Hepatoprotective and Antihyperglycemic Effects of Sea Grapes  
(Caulerpa spp.) as a Potential Therapeutic Approach for NAFLD and  
NASH  
Yasmin Azzahra Arifin1, Annette d’Arqom2*  
1 Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60131, Indonesia.  
2Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine, Universitas Airlangga,  
Surabaya, East Java, 60131, Indonesia.  
Received: 02 October 2025; Accepted: 08 October 2025; Published: 21 November 2025  
ABSTRACT  
Non-alcoholic fatty liver disease (NAFLD) and its advanced form, non-alcoholic steatohepatitis (NASH), are  
increasingly prevalent metabolic liver disorders characterized by hepatic lipid accumulation, oxidative stress,  
inflammation, and insulin resistance. Despite their global burden, effective pharmacological treatments remain  
limited, which highlights the need for safe and multi-targeted natural alternatives. This literature review  
summarizes current evidence from in vitro, in vivo, and early clinical studies investigating the hepatoprotective  
and antihyperglycemic properties of Caulerpa spp., including C. lentillifera, C. racemosa, C. taxifolia, and C.  
prolifera. The review focuses on research published between 2015 and 2025 related to Caulerpa bioactivity,  
liver protection, glucose regulation, oxidative stress, lipid metabolism, and gut microbiota modulation. Across  
the reviewed studies, Caulerpa-derived bioactive compounds such as polysaccharides, carotenoids, peptides,  
and polyphenols consistently demonstrated hepatoprotective and metabolic benefits. Supplementation improved  
antioxidant enzyme activities, including superoxide dismutase, catalase, and glutathione, while reducing  
malondialdehyde levels, thereby protecting hepatocytes from oxidative damage that contributes to NAFLD  
progression. Several studies also reported downregulation of lipogenic genes such as SREBF1, FAS, and ACC,  
together with activation of SIRT1 and AMPK signalling pathways, which reduced hepatic triglyceride  
accumulation. Extracts from C. taxifolia and C. prolifera exhibited strong alpha-amylase and alpha-glucosidase  
inhibition, improving glucose regulation. In addition, Caulerpa supplementation was shown to restore intestinal  
barrier integrity and modify gut microbiota composition, leading to lower endotoxin levels and reduced hepatic  
inflammation. The only available human clinical trial demonstrated a significant reduction in fasting glucose  
following C. racemosa supplementation, supporting its potential for clinical application. Overall, the evidence  
indicates that Caulerpa species exert multi-pathway hepatoprotective and antihyperglycemic effects. However,  
further studies are required to standardize extract preparation, determine optimal dosage, and assess long-term  
safety. Caulerpa spp. demonstrates promising preclinical potential. However, further translational research and  
rigorous clinical trials are warranted to validate these findings and determine its feasibility as a nutraceutical for  
NAFLD, NASH, and metabolic syndrome.  
Keywords: Caulerpa spp, Sea grapes, Hepatoprotective, Anti hyperglycemic, Fatty liver disease, Metabolic  
syndrome  
INTRODUCTION  
Non-alcoholic fatty liver disease (NAFLD) and its more severe form, non-alcoholic steatohepatitis (NASH), are  
major global health problems linked to obesity, insulin resistance, and type 2 diabetes. Affecting nearly one-  
third of the world’s population, NAFLD is considered the hepatic manifestation of metabolic syndrome,  
characterized by excessive lipid accumulation in hepatocytes independent of alcohol consumption. Persistent  
hepatic steatosis can progress to chronic inflammation, fibrosis, and cirrhosis, posing a significant risk for  
hepatocellular carcinoma. The underlying mechanisms of NAFLD and NASH involve oxidative stress,  
mitochondrial dysfunction, endoplasmic reticulum stress, and dysregulation of lipid and glucose metabolism  
Page 3698  
[3][6]. Despite the high prevalence of NAFLD, current pharmacological treatments remain limited. Management  
strategies largely depend on diet and lifestyle modification, as no approved first-line drugs are available.  
