Next Article in Journal
Strategies to Reduce the Consumption of Foods and Drinks with High Sugar Content in the UK: A Rapid Review Approach
Previous Article in Journal
Assessment of Body Mass Index for Obesity Diagnosis in the Mexican Population: A Cross-Sectional Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lycium barbarum for Health and Longevity: A Review of Its Biological Significance

by
Tao Zhang
1,2,3,*,
Elena-Alexandra Alexa
1,2,4,
Gavin Liu
1,
Alois Berisha
1,
Rhys Walsh
1 and
Robbie Kelleher
5
1
School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin 7, D07 ADY7 Dublin, Ireland
2
Sustainability and Health Research Hub (SHRH), Technological University Dublin, Grangegorman, Dublin 7, D07 H6K8 Dublin, Ireland
3
Food Innovation & Health Research Centre, Technological University Dublin, D07 ADY7 Dublin, Ireland
4
Centre for AMR and One Health Research (CAMROH), Technological University Dublin, D07 H6K8 Dublin, Ireland
5
School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02 R590 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Obesities 2025, 5(2), 35; https://doi.org/10.3390/obesities5020035
Submission received: 25 February 2025 / Revised: 4 May 2025 / Accepted: 12 May 2025 / Published: 16 May 2025

Abstract

:
Lycium barbarum (L. barbarum), commonly known as goji berry, is a functional food recognised for its diverse phytochemical composition and health benefits, particularly in metabolic health and disease prevention. This review explores its phytochemistry, focusing on polysaccharides, carotenoids, polyphenols, and alkaloids, which contribute to its extensive biological activities. L. barbarum polysaccharides, the primary bioactive components, exhibit antioxidant, immunomodulatory, and glycaemic-regulating properties, making them promising candidates for managing obesity-related metabolic disorders. Carotenoids, particularly zeaxanthin, play a key role in ocular health, while polyphenols and alkaloids enhance antioxidant, antimicrobial, and hepatoprotective effects. The biological properties of L. barbarum span metabolic health, cardiovascular function, and glycaemic control, alongside neuroprotection and cancer prevention. Its prebiotic effects on gut microbiota modulation offer additional benefits in managing obesity and associated complications. Furthermore, its antioxidant and anti-inflammatory activities support its role in alleviating oxidative stress and chronic inflammation, common in obesity and metabolic syndrome. Despite robust preclinical evidence, further studies are needed to validate its safety, efficacy, and long-term potential in human populations. This review highlights L. barbarum’s promising applications as a nutraceutical and therapeutic agent, particularly for metabolic and obesity-related health challenges.

1. Introduction

Traditional Chinese Medicine (TCM) is a comprehensive medical system developed over more than 2000 years in China. It integrates modalities such as herbal medicine, acupuncture, massage (tui na), dietary therapy, and qigong, and is underpinned by the philosophical frameworks of yin-yang and the five elements, which describe the dynamic balance governing health and disease [1]. TCM has historically been used to manage a wide range of conditions, including musculoskeletal disorders, respiratory illnesses, and constitutional imbalances such as yin deficiency syndrome [2]. In recent decades, interest in TCM has grown internationally, particularly in countries such as the UK, the US, and Australia, where it is increasingly regarded as a complementary approach to conventional medicine [3]. However, broader integration into Western healthcare systems depends on rigorous evidence of efficacy, safety, and quality control [1]. Lycium barbarum (L. barbarum), known commonly as goji berry, is a prominent herb in TCM, traditionally used to nourish the liver and kidneys, improve vision, and promote longevity. Its documented use dates back to the Tang Dynasty, and it remains officially recognised in the Pharmacopoeia of the People’s Republic of China [4]. L. barbarum is a perennial deciduous shrub belonging to the Solanaceae family, with a long-standing history in traditional Chinese medicine. It has been highly valued for its medicinal properties and functional food applications [5]. Commonly known as goji berries or wolfberries, L. barbarum and L. chinense have been widely utilised in Chinese medicine in various forms, including whole-plant preparations, tinctures, teas, herbal remedies, juices, and wines. In recent years, however, the fruits of L. barbarum have gained global recognition as a functional food, frequently consumed in raw or dried forms [6].
Historically, L. barbarum was cultivated in the Ningxia region of China, where the temperate continental semi-arid climate provided optimal growing conditions. However, as global demand increased, extensive cultivation expanded to other regions across China and beyond [7]. Depending on the species, the fruits exhibit variations in size, colour, texture, and taste. For instance, L. barbarum is characterised by its larger, sweeter, and brighter red fruits, whereas L. ruthenicum produces dark-coloured berries [8]. In contrast, L. chinense bears smaller red fruits with distinct variations in taste and sugar content [7,9]. The quality and bioactive content of these species are significantly influenced by geographical distribution, environmental conditions, soil composition, and harvesting periods [9,10,11]. While L. chinense is primarily cultivated in the Hebei region, which has a temperate monsoon climate, L. barbarum demonstrates greater adaptability and is cultivated across diverse regions of China [7].
Today, L. barbarum is widely cultivated and accounts for nearly 90% of all commercially available goji berries. Their production extends beyond Asia to parts of Europe and the United States, and they are among the 31 species cultivated for both food and medicinal applications [6,12]. Rich in essential nutrients, L. barbarum provides an abundant source of proteins, lipids, dietary fibres, vitamins (e.g., riboflavin, thiamine), and minerals (e.g., magnesium, copper, selenium). Its growing scientific interest is largely due to its bioactive compounds, including polysaccharides, polyphenols, carotenoids, alkaloids, and anthocyanins [6]. These bioactive compounds exhibit significant variability influenced by factors such as plant part utilised, geographical origin, climatic conditions, and extraction techniques (Figure 1) [11,13,14,15]. Advanced analytical techniques have enabled the identification of these variations across different L. barbarum varieties, reinforcing the importance of environmental and agronomic factors in shaping their bioactive profile [14,15,16,17].
Beyond its nutritional and phytochemical properties, L. barbarum has been extensively studied for its health benefits and therapeutic potential, earning its reputation as a “superfruit”. It has demonstrated promising effects in the prevention and management of various health conditions, including cardiovascular diseases, liver disorders, immune modulation, and cancer, as well as its role in promoting longevity [18]. This increasing interest aligns with consumer demand for healthier and more sustainable dietary choices, further solidifying L. barbarum’s role as a functional food [6,15]. Currently, goji berries are incorporated into various food applications, including confectionery, baked goods, salads, beer, wine, smoothies, and sauces [19,20]. Furthermore, they are also used as functional ingredients to enhance the sensory and nutritional qualities of foods [21].
Our previous review focused on human clinical studies, pharmacokinetics, and the safety profile of L. barbarum, providing a comprehensive evaluation of its physiological effects in human populations [18]. The current review aims to expand the scope by exploring in vivo and in vitro studies that assess the biological responses of L. barbarum, alongside an examination of its phytochemistry. Specifically, we highlight key bioactive compounds, including polysaccharides, polyphenols, alkaloids, and carotenoids, and discuss how variations in their composition influence the plant’s biological properties. By synthesising the existing literature on phytochemical variability and biological activity, this review seeks to provide a comprehensive resource for researchers investigating the therapeutic potential of L. barbarum.

2. Methodology

The preparation of this review article involved a comprehensive search of the literature on L. barbarum across multiple scientific databases, with coverage from inception until 31 January 2025, and no language restrictions applied. The databases used included Scopus, PubMed, Google Scholar, ISI Web of Science, and the China National Knowledge Infrastructure (CNKI). Both original research articles and review papers focusing on L. barbarum were included to synthesise existing knowledge comprehensively. The primary search terms applied were “Lycium barbarum”, “L. barbarum”, and “goji berries”, combined with additional terms including “health”, “therapeutic effects”, “phytochemistry”, “bioactivities”, and “biological effects”. These terms were selected to ensure a broad coverage of the plant’s phytochemical composition, biological properties, and health applications. The search was supplemented by manually reviewing the reference lists of relevant articles, reviews, and meta-analyses to identify additional sources. No restrictions were imposed on the publication timeframe; however, particular emphasis was placed on studies published in the past 20 years to capture recent advancements. Articles were initially screened based on their titles and abstracts, and full texts were subsequently reviewed if the information in the title or abstract was insufficient to determine their relevance. Figures were designed using Canva (www.canva.com, accessed on 8 February 2025) as schematic illustrations to visually summarise key concepts. All scientific content depicted is based on inclusion of peer-reviewed literature and accurately reflects current understanding in the field. Grey literature, conference abstracts, and unpublished materials were excluded. Studies were selected based on original data provided related to the phytochemistry or biological effects of L. barbarum in in vitro or in vivo models. Review articles were used to support contextual background but not as sources of primary data.

3. Phytochemistry

L. barbarum is recognised for its richness in essential nutrients and numerous health benefits [22]. Phytochemical investigations have revealed a diverse array of compounds in L. barbarum, including polysaccharides, carotenoids, polyphenols, alkaloids, betaine, essential oils, anthocyanins, amino acids, proteins, fibres, organic acids, carbohydrates, lipids, vitamins, and minerals [14,23,24]. Among these, L. barbarum polysaccharides (LBPs) are the primary bioactive components, with the total sugar content of L. barbarum accounting for about 40% of its dry weight, and LBP comprising 5–8% of the total dry weight [25,26].
In addition to LBPs, other notable phytochemicals include kaempferol, caffeic acid, and hydroxyferulic acid in the fruits; rutin and chlorogenic acid in the leaves and flowers; and N-feruloyltyramine derivatives in the stems [27]. These bioactive compounds contribute to a wide range of biological activities, such as antioxidant, anti-inflammatory, immunomodulatory, antimicrobial, anticancer, and neuroprotective effects [28,29,30,31].
Due to its nutritional richness and health-promoting effects, the increasing popularity of L. barbarum as a functional food and dietary supplement has spurred advancements in quality control and industrial applications, fostering sustainable growth within the L. barbarum industry [32,33]. Recent studies have emphasised the structural characterisation, extraction, and purification of these phytochemicals, highlighting their substantial health benefits [19]. The leaves, flowers, and stems of the plant also serve as valuable sources of bioactive compounds, complementing the well-established nutraceutical value of the fruits [27].
The primary analytical techniques used to detect and characterise the bioactive molecules in L. barbarum include high-performance liquid chromatography with ultraviolet detection (HPLC-UV), liquid chromatography–mass spectrometry (LC-MS), liquid chromatography–tandem mass spectrometry (LC-MS/MS), gas chromatography–mass spectrometry (GC-MS), ultra-performance liquid chromatography–triple quadrupole mass spectrometry (UPLC-TQ-MS), and nuclear magnetic resonance (NMR) spectroscopy [34,35]. The following sections will focus specifically on LPBs, polyphenols, alkaloids and carotenoids.

3.1. Polysaccharides

LBPs are composed of glycopeptide chains that include acidic heteropolysaccharides and proteins or polypeptides. Polysaccharides have a sophisticated structural hierarchy, classified into primary, secondary, tertiary, and quaternary levels, with each level built upon the primary structure [25]. The structure and composition of LBPs can vary significantly depending on the extraction and purification techniques used. These methods include water-based extraction, microwave-assisted extraction, ultrasonic-assisted extraction, enzyme-assisted extraction, and supercritical fluid extraction. Despite their differences, all these methods aim to break down the cell wall under specific conditions without compromising the polysaccharides’ activity [36].
Traditional solvent extraction is straightforward and economical, but it has disadvantages such as lengthy extraction times, limited efficiency, and the potential for polysaccharide degradation at high temperatures, which can diminish biological activity [25]. Microwave-assisted extraction, on the other hand, is a physical technique that is both cost-effective and efficient, offering short extraction times and high yields [37]. Ultrasonic-assisted extraction is another rapid method that preserves the integrity of bioactive substances and requires less solvent [38]. Enzymatic extraction has recently gained popularity due to its environmental friendliness, efficiency, mild operating conditions, and low energy requirements [39], although it is sensitive to factors such as enzyme type, temperature, and concentration [40].
In a comprehensive review of LBP extraction, purification, and structural characterisation, Masci et al. analysed various methods, reporting extraction yields ranging from 4% to 8% [41]. Using response surface methodology (RSM), Yin and colleagues identified optimal extraction conditions as 5.5 h at 100 °C, with a water-to-raw material ratio of 31.2 and five extraction cycles, achieving a polysaccharide mass fraction of 23% [42]. However, such extended extraction at elevated temperatures may lead to degradation or structural modifications of LBP, potentially affecting its bioactivity and molecular weight profile [43]. Xia and his team optimised LBP extraction using an amylase method, determining through RSM and orthogonal testing that the best conditions were an enzymolysis temperature of 49.56 °C, 0.3% enzyme concentration, and 140 min of enzymolysis time, resulting in a maximum extraction rate of 13.25% [44]. Amylase is often added during hot-water extraction to hydrolyse starch and improve LBP extraction efficiency. However, enzymatic hydrolysis may also modify the structural features of LBPs by partially degrading high-molecular-weight fractions, leading to reduced polymer size and altered bioactivity [45]. Therefore, LBPs extracted via this method may not fully represent the native polysaccharide composition in L. barbarum, and comparisons with intact plant-derived LBPs should be interpreted with caution.
Zhang et al. utilised the phenol-sulfuric acid method to determine LBP content, finding extraction rates of 6.71% for hot water extraction and 9.62% for microwave-ultrasonic synergistic extraction, respectively [46]. Quan and colleagues also employed a combination of microwave and ultrasound methods for LBP extraction, establishing optimal conditions of 16 min for microwave extraction, 20 min for ultrasound extraction, and a liquid-solid ratio of 55:1 using water as the solvent, yielding a polysaccharide extraction rate of 1.87% [47].
Extensive research on LBPs has identified over 33 different polysaccharides from L. barbarum, with relative molecular weights ranging from 10 to 2300 kDa [43]. However, this number is strongly influenced by the extraction methods used. Milder extraction conditions are more likely to preserve native LBP structures, thereby providing a more accurate representation of the true diversity of polysaccharides in L. barbarum. Structurally, LBPs are composed of various monosaccharides, including D-galactose (Gal), D-glucose, D-mannose, arabinose (Ara), D-xylose, D-ribose, and rhamnose (Rha) (Figure 2), as well as galacturonic acid and 18 amino acids. These monosaccharides are linked by glycosidic bonds, primarily (1→3)-β-Galp, (1→4)-β-Galp, (1→4)-β-Galp, (1→6)-α-glucans, and (1→4)-α polygalacturonans, with numerous branches and terminal groups. The LBPs are notably branched, featuring a main chain of (1→6) Galp-connected galactose [25]. Research has identified around 20 types of polysaccharides, including Rha, fucose (Fuc), Ara, Gal, and galacturonic acid (GalA) [43]. The complex structure of these polysaccharides is crucial for their biological activity. Studies have shown that LBPs with different molecular weights exhibit varied biological functions, such as antioxidant, immunomodulatory, and hypoglycemic effects [35], which will be further discussed in the following sections. Selecting an appropriate extraction and purification method is essential to optimize the yield while preserving the biological activity of LBPs.

3.2. Polyphenols

L. barbarum is rich in polyphenols, renowned for their antioxidant properties. Various compounds, including flavonoids, phenolic acids, and proanthocyanidins, have been identified in the leaves, fruits, and root bark of L. barbarum [13,48]. The polyphenol content in L. barbarum is influenced by factors such as geographical origin, variety, harvest season, and extraction methods [13]. Islam and co-authors reported average total phenolic content (TPC) and total flavonoid content (TFC) in dried L. barbarum fruits from China as 3.16 mg gallic acid equivalents per gram (GAE/g) and 2.83 mg catechin equivalents per gram (CAE/g), respectively [49]. In contrast, Ebeydulla et al. found that after wax removal and hot drying, TPC and TFC were 73.98 ± 5.10 and 63.10 ± 5.10 mg GAE/100 g, respectively, with even higher values observed after natural air drying (76.95 ± 2.28 and 66.78 ± 8.84 mg GAE/100 g) [50]. In Serbia, L. barbarum cultivated by Ilić and his group showed TPC and TFC levels of 162.4 ± 11.5 mg GAE/100 g and 214.2 ± 28.6 mg HE/100 g, respectively [14]. In a similar study, Donno et al. reported TPC of 281.91 mg GAE/100 g FW in L. barbarum fruits from Northern Italy [51].
In a study examining L. barbarum berries from Gansu Province, China, nine phenolic compounds were identified, including quercetin, isoquercitrin, chlorogenic acid, ferulic acid, and rutin [52]. Liu and co-authors characterised 11 polyphenols in L. barbarum fruits, covering phenolic acids (chlorogenic acid, caffeic acid, ellagic acid, p-hydroxycinnamic acid), flavonoids (rutin, morin, quercetin), and coumarins (scopoletin, 5,7-dihydroxy-4-methylcoumarin, esculetin) and curcumin [53]. Similarly, Pires and co-authors reported 19 phenolic compounds in L. barbarum berries from Portugal, with quercetin-3-O-rutinoside (16.6 mg/g dw) and p-coumaric acid (12.3 mg/gDW) being the most abundant [15]. Additionally, among the 15 compounds positively identified by Inbaraj’s team [54], quercetin-rhamno-di-hexoside (438.6 μg/g) and quercetin-3-O-rutinoside (281.3 μg/g) were the most abundant. Interestingly, a novel N-feruloyl tyramine dimer was discovered as the most abundant antioxidant polyphenol in L. barbarum fruit [55]. Rutin and quercetin are suggested as the principal polyphenols in L. barbarum [13,56]. L. barbarum leaves also contain significant amounts of flavonoids and exhibit antioxidant and antimicrobial activities [57]. Zhu et al. identified 20 polyphenolic compounds in L. barbarum leaves, including several chlorogenic acids, with some being reported in Chinese cultivated leaves for the first time [48].

3.3. Alkaloids

Alkaloids also represent a significant group of bioactive compounds found in L. barbarum. Among them, betaine is the most prominent and extensively studied alkaloid, recognised for its roles in osmoregulation, liver protection, and cardiovascular health [58]. Other alkaloids, such as atropine and hyoscyamine, are also present, though they have received less attention in the context of L. barbarum [59]. The accumulation of betaine in L. barbarum fruits involves key enzymes such as phosphatidylethanolamine N-methyltransferase, choline monooxygenase, and betaine aldehyde dehydrogenase [60].
A recent study reported betaine content ranging from 1.45% to 1.65% in 30 L. barbarum samples from various regions of China and Korea [61]. These variations in betaine levels are attributed to differences in regional growth conditions, including growth duration, harvest timing, temperature, and soil quality. Additionally, processing has been shown to reduce betaine content significantly, with dried fruit containing 7.49 g/kg, while fresh juice contained only 1.52 g/kg [62]. Betaine, along with LBPs, has demonstrated potential in attenuating liver fibrosis by inhibiting hepatic stellate cell activation and reducing the expression of fibrotic markers under oxidative stress conditions [63].
Recent research has further expanded our understanding of the alkaloid profile in L. barbarum. Chen and his team identified nine alkaloids in L. barbarum fruits, including a novel compound, lycibarbarspermidine T, with several of these alkaloids exhibiting anti-inflammatory effects by inhibiting nitric oxide (NO) production in RAW 264.7 macrophage cells [64]. In another study, Chen et al. discovered three distinct tetrahydroquinoline alkaloids, lycibarbarines A-C, which demonstrated neuroprotective effects against corticosterone-induced injury in PC12 cells [65]. Additionally, Kokotkiewicz and colleagues developed and validated TLC methods for the rapid analysis of tropane and steroidal alkaloids in L. barbarum [66]. However, when applied to the fruits, leaves, stems, and roots of three L. barbarum varieties cultivated in Pruszków, Poland, none of these alkaloids were detected in the analysed plant parts.

