23%), and at lower percentages, Proteobacteria and Actinobacteria (Serra et al. 2018). Compared to the healthy population, the number of intestinal microorganisms in the population with lipid abnormalities decreases significantly, and the diversity of the microbiota reduces markedly (Li et al. 2022). Bacteroidetes, Fecobacteria, Lactobacillus, Bifidobacteria, Ackermann mucophilia, Spirilloidetes, Eggerthella, and Butyricimonas exert positive effects on regulating the homeostasis of lipid metabolism, while Firmicutes are abundant in the intestine of obese people, which is inconducive to body’s metabolism. This intestinal flora can directly regulate the absorption and digestion of lipids, lipoprotein metabolism, lipid mass spectrometry indicators, and related apolipoproteins in the diet, in turn affecting lipid metabolism.

Environmental and dietary factors can stimulate the intestinal tract to produce metabolites under the action of intestinal microbial-derived metabolites, including lipopolysaccharide (LPS), bile acids, short-chain fatty acids (SCFAs), and secreted hormones, which together play an important role in regulating the intestinal flora (Yu et al. 2019). Among them, LPS interacts with a high-fat diet and is related to lipid metabolism in the liver. SCFAs can regulate lipid metabolism by upregulating the expression of AMPK. Some fatty acids with low molecular weights, like acetic acid and propionic acid, can enter the circulation to regulate the formation of adipose tissue. Intestinal flora regulates the expression of the FXR receptor, GPCR receptor, and the TGR5 signaling pathway by affecting bile acid metabolism, thus regulating lipid absorption. Hormones secreted by intestinal endocrine cells can also regulate the number of intestinal flora through the expression of glp-1/2 receptors (Fig. 4).

Fig. 4
figure 4

Lipid metabolism and intestinal flora. LPS: Lipopolysaccharide; SCFAs: short-chain fatty acids

Regulatory effects and mechanism of peptide action of different origins on lipid metabolism disorders

Bean peptides

The lipid-lowering potential of bean peptides like those from soybean has received a lot of research attention. Inoue et al. (2011) identified three dipeptides with hypolipidemic effects, among which KA, VK, and SY reduced TG synthesis, and Ser-Tyr reduced ApoB secretion in HepG2 cells. Lee et al. (2021) isolated two bioactive peptides (ALEPDHRVESEGGL and SLVNNDDRDSYRLQSGDAL) from soybean hydrolysates and found that these peptides promoted cholesterol excretion by up-regulating the expressions of ABCG5 and ABCG8 in small intestinal cells, and inhibit hepatobiliary acid synthesis by up-regulating the expression of FGF15/19 and down-regulating those of CYP7A1 and CYP8B1. Chickpea peptides (ChPs) inhibit the activities of FAS and HMG-CoA reductase (HMGR). In an ovariectomized rat model, ChPs were found to decrease the body weight, adipose tissue size, TC, TG, LDL-C, atherogenic index (AI) in serum, and liver TC, TG levels. Serum HDL-C, bile acids in the liver and feces, and fecal TC and TG increased significantly. The expressions of peroxisomal PPARγ and SREBP-1c were down-regulated, while that of LXRα was upregulated by ChPs. Finally, VFVRN in ChPs could reduce the expression of HMGR to inhibit cholesterol biosynthesis (Shi et al. 2019a). Bioactive peptides from black bean protein hydrolysates showed strong inhibitory activities against pancreatic lipase, with IC50 values of 3.21 mg/mL and 1.23 mg/mL, respectively. These inhibited 3T3-L1 adipocyte differentiation and prevented lipid accumulation (Moreno et al. 2019). Five kinds of pinto bean peptides have been studied, and their lipase inhibition rates ranged from 23 to 87%. The C5-OH, C11-OH, C15-OH, or C21-OH sites of cholic acid and deoxycholic acid show hydrogen bonding, and the bile acid binding capacity ranges from 18 to 71% (Ngoh et al. 2017).

Cereal and tuber peptides

Cereals and tubers are rich in lipid-lowering peptides. Quinoa protein hydrolysates can inhibit lipid accumulation and suppress 3T3-L1 cell differentiation through the PPAR-γ pathway (Shi et al. 2019b). Millet peptides have the highest pancreatic lipase inhibitory activity (IC50 = 0.03 mg/mL) (Jakubczyk et al. 2019). Peptides harboring the amino acid sequence, SPFWNINAH, were identified in oat protein hydrolysates, and showed good lipase inhibition (IC50 = 85.4 ± 3 µM) (Esfandi et al. 2022). A polypeptide with the amino acid sequence of FYLGYCDY was identified in de-oiled rice bran hydrolysate and showed lipase inhibitory activity with an IC50 of 0.47 ± 0.02 µM (Ketprayoon et al. 2021). The rice bran protein hydrolysate regulates cholesterol levels in two ways—one by restricting the recycling of bile salts to the liver via direct excretion, and two, by exhibiting maximum micellar cholesterol inhibitory capacity and HMGCR inhibition in a dose-dependent manner (Kumar et al. 2021). Oral administration of potato protein hydrolysates and intraperitoneal injection of potato protein-derived peptides can activate the AMPK signaling pathway and reduce liver fat deposition (Ibrahim et al. 2018). Active peptides can also be produced from grains through fermentation and are found in the by-products after fermentation. Three peptides were identified from brewer’s spent grain protein hydrolysates, including a peptide with the highest cholesterol esterase inhibition capacity (WNIHMEHQDLTTME) and two with pancreatic lipase inhibition capacity (DFGIASF and LAAVEALSTNG) (Garzón et al. 2020). Huangjiu peptides exerted positive effects on serum biomarkers, hepatic metabolism, and intestinal dysbiosis in hyperlipidemia diet-induced hyperlipidemia mice. Among the three Huangjiu peptides (HpT1, HpT2, and HpT3), HpT1 and HpT2 reduced the increase in serum TC, TG, and LDL-C levels and abnormal lipid accumulation in the liver in hyperlipidemic mice induced by high-lipid diet. Compared to simvastatin and HpT3, HpT2 and HpT1 restored the diversity and structure of gut microorganisms (including Lactobacillus, Ileibacterium, Faecalibaculum, and Alloprevotella) (Shi et al. 2021).