Therefore, natural marine-derived compounds with antioxidant, metabolic, and gut-modulating effects are  
attractive alternatives. Caulerpa spp. are unique among green macroalgae due to their high sulfated  
polysaccharide and carotenoid content, which have shown strong metabolic regulatory activity in recent  
preclinical studies Species such as C. racemosa, C. lentillifera, C. taxifolia, and C. prolifera contain a rich array  
of bioactive compounds. Preclinical evidence suggests these compounds hold therapeutic promise. Experimental  
studies, using both in vivo animal models and in vitro cell lines, have shown that Caulerpa extracts can modulate  
key molecular pathways associated with lipid metabolism and energy balance, such as the SIRT1/AMPK and  
mTOR signaling cascades. Furthermore, emerging research highlights mechanisms central to NAFLD/NASH  
pathophysiology, including the inhibition of digestive enzymes like α-amylase, the suppression of oxidative  
stress, and the modulation of the gut-liver axis through changes in gut microbiota composition [3][5][7][9].  
This review therefore aims to synthesize and analyze recent findings on the hepatoprotective and  
antihyperglycemic effects of sea grapes (Caulerpa spp.) and relating to NAFLD and NASH treatment. By  
integrating biochemical, molecular, and microbiome perspectives, this paper provides a comprehensive  
understanding of how Caulerpa-derived compounds contribute to metabolic and hepatic protection, establishing  
a foundation for future nutraceutical development and clinical translation.  
MATERIAL AND METHOD  
Research Strategy  
A structured literature search was conducted in Scopus (Elsevier) and PubMed using the keyword phrases “Sea  
grapes” OR “Caulerpa” OR “Caulerpa lentillifera” OR “Macroalgae” AND “Liver disease” OR “NAFLD” OR  
“NASH” OR “Liver steatosis” OR “Fatty liver disease.” OR "blood glucose" OR "anti diabetic" OR "blood sugar  
regulation" The search was performed on 5 November 2025 (Asia/Surabaya) and limited to publications from  
20152025. The initial query returned 95 for Scopus (Elsevier). Titles and abstracts were screened for relevance  
to the hepatoprotective and blood glucose-lowering effects of sea grapes for treating NAFLD and NASH. Only  
English-language publications were included. After applying these criteria, 15 studies were retained for full-text  
review and included in this narrative literature review. The detailed study selection process, outlining study  
identification, screening, eligibility, and inclusion, is summarized in the PRISMA 2020 Flowchart (See Figure  
1)  
Figure 1 Prisma Flowchart  
Identification of studies via databases and registers  
Records removed before screening:  
Records identified from:  
Duplicate records removed (n = 54)  
Databases (n = 149)  
Records removed for other reasons  
Registers (n = 0)  
(n = 0)  
Records screened  
(n = 95)  
Records excluded  
(n = 80)  
Page 3699  
Reports not retrieved  
(n = 0)  
Reports sought for retrieval  
(n = 15)  
Reports excluded: 0  
Reports assessed for eligibility  
(n = 15)  
Studies included in review  
(n = 15)  
Reports of included studies  
(n = 15)  
Inclusion and Exclusion Criteria  
Inclusion criteria: Articles published between 20152025, written in English, involving Caulerpa spp. or sea  
grapes (including extracts or isolated bioactive compounds), and reporting hepatoprotective and/or  
antihyperglycemic effects in NAFLD, NASH, fatty liver disease, liver steatosis, insulin resistance, or  
hyperglycemia in in vivo, in vitro, or clinical studies Exclusion criteria: Non-English publications; articles  
without liver- or glucose-related outcomes; studies on algae not belonging to Caulerpa spp., reviews, editorials,  
or conference abstracts without primary data, studies focusing only on chemical composition without biological  
activity, and studies unrelated to metabolic or liver disease models.  
Quality and Risk-of-Bias Assessment  
The methodological quality of the included studies was independently assessed by reviewers. The risk-of-bias  
for in vivo (animal) studies was conducted using criteria from the SYRCLE (Systematic Review Centre for  
Laboratory animal Experimentation) checklist. For the single clinical trial, the Cochrane Risk-of-Bias 2 (RoB 2)  
tool was used. In vitro studies were assessed using the SciRAP (Science in Risk Assessment and Policy) method,  
which evaluates both reporting quality and methodological quality based on predefined criteria such as clarity  
of test system description, exposure characterization (e.g., concentrations), adequacy of controls, and replication.  