3.4. Carotenoids

Carotenoids are tetraterpene pigments that impart a range of colours, including yellow, orange, red, and purple, to various plants and organisms. They are widely distributed across nature, found in photosynthetic bacteria, certain archaea, fungi, algae, plants, and animals, with over 850 naturally occurring types identified, underlining their diversity and biological significance [67,68]. In plants, carotenoids play vital roles in photosynthesis, photoprotection, and act as antioxidants [67]. In wolfberries (Lycium species), the carotenoid content dictates their colour variations. These include red wolfberries [69], yellow wolfberries [69], and black wolfberries (L. ruthenicum), each classified based on specific pigmentation [70]. Extensive research on L. barbarum fruits reveals that they are rich in carotenoids, with concentrations typically ranging from 120 to 400 mg/100 g of mature fruit [71,72]. The carotenoid profile in these fruits is dominated by zeaxanthin dipalmitate, followed by β-cryptoxanthin monopalmitate, zeaxanthin monopalmitate, β-carotene, and free zeaxanthin [73].
Geographical origin has a notable impact on carotenoid content. A study of 34 batches of L. barbarum samples from Ningxia and Gansu regions in China found that zeaxanthin dipalmitate levels ranged from 0.81 mg/g to 4.05 mg/g, while zeaxanthin levels varied between 5.88 and 28.17 μg/g, and β-carotene approached its limit value [71]. Another study identified four key carotenoids in 21 L. barbarum fruit samples from Wrocław, Poland, with average concentrations of zeaxanthin (845.39 mg/kg), cryptoflavin (722.94 mg/kg), β-carotene (193.53 mg/kg), and neoxanthin (160.35 mg/kg) [74]. Additionally, Hsu et al. identified nine carotenoid components, including all-trans zeaxanthin and its isomers, all-trans β-carotene, neoxanthin, and β-cryptoxanthin, with the highest content of all-trans zeaxanthin and its isomers reaching 1666.3 μg/g, and the lowest neoxanthin at 4.47 μg/g [75].
Carotenoids in L. barbarum, particularly zeaxanthin, are known to play a significant role in preventing and improving vision in early age-related macular degeneration (AMD) [76]. A recent study also highlights their potential in cancer treatment, showing significant inhibition of HT-29 colon cancer cell growth by modulating key regulatory proteins (P53, P21, CDK2, CDK1, cyclin A, and cyclin B) and arresting the cell cycle at G2/M [75]. Despite these benefits, carotenoids in L. barbarum are prone to degradation under high-temperature and high-humidity conditions [71].

4. Biological Properties

The biological properties of L. barbarum encompass a wide spectrum of health-promoting effects, reflecting its diverse bioactive composition. This section introduces the key therapeutic potentials of L. barbarum, focusing on its ability to regulate metabolism, protect cardiovascular health, improve glycaemic control, and support neuroprotection. Additionally, its antioxidant, anti-inflammatory, and antimicrobial activities are highlighted, alongside its roles in modulating the immune system, promoting gut microbiota balance, and mitigating age-related and ocular health challenges. These interconnected benefits are outlined in Figure 3, providing an overview of L. barbarum’s multifaceted biological effects.
To support a more translational and functional understanding, this section is organised by biological activity rather than by compound class. Given that key constituents such as polysaccharides and flavonoids often exert pleiotropic effects across systems, this thematic structure better illustrates their impact on specific health outcomes. To minimise redundancy, overlapping mechanisms are noted briefly where relevant. The following subsections offer a detailed exploration of these biological effects, highlighting their relevance in nutraceutical and therapeutic applications.

4.1. Cardiovascular Benefits

Several studies have demonstrated the cardioprotective effects of LBPs and extracts, particularly through antioxidant, anti-apoptotic, and endothelial-protective mechanisms. Key findings are summarised in Table 1.
For instance, Wu et al. demonstrated LBPs reduced myocardial ischemia–reperfusion (I/R) injury in cardiomyocytes using an in vitro hypoxia/reoxygenation model. The results indicated that LBPs alleviated cell damage and reduced apoptosis in H9c2 cells by upregulating SIRT3 expression and promoting the deacetylation of CypD, thereby protecting mitochondrial function [77]. Inhibition of SIRT3 negated these protective effects, highlighting the significance of the SIRT3/CypD pathway in reducing myocardial injury. Similarly, LBPs were found to reduce infarct size, prevent adverse remodelling, and lower oxidative stress by inhibiting G protein-coupled receptor kinase-2 (GRK2) expression, a key driver of adverse cardiac remodelling following I/R injury [78]. Furthermore, LBPs restored mitochondrial fission/fusion balance and activated the AKT/eNOS signalling pathway, positioning it as a promising therapeutic candidate for cardiomyocyte protection during I/R injury.
Additionally, Xue et al. demonstrated that LBPs protect against oxidative stress-induced injury in rat aortic endothelial cells, a significant factor in endothelial dysfunction and cardiovascular diseases (CVDs) [79]. The results showed enhanced superoxide dismutase (SOD) activity and NO levels, along with reduced malondialdehyde (MDA) levels, indicating potent antioxidant effects. LBP also increased Bcl-2 expression while reducing Bax levels, demonstrating anti-apoptotic properties that could help preserve endothelial function and prevent oxidative stress-related vascular damage. In addition, another study investigated L. barbarum’s antihypertensive effects in a rat model of salt-sensitive hypertension. After 16 weeks of treatment, L. barbarum normalised elevated blood pressure and reduced lncRNA sONE expression while increasing eNOS levels, suggesting a regulatory mechanism for its antihypertensive action [80].

4.2. Glycaemic Control and Anti-Diabetic Activity

The anti-diabetic effects of L. barbarum have been extensively studied, revealing its potential to lower blood glucose levels, improve insulin sensitivity, and regulate lipid metabolism. Its bioactive components, including LBPs, leaf flavonoids and zeaxanthin, target key pathways involved in metabolic health, offering promise for obesity management and diabetes control [81,82]. Key findings are summarised in Table 2.

4.2.1. Obesity and Insulin Sensitivity

Research indicates that LBPs may counteract obesity by modulating lipid-associated pathways in adipocytes. Zhou et al. showed that LBP supplementation (50 and 100 mg/kg/day for 12 weeks) alleviated lipid droplet accumulation in adipose tissue by downregulating Fsp27 expression via an ATF6/SIRT1-dependent pathway [83]. In high-fat diet-fed mice and 3T3-L1 preadipocytes, LBPs reduced lipid droplet buildup, activated SIRT1, and decreased ATF6 and Fsp27 expression, highlighting its regulatory role in adipocyte function. Additionally, a bioactivity-guided purification identified LICP009-2-1 as the key anti-adipogenic compound, which reduced lipid accumulation in adipocytes and hyperlipidaemia in obese mice, positioning it as a potential functional food ingredient for anti-obesity interventions [84].
LBPs have also been shown to reduce glucose levels, improve insulin sensitivity, and increase testosterone levels in high-fat diet-induced obese mice, suggesting its potential to alleviate metabolic disturbances associated with obesity [85].
Recent study has further demonstrated that L. barbarum leaf flavonoids (LBLFs) improve insulin resistance by regulating blood glucose levels and lipid disorders. In a high fructose-induced mouse model, LBLF treatment decreased fasting blood glucose and insulin levels, reduced oxidative stress in the liver, and enhanced liver function. Additionally, LBLF optimised the gut microbiota composition by lowering the Firmicutes/Bacteroidetes ratio, highlighting its therapeutic potential in managing insulin resistance and related metabolic disorders [86]. Furthermore, LBLF treatment modulated gene expression in the mitogen-activated protein kinase (MAPK) and retinol metabolism pathways, further supporting its efficacy in managing insulin resistance and metabolic dysfunctions [86]. Furthermore, emerging evidence suggests that LBP may provide therapeutic benefits in sarcopenic obesity, a condition characterised by the coexistence of obesity and muscle wasting. In a recent study, LBP was shown to improve insulin sensitivity by modulating IRS-1 and GLUT-4 expression and mitigating ectopic fat deposition in skeletal muscle. These effects were mediated through activation of the AMPK/PINK1/Parkin-dependent mitophagy pathway, which restored mitochondrial function and reduced oxidative stress. These findings position LBP as a promising candidate for managing complex metabolic syndromes involving both adipose and muscle tissues [87].

4.2.2. Glycaemic Control and Diabetes Management

Zeaxanthin, a major carotenoid in L. barbarum, demonstrated significant antidiabetic, hypolipidemic, and nephroprotective effects in a high-fat, high-sucrose diet and streptozotocin-induced diabetic rat model. Zeaxanthin improved fasting blood glucose, lipid profiles with a lowered low-density lipoprotein cholesterol, triglycerides, and total cholesterol while increasing high-density lipoprotein cholesterol and kidney function, while also modulating inflammatory markers [e.g., interleukin (IL)-2, IL-6, tumour necrosis factor (TNF)-α, Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB)], and antioxidant enzyme levels [88].
LBPs were also found to improve glucose homeostasis in prediabetic mice by modulating gut motility and increasing short-chain fatty acids, particularly acetic acid, which may regulate duodenal contraction via neuronal pathways. This antiglycaemic effect represents a novel therapeutic approach for managing dysglycaemia [89]. Furthermore, LBPs reduced blood glucose and oxidative stress markers such as MDA and NO while increasing SOD levels in non-insulin-dependent diabetic rats. LBPs also decreased DNA damage in peripheral lymphocytes, highlighting its antioxidative properties as a protective dietary supplement for diabetes complications [90]. In a recent study, the water extract of L. barbarum leaf (LBL) was shown to effectively modulate blood glucose and lipid levels in a type 2 diabetic rat model, while also repairing damage to organs such as the liver, kidneys, and pancreas. This therapeutic effect was linked to the reversal of type 2 diabetes mellitus (T2DM)-induced metabolic disruptions and gut microbiota dysbiosis [91].

4.2.3. Effects on Lipid Metabolism

The ability of L. barbarum to regulate lipid metabolism has been increasingly supported by recent studies. LBPs contribute to lipid homeostasis by modulating gut microbiota composition, improving lipid profiles, and enhancing antioxidant defences.
In a high-fat diet-induced obesity model, supplementation with 150 mg/kg of LBPs significantly reduced serum and hepatic cholesterol and triglyceride levels, promoted weight loss, and shifted gut microbiota composition by decreasing Firmicutes and increasing Bacteroidetes [92]. Similarly, on spotted sea bass (Lateolabrax maculatus) fed high-lipid diets inducing lipid metabolism disorders, LBP supplementation improved weight gain, digestion, and antioxidant capacity while reducing oxidative stress and liver damage. These benefits were associated with upregulated expression of lipid metabolism-related genes, such as FAS, PPAR-α, CPT1, and ATGL, further highlighting the therapeutic potential of LBPs in regulating lipid metabolism [93]. A study on common carp (Cyprinus carpio) fed high-fat diets showed that LBP supplementation (0.5–2.0 g/kg) significantly improved growth performance and antioxidant capacity while alleviating lipid metabolism disorders. LBPs also downregulated the expression of key lipid metabolism-related genes, including acetyl-CoA carboxylase 1 and peroxisome proliferator-activated receptor-γ [94].
Supporting this, a recent study demonstrated that LBP positively modulates gut microbiota by increasing Firmicutes abundance and microbial diversity in obese rats. These changes were accompanied by improved lipid metabolism and alleviation of metabolic disorder symptoms. Over 30 differential metabolites and four metabolic pathways were influenced by LBP intervention, contributing to reduced weight gain, although not fully reversing obesity [95].

4.2.4. Obesity-Related Bone Health

Recent evidence challenges the long-standing belief that obesity protects against fractures, showing instead an increased risk of fractures in patients with higher visceral adipose tissue content [98]. Emerging evidence also highlights the role of L. barbarum in mitigating obesity-related bone health complications. As summarised in Table 2, LBPs have been shown to promote osteoblast survival and activity through both direct cellular pathways and gut microbiota-mediated mechanisms.
Jing et al. demonstrated that LBPs mitigated palmitic acid-induced osteoblast apoptosis in MC3T3-E1 cells by downregulating miR-200b-3p, which upregulated the Chrdl1 gene [96]. The modulation of the miR-200b-3p/Chrdl1/PPARγ pathway promotes osteoblast survival and functionality, suggesting a protective role in skeletal health.
In addition, a study exploring the prebiotic properties of LBPs found that they modulate intestinal microbiota metabolites, leading to increased production of short-chain fatty acids (SCFAs) such as acetic, propionic, and butyric acid. These SCFAs were shown to enhance osteoblast proliferation and differentiation by upregulating bone-alkaline phosphatase (B-ALP) and osteocalcin expression in vitro, thereby supporting bone metabolism in a postmenopausal osteoporosis model [97].
These findings highlight the multifunctionality of LBPs, which not only regulate metabolic health but also mitigate skeletal complications associated with obesity and metabolic disorders. Collectively, these studies position L. barbarum as a promising nutraceutical agent for maintaining bone health in individuals at risk of obesity-related osteoporosis. Further research is warranted to elucidate the molecular mechanisms underlying these effects and to determine optimal dosages for therapeutic use. The broad-ranging benefits of L. barbarum, from improving glucose regulation and lipid metabolism to supporting bone health, highlight its therapeutic potential. However, more targeted investigations are necessary to evaluate its long-term safety, efficacy, and clinical applicability in human populations.

4.3. Antioxidant Activity

Oxidative stress arises from an imbalance between pro-oxidants and antioxidants, typically due to excessive reactive species production. This imbalance contributes to the progression of numerous chronic diseases, including arthritis, atherosclerosis, cancer, diabetes, and neurodegenerative conditions [99,100]. While endogenous antioxidant systems offer a degree of protection, dietary supplementation with natural antioxidants—particularly phenolic compounds found in fruits and vegetables—can play a pivotal role in maintaining redox balance [99].
Among these bioactives, phenolic acids, flavonoids, carotenoids, polysaccharides, tocopherol, ascorbic acid, and condensed tannins are well-established contributors to antioxidant capacity [22]. These compounds act through direct free radical scavenging via hydrogen donation or electron transfer, metal chelation, and by enhancing endogenous antioxidant enzymes such as SOD, catalase (CAT), and glutathione peroxidase (GPx). The antioxidant and anti-ageing potential of Lycium barbarum, largely attributed to its carotenoids, ascorbic acid derivatives, flavonoids, and polysaccharides, has been validated across a broad range of studies [25,43] (Table 3).
Antioxidant activity is typically quantified using chemical assays such as 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) •+ radical scavenging, metal ion reduction [ferric ion-reducing antioxidant potential (FRAP), cupric ion-reducing antioxidant capacity (CUPRAC), Folin–Ciocalteu (FC)], and oxygen radical absorbance capacity (ORAC) [101,102]. These methods have been widely employed to characterise the antioxidant potential of L. barbarum extracts and fractions.
Table 3. Effects of L. barbarum on antioxidant activity.
Table 3. Effects of L. barbarum on antioxidant activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vitroFlavonoids,
Zeaxanthin,
LBP
Effective free radical scavengingH-donation, metal chelation [103]
In vitro Phenolics,
Flavonoids,
Anthocyanins
Strong correlation with antioxidant capacityAssay-based correlation [104]
In vitroPolyphenols,
Organic acids
High TPC, FRAP, TBCC levelsDirect ROS scavenging[51]
In vitroZeaxanthin,
Carotenoids
ABTS/FRAP correlations Antioxidant capacity [74]
In vitroAqueous extract↑ GSH, ↓ lipid peroxidation and protein carbonylsAntioxidant enzyme activation[105]
In vitroLBP↓ Apoptosis, ↑ cell protection against H2O2Nrf2 pathway[106]
In vivoLBP ↑ SOD, ↑ CAT, ↑ GPx, ↓ MDAEnzyme-mediated ROS defence[107]
In vivo LBPProtected liver/kidney from ROS damageAntioxidant defence[108]
CAT: catalase; FRAP: ferric-reducing antioxidant power; GPx: glutathione peroxidase; GSH: glutathione; LBP: L. barbarum polysaccharides; MDA: malondialdehyde; ROS: reactive oxygen species; SOD: superoxide dismutase; TBCC: total biophenolic content capacity; TFC: total flavonoid content.
Wang et al. demonstrated that the flavonoid-rich fraction of L. barbarum was particularly effective in DPPH and ABTS scavenging, metal chelation, and reducing power assays, while the zeaxanthin and polysaccharide fractions showed the most pronounced effect on scavenging hydroxy free radicals and superoxide anions, respectively [103].
Similarly, Islam et al. demonstrated strong positive correlations (0.786–0.856) between phenolic content and antioxidant activity in berry extracts, confirming the significance of total phenolics, flavonoids, tannins, and anthocyanins in antioxidant efficacy [49].
Donno et al. extended these findings by evaluating methanolic extracts of L. barbarum, reporting high TPC (268.5 mg GAE/100 g FW), antioxidant capacity [FRAP 19.36 mmol Fe2+/kg); total bioactive compound content (TBCC, 5806.80 mg/100 g FW)]. Polyphenols (16.2% including cinnamic acids, catechins, flavonols, and benzoic acids) and organic acids (76.8%) were the dominant bioactive groups, both of which showed strong correlations (0.8290 for phenols, 0.8606 for organic acids, 0.9996 for TPC, and 0.8363 for TBCC) with antioxidant capacity [51].
Wojdyło and colleagues corroborated these results, identifying substantial levels of polyphenols (972.32 mg/kg DW), carotenoids (2129.44 mg/kg DW), flavonols (75.3%), and phenolic acids (24.7%). Their study identified significant linear correlations between antioxidant assay performance (ABTS and FRAP) and various bioactive compounds such as total polyphenolic compounds, flavanols, phenolic acids, carotenoids, and specific carotenoids like zeaxanthin, β-carotene, neoxanthin, and cryptoxanthin [74].
Further support comes from in vitro cellular assays. Skenderidis and co-authors demonstrated that aqueous extracts of L. barbarum exhibited potent DPPH and ABTS scavenging (IC50 = 1.29–3.00 mg/mL and 0.39–1.10 mg/mL, respectively). At concentrations of 25 and 100 μg/mL, the extract increased glutathione (GSH) levels by up to 189.5% and decreased lipid peroxidation and protein carbonyls by 21.8% and 29.1%, respectively, in C2C12 muscle cells treated with the extract, highlighting its protective effects against oxidative stress [105]. In arising retinal epithelial (ARPE)-19 cells, LBP pre-treatment, before exposure to H2O2, mitigated oxidative damage, inhibited apoptosis, and modulated apoptotic proteins, potentially through the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signalling pathway, thereby reinforcing the antioxidant properties [106].
In vivo evidence further confirms these antioxidant effects. Pehlivan Karakaçs et al. reported increased antioxidant enzyme levels (SOD, CAT, GPx) and reduced MDA levels in rats following LBP administration, indicating effective physiological antioxidant activity [107]. In the STZ-induced diabetic rat model, LBP supplementation exhibited significant free radical scavenging activities, helping to protect liver and kidney tissue from oxidative damage, reinforcing its therapeutic potential [108].
Overall, these studies collectively highlight the robust antioxidant potential of L. barbarum attributed to its rich content of phenolic compounds, flavonoids, carotenoids, and polysaccharides, making it a promising candidate for the development of natural antioxidants and dietary supplements aimed at combating oxidative stress-related diseases.