Nutty peptides

Walnut meal peptides counteract the high-fat diet-induced increase in body, liver, and epididymal fat weights, and reduce the serum concentrations of TC, TG, and LDL-C, along with hepatic TC and TG content. These increase HDL-C levels while reducing AI. Walnut meal peptides correlate with the normalization of elevated apolipoprotein Apo-B and reduced Apo-A1 levels induced by a high-fat diet, along with favorable changes in the expression of genes associated with lipid metabolism (LCAT, CYP7A1, HMGR, and FAS) (Yang et al. 2021). The IC50 of the cocoa proteolytic peptide for pancreatic lipase is 1.38 mg/mL and reduces the apparent absorption rate of fat by promoting the excretion of TG and TC in rats. Molecular docking analyses show that peptides with sequences EEQR, GGER, QTGVQ and VSTDVNIE have a higher theoretical affinity for pancreatic lipase and can regulate lipid metabolism (Coronado-Cáceres et al. 2020).

Other plant peptides

Olive seed protein hydrolysates can reduce dietary cholesterol absorption or inhibit cholesterol biosynthesis by inhibiting cholesterol esterase and bile acid binding and reducing the solubility of cholesterol in micelles (Prados et al. 2020). Peptides from chia protein with a molecular mass lower than 3 kDa can reduce up to 80.7% of the HMGCR enzymatic reaction velocity (Coelho et al. 2018). Cumin seed peptides show > 50% inhibition of pancreatic lipase activity along with a high affinity for binding to bile acids, in addition to its inhibitory effects (up to 80%) on the formation of cholesterol micelles (Siow et al. 2016). Sunflower protein hydrolysates, to some degree, inhibit cholesterol incorporation into micelles, thus reducing cholesterol absorption in vivo (Megías et al. 2009). Seabuckthorn seed protein hydrolysates inhibit pancreatic lipases in a non-competitive manner. HPLC-MS/MS analysis predicted six peptides (EEAASLR, FR, RDR, VR, APYR, and NLLHR) with pancreatic lipase inhibitory activity through molecular docking (Xiang et al. 2020).

Algal peptides

Chlorella protein hydrolysate can prevent high-fat diet-induced glucose disorders and fatty liver by inhibiting adipocyte hypertrophy and reducing liver TC and TG levels (Noguchi et al. 2016). The lipid-lowering effects of Spirulina platensis protein hydrolysate include weight reduction and serum glucose and TC reduction through the brain-liver axis. The signaling pathways involved include PPAR, adipocytokine, AMPK, and MAPK (Zhao et al. 2019).

Peptides from aquatic animals

Two pancreatic lipase inhibitory peptides were isolated from cod meat hydrolysate, namely GSPPPSG and KLEGDLK, with IC50 values of 0.60 and 1.08 mg/mL (Tian et al. 2022). Fish goby protein hydrolysates fed to high fat and fructose diet-fed rats showed good efficacy in lowering serum TC, TG, and LDL-C levels as well as hepatic TC and TG contents, whilst increasing the serum HDL-C content (Nasri et al. 2018). Peptides from tuna reduced lipid components and adipogenesis in 3T3-L1 cells along with the expressions of CCAAT/enhancer-binding proteinα (C/EBP-α) and PPAR-γ (Kim et al. 2015). Ark shell protein hydrolysates (ASPH) in mesenchymal stem cells (MSCs) led to a decrease in intracellular lipid accumulation and an increase in lipolysis. At the molecular level, ASPH III down-regulated PPAR-γ, C/EBPα, and SREBP-1c, along with the downstream LPL and FAS expression. Moreover, treatment with ASPH III increased HSL and leptin expressions in the differentiated adipocytes from MSCs (Hyung et al. 2017). Sea cucumber collagen peptides can reduce TC, TG contents, and atherogenic index (AI) in rats (Hu et al. 2012). Trachinotus ovatus protein hydrolysates effectively reduce TC, LDL-C, and AI, whilst increasing HDL-C content (Wan et al. 2020).