Any disagreements in assessment were resolved through discussion or consensus. The results of this risk-of-bias  
assessment are summarized and integrated into the synthesis tables (Tables 1-3).  
Data Extraction and Synthesis  
From the 15 studies, we extracted aggregate data on study design (e.g., in vivo animal model, in vitro cell culture,  
or human clinical trial), sample characteristics (e.g., species, strain, or participant demographics), and  
intervention specifics (e.g., Caulerpa species, extract type, dosage, and duration). Key outcome measures were  
categorized by hepatoprotective effects (including changes in liver enzymes, hepatic fat accumulation, oxidative  
stress markers, and fibrosis) and antidiabetic effects (including fasting blood glucose, enzyme inhibition, insulin  
levels, and metabolic regulators like PGC-1). Evidence was synthesized narratively, with harmonization of effect  
measures where feasible. No meta-analysis was performed due to the significant heterogeneity observed across  
Page 3700  
study models, intervention protocols, and outcome measures. Although a meta-analysis was not performed, the  
narrative synthesis includes an evaluation of the overall strength of evidence based on an adapted GRADE  
(Grading of Recommendations, Assessment, Development and Evaluations) framework. The strength of  
evidence was categorized as 'high', 'moderate', 'low', or 'very low' based on study risk of bias, consistency of  
findings, and the precision of the reported preclinical data.  
RESULT AND DISCUSSION  
Hepatoprotective Effects of Caulerpa spp.  
Multiple studies demonstrate Caulerpa spp, possess notable hepatoprotective activities, primarily through  
enhancing endogenous antioxidant defenses, reducing hepatic inflammation, and restoring liver enzyme balance.  
In a diquat-induced liver injury model, supplementation with Caulerpa lentillifera significantly decreased ALT  
levels (from approximately 29 U/mg to 20 U/mg) while increasing SOD activity (from 38 U/mg to 46 U/mg),  
indicating an effective reduction of oxidative stress within hepatocytes [1]. Comparable protective outcomes  
were observed in ethanol-induced liver injury, where C. lentillifera administration alleviated elevations in AST  
and GGT and lowered hepatic inflammation scores. These benefits were accompanied by suppression of TLR4-  
mediated inflammatory signaling and recovery of intestinal tight-junction proteins such as occludin and ZO-1  
[5]. In high-fat diet (HFD) animal models, sulfated polysaccharides derived from C. racemosa significantly  
decreased oxidative stress markers and enhanced the activity of endogenous antioxidant enzymes, further  
confirming its hepatoprotective potential [7]. Supplementation with C. lentillifera in animals with metabolic  
syndrome similarly improved hepatic SOD activity and reduced lipid accumulation in liver tissue [10]. Since  
oxidative stress plays a central role in the development and progression of NAFLD, these consistent findings  
suggest that Caulerpa supplementation may prevent early hepatocellular damage and slow the transition to  
steatohepatitis.  
Antihyperglycemic and Metabolic Effects  
The reviewed studies also reveal strong antihyperglycemic effects across different Caulerpa species. A sulfated  
galactan isolated from C. taxifolia improved insulin sensitivity, reduced fasting blood glucose, and inhibited α-  
amylase activity in diabetic mice [3]. Polyphenolic extracts of C. prolifera demonstrated potent α-glucosidase  
and α-amylase inhibition in vitro, with compounds such as vanillin and kaempferol showing strong enzyme-  
binding affinity [4]. In vivo studies confirmed glucose-lowering effects, as C. racemosa supplementation  
significantly reduced blood glucose levels in obese and high-fat diet animal models [9,12], while C.  
lentillifera improved glucose profiles in metabolic syndrome rats [10]. Notably, the only available double-blind,  
placebo-controlled human trial demonstrated that 4-week supplementation with C. racemosa significantly  
reduced mean fasting blood glucose in obese adults, from a baseline of 113.68 mg/dL to 79.82 mg/dL (p=0.000)  
[11].These findings suggest that Caulerpa acts through multiple mechanisms, including enzyme inhibition,  
increased insulin sensitivity, and improved hepatic glucose metabolism.  
Comparison on Caulerpa spp.  