4.4. Anti-Inflammatory Activity

Inflammation, whether acute or chronic, serves as a fundamental defence mechanism against harmful stimuli such as pathogens, irritants, or injury. While acute inflammation is protective and typically self-limiting, unresolved or repeated inflammatory responses can evolve into chronic inflammation, which underlies many metabolic and degenerative diseases [109]. Central to this process is the NF-κB signalling pathway, which regulates the expression of pro-inflammatory genes and cytokines, including TNF-α, IL-6, IL-1β, MCP-1, adipokines, cell adhesion molecules (sVCAM-1 and sICAM-1), and the acute-phase C-reactive protein (CRP) [110].
Nardi et al. investigated the anti-inflammatory effects of L. barbarum berries using a carrageenan-induced paw oedema model in mice. Oral administration of the extract at 50 and 200 mg/kg over 10 days significantly reduced paw swelling by 38% and 63.8%, respectively. These findings suggest a dose-dependent anti-inflammatory effect, likely mediated through the inhibition of neutrophil infiltration and associated oxidative stress [56].
Building on these findings, Lee et al. demonstrated for the first time that supplementation of non-fermented and fermented L. barbarum protected against high fat-induced metabolic complications, primarily by improving hepatic function and lipid metabolism via combined anti-inflammatory and antioxidant actions [111].
Xiao et al. investigated the effects of LBP in a high-fat diet-induced non-alcoholic steatohepatitis (NASH) rat model, which revealed that LBP administration significantly ameliorated increases in body and liver weight, insulin resistance, glucose metabolic dysfunction, elevated serum aminotransferases, hepatic fat accumulation, fibrosis, oxidative stress, inflammation, and apoptosis [112]. These effects were partially mediated through modulation of the NF-κB, MAPK pathways, and autophagic processes [112].
In a high-fat diet-streptozotocin-induced diabetic rat model, LBP significantly inhibited albuminuria, decreased blood urea nitrogen levels, and serum inflammatory cytokines, including IL-2, IL-6, TNF-α, interferon (IFN)-α, MCP-1, and ICAM-1. Additionally, LBP enhanced serum antioxidant activity (SOD and GSH-Px) and suppressed NF-κB expression in kidney tissues, suggesting both anti-inflammatory and antioxidant mechanisms of renal protection [113].
Further mechanistic insights were provided by Xie and co-authors who proposed that the anti-inflammatory and anti-apoptotic effects of LBP in the kidneys of mice are mediated through activation of the Nrf2 signalling pathway [114]. Similarly, Gan et al. observed that LBP supplementation alleviated CCl4-induced hepatic oxidative injury and inflammatory response in Wistar rats, possibly through inhibiting the toll-like receptors (TLRs)/NF-kB signalling pathway [115]. In a diabetic rabbit model, Zhao’ group found that LBP improved renal function and alleviated kidney inflammation, with a more pronounced preventive effect compared to treatment [116]. The protective effects of LBP on diabetic nephropathy may be attributed to the reduction in MCP-1mRNA and ICAM-1mRNA expression via inhibition of NF-κB and angiotensin II pathways [94].
Taken together, as summarised in Table 4, these findings highlight the robust anti-inflammatory effects of L. barbarum, mediated through inhibition of key inflammatory pathways (NF-κB, MAPK, TLR), enhancement of antioxidant defences, and modulation of cytokine expression. These properties position L. barbarum as a promising nutraceutical for managing inflammation-related conditions.

4.5. Immunomodulatory Effects

L. barbarum has demonstrated diverse immunomodulatory properties in both in vitro and in vivo models, largely attributed to its polysaccharide components (LBPs). These effects encompass the enhancement of innate and adaptive immune responses, modulation of cytokine production, and protection of immune organs under pathological conditions.
In the S180-bearing mice module, LBP effectively inhibited tumour growth, enhanced immune functions such as thymus index, macrophage phagocytic activity, spleen cell antibody secretion, and lymphocyte proliferation, while reducing lipid peroxidation, indicating its tumour-inhibitory mechanism is linked to immune function enhancement [117]. Similar immuno-enhancing effects were observed in H22-bearing mice, where LBP promoted CD8⁺ T-cell infiltration, prevented T-cell exhaustion, and sustained lymphocyte cytotoxicity [118].
Moreover, Feng and his team demonstrated that LBP, specifically the fraction with a molecular weight > 10 kDa, exerts notable immunomodulatory effects by enhancing the viability and polarisation of macrophage RAW264.7 cells [119]. This fraction effectively modulates inflammatory mediators such as NO, TNF-α, IL-6, and reactive oxygen species (ROS) in RAW264.7 cells and exhibits efficient cellular uptake, including transport across intestinal epithelial cells via clathrin-mediated endocytosis, highlighting its potential for therapeutic applications.
In poultry studies, LBP supplementation proved its clinical efficacy in broilers, showing that 4 g/kg LBP significantly improved feed conversion ratio, immune organ indexes, and the CD4+/CD8+ ratio in blood T cells. Meanwhile, 8 g/kg LBP increased serum total protein, globulin, albumin, and lysozyme levels. Additionally, 0.1 and 1.6 mg/mL LBP supplementation significantly promoted blood B and T lymphocyte proliferation and decreased TNF-α mRNA abundance, suggesting its potential as a growth-promoting and immunomodulatory feed additive [120]. Another study found that dietary supplementation of 2 g/kg LBP in broiler chickens significantly improved average daily gain, feed-to-gain ratio, digestive enzyme activities, antioxidant capacity, and immune function. Broilers fed LBP exhibited increased serum IgG, IgA, superoxide dismutase, glutathione peroxidase, and cytokines (TNF-α, IL-6, IFN-γ, IL-4), suggesting LBP as a promising feed additive for broilers [24].
In cyclophosphamide-induced immunosuppressed mice, LBP improved cytokine levels (IL-2, IL-12, TNF-α) and sperm quality (density, motility, normal morphology), suggesting protective effects in reproductive immunology. Optimal effects were observed at a 1.0 g/kg LBP dosage, while a higher dosage (1.5 g/kg) did not yield additional benefits [121]. Other work demonstrated that LBP modulated gut microbiota composition (increase in Bacteroidaceae, Lactobacillaceae, Prevotellaceae, and Verrucomicrobiaceae) and improved short-chain fatty acid production, linking its immunoregulatory effects to gut health [122]. Further, Cao et al. investigated the immunomodulatory effects of L. barbarum juice and its mixtures with other juices in a mouse model. It was found that the antioxidant activity of L. barbarum juice, particularly in combinations with raspberry and blueberry juices, correlates with increased splenic macrophage numbers and spleen weight, suggesting potential enhancements in immune response [28]. Table 5 provides an overview of the immunological activities of L. barbarum across different biological models.

4.6. Anticancer Activity

Cancer remains the second leading cause of death globally, with 19.3 million new cases and 10 million cancer-related deaths reported in 2020 alone [123]. As the search for safer and more effective cancer treatments continues, natural products have garnered significant interest for their ability to suppress tumour growth and enhance immune responses while reducing the side effects commonly associated with conventional therapies [124]. Among these, L. barbarum extracts and polysaccharides (LBPs) have demonstrated promising anticancer effects across a variety of cancer types and models [125]. Key studies detailing these effects are presented in Table 6.

4.6.1. Inhibition of Cancer Cell Proliferation

Several in vitro studies have shown that L. barbarum extracts can inhibit cancer cell proliferation through both direct cytotoxicity and immune-mediated mechanisms. In human colon cancer (LS180) cells, it was demonstrated that without the presence of the ethanolic L. barbarum extract, the most significant anticancer effect (eliminating 91% of cancer cells) occurred with a 1:1 ratio of LS180 to natural killer (NK)-92 cells. When L. barbarum extract was introduced at this ratio, 94.8% of LS180 cells were eliminated, showing that L. barbarum extract reduced LS180 proliferation by 5%, 12.8%, and 20.6% at concentrations of 1, 2.5, and 5 µg/mL, respectively. At a 2:3 ratio of LS180 to NK-92, L. barbarum extract (2.5 and 5 µg/mL) decreased LS180 cell viability to 96.5% and 98.1%, respectively. These results underline L. barbarum extract’s chemo-preventive potential, particularly through dose- and lymphocyte-dependent mechanisms [126].
Wawruszak et al. examined the impact of ethanolic L. barbarum extract on T47D human breast cancer cells, revealing a 70% reduction in cell proliferation at the highest concentration (1 mg/mL) after 24 h, which decreased to 55.7% and 51.4% at 48 and 96 h, respectively, using the MTT assay [31]. This decrease was corroborated by bromodeoxyuridine assays, which showed a sharp decline in proliferation, and Neutral Red assays, indicating a slight reduction in viability. The expression of key regulators such as p21 and p53 was increased, while cyclin-dependent kinase 6 (CDK6) and cyclin D1 levels were marginally decreased. Apoptosis was dose-dependent, with apoptosis rates of 37%, 61.6%, and 88% observed at concentrations of 0.1, 0.5, and 1 mg/mL, respectively, alongside necrotic changes at 0.5 mg/mL. Western blotting confirmed a dose-dependent increase in Bax and a decrease in BclxL, supporting the role of L. barbarum in promoting apoptosis via mitochondrial pathways [31]. Similarly, a pectin-free L. barbarum extract enriched with polyphenols has demonstrated significant inhibition of breast cancer cell proliferation, especially in oestrogen-positive (MCF-7) and triple-negative (MDA-MB-231) subtypes [127].
Expanding to other cancers, Gong et al. reported that various LBP fractions inhibited proliferation across multiple cell lines, including hepatoma (SMMC-7721 and HepG2), cervical cancer (HeLa), gastric carcinoma (SGC-7901), and human breast cancer (MCF-7) cells [128]. The various LBP fractions, isolated via water extraction and ethanol precipitation, exhibited dose-dependent inhibitory effects, with MCF-7 cells being particularly sensitive to the LBGP-I-3 fraction (reducing cell viability to 48.96%). Flow cytometry analysis showed G0/G1 phase arrest in MCF-7 cells treated with LBGP-I-3 at 1 mg/mL. Morphological changes characteristic of apoptosis were confirmed by AO-EB and DAPI staining, and these effects were associated with increased Caspase-3, -8, and -9 protein expression, along with a decreased Bcl-2/Bax ratio and reduced mitochondrial membrane potential. Additionally, LBGP-I-3 treatment reduced T-SOD and CAT activity, alongside a decline in GSH-Px and GSH levels, while enhancing MAPK signalling, as evidenced by p-JNK and p-p38 upregulation [128].
Furthermore, Zhang and colleagues reported that LBP exhibited anticancer activity in human hepatoma cells (QGY7703) by inducing S phase cell cycle arrest, and promoting apoptosis. Treatment with LBP resulted in increased RNA content and elevated intracellular calcium levels, suggesting apoptotic pathways linked to calcium regulation [129]. Similarly, Deng et al. identified LBP-3 (40–350 kDa) as the most potent fraction against H22 murine hepatoma cells, demonstrating its efficacy in inducing apoptosis and S phase arrest in vitro and tumour suppression in vivo without significant toxicity [130]. Follow-up research revealed that LBP could also enhance the anticancer efficacy of doxorubicin while supporting recovery of peripheral blood lymphocytes and bone marrow cell cycles in treated mice [135].
In oral squamous cell carcinoma (CAL-27), the ethanolic L. barbarum extract demonstrated significant antiproliferative and anti-invasive properties by inhibiting CAL-27 cell growth, proliferation, and migration, while modulating key signalling pathways, including downregulation of ERK1/2 and AKT1 phosphorylation, reduced cyclin D1, cadherin 2, and vimentin expression, and increased cadherin [131]. These findings suggest a multifaceted mechanism by which L. barbarum disrupts tumour progression.

4.6.2. Anti-Metastatic Properties

Beyond proliferation, L. barbarum has shown potential in limiting tumour metastasis. In hepatocellular carcinoma (HepG2) cells, L. barbarum extract protected against DNA damage and altered gene expression related to tumour progression. Specifically, the extract downregulated genes associated with tumour migration (CCL5), metastasis (DUSP1), and carcinogenesis (GPx-3, PTGS1), while upregulating the tumour suppressor genes such as MT3 [69]. These effects suggest that L. barbarum may modulate both oxidative stress and metastatic signalling pathways.

4.6.3. Synergistic Effects with Chemotherapy

Several studies have reported synergistic effects when L. barbarum is combined with chemotherapeutic agents. Liu et al. demonstrated that combining LBP and cisplatin significantly enhanced the inhibitory effect on the proliferation of human alveolar adenocarcinoma A549 cells [132]. This combination therapy not only promoted apoptosis but also regulated cell cycle proteins through the cyclin D1-CDK4-Rb pathway, indicating a potential synergistic anticancer effect. The potential of L. barbarum to amplify the effects of traditional chemotherapeutic agents while mitigating side effects presents a promising area of research in cancer treatment.
Another notable study on renal cell carcinoma (RCC) revealed that co-treatment with LBPs and recombinant IFN-α2b markedly inhibited tumour growth and enhanced apoptosis both in vitro and in vivo. This combination therapy led to the downregulation of cyclin D1, c-Myc, and Bcl-2 expression, while upregulating pro-apoptotic Bax. Additionally, the treatment reduced the presence of myeloid-derived suppressor cells (MDSCs) and tumour volume, leading to a stronger anti-tumour immune response than either agent alone [133].

4.6.4. Mitigation of Chemotherapy-Induced Toxicity

Importantly, L. barbarum may also reduce the side effects of chemotherapy. For instance, Xin and colleagues found that LBPs significantly mitigated doxorubicin-induced cardiotoxicity [134]. LBPs reduced oxidative stress and apoptosis in cardiac tissues, improved conduction abnormalities, and lessened myocardial damage. Crucially, these protective effects did not compromise the anticancer efficacy of doxorubicin [136]. This suggests that L. barbarum could serve as a valuable adjunct therapy to enhance treatment tolerability in cancer patients.

4.7. Hepatoprotective Activity

Hepatoprotective effects of L. barbarum have been validated in various chemically induced liver injury models. These benefits are mainly driven by suppression of oxidative stress, inflammasome activity, and inflammatory cytokine secretion. Key hepatoprotective findings are outlined in Table 7A.
In ethanol-induced liver injury, LBP conferred hepatoprotection by inhibiting thioredoxin-interacting protein (TXNIP) and NOD-like receptor protein 3 (NLRP3) inflammasome activation. Pre-treatment with LBP significantly attenuated ethanol-induced overexpression of TXNIP, reduced cellular apoptosis, and inhibited the activation of the NLRP3 inflammasome [137]. These effects were accompanied by a decrease in oxidative stress and inflammatory cytokine secretion, suggesting its potential as a therapeutic agent for liver diseases. Similarly, L. barbarum extract alleviated paracetamol-induced hepatotoxicity in rats, as indicated by improved total antioxidant status and reduced oxidative stress markers: total oxidant status, oxidative stress index, and serum liver function markers (alanine aminotransferase, ALT and aspartate aminotransferase, AST) [138]. Cheng’s group further supported the hepatoprotective effects of LBP in alcohol-induced liver damage. Rats treated with LBP (300 mg/kg body weight/day) showed improved liver function, reduced fatty liver progression, and enhanced antioxidant activity, confirming its potential as a treatment for alcoholic liver disease [139]. Future studies should investigate its efficacy in human models, focusing on chronic liver conditions and hepatotoxicity caused by long-term alcohol consumption.

4.8. Antimicrobial Effect

Recent interest in L. barbarum has been driven by its high content of polyphenols and other bioactive compounds, many of which demonstrate notable antimicrobial activity. The antimicrobial action is primarily attributed to polyphenolic compounds that disrupt microbial cell membranes. This disruption is influenced by factors such as pH and ionic strength, leading to altered membrane permeability and eventual cell death [15,29].
Pires et al. found that hydromethanolic extracts from L. barbarum fruits and stems exhibited stronger antibacterial activity against Gram-positive bacteria than Gram-negative bacteria, with minimum inhibitory concentrations (MICs) ranging from 2.5 to 20 mg/mL [15]. Particularly low MICs (2.5–5 mg/mL) were observed for Enterococcus faecalis, Staphylococcus aureus (including both methicillin-resistant and methicillin-sensitive strains), and Listeria monocytogenes, while higher concentrations (10–20 mg/mL) were required to inhibit Gram-negative bacteria, such as Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii [15]. Supporting this, Ilic et al. reported that yellow L. barbarum extracts exhibited strong antimicrobial activity against Gram-negative bacteria (Salmonella abony, P. aeruginosa, and K. pneumoniae) and Candida albicans (MIC = 2 mg/mL). In contrast, higher concentrations were required to inhibit Gram-positive bacteria, such as E. faecalis, Staphylococcus epidermidis, and S. aureus (MIC > 2 mg/mL) [14].
Chlorogenic acid, a key phenolic compound in L. barbarum, has also been identified as a potent antimicrobial agent against E. coli. Other metabolites, including benzoic, hydroxybenzoic, ferulic, and isoferulic acids, contribute to the reduction in E. coli populations [29], with activity influenced by factors such as time, temperature, and dosage [29].
In addition to systemic antibacterial effects, ethanolic extracts of L. barbarum have shown promise as an alternative to chlorhexidine, commonly used in treating gingivitis. These extracts have been assessed against various periodontal pathogens, including Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, Fusobacterium nucleatum, and Tannerella forsythia [140]. Table 7B summarises the major findings related to the antimicrobial activities of L. barbarum.

4.9. Prebiotic Activity

The prebiotic potential of L. barbarum has attracted growing attention, particularly due to its phenolic and polysaccharide components, which can beneficially modulate gut microbiota and support intestinal health. These properties are especially relevant in the context of rising intestinal disorders such as ulcerative colitis and Crohn’s disease [141].
In a study by Zhu et al., LBPs were shown to alter gut microbial composition by increasing the relative abundance of Firmicutes and Proteobacteria while decreasing Bacteroidetes [141]. Further analysis of the caecal microbiota in mice revealed that supplementation with LBPs led to an increased ratio of beneficial probiotic bacteria, including Lactobacillus, Akkermansia, and members of the Prevotellaceae family. These findings suggest that dietary supplementation with LBPs could serve as an effective strategy to modulate intestinal microbiota and promote gut health [141].
The interplay between gut and liver health has also been explored. In a mouse model of alcohol-induced liver injury, dietary intake of L. barbarum for 14 days resulted in lower serum levels of ALT and AST, enzymes commonly associated with liver damage [142]. L. barbarum supplementation significantly increased the relative abundance of butyric acid-producing bacteria, such as Lachnospiraceae and Ruminococcaceae. This modulation was largely attributed to increased levels of glutathione in the liver, which serves as a nutrient source for these beneficial bacteria [142].
Further evidence of the prebiotic activity of L. barbarum was observed in studies where aqueous extracts of the berries, encapsulated in maltodextrin, stimulated the growth of probiotic bacteria like Bifidobacterium animalis subsp. lactis (Bb12), B. longum (Bb46), and Lactobacillus casei, with increases of 2, 0.26, and 1.34 log CFU/mL, respectively [143]. Similarly, an in vitro assessment of LBPs demonstrated a significant enhancement in the growth of Lactobacillus acidophilus and Bifidobacterium longum, with log CFU/mL values increasing relative to the control, regardless of the polysaccharide concentration used (ranging from 2.5% to 15%) [141].
Together, these studies support the role of L. barbarum as a functional prebiotic capable of positively influencing gut microbiota composition, intestinal function, and host health. Table 7C compiles the key findings related to the prebiotic effects of L. barbarum.