Egg and milk-derived peptides

Egg white protein hydrolysates improve fat accumulation and dyslipidemia in obese rats. Analysis of microbial composition and metabolic compounds in feces revealed that the hydrolysates of egg white could regulate intestinal microorganisms (Requena et al. 2017). Two bioactive peptides in casein hydrolysates (SQSKVLPVPQK and HPHPHLSF) induced TICE through the expression of ABCG5 in enterocytes and suppressed hepatic mRNA levels of CYP7A1 and CYP8B1 by ileal FGF19 expression in an LXRα-dependent manner. Oral administration of casein peptides in the hyperlipidemic mouse model reduced serum cholesterol levels and elevated ABCG5 expression and fecal cholesterol secretion (Lee & Youn 2020). Peptides, MMML, FDML, and HLPGRG from cow and AAGF, MSNYF, and FLWPEYGAL from camel casein hydrolysates are likely the most active pancreatic lipase inhibitory peptides. Peptide LPs found in both the cow and camel casein hydrolysates may be active cholesterol esterase inhibitors (Mudgi et al., 2022). Kefir is produced by the symbiotic fermentation of milk by lactic acid bacteria and yeast. Kefir peptides can improve weight gain, fat deposition in adipose tissue around the epididymis and kidney, and adipocyte size in obese rats induced by a high-fat diet. Kefir peptide blocks adipogenesis in the liver by reducing the FAS protein levels and increasing those of p-acetyl-CoA carboxylase. Kefir peptide increases fatty acid oxidation by increasing protein expression of phosphorylated AMP-activated protein kinase, peroxisome proliferator-activated receptor ɑ and carnitine palmitoyltransferase-1 (CPT-1) in the liver (Tung et al. 2018).

Livestock organ or tissue peptides

Pork and chicken skin collagen extracts have good inhibitory effects on pancreatic lipase, and the ultrafiltration component > 5 kDa shows the highest inhibitory rate (IC50 = 4.33 mg/mL) (González-Noriega et al. 2022). Collagen can reduce visceral fat content but is less effective in reducing body weight (Watanabe et al. 2021). A novel cholesterol-lowering dipeptide, Phe-Pro (FP), was isolated from cattle heart protein hydrolysate and could reduce TC and non-HDL-C in rat serum, along with hepatic cholesterol levels. FP significantly increased serum HDL-C, together with a significant decrease in AI and increased fecal cholesterol and acidic steroid excretion. Moreover, FP decreased ABCA1 expression in rat jejunum and reduced cholesterol absorption in Caco-2 cells (Banno et al. 2019).

Synthetic peptides

In Wistar rats with experimental hyperlipidemia and diabetes mellitus, KEW-NH2 tripeptide produced a hypolipidemic effect on atherogenic lipoproteins, decreased the intensity of plasma lipid peroxide (LPO), and normalized HDL levels (Malinin et al. 2014). A 9-amino-acid peptide, D3 has been designed. Ileal transcriptome and molecular analyses showed that D3 treatment could significantly reduce the body weight of mice, improve leptin resistance, up-regulate the expression of urinary guanosine (UGN), and inhibit appetite through the UGN-GUCY2C endocrine axis. Following D3 treatment, IFN-γ in the IRGM1 axis and the number of enteromyxophilic Ackermann bacteria increased about 100-fold, further inhibiting fat absorption by down-regulating CD36 (Li et al. 2022). Guru et al. (2022) showed that RF13 polypeptide derived from vacuolar sortilin (VPS26B) could significantly down-regulate lipid metabolism-related genes, including C/EBPα, SREBP1, and FAS, in zebrafish larvae induced by a high-fat diet. Esquejo et al. (2018) found that PF-06409577 could reduce the level of mevalonate (a direct product of HMGCR) in plasma and plasma TC, whilst increasing the level of plasma HDL-C in obese male rats. The contents of TC and LDL-C in the plasma of cynomolgus monkeys fed on PF-06409577 for 6 weeks decreased. Therefore, experiments based on rats and primates show that PF-06409577 has lipid-lowering effects that should be explored further in clinical settings.

Existing studies have targeted AMPK and ASGR1 to reduce blood lipid levels. Abbott laboratories identified the first direct AMPK activator, A-769,662, that could bind to the ADaM site (Cameron & Kurumbail 2016; Cool et al. 2006). ASGR1 can be targeted to upregulate LXRα, ABCA1, and ABCG5/G8, promote cholesterol excretion and inhibit SREBP1 and lipogenesis, thus reducing blood lipid levels (Wang et al. 2022). These studies raise important questions about the role of AMPK and ASGR1 and the potential targets involved in exerting these beneficial effects. However, whether these should be used alone or in combination with currently available cholesterol-lowering drugs (like statins, ezetimibe, and PCKS9 inhibitors) (Wang et al. 2022; Patil et al. 2022), the biological effectiveness, and safety should be considered for preventing and/or treating lipid metabolism disorders. Long-term research is needed to supplement the shortcomings of clinical trials.

In summary, the peptides extracted or prepared from the above-mentioned sources can inhibit enzymatic activities related to lipid metabolism, regulate the relevant abnormal indicators of lipid disorders in animal models, and show good lipid-lowering activities in cellular models. Some of these peptides can also regulate lipid metabolism through intestinal microorganisms.

Conclusion

Herein, lipid metabolism pathways and the lipid-lowering effect of peptides from different sources were reviewed, and the regulatory effects and lipid-lowering mechanisms of bioactive peptides in lipid metabolic disorders were elaborated, thus providing a reference for further development of bioactive peptides as potential lipid-lowering functional foods or drugs. Although some progress has been made in the extraction, preparation, and synthesis of bioactive peptides, existing research on the lipid-lowering activity of bioactive peptides mainly focuses on their preparation, determination of activity in vitro, and the evaluation of lipid-lowering activity in animal models. Compared to the absorption and transport mechanism in the human body, its bioavailability and regulation mode remain largely unclear, and there is a lack of corresponding clinical experiments. The content of active peptides is relatively low and the preparation cycle is long; even after the characterization of polypeptide sequence and spatial structure, their safety and stability should be considered when large-scale preparation is by synthesis and other methods. Therefore, translating the research results for bioactive peptides to regulate lipid levels into industrial achievements with strong safety and high availability remains an arduous task.

Availability of data and materials

The datasets used and/or analyzed in the present study are available from the corresponding author upon reasonable request.