Although hepatoprotective and antidiabetic effects were observed across all species, different Caulerpa types  
demonstrated distinct therapeutic strengths. C. lentillifera consistently showed antioxidant and anti-steatosis  
activity, preventing hepatic lipid accumulation and reducing oxidative injury in zebrafish, rats, and HepG2 cells  
[1,5,6]. C. racemosa produced the strongest antihyperglycemic effects in both preclinical and clinical studies  
and significantly improved gut microbiota composition in high-fat diet models [7,9,11]. C. taxifolia and C.  
prolifera exhibited potent α-amylase and α-glucosidase inhibition due to high phenolic and polysaccharide  
content [3,4]. These differences suggest the possibility of species-specific application, where C. lentillifera may  
be more suitable as an antioxidant hepatoprotective agent, while C. racemosa and C. taxifolia may serve as  
effective glucose-lowering and metabolic regulators.  
Table 1 Summary Of In Vitro Studies on The Metabolic Effects of Caulerpa Spp.  
Page 3701  
Study  
(Author,  
Year)  
Caulerpa  
Species  
Model Cell /  
Assay  
Key Finding  
Mechanism  
Risk of Bias  
Assessment  
Sangpairoj et C. lentillifera  
HepG2  
Lipid accumulation;  
SIRT1/AMPK  
Low Risk  
hepatocytes;  
assays: MTT  
cytotoxicity,  
Oil Red O  
al. (2024)  
Trigliserida  
Pathway,  
Lipogenesis genes  
(SREBF1, FAS, ACC)  
Potential binding of  
major compounds (dl-2-  
phenyltryptophane,  
benzoic acid) to SIRT1  
and AMPK (via  
staining,  
Triglyceride  
assay, qPCR,  
Western blot  
docking)  
Ouahabi S. et  
al. (2025)  
C. prolifera  
Assay α -  
mylase & α-  
glucosidase  
Strong inhibitory activity;  
some extracts (ME,  
Digestive enzyme  
inhibition (slower carb  
breakdown & glucose  
absorption  
Low Risk  
Low Risk  
Low Risk  
Low Risk  
AQE) comparable or  
superior to acarbose  
DPPH & β-  
carotene  
bleaching  
Aqueous/methanolic  
extracts show strong  
radical scavenging; EA  
strongest in lipid  
Antioxidant  
activity (radical  
scavenging & lipid  
oxidation inhibition)  
peroxidation inhibition  
Dissanayake,  
I.H et al.  
(2022)  
C. racemosa  
DPPH, FRAP  
Moderatestrong  
antioxidant activity;  
strong correlation with  
phenolic & flavonoid  
content  
Free-radical scavenging  
& reducing activity  
α-amylase &  
α-glucosidase  
inhibition;  
Anti-glycation  
assays  
Strong inhibition of α-  
amylase (CPE) and α-  
glucosidase (EA  
fraction); clear dose–  
response; notable anti-  
glycation effects  
Digestive enzyme  
inhibition → potential  
postprandial glucose  
control  
Nurkolis et  
al. (2023)  
C. lentillifera  
HepG2  
hepatocytes  
(lipid  
↓Lipid accumulation;  
↓TG; ↑SIRT1/AMPK  
Lipid-lowering  
via SIRT1AMPK  
activation  
Moderate  
Risk  
accumulation  
assays)  
Table 2 Summary of In Vivo (Animal Model) Studies on The Hepatoprotective And Antidiabetic Effects of  
Caulerpa Spp  
Page 3702  
Study  
(Author,  
Year)  
Key Findings  
Animal  
Model  
Risk of Bias  
Assessment  
(Liver &  
Glucose)  
Caulerpa Species  
Dose  
Duration  
Glucose:  
↓ Blood Glucose  
↑ glut2 & akt  
mRNA (Insulin  
signaling)  
Zebrafish  
(Danio rerio)  
(Diquat-  
induced  
oxidative  
damage)  
Liver:  
20 g/kg &  
50 g/kg in  
diet  
C. lentillifera  
Lin X et al.  
(2024)  
Medium  
Risk  
↓ Liver MDA  
(Oxidative stress)  
56 days  
(Dried powder)  
↑ SOD activity  
(Antioxidant)  
↓ ALT & AST  
(Liver enzymes)  
↓ Hepatic TG  
↓Histopathological  
lesions  
Glucose &  
Metabolism:  
↓ Blood Glucose  
↓ α-glucosidase &  
α-amylase activity  
↓ Lipase activity  
22.5  
mg/kg &  
45 mg/kg  
BW  
Kurniawan,  
R. et al.  
C. racemosa  
(Carotenoids) & C.  
lentillifera (Peptide).  