4.10. Neuroprotective Effects

Neurodegenerative disorders, including Alzheimer’s disease and Parkinson’s disease, are characterised by the progressive loss of neuronal structure or function, leading to symptoms such as memory loss, motor dysfunction, and cognitive decline [144]. These disorders are triggered by a complex interplay of genetic, environmental, and lifestyle factors, sharing common pathological features such as protein misfolding, oxidative stress, and neuroinflammation [145]. Addressing these complex mechanisms remains a major challenge in therapeutic development.
Recent research has increasingly highlighted the neuroprotective effects of L. barbarum in various experimental models. The key experimental findings supporting these neuroprotective effects are summarised in Table 8.
In both ischemic stroke and radiation-induced neuronal damage, L. barbarum has been shown to modulate neuroinflammatory factors (cytokines and chemokines), reduce oxidative stress, and prevent hippocampal interneuron loss. These protective effects are mediated through multiple signalling pathways, including PI3K/Akt/GSK-3β, PI3K/Akt/mTOR, PKCε/Nrf2/haem oxygenase-1 (HO-1), keap1-Nrf2/HO-1, and NR2A/NR2B receptor-related mechanisms [146]. Such findings suggest that L. barbarum could serve as a valuable adjunct therapy in treating brain tumour during radiotherapy as well as in managing ischemic stroke.
The neuroprotective potential of L. barbarum has also been explored in chronic intermittent hypoxia (CIH)-induced spatial memory deficits, which mimic the conditions of obstructive sleep apnoea. In this model, LBP administration mitigated oxidative stress, inflammation, endoplasmic reticulum stress, and apoptosis in the hippocampus. Additionally, LBPs enhanced hippocampal neurogenesis and improved spatial memory performance, demonstrating a robust neuroprotective role [147]. In a separate study, Cao and his team further explored the neuroprotective effects of LBP using H2O2-treated PC12 cells and CoCl2-treated rats [148]. LBP demonstrated a concentration-dependent mitigation of oxidative stress-induced cellular damage, demonstrated by reduced ROS levels, decreased cell viability loss, and increased TUNEL-stained cells, as well as enhanced caspase-3 and -9 activity and mitochondrial apoptosis markers. Furthermore, LBP upregulated the of Nrf2 and HO-1 in both models, providing further evidence of its antioxidative and anti-apoptotic properties.
A recent investigation also revealed that L. barbarum could reverse cognitive impairment and depressive-like behaviour induced by light at night (LAN) exposure in mice. This was accompanied by reduced apoptosis and mitochondrial damage in HT-22 cells, along with enhanced antioxidant defences via activation of the Nrf2/HO-1 signalling pathway. Notably, the Nrf2 antagonist ML385 significantly diminished these neuroprotective effects, indicating the critical role of Nrf2 activation in L. barbarum’s mode of action [149]. In another study, L. barbarum water extract demonstrated significant neuroprotective effects in cellular and animal models of neurodegeneration. Using a differentiated PC12 cell model exposed to L-glutamic acid, L. barbarum enhanced cell survival, reduced apoptosis, suppressed intracellular ROS accumulation, blocked Ca2⁺ overload, and maintained mitochondrial membrane potential. Similarly, in an Alzheimer’s disease mouse model induced by AlCl₃ and D-galactose, L. barbarum improved cognitive function and increased levels of neurotransmitters, such as acetylcholine and choline acetyltransferase [150]. These findings suggest that L. barbarum may offer neuroprotective benefits against neurodegenerative diseases such as Alzheimer’s and Parkinson’s.
Further evidence supporting L. barbarum’s neuroprotective properties comes from a review by Lp and colleagues [152], which suggested that L. barbarum may be a potential therapy for ischemic stroke by inhibiting glutamate excitotoxicity and regulate signalling pathways. Moreover, an alkaline extract from L. barbarum has been found to protect primary cortical neurons against β-amyloid (Aβ)-induced neurotoxicity, a key feature of Alzheimer’s diseases. L. barbarum significantly reduced lactate dehydrogenase release and caspase-3 activity triggered by Aβ, with further analysis revealing that subfractions of L. barbarum, particularly subfraction-I and subfraction-II, exhibited differential neuroprotective effects through enhanced Akt phosphorylation. This suggests that the glycoconjugate isolated from L. barbarum using a novel alkaline extraction method may be promising for drug development in neurodegenerative diseases [30].
Similarly, Yao and co-authors examined the neuroprotective effects of L. barbarum extract in both Caenorhabditis elegans and PC12 cells treated with MPP(+), a neurotoxin that induces Parkinson-like symptom [151]. L. barbarum provided dose-dependent protection against dopamine neurodegeneration in C. elegans, while also attenuating cell damage, reducing intracellular ROS accumulation, stabilising mitochondrial membrane potential, and restoring total GSH levels in PC12 cells. These findings indicate that L. barbarum exerts neuroprotective effects primarily through its antioxidative mechanisms.

4.11. Anti-Aging Activity

Aging is characterised by the gradual accumulation of cellular and tissue damage, with oxidative stress playing a pivotal role [4]. Various signalling pathways influenced by environmental, hormonal, and nutritional factors contribute to this complex biological process.

4.11.1. Glycation and Oxidative Stress Reduction

Achyranthes bidentata polysaccharide (ABP) and LBP have demonstrated anti-aging effects in a D-galactose-induced mouse aging model. Both polysaccharides significantly reduced serum advanced glycation end-products and hydroxyproline concentrations in the skin while enhancing lymphocyte proliferation, IL-2 activity, and neurocognitive functions, including improved learning and memory. Additionally, spontaneous motor activity and SOD levels increased. Remarkably, ABP exhibited stronger inhibitory effects on non-enzymatic glycation than LBP, highlighting its potential in age-related treatments [153].
In a separate study on Drosophila melanogaster, LBPs were found to significantly extend the average lifespan of fruit flies. This effect was linked to enhanced antioxidant activity, characterised by elevated SOD and CAT levels and reduced MDA concentrations, indicating lower oxidative stress. Particularly, the LBP-2 fraction, rich in arabinogalactan, showed the strongest anti-aging properties, likely due to its modulation of MAPK, target of rapamycin (TOR), and S6K pathways, and upregulation of longevity genes [154].
Further research explored LBP’s photoprotective properties, particularly against UVB-induced skin damage. LBP pretreatment increased cell viability, reduced ROS, and mitigated DNA damage in immortalised human keratinocytes (HaCaT cells). The protection was attributed to the inhibition of UVB-induced p38 MAPK activation, reduced caspase-3 activation, and decreased MMP-9 expression. LBP also activated the Nrf2/ARE signalling pathway, promoting antioxidant gene expression [155].

4.11.2. Stem Cell Promotion and Tissue Regeneration

Emerging evidence highlights LBP’s role in promoting stem cell function and tissue regeneration. In particular, the LBP1C-2 fraction, a homogeneous LBP isolated from the mice, was found to increase satellite cell (muscle stem cells) activation and self-renewal, promoting muscle repair in both adult and aging mice. This mechanism is mediated by the binding of LBP to FGFR1, which upregulates Spry1, enhancing stem cell renewal [156].
LBP has also shown potential in periodontal therapy by promoting the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) and reducing bone loss in periodontitis models. In vitro, LBP increased the proliferation and migration of hPDLSCs while enhancing the expression of key osteogenic markers (e.g., RUNX2, ALP, collagen I, and osteocalcin) via activation of the ERK1/2 signalling pathway. In vivo, LBP significantly reduced alveolar bone resorption and osteoclast activity in a rat periodontal model [157]. Additionally, in zebrafish models, LBP treatment reduced cell senescence and apoptosis through modulation of the p53 signalling pathway, further highlighting its anti-aging effects [158].

4.11.3. Anti-Apoptotic Effects

L. barbarum polysaccharides from Fructus Lycii have demonstrated protective effects against oxidative stress-induced apoptosis, particularly in murine seminiferous epithelium. LBP treatment reduced lipid peroxidation and delayed apoptosis under hyperthermic and normothermic conditions, showing potential for both fertility and anti-aging applications [159]. LBP’s neuroprotective and anti-aging effects were further highlighted by Yu and colleagues, where LBP treatment reduced oxidative stress, apoptosis, and autophagic cell death against oxygen glucose deprivation/reperfusion (OGD/R)-induced injury in hippocampal neurons while modulating key apoptosis-related proteins, including Bcl-2/Bax and cleaved Caspase-3 [160]. The protective effects were attributed to the activation of the PI3K/Akt/mTOR signalling pathway [160]. Similarly, LBP alleviated myocardial I/R injury by upregulating SIRT3 expression, promoting the deacetylation of Cyclophilin D, enhancing mitochondrial protection and improving cardiomyocyte survival [77]. This connection between LBP and SIRT3 aligns with exercise-induced benefits, suggesting a shared mechanism for combating age-related mitochondrial decline [161].
The studies collectively demonstrate the significant anti-aging potential of LBPs. Through their antioxidant, anti-inflammatory, and anti-apoptotic properties, LBPs have been shown to modulate key signalling pathways involved in aging, including PI3K/Akt/mTOR and Nrf2/ARE. These mechanisms suggest that LBPs could be developed into effective therapeutic agents or supplements to mitigate age-related cellular damage and promote tissue regeneration. The key experimental findings supporting these anti-aging effects are summarised in Table 9.

4.12. Ocular Health

Lycium barbarum polysaccharides (LBPs) have shown therapeutic potential in a variety of ocular disorders, including glaucoma, retinitis pigmentosa (RP), AMD, ocular hypertension, and retinal ischaemia (Table 10).

4.12.1. Glaucoma

L. barbarum polysaccharides have demonstrated promising neuroprotective effects in glaucoma-related models, primarily through modulation of oxidative stress, inhibition of apoptosis, and preservation of retinal ganglion cells (RGCs). In an ischaemia/reperfusion (I/R) rat model, LBP treatment activated the Nrf2/HO-1 antioxidant pathway, significantly enhancing nuclear translocation of Nrf2 and expression of HO-1, which led to reduced retinal cell apoptosis. The protective effect was diminished upon administration of zinc protoporphyrin, an HO-1 inhibitor, indicating the essential role of the Nrf2/HO-1 axis in mediating retinal protection [162].
In models of optic nerve injury, particularly the partial optic nerve transection (PONT) model, LBPs were effective in delaying secondary degeneration of retinal ganglion cells (RGCs). While LBPs did not influence primary degeneration following complete optic nerve transection (CONT), it effectively inhibited oxidative stress and the c-jun N-terminal kinase (JNK) pathway, contributing to neuroprotection [163]. Additionally, LBP treatment was associated with a transient increase in insulin-like growth factor-1 (IGF-1), suggesting involvement of neurotrophic support mechanisms in glaucomatous protection [163].
Functionally, LBPs have been shown to enhance visual function, as evidenced by significant preservation of retinal function in Sprague Dawley rats. This was demonstrated by the recovery of multifocal electroretinogram (mfERG) amplitudes in the superior retina corresponding to the transacted area, indicating LBP modulates retinal signalling and mitigates functional decline associated with optic nerve injury, thereby highlighting its potential therapeutic role in vision restoration [164].
The reliability of the PONT model for studying secondary degeneration of RGCs has been further confirmed in recent investigations, which revealed significant changes in microglia/macrophage polarisation and autophagy levels following injury, with LBP treatment found to delay secondary RGC degeneration, promote M2 polarisation of microglia/macrophages, and reduce autophagy. These findings demonstrate the therapeutic potential of LBP in modulating retinal responses to optic nerve injury [165].

4.12.2. Retinitis Pigmentosa

Retinitis pigmentosa (RP) is an inherited disorder that primarily affects rod photoreceptors, leading to progressive vision loss. A study investigated the neuroprotective effects of LBP in the rd10 mouse model of RP, revealing that these polysaccharides significantly preserved both the morphology and function of photoreceptors while enhancing visual behaviour. The protective effects were attributed to their antioxidant, anti-inflammatory, and anti-apoptotic properties, with modulation of inflammation and apoptosis occurring through the inhibition of NF-κB and HIF-1α expressions, suggesting potential therapeutic avenues for RP patients [166].

4.12.3. Age-Related Macular Degeneration

Age-related macular degeneration (AMD) leads to central vision impairment due to progressive damage to the macula. Zeaxanthin and lutein—oxygenated carotenoids concentrated in the macula—are well known for their protective effects against AMD owing to their antioxidant capacity and ability to absorb blue light. L. barbarum berries are a rich source of zeaxanthin, and ethanol-extracted fractions have shown potential in mitigating AMD-related damage.
In a mouse AMD model, the 95% ethanol extract LBW-95E significantly improved retinal morphology, enhanced antioxidant defences via upregulation of SOD and GSH, and promoted nuclear translocation of Nrf2, while simultaneously reducing ROS and inflammation. These findings support the potential of L. barbarum in protecting against oxidative stress-related retinal degeneration associated with AMD [167].

4.12.4. Ocular Hypertension

Ocular hypertension (OHT), including both acute and chronic forms, contributes to RGC damage and is a major risk factor for glaucoma. In an acute ocular hypertension (AOH) mouse model, LBP treatment significantly mitigated RGC loss and preserved blood–retinal barrier integrity [168]. This was associated with the downregulation of key inflammatory mediators, indicating its potential as an anti-inflammatory neuroprotectant in ischaemic retinal injury [168].
In another study, LBP administration post-AOH significantly preserved inner retinal layer thickness and improved retinal function over time, suggesting its potential role in arresting secondary degeneration of RGCs following acute injury [169]. Moreover, recent investigations demonstrated L. barbarum’s potential to modulate the endothelin-1 (ET-1) signalling axis in a chronic ocular hypertension (COH) model. In a study carried out by Mi and colleagues [170], LBP administration significantly reduced ET-1 expression while altering the balance of its receptors, enhancing endothelin type A (ETA) receptor and diminishing the ETB receptor expression, particularly in RGCs, providing mechanistic insight into its pressure-lowering neuroprotective effect [170].
Importantly, LBPs also modulated retinal glial activity following AOH. Elevated levels of astrocytes and microglia and associated decreases in glial biomarkers were attenuated by LBP treatment. This resulted in preserved blood–retinal barrier function and improved neuronal survival, underscoring LBPs’ potential to stabilise the retinal microenvironment under hypertensive stress [171].

4.12.5. Transient Retinal Ischaemia

LBPs have shown substantial neuroprotective benefits in models of retinal ischemia. In a model involving two hours of retinal ischemia followed by reperfusion, LBP treatment led to significantly improved electroretinogram responses and increased numbers of viable retinal cells, along with enhanced expression of neuroprotective markers and reduced glial activation over a seven-day reperfusion period. These findings highlight the potential of LBPs as a therapeutic agent in managing ischemic retinopathies, sustaining retinal health and function long after the initial injury [172].
Overall, the findings across various studies highlight the potential of L. barbarum as a therapeutic agent for a range of ocular conditions, such as glaucoma, RP, AMD, ocular and hypertension. By reducing oxidative stress, inflammation, and retinal degeneration, LBPs show potential as therapeutic agents for retinal protection.

5. Conclusions

L. barbarum (goji berry) is endowed with a diverse array of bioactive compounds, including LBPs, carotenoids (notably zeaxanthin), polyphenols, and alkaloids, which exhibit potent antioxidant, anti-inflammatory, immunomodulatory, and anticancer activities. Preclinical studies have elucidated key mechanisms, including modulation of signalling pathways (e.g., NF-κB and MAPK), free-radical scavenging, and enhancement of host defences. In metabolic disorders, LBPs improve glycaemic control, regulate lipid metabolism, and beneficially modulate gut microbiota. They also confer protection in cardiovascular, neurodegenerative, and oncology models. Recent studies further expand LBP’s potential to address obesity-related comorbidities. LBP has been shown to attenuate weight gain by improving lipid parameters and enhancing gut microbiota diversity. Notably, its efficacy in sarcopenic obesity is emerging, where it mitigates skeletal muscle atrophy via the AMPK/PINK1/Parkin-mediated mitophagy pathway.
Despite this robust preclinical foundation, evidence from human studies remains scarce. Available clinical trials, summarised in our earlier review [18], suggest that L. barbarum formulations are generally safe and may confer benefits in metabolic and ocular health, but these investigations are limited by small sample sizes and heterogenous designs. Consequently, the translation of laboratory findings into clinical practice must proceed with caution. Future work should therefore (1) standardise extraction and formulation methods to ensure batch-to-batch consistency; (2) delineate dose–response relationships and pharmacokinetics; and (3) undertake adequately powered, randomised, placebo-controlled trials to confirm efficacy and safety in target populations. By distinguishing clearly between preclinical insights and the nascent clinical evidence, this review delineates both the promise of L. barbarum and the critical steps required for its development as an evidence-based nutraceutical or therapeutic agent.