References

  • Ahmadi, Y., Haghjoo, A. G., Dastmalchi, S., Nemati, M., & Bargahi, N. (2018). Effects of Rosuvastatin on the expression of the genes involved in cholesterol metabolism in rats: adaptive responses by extrahepatic tissues. Gene,661, 45–50. https://doi.org/10.1016/j.gene.2018.03.092

    Article  CAS  Google Scholar 

  • Ajeigbe, O. F., Oboh, G., Ademosun, A. O., & Oyagbemi, A. A. (2022). Ficus asperifolia miq-enriched biscuit diet protects against l-NAME induced hyperlipidemia and hypertension in rats. Food Frontiers, 3, 150–160. https://doi.org/10.1002/fft2.101.

    Article  CAS  Google Scholar 

  • Banno, A., Wang, J., Okada, K., Mori, R., Mijiti, M., & Nagaoka, S. (2019). Identification of a novel cholesterol-lowering dipeptide, phenylalanine-proline (FP), and its down-regulation of intestinal ABCA1 in hypercholesterolemic rats and Caco-2 cells. Scientific Reports, 9(1), 19416. https://doi.org/10.1038/s41598-019-56031-8.

    Article  CAS  Google Scholar 

  • Cagen, L. M., Deng, X., Wilcox, H. G., Park, E. A., Raghow, R., & Elam, M. B. (2005). Insulin activates the rat sterol-regulatory-element-binding protein 1c (SREBP-1c) promoter through the combinatorial actions of SREBP, LXR, Sp-1 and NF-Y cis-acting elements. The Biochemical Journal, 385(Pt 1), 207–216. https://doi.org/10.1042/BJ20040162.

    Article  CAS  Google Scholar 

  • Cameron, K. O., & Kurumbail, R. G. (2016). Recent progress in the identification of adenosine monophosphate-activated protein kinase (AMPK) activators. Bioorganic & Medicinal Chemistry Letters, 26(21), 5139–5148. https://doi.org/10.1016/j.bmcl.2016.09.065.

    Article  CAS  Google Scholar 

  • Catalkaya, G., Venema, K., & Lucini, L. (2020). Interaction of dietary polyphenols and gut microbiota: microbial metabolism of polyphenols, influence on the gut microbiota and implications on host health. Food Frontiers, 1, 109–133. https://doi.org/10.1002/fft2.25.

    Article  Google Scholar 

  • Chakrabarti, S., Guha, S., & Majumder, K. (2018). Food-derived bioactive peptides in human health: challenges and opportunities. Nutrients, 10(11), 1738. https://doi.org/10.3390/nu10111738.

    Article  CAS  Google Scholar 

  • Chalamaiah, M., Yu, W., & Wu, J. (2018). Immunomodulatory and anticancer protein hydrolysates (peptides) from food proteins: a review. Food Chemistry, 245, 205–222. https://doi.org/10.1016/j.foodchem.2017.10.087.

    Article  CAS  Google Scholar 

  • Chen, Y., Wu, W. H., Ni, X., Farag, M. A., Capanoglu, E., & Zhao, C. (2022). Regulatory mechanisms of the green alga Ulva lactuca oligosaccharide via the metabolomics and gut microbiome in diabetic mice. Current Research in Food Science, 5, 1127–1139. https://doi.org/10.1016/j.crfs.2022.07.003.

    Article  CAS  Google Scholar 

  • Cian, R. E., Garzón, A. G., Ancona, D. B., Guerrero, L. C., & Drago, S. R. (2015). Hydrolyzates from pyropia columbina seaweed have antiplatelet aggregation, antioxidant and ACE inhibitory peptides which maintain bioactivity after simulated gastrointestinal digestion. LWT-Food Science and Technology, 64(2), 881–888. https://doi.org/10.1016/j.lwt.2015.06.043.

    Article  CAS  Google Scholar 

  • Coelho, M. S., Soares-Freitas, R., Arêas, J., Gandra, E. A., & Salas-Mellado, M. (2018). Peptides from Chia present antibacterial activity and inhibit cholesterol synthesis. Plant Foods for Human Nutrition, 73(2), 101–107. https://doi.org/10.1007/s11130-018-0668-z.

    Article  CAS  Google Scholar 

  • Cool, B., Zinker, B., Chiou, W., Kifle, L., Cao, N., Perham, M., Dickinson, R., Adler, A., Gagne, G., Iyengar, R., Zhao, G., Marsh, K., Kym, P., Jung, P., Camp, H. S., & Frevert, E. (2006). Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metabolism, 3(6), 403–416. https://doi.org/10.1016/j.cmet.2006.05.005.

    Article  CAS  Google Scholar 

  • Coronado-Cáceres, L. J., Rabadán-Chávez, G., Mojica, L., Hernández-Ledesma, B., Quevedo-Corona, L., & Cervantes, L., E (2020). Cocoa (Theobroma cacao L.) seed proteins’ anti-obesity potential through lipase inhibition using in silico, in vitro and in vivo models. Foods, 9(10), 1359. https://doi.org/10.3390/foods9101359.

  • Durkar, A. M., Patil, R. R., & Naik, S. R. (2014). Hypolipidemic and antioxidant activity of ethanolic extract of Symplocos racemosa Roxb. In hyperlipidemic rats: an evidence of participation of oxidative stress in hyperlipidemia. Indian Journal of Experimental Biology, 52(1), 36–45.