Male Rattus  
norvegicus  
fed a CFED  
Liver & Lipids:  
↓ TG, TC, LDL  
↑ HDL  
4 weeks  
Low Risk  
(2025)  
↓ AST (Liver  
enzyme)  
↓ TNF-α,  
↑ IL-10  
Male  
C57BL/6J  
mice (T2DM  
Liu, S. et  
al. (2025)  
C. taxifolia (Sulfated  
Medium  
Risk  
100, 200,  
400  
Glucose & Insulin:  
↓ Fasting Blood  
5 weeks  
galactan - SGC)  
Page 3703  
model)  
mg/kg/day  
Glucose  
↓ HOMA-IR  
(Insulin  
Resistance)  
↑ Glucose  
Tolerance (OGTT)  
Liver & Oxidative  
Stress:  
↓ Liver damage  
(vacuolation)  
↑ Antioxidants  
(SOD, CAT, GSH)  
↓ MDA (Oxidative  
stress marker)  
Glucose:  
No glucose data  
measured.  
Liver & Gut:  
↓ Serum AST &  
GGT  
↓ Liver  
inflammation  
score ↔ Liver  
steatosis, Hepatic  
Male Wistar  
rats (Chronic  
ethanol  
5% in  
Lin K.Y. et  
al. (2023)  
C. lentillifera (Dried  
liquid diet 12 weeks TC & TG (No  
Low Risk  
powder)  
(8.4 g/L)  
change)  
exposure)  
↓ Circulatory  
endotoxin  
↓ Hepatic TNF-α  
& IL-1β  
↓ TLR4 pathway  
(Inflammation)  
↑ Intestinal  
Occludin & ZO-1  
↓ F/B ratio;  
↑ Akkermansia  
Mayulu, N.  
et al.  
Male Rattus  
norvegicus  
fed a CFED  
65 & 130  
mg/kg  
BW  
Glucose &  
6 weeks Metabolism:  
C. racemosa (Sulfated  
Polysaccharide - SPCr)  
Medium  
Risk  
(2023)  
↓ Blood Glucose  
Page 3704  
(High dose most  
effective)  
↓ Serum Lipase &  
Amylase Liver &  
Lipids: ↔ AST &  
ALT (No  
significant change)  
↓ HMG-CoA  
Reductase ↑ SOD  
Cardio  
Glucose &  
Metabolism:  
↓ Blood Glucose  
↓ Serum Amylase  
& Lipase  
Male albino  
Swiss mice  
fed a CFED  
65 & 130  
mg/kg  
BW  
Nurkolis et  
al. (2023)  
C. racemosa (Aqueous  
Liver & Lipids:  
↓ TG, TC, LDL;  
↑ HDL  
Medium  
Risk  
6 weeks  
extract - AEC)  
↑ Cardio SOD  
↓ PRMT-1 &  
ADMA  
(Cardiometabolic  
markers)  
Glucose:  
↓ Blood Glucose  
Liver & Lipids:  
Manoppo,  
J.I.C. et  
al. (2022)  
Male Rattus 150 & 450  
Medium  
Risk  
C. lentillifera (Extract)  
norvegicus  
mg/kg  
BW  
4 weeks  
↓ Total  
fed a CFED  
Cholesterol ↑  
Liver SOD  
(Antioxidant)  
↑ PGC-1α  
(Mitochondrial  
biogenesis)  
Glucose:  