Author Contributions

T.Z.: Conceptualization, Investigation, Writing—original draft, Writing—review and editing, Supervision. E.-A.A.: Investigation, Supervision, Writing—original draft, Writing—review and editing. G.L.: Investigation, Writing—original draft, Writing—review and editing. A.B.: Investigation, Writing—original draft, Writing—review and editing. R.W.: Investigation, Writing—original draft, Writing—review and editing. R.K.: Investigation, Writing—original draft, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This review builds upon the final-year research project completed as part of the BSc (Hons) Pharmaceutical Healthcare programme, with further investigations and contributions from the authors. The authors would like to express their sincere gratitude to the School of Food Science and Environmental Health at Technological University Dublin, Grangegorman, for providing the resources and support necessary for this research.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABPAchyranthes bidentata polysaccharide
ABTS2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid
ALTAlanine aminotransferase
AMDAge-related macular degeneration
AOHAcute ocular hypertension
AREAntioxidant response element
ARPEArising retinal pigment epithelia
ASTAspartate aminotransferase
ATF6Activating transcription factor 6
ATGLAdipose triglyceride lipase
ATKProtein kinase B (also known as Akt)
B-ALPBone-alkaline phosphatase
BaxBcl-2-associated X protein
BclxLB-cell lymphoma-extra large
CATCatalase
CDKCyclin-dependent kinase
Chrdl1Chordin-like 1
CIHChronic intermittent hypoxia
CONTComplete optic nerve transection
CPT1Carnitine palmitoyltransferase 1
CVDCardiovascular disease
CypDCyclophilin D
CRPC-reactive protein
CUPRACCupric ion reducing antioxidant capacity
DPPH2,2-Diphenyl-1-picrylhydrazyl
eNOSEndothelial nitric oxide synthase
ERK1/2Extracellular signal-regulated kinases 1 and 2
FASFatty acid synthase
FCFolin–Ciocalteu
FGFR1Fibroblast growth factor receptor 1
Fsp27Fat-specific protein 27
FRAPFerric reducing antioxidant power
GAEGallic acid equivalents
GCGas chromatography
GSHGlutathione
GPxGlutathione peroxidase
GRK2G protein-coupled receptor kinase 2
HO-1Haem oxygenase 1
hPDLSCsHuman periodontal ligament stem cells
HPLCHigh-performance liquid chromatography
IFNInterferon
IGFInsulin-like growth factor
ILInterleukin
I/RIschaemia/reperfusion
JNKc-Jun N-terminal kinase
L. BarbarumLycium Barbarum
LBLFL. Barbarum leaf flavonoids
LBPL. Barbarum polysaccharide
LCLiquid chromatography
MAPKMitogen-activated protein kinase
MDAMalondialdehyde
MDSCsMyeloid-derived suppressor cells
mfERGMultifocal electroretinography
MICMinimum inhibitory concentration
MMP-9Matrix metalloproteinase 9
MS/MSTandem mass spectrometry
NASHNon-alcoholic steatohepatitis
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NOD-like receptor protein 3
NMRNuclear magnetic resonance
NONitric oxide
Nrf2Nuclear factor erythroid 2-related factor 2
PI3KPhosphoinositide 3-kinase
PONTPartial optic nerve transection
PPAR-αPeroxisome proliferator-activated receptor alpha
PPARγPeroxisome proliferator-activated receptor gamma
RCCRenal cell carcinoma
RGCRetinal ganglion cell
ROSReactive oxygen species
RSMResponse surface methodology
RPRetinitis pigmentosa
SCFAsShort-chain fatty acids
SIRT1Sirtuin 1
SIRT3Sirtuin 3
sONEAn antisense mRNA
SODSuperoxide dismutase
STZStreptozotocin
TBCCTotal bioactive compound content
TFCTotal flavonoid content
TLRToll-like receptor
TNFTumour necrosis factor
TORTarget of rapamycin
TPCTotal phenolic content
TQTriple quadrupole
TXNIPThioredoxin-interacting protein
UPLCUltra performance liquid chromatography
UVBUltraviolet B radiation