    CAS  Google Scholar 

  • Esfandi, R., Seidu, I., Willmore, W., & Tsopmo, A. (2022). Antioxidant, pancreatic lipase, and α-amylase inhibitory properties of oat bran hydrolyzed proteins and peptides. Journal of Food Biochemistry, 46(4), e13762. https://doi.org/10.1111/jfbc.13762.

    Article  CAS  Google Scholar 

  • Esquejo, R. M., Salatto, C. T., Delmore, J., Albuquerque, B., Reyes, A., Shi, Y., Moccia, R., Cokorinos, E., Peloquin, M., Monetti, M., Barricklow, J., Bollinger, E., Smith, B. K., Day, E. A., Nguyen, C., Geoghegan, K. F., Kreeger, J. M., Opsahl, A., Ward, J., Kalgutkar, A. S., & Miller, R. A. (2018). Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate preclinical models. EBioMedicine, 31, 122–132. https://doi.org/10.1016/j.ebiom.2018.04.009.

    Article  Google Scholar 

  • Fleischmann, M., & Iynedjian, P. B. (2000). Regulation of sterol regulatory-element binding protein 1 gene expression in liver: role of insulin and protein kinase B/cAkt. The Biochemical Journal, 349(Pt 1), 13–17. https://doi.org/10.1042/0264-6021:3490013.

    Article  CAS  Google Scholar 

  • Fogarty, S., & Hardie, D. G. (2010). Development of protein kinase activators: AMPK as a target in metabolic disorders and cancer. Biochimica et Biophysica Acta Proteins & Proteomics,1804(3), 581–591. https://doi.org/10.1016/j.bbapap.2009.09.012

    Article  CAS  Google Scholar 

  • Garzón, A. G., Cian, R. E., Aquino, M. E., & Drago, S. R. (2020). Isolation and identification of cholesterol esterase and pancreatic lipase inhibitory peptides from brewer’s spent grain by consecutive chromatography and mass spectrometry. Food & Function, 11(6), 4994–5003. https://doi.org/10.1039/d0fo00880j.

    Article  CAS  Google Scholar 

  • Ge, Y., Zhang, Y., Li, R., Chen, W., Li, Y., & Chen, G. (2011). Berberine regulated GCK, G6PC, PCK1 and SREBP-1c expression and activated AMP-activated protein kinase in primary rat hepatocytes. International Journal of Biological Sciences, 7(5), 673–684. https://doi.org/10.7150/ijbs.7.673.

    Article  CAS  Google Scholar 

  • González-Noriega, J. A., Valenzuela-Melendres, M., Hernández-Mendoza, A., Astiazarán-García, H., Mazorra-Manzano, M., & Peña-Ramos, E. A. (2022). Hydrolysates and peptide fractions from pork and chicken skin collagen as pancreatic lipase inhibitors. Food Chemistry-X, 13, 100247. https://doi.org/10.1016/j.fochx.2022.100247.

    Article  CAS  Google Scholar 

  • Guru, A., Velayutham, M., & Arockiaraj, J. (2022). Lipid-lowering and antioxidant activity of RF13 peptide from vacuolar protein sorting-associated protein 26B (VPS26B) by modulating lipid metabolism and oxidative stress in HFD induced obesity in zebrafish larvae. International Journal of Peptide Research and Therapeutics, 28(2), 1–15. https://doi.org/10.1007/s10989-022-10376-3.

    Article  CAS  Google Scholar 

  • Hu, X. Q., Xu, J., Xue, Y., Li, Z. J., Wang, J. F., Wang, J. H., Xue, C. H., & Wang, Y. M. (2012). Effects of bioactive components of sea cucumber on the serum, liver lipid profile and lipid absorption. Bioscience Biotechnology and Biochemistry, 76(12), 2214–2218. https://doi.org/10.1271/bbb.120495.

    Article  CAS  Google Scholar 

  • Huang, Z., Wang, C. E., Chen, J., He, X., Chen, K., Jiang, X., Zhao, C., & Liu, B. (2022). Ameliorating effect on glycolipid metabolism of Spirulina functional formulation combination from traditional Chinese medicine. Oxidative Medicine and Cellular Longevity,22, 3910116. https://doi.org/10.1155/2022/3910116

    Article  CAS  Google Scholar 

  • Hyung, J. H., Ahn, C. B., & Je, J. Y. (2017). Ark shell protein hydrolysates inhibit adipogenesis in mouse mesenchymal stem cells through the down-regulation of transcriptional factors. RSC Advances,7, 6223–6228. https://doi.org/10.1039/C6RA24995G

    Article  CAS  Google Scholar 

  • Ibrahim, M. A., Bester, M. J., Neitz, A. W., & Gaspar, A. R. M. (2018). Tuber storage proteins as potential precursors of bioactive peptides: an in silico analysis. International Journal of Peptide Research & Therapeutics, 25, 437–446. https://doi.org/10.1007/s10989-018-9688-7.