Kuswari  
M. et al.  
(2021)  
Male Wistar 150 & 450  
↓ Blood Glucose  
(150 mg/kg > 450  
mg/kg)  
albino rats  
mg/kg  
BW  
C. racemosa (Extract)  
4 weeks  
High Risk  
fed a CFED  
Page 3705  
Liver & Lipids: ↓  
Total Cholesterol  
Glucose & Insulin:  
↓ Fasting Blood  
Glucose  
↑ Serum Insulin  
Liver/Kidney  
(Hepatoprotective)  
:
Male Wistar  
albino rats  
(Diabetic  
Nephropathy:  
high fructose  
+ STZ)  
C. racemosa  
↓ Serum TC, TG,  
LDL-C  
100 & 400  
mg/kg  
Cao, M. et  
al. (2021)  
Medium  
Risk  
8 weeks  
(Polysaccharide extract -  
PCR)  
BW  
↓ Renal MDA  
(Oxidative stress)  
↑ Renal  
Antioxidants  
(SOD, CAT, GSH-  
Px)  
↓ Renal Cytokines  
(IL-1β, IL-6, TNF-  
α)  
↓ Renal Fibrosis &  
Lesions  
Glucose: ↔ Basal  
Blood Glucose  
(No reduction) ↔  
Glucose Tolerance  
(No improvement)  
Male Wistar  
rats fed  
High-Carb  
High-Fat diet  
C. lentillifera  
Liver & Lipids:  
du Preez, R  
et al. (2020  
Medium  
Risk  
5% in diet 8 weeks  
(Dried, whole)  
↓ Liver Fat  
Deposition &  
Inflammation ↔  
Plasma ALT &  
AST (No change)  
↓ Total  
Cholesterol &  
NEFA  
Glucose & Insulin:  
↓ Fasting Blood  
Glucose ↑ Glucose  
& Insulin  
C57BL/KsJ-  
db/db mice  
(Genetic  
250 & 500  
mg/kg  
BW  
C.lentillifera  
Sharma et  
al. (2015)  
Medium  
Risk  
6 weeks  
(Ethanol extract - CLE)  
T2DM)  
Tolerance  
(OGTT/IPITT) ↓  
Page 3706  
HOMA-IR  
Liver &  
Metabolism: ↑  
Hepatic Glycogen  
↑ GK activity,  
↓ G-6Pase activity  
Table 3 Summary of Clinical (Human) Studies on The Metabolic Effects of Caulerpa Spp.  
Study  
(Author,  
Year)  
Caulerpa  
Species  
Study  
Design  
Participants  
(n)  
Key Quantitative  
Findings  
Risk of Bias  
Assessment  
Dose  
Duration  
↓ Blood  
69 obese  
men (BMI  
2530  
Glucose: Significantly  
lower (p<0.0001)  
compared to placebo  
RCT,  
double-  
blind,  
1.68  
g/day of  
sea  
grape  
extract  
or  
kg/m²)  
Permatasari  
et al. (2022)  
completed  
the study (35  
in extract  
group, 34 in  
placebo  
Some  
Concerns  
C. racemosa placebo-  
4 weeks  
↑ PGC-  
controlled  
clinical  
trial  
1α: Significantly  
higher (p=0.000)  
compared to placebo  
placebo  
group)  
Caulerpa spp. as a Potential Therapy for NAFLD and NASH  
Non-alcoholic fatty liver disease (NAFLD) progresses to non-alcoholic steatohepatitis (NASH) through a series  
of interrelated metabolic and inflammatory disturbances. Persistent insulin resistance promotes excessive  
delivery of free fatty acids to the liver and stimulates de novo lipogenesis via regulatory factors such as SREBP-  
1c, FAS, and ACC, resulting in hepatocellular triglyceride accumulation and lipotoxicity [16][17]. As lipid  
burden increases, mitochondrial β-oxidation becomes impaired, generating reactive oxygen species and lipid  
peroxidation products such as malondialdehyde. These oxidative insults induce hepatocyte injury and activate  
inflammatory cascades, including TLR4NF-κB signaling, which further amplify cytokine production and  
immune cell recruitment [16][19]. Concurrently, gut dysbiosis and disruption of intestinal barrier function  
increase endotoxin translocation into portal circulation, promoting hepatic inflammation, stellate-cell activation,  
and fibrogenesis through the gutliver axis [17][18]. Collectively, lipid overload, oxidative stress, inflammation,  
and microbiome-mediated hepatic injury constitute the major mechanisms driving progression from simple  
steatosis to NASH.  