References

  1. Matos, L.C.; Machado, J.P.; Monteiro, F.J.; Greten, H.J. Understanding traditional chinese medicine therapeutics: An overview of the basics and clinical applications. Healthcare 2021, 9, 257. [Google Scholar] [CrossRef] [PubMed]
  2. Smith, H.F.; Laitman, J.T. The yin, yang, and anatomy of traditional Chinese medicine in The Anatomical Record. Anat. Rec. 2023, 306, 2915–2919. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, M.; Fang, M.; Hu, Y.; Wang, X. Four types of traditional Chinese medicine inducing epileptic seizures. Seizure 2012, 21, 311–315. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, Y.; Wei, Y.; Wang, Y.; Gao, F.; Chen, Z. Lycium barbarum: A traditional Chinese herb and a promising anti-aging agent. Aging Dis. 2017, 8, 778–791. [Google Scholar] [CrossRef]
  5. German Federal Institute for Risk Assessment. Superfoods—Super Good? German Federal Institute for Risk Assessment: Berlin, Germany, 2020. [Google Scholar]
  6. Vidović, B.B.; Milinčić, D.D.; Marčetić, M.D.; Djuriš, J.D.; Ilić, T.D.; Kostić, A.Ž.; Pešić, M.B. Health benefits and applications of goji berries in functional food products development: A review. Antioxidants 2022, 11, 248. [Google Scholar] [CrossRef]
  7. Yao, R.; Heinrich, M.; Zou, Y.; Reich, E.; Zhang, X.; Chen, Y.; Weckerle, C.S. Quality variation of Goji (fruits of Lycium spp.) in China: A comparative morphological and metabolomic analysis. Front. Pharmacol. 2018, 9, 151. [Google Scholar] [CrossRef]
  8. Wang, H.; Li, J.; Tao, W.; Zhang, X.; Gao, X.; Yong, J.; Zhao, J.; Zhang, L.; Li, Y.; Duan, J.-A. Lycium ruthenicum studies: Molecular biology, phytochemistry and pharmacology. Food Chem. 2018, 240, 759–766. [Google Scholar] [CrossRef]
  9. Rubio, R.M.; García, M.R.; Barroso, N.N.; Iñiguez, F.M.S.; Gómez, M.J.R.; Magro, P.C. Classification of goji berry (Lycium barbarum L.) varieties according to physicochemical and bioactive signature. Eur. Food Res. Technol. 2024, 251, 355–365. [Google Scholar] [CrossRef]
  10. Ju, Y.; Liu, H.; Niu, S.; Kang, L.; Ma, L.; Li, A.; Zhao, Y.; Yuan, Y.; Zhao, D. Optimizing geographical traceability models of Chinese Lycium barbarum: Investigating effects of region, cultivar, and harvest year on nutrients, bioactives, elements and stable isotope composition. Food Chem. 2025, 467, 142286. [Google Scholar] [CrossRef]
  11. Bertoldi, D.; Cossignani, L.; Blasi, F.; Perini, M.; Barbero, A.; Pianezze, S.; Montesano, D. Characterisation and geographical traceability of Italian goji berries. Food Chem. 2019, 275, 585–593. [Google Scholar] [CrossRef]
  12. Yao, R.; Heinrich, M.; Weckerle, C.S. The genus Lycium as food and medicine: A botanical, ethnobotanical and historical review. J. Ethnopharmacol. 2018, 212, 50–66. [Google Scholar] [CrossRef] [PubMed]
  13. Ju, Y.; Wang, Y.; Ma, L.; Kang, L.; Liu, H.; Ma, X.; Zhao, D. Comparative analysis of polyphenols in Lycium barbarum fruits using UPLC-IM-QTOF-MS. Molecules 2023, 28, 4930. [Google Scholar] [CrossRef] [PubMed]
  14. Ilić, T.; Dodevska, M.; Marčetić, M.; Božić, D.; Kodranov, I.; Vidović, B. Chemical characterization, antioxidant and antimicrobial properties of goji berries cultivated in Serbia. Foods 2020, 9, 1614. [Google Scholar] [CrossRef] [PubMed]
  15. Pires, T.C.S.P.; Dias, M.I.; Barros, L.; Calhelha, R.C.; Alves, M.J.; Santos-Buelga, C.; Ferreira, I.C.F.R. Phenolic compounds profile, nutritional compounds and bioactive properties of Lycium barbarum L.: A comparative study with stems and fruits. Ind. Crops Prod. 2018, 122, 574–581. [Google Scholar] [CrossRef]
  16. Lu, Y.; Guo, S.; Zhang, F.; Yan, H.; Qian, D.-W.; Wang, H.-Q.; Jin, L.; Duan, J.-A. Comparison of functional components and antioxidant activity of Lycium barbarum L. fruits from different regions in China. Molecules 2019, 24, 2228. [Google Scholar] [CrossRef]
  17. Liu, J.; Shi, X.; Lin, H.; He, C.; Li, Q.; Shen, G.; Feng, J. Geographical origin identification and quality comparison of Ningxia goji berries (Lycium barbarum L.) by NMR-based techniques. J. Food Compos. Anal. 2023, 119, 105258. [Google Scholar] [CrossRef]
  18. Berisha, A.; Alexa, E.-A.; Kelleher, R.; Zhang, T. From botany to bedside: A review of the health benefits of Lycium barbarum as a functional food. Explor. Foods Foodomics 2025, 3, 101070. [Google Scholar] [CrossRef]
  19. Ma, R.-H.; Zhang, X.-X.; Thakur, K.; Zhang, J.-G.; Wei, Z.-J. Research progress of Lycium barbarum L. as functional food: Phytochemical composition and health benefits. Curr. Opin. Food Sci. 2022, 47, 100871. [Google Scholar] [CrossRef]
  20. Sicari, V.; Romeo, R.; Mincione, A.; Santacaterina, S.; Tundis, R.; Loizzo, M.R. Ciabatta bread incorporating goji (Lycium barbarum L.): A new potential functional product with impact on human health. Foods 2023, 12, 566. [Google Scholar] [CrossRef]
  21. Blasi, F.; Rocchetti, G.; Montesano, D.; Lucini, L.; Chiodelli, G.; Ghisoni, S.; Baccolo, G.; Simonetti, M.S.; Cossignani, L. Changes in extra-virgin olive oil added with Lycium barbarum L. carotenoids during frying: Chemical analyses and metabolomic approach. Food Res. Int. 2018, 105, 507–516. [Google Scholar] [CrossRef]
  22. Teixeira, F.; Silva, A.M.; Delerue-Matos, C.; Rodrigues, F. Lycium barbarum berries (Solanaceae) as source of bioactive compounds for healthy purposes: A review. Int. J. Mol. Sci. 2023, 24, 4777. [Google Scholar] [CrossRef] [PubMed]
  23. Rjeibi, I.; Feriani, A.; Ben Saad, A.; Ncib, S.; Sdayria, J.; Saidi, I.; Souid, S.; Hfaiedh, N.; Allagui, M.S. Phytochemical characterization and bioactivity of Lycium europaeum: A focus on antioxidant, antinociceptive, hepatoprotective and nephroprotective effects. Biomed. Pharmacother. 2017, 95, 1441–1450. [Google Scholar] [CrossRef] [PubMed]
  24. Long, L.N.; Kang, B.J.; Jiang, Q.; Chen, J.S. Effects of dietary Lycium barbarum polysaccharides on growth performance, digestive enzyme activities, antioxidant status, and immunity of broiler chickens. Poult. Sci. 2020, 99, 744–751. [Google Scholar] [CrossRef] [PubMed]
  25. Niu, Y.; Zhang, G.; Sun, X.; He, S.; Dou, G. Distinct role of Lycium barbarum L. polysaccharides in oxidative stress-related ocular diseases. Pharmaceuticals 2023, 16, 215. [Google Scholar] [CrossRef]
  26. Potterat, O. Goji (Lycium barbarum and L. chinense): Phytochemistry, pharmacology and safety in the perspective of traditional uses and recent popularity. Planta Med. 2010, 76, 7–19. [Google Scholar] [CrossRef]
  27. Lopatriello, A.; Previtera, R.; Pace, S.; Werner, M.; Rubino, L.; Werz, O.; Taglialatela-Scafati, O.; Forino, M. NMR-based identification of the major bioactive molecules from an Italian cultivar of Lycium barbarum. Phytochemistry 2017, 144, 52–57. [Google Scholar] [CrossRef]
  28. Chao, S.; Schreuder, M.; Young, G.; Nakaoka, K.; Moyes, L.; Oberg, C. Preclinical study: Antioxidant levels and immunomodulatory effects of wolfberry juice and other juice mixtures in mice. Jana 2004, 7, 2–8. [Google Scholar]
  29. Kabir, F.; Katayama, S.; Tanji, N.; Nakamura, S. Antimicrobial effects of chlorogenic acid and related compounds. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 359–365. [Google Scholar] [CrossRef]
  30. Ho, Y.-S.; Yu, M.-S.; Lai, C.S.-W.; So, K.-F.; Yuen, W.-H.; Chang, R.C.-C. Characterizing the neuroprotective effects of alkaline extract of Lycium barbarum on β-amyloid peptide neurotoxicity. Brain Res. 2007, 1158, 123–134. [Google Scholar] [CrossRef]
  31. Wawruszak, A.; Czerwonka, A.; Okła, K.; Rzeski, W. Anticancer effect of ethanol Lycium barbarum (Goji berry) extract on human breast cancer T47D cell line. Nat. Prod. Res. 2016, 30, 1993–1996. [Google Scholar] [CrossRef]
  32. Yu, Z.; Xia, M.; Lan, J.; Yang, L.; Wang, Z.; Wang, R.; Tao, H.; Shi, Y. A comprehensive review on the ethnobotany, phytochemistry, pharmacology and quality control of the genus Lycium in China. Food Funct. 2023, 14, 2998–3025. [Google Scholar] [CrossRef] [PubMed]
  33. Ma, R.H.; Zhang, X.X.; Ni, Z.J.; Thakur, K.; Wang, W.; Yan, Y.M.; Cao, Y.L.; Zhang, J.G.; Rengasamy, K.R.R.; Wei, Z.J. Lycium barbarum (Goji) as functional food: A review of its nutrition, phytochemical structure, biological features, and food industry prospects. Crit. Rev. Food Sci. Nutr. 2023, 63, 10621–10635. [Google Scholar] [CrossRef] [PubMed]
  34. Magiera, S.; Zaręba, M. Chromatographic Determination of Phenolic Acids and Flavonoids in Lycium barbarum L. and Evaluation of Antioxidant Activity. Food Anal. Methods 2015, 8, 2665–2674. [Google Scholar] [CrossRef]
  35. Qiang, X.; Xia, T.; Geng, B.; Zhao, M.; Li, X.; Zheng, Y.; Wang, M. Bioactive components of Lycium barbarum and deep-processing fermentation products. Molecules 2023, 28, 8044. [Google Scholar] [CrossRef]
  36. Chen, Y.; Yao, F.; Ming, K.; Wang, D.; Hu, Y.; Liu, J. Polysaccharides from Traditional Chinese Medicines: Extraction, Purification, Modification, and Biological Activity. Molecules 2016, 21, 1705. [Google Scholar] [CrossRef]
  37. Wang, Z.; Pan, H.; Xu, J.; Chang, Y.; Liu, C.; Zhang, Y.; Yang, H.; Duan, C.; Huang, J.; Fu, Y. A sustainable and integrated natural surfactant mediated microwave-assisted extraction technique enhances the extraction of phytochemicals from plants. Ind. Crops Prod. 2022, 184, 115043. [Google Scholar] [CrossRef]
  38. Perera, C.O.; Alzahrani, M.A.J. Ultrasound as a pre-treatment for extraction of bioactive compounds and food safety: A review. LWT 2021, 142, 111114. [Google Scholar] [CrossRef]
  39. Song, Y.-R.; Han, A.-R.; Park, S.-G.; Cho, C.-W.; Rhee, Y.-K.; Hong, H.-D. Effect of enzyme-assisted extraction on the physicochemical properties and bioactive potential of lotus leaf polysaccharides. Int. J. Biol. Macromol. 2020, 153, 169–179. [Google Scholar] [CrossRef]
  40. Gao, S.; Yan, S.; Zhou, Y.; Feng, Y.; Xie, X.; Guo, W.; Shen, Q.; Chen, C. Optimisation of enzyme-assisted extraction of Erythronium sibiricum bulb polysaccharide and its effects on immunomodulation. Glycoconj. J. 2022, 39, 357–368. [Google Scholar] [CrossRef]
  41. Masci, A.; Carradori, S.; Casadei, M.A.; Paolicelli, P.; Petralito, S.; Ragno, R.; Cesa, S. Lycium barbarum polysaccharides: Extraction, purification, structural characterisation and evidence about hypoglycaemic and hypolipidaemic effects. A review. Food Chem. 2018, 254, 377–389. [Google Scholar] [CrossRef]
  42. Yin, G.; Dang, Y. Optimization of extraction technology of the Lycium barbarum polysaccharides by Box–Behnken statistical design. Carbohydr. Polym. 2008, 74, 603–610. [Google Scholar] [CrossRef]
  43. Tian, X.; Liang, T.; Liu, Y.; Ding, G.; Zhang, F.; Ma, Z. Extraction, structural characterization, and biological functions of Lycium barbarum polysaccharides: A review. Biomolecules 2019, 9, 389. [Google Scholar] [CrossRef] [PubMed]
  44. Xia, F.; Zhou, S.; Sun, D. Surface optimization method to optimize the enzymatic extraction of Lycium barbarum polysaccharides. Chin. Wild Plant Resour. 2017, 36, 27–33. [Google Scholar]
  45. Kou, R.; Zuo, G.; Liu, J.; Di, D.; Guo, M. Structural properties and hypoglycaemic activity of polysaccharides extracted from the fruits of Lycium barbarum L. using various extraction media. Ind. Crops Prod. 2022, 188, 115725. [Google Scholar] [CrossRef]
  46. Zhang, Q.; Li, S. Effects of extraction methods on extraction ratio and antioxidant activity of polysaccharide from Lycium barbarum. Agricul. Sci. 2019, 47, 169–173. [Google Scholar]
  47. Quan, N.; Wang, Y.-D.; Li, G.-R.; Liu, Z.-Q.; Feng, J.; Qiao, C.-L.; Zhang, H.-F. Ultrasound–microwave combined extraction of novel polysaccharide fractions from Lycium barbarum leaves and their in vitro hypoglycemic and antioxidant activities. Molecules 2023, 28, 3880. [Google Scholar] [CrossRef]
  48. Zhu, L.; Peng, Z.; Zhang, X.; Yang, J.; Lai, X.; Yang, G. Determination of polyphenols in Lycium barbarum leaves by high-performance liquid chromatography–tandem mass spectrometry. Anal. Lett. 2017, 50, 761–776. [Google Scholar] [CrossRef]
  49. Islam, T.; Yu, X.; Badwal, T.S.; Xu, B. Comparative studies on phenolic profiles, antioxidant capacities and carotenoid contents of red goji berry (Lycium barbarum) and black goji berry (Lycium ruthenicum). Chem. Cent. J. 2017, 11, 59. [Google Scholar] [CrossRef]
  50. Ebeydulla, R.; Wang, X.; Zhao, L.; Xu, D.; Sun, H.; Ji, B.; Zhou, F. Nutritional and functional components of Chinese wolfberry dried by different drying methods. Food Sci. 2017, 38, 138–142. [Google Scholar]
  51. Donno, D.; Beccaro, G.L.; Mellano, M.G.; Cerutti, A.K.; Bounous, G. Goji berry fruit (Lycium spp.): Antioxidant compound fingerprint and bioactivity evaluation. J. Funct. Foods 2015, 18, 1070–1085. [Google Scholar] [CrossRef]
  52. Zhao, W.-H.; Shi, Y.-P. Comprehensive analysis of phenolic compounds in four varieties of goji berries at different ripening stages by UPLC–MS/MS. J. Food Compos. Anal. 2022, 106, 104279. [Google Scholar] [CrossRef]
  53. Liu, G.-T.; Li, Y.-L.; Wang, J.; Dong, C.-Z.; Deng, M.; Tai, M.; Deng, L.; Che, B.; Lin, L.; Du, Z.-Y.; et al. Improvement of skin barrier dysfunction by phenolic-containing extracts of Lycium barbarum via Nrf2/HO-1 regulation. Photochem. Photobiol. 2022, 98, 262–272. [Google Scholar] [CrossRef] [PubMed]
  54. Inbaraj, B.S.; Lu, H.; Kao, T.H.; Chen, B.H. Simultaneous determination of phenolic acids and flavonoids in Lycium barbarum Linnaeus by HPLC–DAD–ESI-MS. J. Pharm. Biomed. Anal. 2010, 51, 549–556. [Google Scholar] [CrossRef] [PubMed]
  55. Forino, M.; Tartaglione, L.; Dell’Aversano, C.; Ciminiello, P. NMR-based identification of the phenolic profile of fruits of Lycium barbarum (goji berries). Isolation and structural determination of a novel N-feruloyl tyramine dimer as the most abundant antioxidant polyphenol of goji berries. Food Chem. 2016, 194, 1254–1259. [Google Scholar] [CrossRef]
  56. Nardi, G.M.; Farias Januario, A.G.; Freire, C.G.; Megiolaro, F.; Schneider, K.; Perazzoli, M.R.; Do Nascimento, S.R.; Gon, A.C.; Mariano, L.N.; Wagner, G.; et al. Anti-inflammatory activity of berry fruits in mice model of inflammation is based on oxidative stress modulation. Pharmacognosy. Res. 2016, 8, S42–S49. [Google Scholar] [CrossRef]
  57. Mocan, A.; Vlase, L.; Vodnar, D.C.; Bischin, C.; Hanganu, D.; Gheldiu, A.M.; Oprean, R.; Silaghi-Dumitrescu, R.; Crișan, G. Polyphenolic content, antioxidant and antimicrobial activities of Lycium barbarum L. and Lycium chinense Mill. leaves. Molecules 2014, 19, 10056–10073. [Google Scholar] [CrossRef]
  58. Arumugam, M.K.; Paal, M.C.; Donohue, T.M.; Ganesan, M.; Osna, N.A.; Kharbanda, K.K. Beneficial effects of betaine: A comprehensive review. Biology 2021, 10, 456. [Google Scholar] [CrossRef]
  59. Qian, D.; Zhao, Y.; Yang, G.; Huang, L. Systematic review of chemical constituents in the genus Lycium (Solanaceae). Molecules 2017, 22, 911. [Google Scholar] [CrossRef]
  60. Tian, L.; Zhao, X.; Hu, Z.; Liu, J.; Ma, J.; Fan, Y.; Liu, D. iTRAQ-based proteomics identifies proteins associated with betaine accumulation in Lycium barbarum L. J. Proteom. 2024, 290, 105033. [Google Scholar] [CrossRef]
  61. Liu, W.; Xia, M.; Yang, L.; Wang, Z.; Wang, R.; Shi, Y. Development and optimization of a method for determining betaine and trigonelline in the fruits of Lycium species by using solid-phase extraction combined with high-performance liquid chromatography-diode array detector. J. Sep. Sci. 2020, 43, 2073–2078. [Google Scholar] [CrossRef]
  62. Yan, X.; Meng, X.; Pan, S.; Li, Z.; Song, Y. Determination of betaine in dried wolfberry by ion chromatography with pulsed amperometric detection. Food Sci. Technol. 2017, 42, 278–281. [Google Scholar]
  63. Chao, J.; Li, B.Y. Effects of Lycium barbarum polysaccharides extract and betaine on cell activation and liver fibrosis in rat hepatic stellate HSC-T6 cells. FASEB J. 2018, 31, 793–795. [Google Scholar] [CrossRef]
  64. Chen, D.; Guo, S.; Zhou, J.; Zhu, Y.; Zhang, F.; Zeng, F.; Duan, R.; Xu, M.; Duan, J.-A. Chemical constituents from Lycium barbarum (Solanaceae) and their chemophenetic significance. Biochem. Syst. Ecol. 2021, 97, 104292. [Google Scholar] [CrossRef]
  65. Chen, H.; Kong, J.-B.; Zhang, L.; Wang, H.-H.; Cao, Y.-G.; Zeng, M.-N.; Li, M.; Sun, Y.-J.; Du, K.; Xue, G.-M.; et al. Lycibarbarines A–C, three tetrahydroquinoline alkaloids possessing a spiro-heterocycle moiety from the fruits of Lycium barbarum. Org. Lett. 2021, 23, 858–862. [Google Scholar] [CrossRef]
  66. Kokotkiewicz, A.; Migas, P.; Stefanowicz, J.; Luczkiewicz, M.; Krauze-Baranowska, M. Densitometric TLC analysis for the control of tropane and steroidal alkaloids in Lycium barbarum. Food Chem. 2017, 221, 535–540. [Google Scholar] [CrossRef]
  67. Maoka, T. Carotenoids as natural functional pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef]
  68. Böhm, V. Carotenoids. Antioxidants 2019, 8, 516. [Google Scholar] [CrossRef]
  69. Ceccarini, M.R.; Vannini, S.; Cataldi, S.; Moretti, M.; Villarini, M.; Fioretti, B.; Albi, E.; Beccari, T.; Codini, M. In vitro protective effects of Lycium barbarum berries cultivated in Umbria (Italy) on human hepatocellular carcinoma cells. Biomed. Res. Int. 2016, 2016, 7529521. [Google Scholar] [CrossRef]
  70. Long, J.T.; Fan, H.X.; Zhou, Z.Q.; Sun, W.Y.; Li, Q.W.; Wang, Y.; Ma, M.; Gao, H.; Zhi, H. The major zeaxanthin dipalmitate derivatives from wolfberry. J. Asian Nat. Prod. Res. 2020, 22, 746–753. [Google Scholar] [CrossRef]
  71. Ren, R.; Li, Y.; Chen, H.; Wang, Y.; Yang, L.; Su, C.; Zhao, X.; Chen, J.; Ma, X. Carotenoid contents of Lycium barbarum: A novel QAMS analysis, geographical origins discriminant evaluation, and storage stability assessment. Molecules 2021, 26, 5374. [Google Scholar] [CrossRef]
  72. Yu, J.; Yan, Y.; Zhang, L.; Mi, J.; Yu, L.; Zhang, F.; Lu, L.; Luo, Q.; Li, X.; Zhou, X.; et al. A comprehensive review of goji berry processing and utilization. Food Sci. Nutr. 2023, 11, 7445–7457. [Google Scholar] [CrossRef] [PubMed]
  73. Inbaraj, B.S.; Lu, H.; Hung, C.F.; Wu, W.B.; Lin, C.L.; Chen, B.H. Determination of carotenoids and their esters in fruits of Lycium barbarum Linnaeus by HPLC-DAD-APCI-MS. J. Pharm. Biomed. Anal. 2008, 47, 812–818. [Google Scholar] [CrossRef] [PubMed]
  74. Wojdyło, A.; Nowicka, P.; Bąbelewski, P. Phenolic and carotenoid profile of new goji cultivars and their anti-hyperglycemic, anti-aging and antioxidant properties. J. Funct. Foods 2018, 48, 632–642. [Google Scholar] [CrossRef]
  75. Hsu, H.J.; Huang, R.F.; Kao, T.H.; Inbaraj, B.S.; Chen, B.H. Preparation of carotenoid extracts and nanoemulsions from Lycium barbarum L. and their effects on growth of HT-29 colon cancer cells. Nanotechnology 2017, 28, 135103. [Google Scholar] [CrossRef]
  76. Langi, P.; Kiokias, S.; Varzakas, T.; Proestos, C. Carotenoids: From plants to food and feed industries. In Microbial Carotenoids: Methods and Protocols; Barreiro, C., Barredo, J.-L., Eds.; Springer: New York, NY, USA, 2018; pp. 57–71. [Google Scholar]
  77. Wu, H.; Liu, Y.; Hao, Y.; Hou, D.; Yang, R. Lycium barbarum polysaccharide protects cardiomyocytes from hypoxia/reoxygenation injury via activation of SIRT3/CypD signaling. Ann. Transl. Med. 2023, 11, 72. [Google Scholar] [CrossRef]
  78. Li, Y.; Yang, B.; Zhang, X.; Shen, X.; Ma, Y.; Jing, L. Lycium barbarum polysaccharide antagonizes cardiomyocyte apoptosis by inhibiting the upregulation of GRK2 induced by I/R injury, and salvage mitochondrial fission/fusion imbalance and AKT/eNOS signaling. Cell. Signal. 2022, 92, 110252. [Google Scholar] [CrossRef]
  79. Xue, S.; Hu, X.; Zhu, L.; Nie, L.; Li, G. Protective functions of Lycium barbarum polysaccharides in H(2)O(2)-injured vascular endothelial cells through anti-oxidation and anti-apoptosis effects. Biomed. Rep. 2019, 11, 207–214. [Google Scholar] [CrossRef]
  80. Zhang, X.; Yang, X.; Lin, Y.; Suo, M.; Gong, L.; Chen, J.; Hui, R. Anti-hypertensive effect of Lycium barbarum L. with down-regulated expression of renal endothelial lncRNA sONE in a rat model of salt-sensitive hypertension. Int. J. Clin. Exp. Pathol. 2015, 8, 6981–6987. [Google Scholar]