    Article  CAS  Google Scholar 

  • Inoue, N., Nagao, K., Sakata, K., Yamano, N., Gunawardena, P. E., Han, S. Y., Matsui, T., Nakamori, T., Furuta, H., Takamatsu, K., & Yanagita, T. (2011). Screening of soy protein-derived hypotriglyceridemic di-peptides in vitro and in vivo. Lipids in Health and Disease,10, 85. https://doi.org/10.1186/1476-511X-10-85

    Article  CAS  Google Scholar 

  • Jakubczyk, A., Szymanowska, U., Karaś, M., Złotek, U., & Kowalczyk, D. (2019). Potential anti-inflammatory and lipase inhibitory peptides generated by in vitro gastrointestinal hydrolysis of heat treated millet grains. CyTA-Journal of Food,17(1), 324–333. https://doi.org/10.1080/19476337.2019.1580317

    Article  CAS  Google Scholar 

  • Ketprayoon, T., Noitang, S., Sangtanoo, P., Srimongkol, P., Saisavoey, T., Reamtong, O., Choowongkomon, K., & Karnchanatat, A. (2021). An in vitro study of lipase inhibitory peptides obtained from de-oiled rice bran. RSC Advances,11(31), 18915–18929. https://doi.org/10.1039/d1ra01411k

    Article  CAS  Google Scholar 

  • Kim, Y. M., Kim, E. Y., Kim, I. H., & Nam, T. J. (2015). Peptide derived from desalinated boiled tuna extract inhibits adipogenesis through the downregulation of C/EBP-α and PPAR-γ in 3T3-L1 adipocytes. International Journal of Molecular Medicine,35(5), 1362–1368. https://doi.org/10.3892/ijmm.2015.2127

    Article  CAS  Google Scholar 

  • Kumar, V., Kurup, L. V., & Tiku, P. K. (2021). The modulatory effect of cholesterol synthesis by Oryza sativa derived bioactive peptide fractions: an in vitro investigation. International Journal of Peptide Research and Therapeutics, 27(1), 245–251. https://doi.org/10.1007/s10989-020-10079-7.

    Article  CAS  Google Scholar 

  • Lee, H., Shin, E., Kang, H., Youn, H., & Youn, B. (2021). Soybean-derived peptides attenuate hyperlipidemia by regulating trans-intestinal cholesterol excretion and bile acid synthesis. Nutrients, 14(1), 95. https://doi.org/10.3390/nu14010095.

    Article  CAS  Google Scholar 

  • Lee, S., & Youn, B. (2020). Hypolipidemic roles of casein-derived peptides by regulation of trans-intestinal cholesterol excretion and bile acid synthesis. Nutrients, 12(10), 3058. https://doi.org/10.3390/nu12103058.

    Article  CAS  Google Scholar 

  • Li, T. T., Huang, Z. R., Jia, R. B., Lv, X. C., Zhao, C., & Liu, B. (2021). Spirulina platensis polysaccharides attenuate lipid and carbohydrate metabolism disorder in high-sucrose and high-fat diet-fed rats in association with intestinal microbiota. Food Research International, 147, 110530. https://doi.org/10.1016/j.foodres.2021.110530.

    Article  CAS  Google Scholar 

  • Li, Z., Zhang, B., Wang, N., Zuo, Z., Wei, H., & Zhao, F. (2022). A novel peptide protects against diet-induced obesity by suppressing appetite and modulating the gut microbiota. Gut. https://doi.org/10.1136/gutjnl-2022-328035.

    Article  Google Scholar 

  • Liu, Z., Zhang, Y., Zhang, R., Gu, L., & Chen, X. (2017). Promotion of classic neutral bile acids synthesis pathway is responsible for cholesterol-lowing effect of Si-miao-yong-an decoction: application of LC-MS/MS method to determine 6 major bile acids in rat liver and plasma. Journal of Pharmaceutical and Biomedical Analysis,135, 167–175. https://doi.org/10.1016/j.jpba.2016.12.021

    Article  CAS  Google Scholar 

  • Malinin, V. V., Savateeva-Lyubimova, T. N., & Sivak, K. V. (2014). Hypolipidemic and hypoglycemic effect of peptide Lys-Glu-Trp-NH(2) in rats with associated metabolic disorders. Bulletin of Experimental Biology and Medicine,156(3), 338–341. https://doi.org/10.1007/s10517-014-2344-0

    Article  CAS  Google Scholar 

  • Megías, C., Pedroche, J., Del Mar Yust, M., Alaiz, M., Girón-Calle, J., Millán, F., & Vioque, J. (2009). Sunflower protein hydrolysates reduce cholesterol micellar solubility. Plant Foods for Human Mutrition, 64(2), 86–93. https://doi.org/10.1007/s11130-009-0108-1.

    Article  CAS  Google Scholar 

  • Moreno, V. C., De, M., Luis, M., Diego, L. V., & Rosa, C. (2019). Bioactive peptides from black bean proteins play a potential role in the prevention of adipogenesis (P06-119-19). Current Developments in Nutrition. https://doi.org/10.1093/cdn/nzz031.P06-119-19. 3.

  • Nasri, R., Abdelhedi, O., Jemil, I., Ben Amor, I., Elfeki, A., Gargouri, J., Boualga, A., Karra-Châabouni, M., & Nasri, M. (2018). Preventive effect of goby fish protein hydrolysates on hyperlipidemia and cardiovascular disease in Wistar rats f ed a high-fat/fructose diet. RSC Advances, 8(17), 9383–9393. https://doi.org/10.1039/c7ra13102j.

    Article  CAS  Google Scholar 

  • Ngoh, Y. Y., Choi, S. B., & Gan, C. Y. (2017). The potential roles of pinto bean (Phaseolus vulgaris cv. Pinto) bioactive peptides in regulating physiological functions: protease activating, lipase inhibiting and bile acid binding activities. Journal of Functional Foods, 33, 67–75. https://doi.org/10.1016/j.jff.2017.03.029.

    Article  CAS  Google Scholar 

  • Noguchi, N., Yanagita, T., Rahman, S. M., & Ando, Y. (2016). Chlorella protein hydrolysate attenuates glucose metabolic disorder and fatty liver in high-fat diet-induced obese mice. Journal of Oleo Science, 65(7), 613–620. https://doi.org/10.5650/jos.ess16034.

    Article  CAS  Google Scholar 

  • Oluwajuyitan, T. D., Ijarotimi, O. S., & Fagbemi, T. N. (2021). Plantain based dough meal: nutritional property, antioxidant activity and dyslipidemia ameliorating potential in high-fat induced rats. Clinical Phytoscience, 7(1), 1–16. https://doi.org/10.1186/s40816-021-00327-8.

    Article  CAS  Google Scholar 

  • Patil, P. J., Usman, M., Zhang, C., Mehmood, A., Zhou, M., Teng, C., & Li, X. (2022). An updated review on food-derived bioactive peptides: focus on the regulatory requirements, safety, and bioavailability. Comprehensive Reviews in Food Science and Food Safety, 21(2), 1732–1776. https://doi.org/10.1111/1541-4337.12911.

    Article  Google Scholar 

  • Prados, I. M., Plaza, M., Marina, M. L., & García, M. C. (2020). Evaluation of the relationship between the peptide profiles and the lipid-lowering properties of olive seed hydrolysates as a tool for tuning hypocholesterolemic functionality. Food & Function,11(6), 4973–4981. https://doi.org/10.1039/d0fo00576b

    Article  CAS  Google Scholar 

  • Requena, T., Miguel, M., Garcés-Rimón, M., Martínez-Cuesta, M. C., López-Fandiño, R., & Peláez, C. (2017). Pepsin egg white hydrolysate modulates gut microbiota in Zucker obese rats. Food & Function, 8(1), 437–443. https://doi.org/10.103 9/c6fo01571a.

  • Ross, S. M. (2017). Red yeast rice: the efficacy of Monascus purpureus yeast for treatment of hyperlipidemia a modifiable risk factor of cardiovascular disease. Holistic Nursing Practice, 31(1), 52–58. https://doi.org/10.1097/HNP.0000000000000192.

    Article  Google Scholar 

  • Serra, D., Almeida, L. M., & Dinis, T. C. (2018). Dietary polyphenols: a novel strategy to modulate microbiota-gut-brain axis. Trends in Food Science & Technology, 78, 224–233. https://doi.org/10.1016/j.tifs.2018.06.007.

    Article  CAS  Google Scholar 

  • Shi, W., Hou, T., Liu, W., Guo, D., & He, H. (2019a). The hypolipidemic effects of peptides prepared from Cicer arietinum in ovariectomized rats and HepG2 cells. Journal of the Science of Food and Agriculture,99(2), 576–586. https://doi.org/10.1002/jsfa.9218

    Article  CAS  Google Scholar 

  • Shi, Y., Feng, R., Mao, J., Liu, S., Zhou, Z., Ji, Z., Chen, S., & Mao, J. (2021). Structural characterization of peptides from huangjiu and their regulation of hepatic steatosis and gut microbiota dysbiosis in hyperlipidemia mice. Frontiers in Pharmacology, 12, 689092. https://doi.org/10.3389/fphar.2021.689092.

    Article  CAS  Google Scholar 

  • Shi, Z., Hao, Y., Teng, C., Yao, Y., & Ren, G. (2019b). Functional properties and adipogenesis inhibitory activity of protein hydrolysates from quinoa (Chenopodium quinoa Willd.). Food Science & Nutrition, 7(6), 2103–2112. https://doi.org/10.10 02/fsn3.1052.

  • Shuai, M., Fu, Y., Zhong, H. L., Gou, W., Jiang, Z., Liang, Y., Miao, Z., Xu, J. J., Huynh, T., Wahlqvist, M. L., Chen, Y. M., & Zheng, J. S. (2022). Mapping the human gut mycobiome in middle-aged and elderly adults: multiomics insights and implications for host metabolic health. Gut, 71(9), 1812–1820. https://doi.org/10.1136/gutjnl-2021-326298.

    Article  CAS  Google Scholar 

  • Siow, H. L., Choi, S. B., & Gan, C. Y. (2016). Structure-activity studies of protease activating, lipase inhibiting, bile acid binding and cholesterol-lowering effects of pre-screened cumin seed bioactive peptides. Journal of Functional Foods, 27, 600–611. https://doi.org/10.1016/j.jff.2016.10.013.

    Article  CAS  Google Scholar 

  • Stoye, A., Nagalingam, G., Britton, W. J., & Payne, R. J. (2017). Synthesis of norfijimycin a with activity against mycobacterium tuberculosis. Australian Journal of Chemistry,70(2), 229–232. https://doi.org/10.1071/CH16559

    Article  CAS  Google Scholar 

  • Tian, Y., Liu, C., Wang, S., Du, M., & Zhu, B. (2022). Efficient screening of pancreatic lipase inhibitors from cod meat hydrolysate through ligand fishing strategy. Frontiers in Nutrition, 9, 969558. https://doi.org/10.3389/fnut.2022.969558.

    Article  CAS  Google Scholar 

  • Tung, Y. T., Chen, H. L., Wu, H. S., Ho, M. H., Chong, K. Y., & Chen, C. M. (2018). Kefir peptides prevent hyperlipidemia and obesity in high-fat-diet-induced obese rats via lipid metabolism modulation. Molecular Nutrition & Food Research, 62(3), 1700505. https://doi.org/10.1002/mnfr.201700505.

    Article  CAS  Google Scholar 

  • van de Beek, M. C., Dijkstra, I. M., & Kemp, S. (2017). Method for measurement of peroxisomal very long-chain fatty acid beta-oxidation and de novo C26:0 synthesis activity in living cells using stable-isotope labeled docosanoic acid. Methods in Molecular Biology, 1595, 45–54. https://doi.org/10.1007/978-1-4939-6937-1_5.