  81. Huang, R.; Wu, E.; Deng, X. Potential of Lycium barbarum polysaccharide for the control of glucose and lipid metabolism disorders: A review. Int. J. Food Prop. 2022, 25, 673–680. [Google Scholar] [CrossRef]
  82. Liu, H.; Cui, B.; Zhang, Z. Mechanism of glycometabolism regulation by bioactive compounds from the fruits of Lycium barbarum: A review. Food Res. Int. 2022, 159, 111408. [Google Scholar] [CrossRef]
  83. Zhou, R.; Liu, Y.; Hu, W.; Yang, J.; Lin, B.; Zhang, Z.; Chen, M.; Yi, J.; Zhu, C. Lycium barbarum polysaccharide ameliorates the accumulation of lipid droplets in adipose tissue via an ATF6/SIRT1-dependent mechanism. Acta Biochim. Biophys. Sin. 2024, 56, 844–856. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, H.; Li, Y.; Dai, Y.; Ma, L.; Di, D.; Liu, J. Screening, structural characterization and anti-adipogenesis effect of a water-soluble polysaccharide from Lycium barbarum L. by an activity-oriented approach. Food Biosci. 2023, 53, 102502. [Google Scholar] [CrossRef]
  85. Yang, F.L.; Wei, Y.X.; Liao, B.Y.; Wei, G.J.; Qin, H.M.; Pang, X.X.; Wang, J.L. Effects of Lycium barbarum polysaccharide on endoplasmic reticulum stress and oxidative stress in obese mice. Front. Pharmacol. 2020, 11, 742. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, C.; Ma, L.; Ma, J.; Liu, S.; Fu, J.; Fan, Y.; Liu, Y. Lycium barbarum leaf flavonoids ameliorate high fructose induced insulin resistance in mice by regulating blood glucose and gut microbiota composition. Food Biosci. 2024, 62, 105087. [Google Scholar] [CrossRef]
  87. Ren, Y.; Wang, K.; Wu, Y.; Li, J.; Ma, J.; Wang, L.; Zhang, C.; Li, J.; Wei, Y.; Yang, Y. Lycium barbarum polysaccharide mitigates high-fat-diet-induced skeletal muscle atrophy by promoting AMPK/PINK1/Parkin-mediated mitophagy. Int. J. Biol. Macromol. 2025, 301, 140488. [Google Scholar] [CrossRef]
  88. Kou, L.; Du, M.; Zhang, C.; Dai, Z.; Li, X.; Zhang, B. The Hypoglycemic, Hypolipidemic, and Anti-Diabetic Nephritic Activities of Zeaxanthin in Diet-Streptozotocin-Induced Diabetic Sprague Dawley Rats. Appl. Biochem. Biotechnol. 2017, 182, 944–955. [Google Scholar] [CrossRef]
  89. Li, D.; Zhang, X.; Fan, Y.; Zhang, Y.; Tao, X.; Yang, J. Lycium barbarum polysaccharides improved glucose metabolism in prediabetic mice by regulating duodenal contraction. Nutrients 2023, 15, 4437. [Google Scholar] [CrossRef]
  90. Wu, H.; Guo, H.; Zhao, R. Effect of Lycium barbarum polysaccharide on the improvement of antioxidant ability and DNA damage in NIDDM rats. Yakugaku Zasshi 2006, 126, 365–371. [Google Scholar] [CrossRef]
  91. Zhao, X.-q.; Guo, S.; Lu, Y.-y.; Hua, Y.; Zhang, F.; Yan, H.; Shang, E.-x.; Wang, H.-q.; Zhang, W.-h.; Duan, J.-a. Lycium barbarum L. leaves ameliorate type 2 diabetes in rats by modulating metabolic profiles and gut microbiota composition. Biomed. Pharmacother. 2020, 121, 109559. [Google Scholar] [CrossRef]
  92. Xia, H.; Zhou, B.; Sui, J.; Ma, W.; Wang, S.; Yang, L.; Sun, G. Lycium barbarum polysaccharide regulates lipid metabolism and alters gut microbiota in high-fat diet-induced obese mice. Int. J. Environ. Res. Public Health 2022, 19, 12093. [Google Scholar] [CrossRef]
  93. Huang, Z.; Ye, Y.; Long, Z.; Qin, H.; Liu, L.; Xu, A.; Li, Z. Lycium barbarum polysaccharides improve lipid metabolism disorders of spotted sea bass Lateolabrax maculatus induced by high lipid diet. Int. J. Biol. Macromol. 2023, 242, 125122. [Google Scholar] [CrossRef] [PubMed]
  94. Wu, D.; Li, J.; Fan, Z.; Sun, Z.; Zheng, X.; Zhang, H.; Xu, H.; Wang, L. Dietary Lycium barbarum polysaccharide modulates growth performance, antioxidant capacity, and lipid metabolism in common carp (Cyprinus carpio) fed with high-fat diet. Antioxidants 2024, 13, 540. [Google Scholar] [CrossRef] [PubMed]
  95. Fan, Y.N.; Chi, X.; Yan, L.; Pu, Z.Y.; Yang, J.J.; Zhang, Y.N. Lycium barbarum polysaccharides regulate the gut microbiota to modulate metabolites in high-fat diet-induced obese rats. J. Asian Nat. Prod. Res. 2024, 26, 1115–1129. [Google Scholar] [CrossRef] [PubMed]
  96. Jing, L.; Hu, B.; Song, Q.H. Lycium barbarum polysaccharide (LBP) inhibits palmitic acid (PA)-induced MC3T3-E1 cell apoptosis by regulating miR-200b-3p/Chrdl1/PPARγ. Food Nutr. Res. 2020, 64, 10-29219. [Google Scholar] [CrossRef]
  97. Li, Z.-X.; Zhuo, J.-L.; Yang, N.; Gao, M.-B.; Qu, Z.-H.; Han, T. Effect of Lycium barbarum polysaccharide on osteoblast proliferation and differentiation in postmenopausal osteoporosis. Int. J. Mol. Sci. 2024, 271, 132415. [Google Scholar] [CrossRef]
  98. Piñar-Gutierrez, A.; García-Fontana, C.; García-Fontana, B.; Muñoz-Torres, M. Obesity and bone health: A complex relationship. Int. J. Mol. Sci. 2022, 23, 8303. [Google Scholar] [CrossRef]
  99. Tan, B.L.; Norhaizan, M.E.; Liew, W.P.; Sulaiman Rahman, H. Antioxidant and oxidative stress: A mutual interplay in age-related diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef]
  100. Yun, D.; Yan, Y.; Liu, J. Isolation, structure and biological activity of polysaccharides from the fruits of Lycium ruthenicum Murr: A review. Carbohydr. Polym. 2022, 291, 119618. [Google Scholar] [CrossRef]
  101. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  102. Rumpf, J.; Burger, R.; Schulze, M. Statistical evaluation of DPPH, ABTS, FRAP, and Folin-Ciocalteu assays to assess the antioxidant capacity of lignins. Int. J. Biol. Macromol. 2023, 233, 123470. [Google Scholar] [CrossRef]
  103. Wang, C.C.; Chang, S.C.; Inbaraj, B.S.; Chen, B.H. Isolation of carotenoids, flavonoids and polysaccharides from Lycium barbarum L. and evaluation of antioxidant activity. Food Chem. 2010, 120, 184–192. [Google Scholar] [CrossRef]
  104. de Nijs, M.; Crews, C.; Dorgelo, F.; MacDonald, S.; Mulder, P.P.J. Emerging issues on tropane alkaloid contamination of food in Europe. Toxins 2023, 15, 98. [Google Scholar] [CrossRef] [PubMed]
  105. Skenderidis, P.; Kerasioti, E.; Karkanta, E.; Stagos, D.; Kouretas, D.; Petrotos, K.; Hadjichristodoulou, C.; Tsakalof, A. Assessment of the antioxidant and antimutagenic activity of extracts from goji berry of Greek cultivation. Toxicol. Rep. 2018, 5, 251–257. [Google Scholar] [CrossRef] [PubMed]
  106. Liang, R.; Zhao, Q.; Zhu, Q.; He, X.; Gao, M.; Wang, Y. Lycium barbarum polysaccharide protects ARPE-19 cells against H2O2-induced oxidative stress via the Nrf2/HO-1 pathway. Mol. Med. Rep. 2021, 24, 769. [Google Scholar] [CrossRef]
  107. Pehlİvan KarakaŞ, F.; CoŞkun, H.; SoytÜrk, H.; Bozat, B.G. Anxiolytic, antioxidant, and neuroprotective effects of goji berry polysaccharides in ovariectomized rats: Experimental evidence from behavioral, biochemical, and immunohistochemical analyses. Turk. J. Biol. 2020, 44, 238–251. [Google Scholar] [CrossRef]
  108. Li, X.M. Protective effect of Lycium barbarum polysaccharides on streptozotocin-induced oxidative stress in rats. Int. J. Biol. Macromol. 2007, 40, 461–465. [Google Scholar] [CrossRef]
  109. Ma, Z.; Du, B.; Li, J.; Yang, Y.; Zhu, F. An insight into anti-inflammatory activities and inflammation related diseases of anthocyanins: A review of both in vivo and in vitro investigations. Int. J. Mol. Sci. 2021, 22, 11076. [Google Scholar] [CrossRef]
  110. Mitchell, S.; Vargas, J.; Hoffmann, A. Signaling via the NFκB system. Wiley Interdiscip. Rev. Syst. Biol. Med. 2016, 8, 227–241. [Google Scholar] [CrossRef]
  111. Lee, S.; Jeong, S.; Park, Y.; Seo, H.; You, C.; Hwang, U.; Park, H.; Suh, H.-j. Supplementation of non-fermented and fermented goji berry (Lycium barbarum) improves hepatic function and corresponding lipid metabolism via their anti-inflammatory and antioxidant properties in high fat-fed rats. Appl. Biol. Chem. 2021, 64, 70. [Google Scholar] [CrossRef]
  112. Xiao, J.; Xing, F.; Huo, J.; Fung, M.L.; Liong, E.C.; Ching, Y.P.; Xu, A.; Chang, R.C.C.; So, K.F.; Tipoe, G.L. Lycium barbarum polysaccharides therapeutically improve hepatic functions in non-alcoholic steatohepatitis rats and cellular steatosis model. Sci. Rep. 2014, 4, 5587. [Google Scholar] [CrossRef]
  113. Du, M.; Hu, X.; Kou, L.; Zhang, B.; Zhang, C. Lycium barbarum polysaccharide mediated the antidiabetic and antinephritic effects in diet-streptozotocin-induced diabetic Sprague Dawley rats via regulation of NF-κB. Biomed. Res. Int. 2016, 2016, 3140290. [Google Scholar] [CrossRef] [PubMed]
  114. Xie, W.; Huang, Y.Y.; Chen, H.G.; Zhou, X. Study on the efficacy and mechanism of Lycium barbarum polysaccharide against lead-induced renal Iinjury in mice. Nutrients 2021, 13, 2945. [Google Scholar] [CrossRef] [PubMed]
  115. Gan, F.; Liu, Q.; Liu, Y.; Huang, D.; Pan, C.; Song, S.; Huang, K. Lycium barbarum polysaccharides improve CCl(4)-induced liver fibrosis, inflammatory response and TLRs/NF-kB signaling pathway expression in wistar rats. Life Sci. 2018, 192, 205–212. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, Q.; Li, J.; Yan, J.; Liu, S.; Guo, Y.; Chen, D.; Luo, Q. Lycium barbarum polysaccharides ameliorates renal injury and inflammatory reaction in alloxan-induced diabetic nephropathy rabbits. Life Sci. 2016, 157, 82–90. [Google Scholar] [CrossRef]
  117. Gan, L.; Zhang, S.H. Effect of Lycium barbarum polysaccharides on anti-tumor activity and immune function. Acta Nutr. Sin. 2003, 25, 200–202. [Google Scholar]
  118. Deng, X.; Luo, S.; Luo, X.; Hu, M.; Ma, F.; Wang, Y.; Lai, X.; Zhou, L. Polysaccharides from Chinese herbal Lycium barbarum induced systemic and local immune responses in H22 tumor-bearing Mice. J. Immunol. Res. 2018, 2018, 3431782. [Google Scholar] [CrossRef]
  119. Feng, L.; Xiao, X.; Liu, J.; Wang, J.; Zhang, N.; Bing, T.; Liu, X.; Zhang, Z.; Shangguan, D. Immunomodulatory effects of Lycium barbarum polysaccharide extract and its uptake behaviors at the cellular level. Molecules 2020, 25, 1351. [Google Scholar] [CrossRef]
  120. Liu, Y.L.; Yin, R.Q.; Liang, S.S.; Duan, Y.L.; Yao, J.H.; Duan, Y.L.; Yang, X.J. Effect of dietary Lycium barbarum polysaccharide on growth performance and immune function of broilers. J. Appl. Poult. Res. 2017, 26, 200–208. [Google Scholar] [CrossRef]
  121. Qian, L. Modulation of cytokine level and sperm quality of mice by Lycium barbarum polysaccharides. Int. J. Biol. Macromol. 2019, 126, 475–477. [Google Scholar] [CrossRef]
  122. Ding, Y.; Yan, Y.; Chen, D.; Ran, L.; Mi, J.; Lu, L.; Jing, B.; Li, X.; Zeng, X.; Cao, Y. Modulating effects of polysaccharides from the fruits of Lycium barbarum on the immune response and gut microbiota in cyclophosphamide-treated mice. Food Funct. 2019, 10, 3671–3683. [Google Scholar] [CrossRef]
  123. Guo, M.; Jin, J.; Zhao, D.; Rong, Z.; Cao, L.-Q.; Li, A.-H.; Sun, X.-Y.; Jia, L.-Y.; Wang, Y.-D.; Huang, L.; et al. Research advances on anti-cancer natural products. Front. Oncol. 2022, 12, 866154. [Google Scholar] [CrossRef] [PubMed]
  124. Naeem, A.; Hu, P.; Yang, M.; Zhang, J.; Liu, Y.; Zhu, W.; Zheng, Q. Natural products as anticancer agents: Current status and future perspectives. Molecules 2022, 27, 8367. [Google Scholar] [CrossRef] [PubMed]
  125. Miranda, M.R.; Vestuto, V.; Amodio, G.; Manfra, M.; Pepe, G.; Campiglia, P. Antitumor mechanisms of Lycium barbarum fruit: An overview of in vitro and in vivo potential. Life 2024, 14, 420. [Google Scholar] [CrossRef] [PubMed]
  126. Kwaśnik, P.; Lemieszek, M.K.; Rzeski, W. Impact of phytochemicals and plant extracts on viability and proliferation of NK cell line NK-92—A closer look at immunomodulatory properties of goji berries extract in human colon cancer cells. Ann. Agric. Environ. Med. 2021, 28, 291–299. [Google Scholar] [CrossRef] [PubMed]
  127. Georgiev, K.D.; Slavov, I.J.; Iliev, I.A. Antioxidant activity and antiproliferative effects of Lycium barbarum’s (Goji berry) fractions on breast cancer cell lines. Folia Med. 2019, 61, 104–112. [Google Scholar] [CrossRef]
  128. Gong, G.; Liu, Q.; Deng, Y.; Dang, T.; Dai, W.; Liu, T.; Liu, Y.; Sun, J.; Wang, L.; Liu, Y.; et al. Arabinogalactan derived from Lycium barbarum fruit inhibits cancer cell growth via cell cycle arrest and apoptosis. Int. J. Biol. Macromol. 2020, 149, 639–650. [Google Scholar] [CrossRef]
  129. Zhang, M.; Chen, H.; Huang, J.; Li, Z.; Zhu, C.; Zhang, S. Effect of Lycium barbarum polysaccharide on human hepatoma QGY7703 cells: Inhibition of proliferation and induction of apoptosis. Life Sci. 2005, 76, 2115–2124. [Google Scholar] [CrossRef]
  130. Deng, X.; Li, X.; Luo, S.; Zheng, Y.; Luo, X.; Zhou, L. Antitumor activity of Lycium barbarum polysaccharides with different molecular weights: An in vitro and in vivo study. Food Nutr. Res. 2017, 61, 1399770. [Google Scholar] [CrossRef]
  131. Sanghavi, A.; Srivatsa, A.; Adiga, D.; Chopra, A.; Lobo, R.; Kabekkodu, S.P.; Gadag, S.; Nayak, U.; Sivaraman, K.; Shah, A. Goji berry (Lycium barbarum) inhibits the proliferation, adhesion, and migration of oral cancer cells by inhibiting the ERK, AKT, and CyclinD cell signaling pathways: An in-vitro study. F1000Research 2022, 11, 1563. [Google Scholar] [CrossRef]
  132. Liu, Y.; Du, Y.; Gao, L.; Ma, L. Effect of Lycium barbarum polysaccharide combined with cisplatin on the proliferation of human lung cancer cells. Pharmacogn. Mag. 2022, 18, 378–385. [Google Scholar]
  133. Chen, S.; Liang, L.; Wang, Y.; Diao, J.; Zhao, C.; Chen, G.; He, Y.; Luo, C.; Wu, X.; Zhang, Y. Synergistic immunotherapeutic effects of Lycium barbarum polysaccharide and interferon-α2b on the murine Renca renal cell carcinoma cell line in vitro and in vivo. Mol. Med. Rep. 2015, 12, 6727–6737. [Google Scholar] [CrossRef] [PubMed]
  134. Xin, Y.-F.; Wan, L.-L.; Peng, J.-L.; Guo, C. Alleviation of the acute doxorubicin-induced cardiotoxicity by Lycium barbarum polysaccharides through the suppression of oxidative stress. Food Chem. Toxicol. 2011, 49, 259–264. [Google Scholar] [CrossRef] [PubMed]
  135. Deng, X.; Luo, S.; Luo, X.; Hu, M.; Ma, F.; Wang, Y.; Zhou, L.; Huang, R. Fraction from Lycium barbarum polysaccharides reduces immunotoxicity and enhances antitumor activity of doxorubicin in mice. Integr. Cancer Ther. 2018, 17, 860–866. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Y.; Liang, X.; Li, Y.; Fan, Y.; Li, Y.; Cao, Y.; An, W.; Shi, Z.; Zhao, J.; Guo, S. Changes in metabolome and nutritional quality of Lycium barbarum fruits from three typical growing areas of China as revealed by widely targeted metabolomics. Metabolites 2020, 10, 46. [Google Scholar] [CrossRef]
  137. Xiao, J.; Zhu, Y.; Liu, Y.; Tipoe, G.L.; Xing, F.; So, K.-F. Lycium barbarum polysaccharide attenuates alcoholic cellular injury through TXNIP-NLRP3 inflammasome pathway. Int. J. Biol. Macromol. 2014, 69, 73–78. [Google Scholar] [CrossRef]
  138. Gündüz, E.; Dursun, R.; Zengin, Y.; İçer, M.; Durgun, H.M.; Kanıcı, A.; Kaplan, İ.; Alabalık, U.; Gürbüz, H.; Güloğlu, C. Lycium barbarum extract provides effective protection against paracetamol-induced acute hepatotoxicity in rats. Int. J. Clin. Exp. Med. 2015, 8, 7898–7905. [Google Scholar]
  139. Cheng, D.; Kong, H. The effect of Lycium barbarum polysaccharide on alcohol-induced oxidative stress in rats. Molecules 2011, 16, 2542–2550. [Google Scholar] [CrossRef]
  140. Sanghavi, A.D.; Chopra, A.; Shah, A.; Lobo, R.; Shenoy, P.A. Antimicrobial, anti-adhesion, anti-biofilm properties of goji berry (Lycium barbarum) against periodontal bacteria: Potential benefits for periodontal diseases. J. Complement. Integr. Med. 2023, 20, 129–136. [Google Scholar] [CrossRef]
  141. Zhu, W.; Zhou, S.; Liu, J.; McLean, R.J.C.; Chu, W. Prebiotic, immuno-stimulating and gut microbiota-modulating effects of Lycium barbarum polysaccharide. Biomed. Pharmacother. 2020, 121, 109591. [Google Scholar] [CrossRef]
  142. Guo, L.; Guan, Q.; Duan, W.; Ren, Y.; Zhang, X.J.; Xu, H.Y.; Shi, J.S.; Wang, F.Z.; Lu, R.; Zhang, H.L.; et al. Dietary goji shapes the gut microbiota to prevent liver injury induced by acute alcohol intake. Front. Nutr. 2022, 9, 929776. [Google Scholar] [CrossRef]
  143. Skenderidis, P.; Mitsagga, C.; Lampakis, D.; Petrotos, K.; Giavasis, I. The effect of encapsulated powder of goji berry (Lycium barbarum) on growth and survival of probiotic bacteria. Microorganisms 2020, 8, 57. [Google Scholar] [CrossRef] [PubMed]
  144. Orsini, M.; Nascimento, O.J.; Matta, A.P.; Reis, C.H.; de Souza, O.G.; Bastos, V.H.; Moreira, R.; Ribeiro, P.; Fiorelli, S.; Novellino, P.; et al. Revisiting the term neuroprotection in chronic and degenerative diseases. Neurol. Int. 2016, 8, 6311. [Google Scholar] [CrossRef] [PubMed]
  145. Karvandi, M.S.; Sheikhzadeh Hesari, F.; Aref, A.R.; Mahdavi, M. The neuroprotective effects of targeting key factors of neuronal cell death in neurodegenerative diseases: The role of ER stress, oxidative stress, and neuroinflammation. Front. Cell. Neurosci. 2023, 17, 1105247. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, Y.; Zhang, X.; Chen, L.; Ren, B.X.; Tang, F.R. Lycium barbarum ameliorates neural damage induced by experimental ischemic stroke and radiation exposure. Front. Biosci. 2023, 28, 38. [Google Scholar] [CrossRef]
  147. Lam, C.S.; Tipoe, G.L.; So, K.F.; Fung, M.L. Neuroprotective mechanism of Lycium barbarum polysaccharides against hippocampal-dependent spatial memory deficits in a rat model of obstructive sleep apnea. PLoS ONE 2015, 10, e0117990. [Google Scholar] [CrossRef]
  148. Cao, S.; Du, J.; Hei, Q. Lycium barbarum polysaccharide protects against neurotoxicity via the Nrf2-HO-1 pathway. Exp. Ther. Med. 2017, 14, 4919–4927. [Google Scholar] [CrossRef]
  149. Yang, Y.; Yu, L.; Zhu, T.; Xu, S.; He, J.; Mao, N.; Liu, Z.; Wang, D. Neuroprotective effects of Lycium barbarum polysaccharide on light-induced oxidative stress and mitochondrial damage via the Nrf2/HO-1 pathway in mouse hippocampal neurons. Int. J. Biol. Macromol. 2023, 251, 126315. [Google Scholar] [CrossRef]
  150. Hu, X.; Qu, Y.; Chu, Q.; Li, W.; He, J. Investigation of the neuroprotective effects of Lycium barbarum water extract in apoptotic cells and Alzheimer’s disease mice. Mol. Med. Rep. 2018, 17, 3599–3606. [Google Scholar] [CrossRef]
  151. Yao, X.-l.; Wu, W.-l.; Zheng, M.-y.; Li, W.; Ye, C.-h.; Lu, X.-l. Protective effects of Lycium barbarum extract against MPP(+) -induced neurotoxicity in Caenorhabditis elegans and PC12 cells. Zhong Yao Cai 2011, 34, 1241–1246. [Google Scholar]
  152. Lp, S.; Rr, U.; Mardhekar, V. Neuroprotective effects of Lycium barbarum in ischemic stroke: Current perspectives. Int. J. Pharmacogn. Chin. Med. 2019, 3, 126315. [Google Scholar] [CrossRef]
  153. Deng, H.B.; Cui, D.P.; Jiang, J.M.; Feng, Y.C.; Cai, N.S.; Li, D.D. Inhibiting effects of Achyranthes bidentata polysaccharide and Lycium barbarum polysaccharide on nonenzyme glycation in D-galactose induced mouse aging model. Biomed. Environ. Sci. 2003, 16, 267–275. [Google Scholar]
  154. Tang, R.; Chen, X.; Dang, T.; Deng, Y.; Zou, Z.; Liu, Q.; Gong, G.; Song, S.; Ma, F.; Huang, L.; et al. Lycium barbarum polysaccharides extend the mean lifespan of Drosophila melanogaster. Food Funct. 2019, 10, 4231–4241. [Google Scholar] [CrossRef]
  155. Li, H.; Li, Z.; Peng, L.; Jiang, N.; Liu, Q.; Zhang, E.; Liang, B.; Li, R.; Zhu, H. Lycium barbarum polysaccharide protects human keratinocytes against UVB-induced photo-damage. Free Radic. Res. 2017, 51, 200–210. [Google Scholar] [CrossRef]
  156. Meng, J.; Lv, Z.; Chen, X.; Sun, C.; Jin, C.; Ding, K.; Chen, C. LBP1C-2 from Lycium barbarum maintains skeletal muscle satellite cell pool by interaction with FGFR1. iScience 2023, 26, 106573. [Google Scholar] [CrossRef]
  157. Lai, S.; Liu, C.; Liu, C.; Fan, L.; Li, X.; Yang, Y.; Zhu, Y.; Deng, L.; Xiao, L.; Mu, Y. Lycium barbarum polysaccharide-glycoprotein promotes osteogenesis in hPDLSCs via ERK activation. Oral Dis. 2023, 29, 3503–3513. [Google Scholar] [CrossRef]