    Article  CAS  Google Scholar 

  • Wa, Y., Yin, B., He, Y., Xi, W., Huang, Y., Wang, C., Guo, F., & Gu, R. (2019). Effects of single probiotic and combined probiotic-fermented milk on lipid metabolism in hyperlipidemic rats. Frontiers in Microbiology,10, 1312. https://doi.org/10.3389/fmicb.2019.01312

    Article  Google Scholar 

  • Wan, P., Chen, D., Chen, H., Zhu, X., Chen, X., Sun, H., Pan, J., & Cai, B. (2020). Hypolipidemic effects of protein hydrolysates from Trachinotus ovatus and identification of peptides implied in bile acid-binding activity using LC-ESI-Q-TOF-MS/MS. RSC Advances, 10(34), 20098–20109. https://doi.org/10.1039/d0ra02428g.

    Article  CAS  Google Scholar 

  • Wang, J. Q., Li, L. L., Hu, A., Deng, G., Wei, J., Li, Y. F., Liu, Y. B., Lu, X. Y., Qiu, Z. P., Shi, X. J., Zhao, X., Luo, J., & Song, B. L. (2022). Inhibition of ASGR1 decreases lipid levels by promoting cholesterol excretion. Nature, 608(7922), 413–420. https://doi.org/10.1038/s41586-022-05006-3.

    Article  CAS  Google Scholar 

  • Watanabe, R., Yamaguchi, M., Watanabe, K., Shimizu, M., Azusa, T., Sone, H., & Kamiyama, S. (2021). Effects of collagen peptide administration on visceral fat content in high-fat diet-induced obese mice. Journal of Nutritional Science and Vitaminology, 67(1), 57–62. https://doi.org/10.3177/jnsv.67.57.

    Article  CAS  Google Scholar 

  • Wu, H., Xu, N., Sun, X., Yu, H., & Zhou, C. (2015). Hydrolysis and purification of ace inhibitory peptides from the marine microalga Isochrysis galbana. Journal of Applied Phycology, 27(1), 351–361. https://doi.org/10.1007/s10811-014-0347-x.

    Article  CAS  Google Scholar 

  • Xiang, H., Waterhouse, D. S., Liu, P., Waterhouse, G. I., Li, J., & Cui, C. (2020). Pancreatic lipase-inhibiting protein hydrolysate and peptides from seabuckthorn seed meal: Preparation optimization and inhibitory mechanism. LWT-Food Science and Technology,134, 109870. https://doi.org/10.1016/j.lwt.2020.109870

    Article  CAS  Google Scholar 

  • Xu, Y., Zhao, J., Liu, X., Zhang, C., Zhao, Z., Li, X., & Sun, B. (2022). Flavor mystery of chinese traditional fermented baijiu: the great contribution of ester compounds. Food Chemistry, 369, 130920. https://doi.org/10.1016/j.foodchem.2021.130920.

    Article  CAS  Google Scholar 

  • Yang, X. Y., Zhong, D. Y., Wang, G. L., Zhang, R. G., & Zhang, Y. L. (2021). Effect of walnut meal peptides on hyperlipidemia and hepatic lipid metabolism in rats fed a high-fat diet. Nutrients, 13(5), 1410. https://doi.org/10.3390/nu13051410.

    Article  CAS  Google Scholar 

  • Yu, Y., Raka, F., & Adeli, K. (2019). The role of the gut microbiota in lipid and lipoprotein metabolism. Journal of Clinical Medicine,8(12), 2227. https://doi.org/10.3390/jcm8122227

    Article  CAS  Google Scholar 

  • Zhao, B., Cui, Y., Fan, X., Qi, P., Liu, C., Zhou, X., & Zhang, X. (2019). Anti-obesity effects of Spirulina platensis protein hydrolysate by modulating brain-liver axis in high-fat diet fed mice. PLoS One, 14(6), e0218543. https://doi.org/10.1371/journal.pone.0218543.

    Article  CAS  Google Scholar 

  • Zheng, Y., Zhang, Y., & San, S. (2020). Efficacy of a novel ACE-inhibitory peptide from Sargassum maclurei in hypertension and reduction of intracellular endothelin-1. Nutrients, 12(3), 653. https://doi.org/10.3390/nu12030653.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the reviewers and the journal’s Editor for their thorough review of the manuscript and constructive comments.

Funding

This research was funded by grants from the Fujian Provincial Natural Science Foundation of China (2022J011109) and the Quanzhou Municipal Science and Technology Planning Project (2022N040). This work was also supported by Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae (2022-KF11).

Author information

Authors and Affiliations

Authors

Contributions

Wenshu Ma and Na Li: Formal analysis, Investigation, Resources, Writing- Original Draft, Writing- Review & Editing, Visualization. Luan Lin and Jiahui Wen: Writing- Review & Editing. Chao Zhao and Fang Wang: Conceptualization, Resources, Writing- Review & Editing, Visualization, Supervision, Funding acquisition. All authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Chao Zhao or Fang Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Author Dr. Chao Zhao is guest editor of special issue “Natural Products and Bioactive Compounds in food” of Food Production, Processing and Nutrition and he was not involved in the journal's review of, or decisions related to this manuscript.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, W., Li, N., Lin, L. et al. Research progress in lipid metabolic regulation of bioactive peptides. Food Prod Process and Nutr 5, 10 (2023). https://doi.org/10.1186/s43014-022-00123-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43014-022-00123-y

Keywords