  158. Xia, G.; Xin, N.; Liu, W.; Yao, H.; Hou, Y.; Qi, J. Inhibitory effect of Lycium barbarum polysaccharides on cell apoptosis and senescence is potentially mediated by the p53 signaling pathway. Mol. Med. Rep. 2014, 9, 1237–1241. [Google Scholar] [CrossRef]
  159. Wang, Y.; Zhao, H.; Sheng, X.; Gambino, P.E.; Costello, B.; Bojanowski, K. Protective effect of Fructus Lycii polysaccharides against time and hyperthermia-induced damage in cultured seminiferous epithelium. J. Ethnopharmacol. 2002, 82, 169–175. [Google Scholar] [CrossRef]
  160. Yu, Y.; Wu, X.; Pu, J.; Luo, P.; Ma, W.; Wang, J.; Wei, J.; Wang, Y.; Fei, Z. Lycium barbarum polysaccharide protects against oxygen glucose deprivation/reoxygenation-induced apoptosis and autophagic cell death via the PI3K/Akt/mTOR signaling pathway in primary cultured hippocampal neurons. Biochem. Biophys. Res. Commun. 2018, 495, 1187–1194. [Google Scholar] [CrossRef]
  161. Zhou, L.; Pinho, R.; Gu, Y.; Radak, Z. The role of SIRT3 in exercise and aging. Cells 2022, 11, 2596. [Google Scholar] [CrossRef]
  162. He, M.; Pan, H.; Chang, R.C.; So, K.F.; Brecha, N.C.; Pu, M. Activation of the Nrf2/HO-1 antioxidant pathway contributes to the protective effects of Lycium barbarum polysaccharides in the rodent retina after ischemia-reperfusion-induced damage. PLoS ONE 2014, 9, e84800. [Google Scholar] [CrossRef]
  163. Li, H.; Liang, Y.; Chiu, K.; Yuan, Q.; Lin, B.; Chang, R.C.; So, K.F. Lycium barbarum (wolfberry) reduces secondary degeneration and oxidative stress, and inhibits JNK pathway in retina after partial optic nerve transection. PLoS ONE 2013, 8, e68881. [Google Scholar] [CrossRef]
  164. Chu, P.H.; Li, H.Y.; Chin, M.P.; So, K.F.; Chan, H.H. Effect of Lycium barbarum (wolfberry) polysaccharides on preserving retinal function after partial optic nerve transection. PLoS ONE 2013, 8, e81339. [Google Scholar] [CrossRef]
  165. Li, H.Y.; Huang, M.; Luo, Q.Y.; Hong, X.; Ramakrishna, S.; So, K.F. Lycium barbarum (wolfberry) increases retinal ganglion cell survival and affects both microglia/macrophage polarization and autophagy after rat partial optic nerve transection. Cell Transpl. 2019, 28, 607–618. [Google Scholar] [CrossRef]
  166. Wang, K.; Xiao, J.; Peng, B.; Xing, F.; So, K.F.; Tipoe, G.L.; Lin, B. Retinal structure and function preservation by polysaccharides of wolfberry in a mouse model of retinal degeneration. Sci. Rep. 2014, 4, 7601. [Google Scholar] [CrossRef]
  167. Zheng, H.-l.; Li, M.-t.; Zhou, T.; Wang, Y.-y.; Shang, E.-X.; Hua, Y.-q.; Duan, J.-a.; Zhu, Y. Protective effects of Lycium barbarum L. berry extracts against oxidative stress-induced damage of the retina of aging mouse and ARPE-19 cells. Food Funct. 2023, 14, 399–412. [Google Scholar] [CrossRef]
  168. Mi, X.S.; Feng, Q.; Lo, A.C.; Chang, R.C.; Lin, B.; Chung, S.K.; So, K.F. Protection of retinal ganglion cells and retinal vasculature by Lycium barbarum polysaccharides in a mouse model of acute ocular hypertension. PLoS ONE 2012, 7, e45469. [Google Scholar] [CrossRef]
  169. Lakshmanan, Y.; Wong, F.S.-Y.; Yu, W.-Y.; Li, S.Z.-C.; Choi, K.-Y.; So, K.-F.; Chan, H.H.-L. Lycium brbarum polysaccharides rescue neurodegeneration in an acute ocular hypertension rat model under pre- and posttreatment conditions. Investig. Ophthalmol. Vis. Sci. 2019, 60, 2023–2033. [Google Scholar] [CrossRef]
  170. Mi, X.S.; Chiu, K.; Van, G.; Leung, J.W.; Lo, A.C.; Chung, S.K.; Chang, R.C.; So, K.F. Effect of Lycium barbarum Polysaccharides on the expression of endothelin-1 and its receptors in an ocular hypertension model of rat glaucoma. Neural. Regen. Res. 2012, 7, 645–651. [Google Scholar] [CrossRef]
  171. Mi, X.S.; Feng, Q.; Lo, A.C.Y.; Chang, R.C.; Chung, S.K.; So, K.F. Lycium barbarum polysaccharides related RAGE and Aβ levels in the retina of mice with acute ocular hypertension and promote maintenance of blood retinal barrier. Neural. Regen. Res. 2020, 15, 2344–2352. [Google Scholar] [CrossRef]
  172. Yang, D.; So, K.-F.; Lo, A.C. Lycium barbarum polysaccharide extracts preserve retinal function and attenuate inner retinal neuronal damage in a mouse model of transient retinal ischaemia. Clin. Exp. Ophthalmol. 2017, 45, 717–729. [Google Scholar] [CrossRef]
Figure 1. Factors influencing the phytochemistry and biological properties of L. barbarum.
Figure 1. Factors influencing the phytochemistry and biological properties of L. barbarum.
Obesities 05 00035 g001
Figure 2. Seven main monosaccharides present in L. barbarum polysaccharides (LBPs).
Figure 2. Seven main monosaccharides present in L. barbarum polysaccharides (LBPs).
Obesities 05 00035 g002
Figure 3. Biological Properties of L. barbarum.
Figure 3. Biological Properties of L. barbarum.
Obesities 05 00035 g003
Table 1. Effects of L. barbarum on cardiovascular health.
Table 1. Effects of L. barbarum on cardiovascular health.
Model Main BioactivesKey FindingsMechanism/PathwayRefs.
In vitro LBP↓ Myocardial damage, ↓ apoptosis, preserved mitochondrial functionSIRT3/CypD pathway[77]
In vivo LBP↓ Infarct size, prevented adverse cardiac remodelling, lowered oxidative stress, improved mitochondrial dynamicsGRK2 expression inhibition; restoration of mitochondrial fission/fusion balance; activation of AKT/eNOS signalling[78]
In vitro LBPEnhanced antioxidant defences (↑ SOD, ↑ NO), ↓ oxidative damage (↓ MDA), anti-apoptotic effectsOxidative stress regulation[79]
In vivo L. barbarum extractNormalised blood pressure, ↑ eNOS, ↓ sONE expressionsONE/eNOS pathway modulation[80]
AKT: protein kinase B; GRK2: G protein-coupled receptor kinase 2; eNOS: endothelial nitric oxide synthase; LBP: L. Barbarum polysaccharide; MDA: malondialdehyde; NO: nitric oxide; SIRT: sirtuin; SOD: superoxide dismutase; sONE: an antisense mRNA.
Table 2. Effects of L. barbarum on glycaemic control and anti-diabetic activity (A: obesity and insulin sensitivity; B: glycaemic control and diabetes management; C: lipid metabolism; D: obesity-related bone health).
Table 2. Effects of L. barbarum on glycaemic control and anti-diabetic activity (A: obesity and insulin sensitivity; B: glycaemic control and diabetes management; C: lipid metabolism; D: obesity-related bone health).
Model Main Bioactive(s)Key FindingsMechanism/PathwayRefs.
AIn vivoLBP↓ Adipocyte lipid accumulation, ↑ insulin sensitivityATF6/SIRT1-dependent downregulation of Fsp27[83]
In vivo LICP009-2-1↓ Lipid accumulation and hyperlipidemiaAnti-adipogenic activity[84]
In vivo LBP↓ Blood glucose, ↑ insulin sensitivity, ↑ testosteroneAnti-metabolic disturbance[85]
In vivo LBLF↓ Oxidative stress, ↑ liver function Improved insulin resistance, optimised gut microbiota MAPK and retinol metabolism pathways modulation[86]
In vitro
In vivo
LBP↓ Obesity-related factors and ↑ muscle-related factors; ↑ glucose metabolism; mitigated ectopic fat and mitochondrial dysfunctionAMPK/PINK1/Parkin-mediated mitophagy; ↑ mitochondrial membrane potential and ATP, ↓ROS[87]
BIn vivo ZeaxanthinImproved blood glucose, lipid profile, nephroprotectionModulation of inflammatory cytokines and antioxidant enzymes[88]
In vivo LBPImproved gut motility, ↑ SCFA productionNeuronal regulation of duodenal contraction[89]
In vivo LBP↓ Oxidative stress, protected DNA in lymphocytesAntioxidant protection (↑ SOD, ↓ MDA/NO)[90]
In vivo LBLRestored organ function, improved lipid and glucose metabolismModulation of gut microbiota and metabolic disruption reversal[91]
CIn vivoLBP↓ Cholesterol and triglycerides, promoted weight lossGut microbiota modulation (↑ Bacteroidetes, ↓ Firmicutes)[92]
In vivo LBPImproved lipid metabolism, ↑ antioxidant capacityUpregulation of lipid metabolism genes (FAS, PPAR-α, CPT1, ATGL)[93]
In vivo LBPImproved growth, lipid metabolism, antioxidant capacityDownregulation of lipid metabolism genes (ACC1, PPAR-γ)[94]
In vivo LBPImproved lipid metabolism, alleviated metabolic disorder symptoms, ↓weight gain, ↑ microbial diversityModulation of gut microbiota (↑ Firmicutes, ↑ microbial diversity), regulation of >30 differential metabolites and 4 metabolic pathways[95]
DIn vitro LBPMitigated osteoblast apoptosismiR-200b-3p/Chrdl1/PPARγ pathway modulation[96]
In vitro LBPPromoted osteoblast proliferation via SCFA productionUpregulation of B-ALP and osteocalcin expression[97]
ATF6: activating transcription factor 6; ATGL: adipose triglyceride lipase; B-ALP: bone-alkaline phosphatase; Chrdl1: chordin-like 1; CPT1: carnitine palmitoyltransferase 1; DNA: deoxyribonucleic acid; FAS: fatty acid synthase; LBLF: lBP leaf flavonoids; LBP: L. barbarum polysaccharides; LICP009-2-1: purified LBP fraction; LLB: L. barbarum leaf extract MAPK: mitogen-activated protein kinase; MDA: malondialdehyde; NO: nitric oxide; PPAR: peroxisome proliferator-activated receptor; SCFAs: short-chain fatty acids; SIRT1: sirtuin 1; SOD: superoxide dismutase.
Table 4. Effects of L. barbarum on anti-inflammatory activity.
Table 4. Effects of L. barbarum on anti-inflammatory activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vivoMethanol extract↓ Paw swelling, ↓ ROS, neutrophil migrationMyeloperoxidase inhibition[56]
In vivoFermented/non-fermented extractsImproved hepatic function, lipid metabolismAnti-inflammatory + antioxidant[111]
In vivoLBP↓ Fat accumulation, fibrosis, inflammationNF-κB, MAPK, autophagy[112]
In vivoLBP↓ Albuminuria, ↓ inflammatory, ↑ SOD ↓ Cytokines, ↓NF-κBRenal protection[113]
In vivoLBPAnti-inflammatory, anti-apoptoticNrf2 pathway[114]
In vivoLBP↓ Oxidative injury, ↓ inflammation markersTLR/NF-κB inhibition[115]
In vivoLBP↓ ROS, 1CAM-1, improved kidney functionNF-κB and angiotensin downregulation[116]
LBP: L. Barbarum polysaccharide; MAPK: mitogen-activated protein kinase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2: nuclear factor erythroid 2-related factor 2; ROS: reactive oxygen species; SOD: superoxide dismutase; TLR: toll-like receptor.
Table 5. Effects of L. barbarum on immunomodulatory activity.
Table 5. Effects of L. barbarum on immunomodulatory activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vivo LBP↓ Tumour size, ↑ macrophage and lymphocyte activityImmune activation, ↓ lipid peroxidation[117]
In vivo LBP↑ CD8+ T cell infiltration, ↓ T cell exhaustionEnhanced systemic/local antitumour immunity[118]
In vitro LBP (>10 kDa)↑ Macrophage viability and NO, TNF-α, IL-6Cellular uptake via clathrin-mediated endocytosis[119]
In vivo LBP↑ Immune organ indices, IgG, CD4+/CD8+ ratioImmune stimulation and improved feed efficiency[24,120]
In vivo LBP↑ Cytokines, improved sperm parametersImmuno-protection in cyclophosphamide model[121]
In vivo LBP↓ Hepatotoxicity, ↑ SCFAs, modulated gut microbiotaGut–immune axis modulation[122]
In vivo Juice blends↑ Splenic macrophages, spleen weightSynergistic antioxidant-immune enhancement[28]
IFN-α: interferon-alpha; IL: interleukin; IgG: immunoglobulin G; LBP: L. Barbarum polysaccharide; NO: nitric oxide; SCFAs: short chain fatty acids.
Table 6. Effects of L. barbarum on anticancer activity.
Table 6. Effects of L. barbarum on anticancer activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vitro Ethanol extract↑ NK cytotoxicity, ↓ProliferationNK cell activation[126]
In vitro Ethanol extract↓ Proliferation, ↑ Apoptosis, ↑ Bax, ↓ BclxLMitochondrial pathway[31]
In vitroPolyphenol-rich extractInhibited proliferation in MCF-7 and MDA-MB-231 cancer cellsDose-dependent inhibition[127]
In vitro LBP fractionsInduced G0/G1 arrest, apoptosis, ↓ mito. potential↑ Caspase, ↑ MAPK, ↓ Bcl-2[128]
In vitro LBPInduced S phase arrest and apoptosis, ↑ RNA/Ca2+Calcium-regulated apoptotic pathways[129]
In vivoLBP-3 (40–350 kDa)Tumour suppression, S phase arrest immune supportImmune modulation [130]
In vitro Ethanol extract↓ Proliferation, ↓ migrationERK1/2, AKT suppression[131]
In vitro LLB extract No cytotoxicity or genotoxicity; ↓ DNA damage; ↓ pro-metastatic and ↑tumour suppressor genesModulation of oxidative stress, apoptosis, and cancer-related gene expression[69]
In vitro LBPCombined LBP and cisplatin inhibited cell proliferation; ↑ apoptosis, ↓ ROS; modulated S/G2-M cell cycle phasesEnhanced apoptosis and cell cycle arrest via cyclin D1-CDK4-Rb pathway[132]
In vitro
In vivo
LBPCombined LBP and IFN-α2b inhibited proliferation, induced apoptosis, ↓ tumour volume, and ↓ MDSC ratioSynergistic regulation of apoptosis and immune suppression via cyclin D1/c-Myc/Bcl-2 pathway and MDSC modulation[133]
In vitro
In vivo
LBPReduced cardiac damage (↓ myofibrillar disarrangement), improved conduction abnormalities, and preserved anti-tumour activity of DoxorubicinSuppression of oxidative stress (↑ SOD, ↑ GSH-Px, ↓ MDA)[134]
AKT: protein kinase B; Bax: Bcl-2-associated X protein; BclxL: B-cell lymphoma-extra large; CDK: cyclin-dependent kinase; ERK: extracellular signal-regulated kinases; GSH-Px: glutathione peroxidase; IFN: interferon; LBP: L. Barbarum polysaccharide; MAPK: mitogen-activated protein kinase; MDA: malondialdehyde; MDSC: myeloid-derived suppressor cell; NK: natural killer; RNA: ribonucleic acid; ROS: reactive oxygen species; SOD: superoxide dismutase.
Table 7. Hepatoprotective (A), antimicrobial (B) and prebiotic (C) effects of L. barbarum.
Table 7. Hepatoprotective (A), antimicrobial (B) and prebiotic (C) effects of L. barbarum.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
AIn vitro LBP↓ Oxidative stress, ↓ apoptosis, ↓ inflammation TXNIP-NLRP3[137]
In vivo L. barbarum
extract
↓ ALT, ↓ASTAntioxidant enhancement[138]
In vivo LBP↓ Fatty liver, ↑ liver enzymes, ↑ antioxidant activityHepatic protection and redox modulation[139]
BIn vitroYellow L. barbarum extractStrong activity against Gram– and CandidaDose-dependent[14]
In vitro Hydromethanolic extract↑ Antibacterial activity (Gram+ > Gram) Disruption of cell membrane integrity[15]
In vitroPolyphenols (e.g., chlorogenic acid)E. coli growthTime/dose/temperature-dependent[29]
In vitroEthanolic extractEffective against periodontal pathogensAlternative to chlorhexidine[140]
CIn vivo LBP↑ Firmicutes/Proteobacteria, ↓ Bacteroidetes
Lactobacillus/Akkermansia/Prevotellaceae
Microbiota modulation
Gut microbiota enrichment
[141]
In vitro LBPL. acidophilus, ↑B. longum (2.5–15% LBP)Prebiotic-enhanced bacterial growth[141]
In vivo L. barbarum↓ ALT/AST ↑ Lachnospiraceae, ↑ Ruminococcaceae Gut–liver axis modulation[142]
In vitroAqueous extractLactobacillus, Bifidobacterium growthStimulated probiotic proliferation[143]
ALT: alanine aminotransferase; AST: aspartate aminotransferase; LBP: L. barbarum polysaccharides; NLRP3: NOD-like receptor protein 3; TXNIP: thioredoxin-interacting protein.
Table 8. Effects of L. barbarum on neuroprotective activity.
Table 8. Effects of L. barbarum on neuroprotective activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vivo L. barbarum extract↓ Oxidative stress and cytokines; ↑ hippocampal neuron survivalPI3K/Akt/GSK-3β, PKCε/Nrf2/HO-1, NR2A/NR2B[146]
In vivo LBP↑ Spatial memory, ↑ neurogenesis; ↓apoptosis and ER stressNrf2/HO-1 signalling[147]
In vitro
In vivo
LBP↓ ROS, ↓ mitochondrial damage; ↑ caspase-3/-9 activity↑ Nrf2 and HO-1 expression[148]
In vivo
In vitro
L. barbarumReversed cognitive impairment; ↓ apoptosis; ↑ antioxidant defenceNrf2/HO-1 [149]
In vitro
In vivo
Water extract↑ Cell survival; ↓ROS; ↑ acetylcholine, choline acetyltransferaseMitochondrial protection and neurotransmitter regulation[150]
In vitro Alkaline extract ↓ Caspase-3 activity; ↑ Akt phosphorylationAnti-apoptotic via Akt signalling[30]
In vivo
In vitro
L. barbarum extract↓ Dopaminergic neuron loss; ↓ROS; ↑ GSHMitochondrial stabilisation, antioxidative action[151]
Akt: protein kinase B; GRK: G protein-coupled receptor kinase; GSH: glutathione; HO-1: haem oxygenase 1; LBP: L. Barbarum polysaccharide; Nrf2: nuclear factor erythroid 2-related factor 2; PI3K: phosphoinositide 3-kinase; ROS: reactive oxygen species.
Table 9. Effects of L. barbarum on anti-aging activity.
Table 9. Effects of L. barbarum on anti-aging activity.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vivo ABP and LBP↓ glycation end-products, ↑ IL-2, ↑ SOD, ↑ cognitive and motor functionGlycation inhibition; immune and oxidative modulation[153]
In vivo LBP, LBP-2 (arabinogalactan)↑ Lifespan, ↑ SOD/CAT, ↓ MDAMAPK/TOR/S6K pathway; longevity gene upregulation[154]
In vitro LBP↓ UVB-induced DNA damage and ROS, ↑ Nrf2 activation, ↓ p38 MAPKNrf2/ARE; caspase-3, MMP-9[155]
In vivo
In vitro
LBP1C-2↑ Muscle stem cell self-renewal and repairFGFR1 binding; Spry1 upregulation[156]
In vivo
In vitro
LBP↑ Osteogenic markers; ↓bone resorption and osteoclastsERK1/2 pathway activation[157]
In vivo LBP↓ Senescence, ↓ apoptosisp53 signalling modulation[158]
In vitro LBP↓ Lipid peroxidation, delayed apoptosisOxidative stress reduction[159]
In vitro LBP↓ Oxidative stress, ↓ apoptosis and autophagyPI3K/Akt/mTOR, Bcl-2/Bax, Caspase-3[160]
In vivo LBP↑ SIRT3, ↓ CypD acetylation, ↑ mitochondrial protectionSIRT3/CypD pathway activation[77]
ARE: antioxidant response element; Bax: Bcl-2-associated X protein; CAT: catalase; FGFR1: fibroblast growth factor receptor 1; IL: interleukin; MAPK: mitogen-activated protein kinase; MDA: malondialdehyde; MMP-9: matrix metalloproteinase 9; Nrf2: nuclear factor erythroid 2-related factor 2; ROS: reactive oxygen species; SIRT: sirtuin; SOD: superoxide dismutase; TOR: target of rapamycin; UAB: ultraviolet B radiation.
Table 10. Effects of L. barbarum on ocular health.
Table 10. Effects of L. barbarum on ocular health.
ModelMain Bioactive(s)Key FindingsMechanism/PathwayRefs.
In vivoLBP↑ Nrf2 and HO-1; ↓ apoptosis; ↑ survival of ganglion cellsNrf2/HO-1 antioxidant pathway[162]
In vivoLBP↓ oxidative stress; ↓ JNK; ↑ IGF-1; ↓ secondary RGC degenerationJNK pathway, oxidative stress inhibition[163]
In vivoLBP↑ retinal function and visual signallingRetinal functional recovery[164]
In vivoLBP↑ M2 microglia/macrophage polarisation; ↓ autophagy; ↑ RGC survivalImmune modulation[165]
In vivoLBPPreserved photoreceptor morphology and visual behaviourNF-κB and HIF-1α inhibition[166]
In vivoLBW-95E↑SOD, ↑GSH, ↑Nrf2; ↓ ROS, inflammationAntioxidant and anti-inflammatory[167]
In vivoLBP↓ RGC loss; preserved blood-retinal barrierDownregulation of inflammatory mediators[168]
In vivoLBPPreserved inner retinal layer thickness; ↑retinal functionProtection from secondary degeneration[169]
In vivoLBP↓ ET-1 expression; ↑ ETA, ↓ ETB in RGCsET-1 signalling modulation[170]
In vivoLBP↓ astrocyte/microglia activation; preserved barrier integrityGlial reactivity modulation[171]
In vivoLBP↑ viable retinal cells, ERG, ↓ glial activityNeuroprotection[172]
ETA: endothelin type A; ET-1: endothelin-1; GSH: glutathione; HIF-1α: hypoxia-inducible factor 1-alpha; HO-1: haem oxygenase 1; IGF-1: insulin-like growth factor 1; LBP: L. Barbarum polysaccharide; LBW-95E: LBP fraction; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2: nuclear factor erythroid 2-related factor 2; JNK: c-Jun N-terminal kinase; RGC: retinal ganglion cell; ROS: reactive oxygen species; SOD: superoxide dismutase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, T.; Alexa, E.-A.; Liu, G.; Berisha, A.; Walsh, R.; Kelleher, R. Lycium barbarum for Health and Longevity: A Review of Its Biological Significance. Obesities 2025, 5, 35. https://doi.org/10.3390/obesities5020035

AMA Style

Zhang T, Alexa E-A, Liu G, Berisha A, Walsh R, Kelleher R. Lycium barbarum for Health and Longevity: A Review of Its Biological Significance. Obesities. 2025; 5(2):35. https://doi.org/10.3390/obesities5020035

Chicago/Turabian Style

Zhang, Tao, Elena-Alexandra Alexa, Gavin Liu, Alois Berisha, Rhys Walsh, and Robbie Kelleher. 2025. "Lycium barbarum for Health and Longevity: A Review of Its Biological Significance" Obesities 5, no. 2: 35. https://doi.org/10.3390/obesities5020035

APA Style

Zhang, T., Alexa, E.-A., Liu, G., Berisha, A., Walsh, R., & Kelleher, R. (2025). Lycium barbarum for Health and Longevity: A Review of Its Biological Significance. Obesities, 5(2), 35. https://doi.org/10.3390/obesities5020035

Article Metrics

Back to TopTop