Within this pathogenic framework, evidence from in vitro and in vivo studies demonstrates that species  
of Caulerpa exert hepatoprotective and antihyperglycemic effects through multiple biochemical  
pathways. Caulerpa lentillifera supplementation has been shown to restore endogenous antioxidant defense  
systems. For instance, in a diquat-induced zebrafish model, 50 g/kg supplementation restored the protective  
enzyme SOD activity (from ~38 U/mg to ~46 U/mg) while concurrently lowering the liver damage marker ALT  
(from ~29 U/mg to ~20 U/mg). In zebrafish and HepG2 hepatocyte models, C. lentillifera downregulated  
SREBF1, FAS, and ACC, and activated SIRT1AMPK signaling, indicating a metabolic shift that reduces  
lipogenesis and enhances fatty-acid oxidation [1][6]. These mechanisms directly counteract oxidative injury and  
metabolic stress, key pathological drivers of NAFLD progression.  
Comparable outcomes have been reported for Caulerpa racemosa. Dietary supplementation and sulfated  
polysaccharide fractions significantly reduced triglycerides, LDL cholesterol, and fasting glucose while  
Page 3707  
increasing HDL concentrations in cholesterol-fat enriched diet models [7][9]. Mechanistic analysis revealed  
modulation of the PRMT-1/DDAH/ADMA and mTORSIRT1AMPK pathways, further supporting its role in  
restoring metabolic and endothelial homeostasis [7][9]. These targets overlap with those of established metabolic  
therapies, suggesting that Caulerpa may offer pharmacologically relevant benefits derived from natural sources.  
A distinct advantage of C. lentillifera is its regulatory effect on the gutliver axis, a central component in the  
transition from NAFLD to NASH. Findings from ethanol-induced liver injury models reported three major  
protective mechanisms [5]:  
1. Anti-inflammatory regulation: C. lentillifera supplementation suppressed hepatic TLR4 signaling by  
downregulating TLR4, MyD88, and TRIF, which reduced the p-NF-κB/NF-κB ratio and lowered pro-  
inflammatory cytokines TNF-α and IL-1β.  
2. Barrier function: Supplementation reversed the ethanol-induced decline of tight-junction proteins,  
strengthening intestinal barrier integrity and significantly decreasing circulating endotoxin.  
3. Microbiota modulation: C. lentillifera normalized the Firmicutes-to-Bacteroidetes ratio and increased  
beneficial taxa such as Akkermansia, which correlated positively with occludin and ZO-1 expression.  
Additional studies involving C.taxifolia and C.prolifera demonstrate complementary antidiabetic and  
hepatoprotective effects. Sulfated galactans and phenolic constituents, including vanillin and kaempferol,  
inhibited α-amylase and α-glucosidase activity, improved insulin sensitivity, and reduced postprandial glucose  
[3][4]. Carotenoid- and peptide-rich fractions of Caulerpa also reduced body weight, hepatic lipid deposition,  
and blood glucose levels while enhancing microbiota diversity [2]. These combined antioxidant,  
antihyperglycemic, and microbiota-mediated effects align closely with the mechanistic requirements for slowing  
NAFLD progression and preventing transition to NASH.  
Overall, Caulerpa spp. intervene at multiple critical stages of NAFLD pathophysiology, including metabolic  
regulation, oxidative stress reduction, inflammatory suppression, and stabilization of the gut liver axis., the  
inhibition of digestive enzymes such as C. racemosa extract demonstrating 81.67% inhibition of α- glucosidase  
and 84.07% inhibition of α- amylase, indicates significant antidiabetic potential. Other studies found C.  
racemosa's ethyl acetate fraction effectively inhibited alpha-glucosidase with an IC50 value of 153.87 μg/ml,  
aligning with the therapeutic actions of standard antidiabetic drugs like acarbose.  
Integrative Hepatoprotective Mechanisms of Caulerpa spp. Against NAFLD/NASH  
Figure 2 Integrative Hepatoprotective Mechanisms  
Given the multi-target biological activities demonstrated by Caulerpa spp. across the reviewed literature, an  
integrative mechanistic pathway diagram is essential to improve conceptual clarity and visually synthesize the  
Page 3708  
interconnected hepatoprotective and antihyperglycemic mechanisms. Prior figures in the manuscript separately  
illustrate oxidative stress reduction, modulation of lipid metabolism, enzyme inhibition, and microbiota  
regulation; however, presenting these mechanisms collectively will provide a more cohesive visualization of  
how Caulerpa-derived compounds act simultaneously on multiple organ systems. This integrative figure is  
particularly relevant for NAFLD/NASH, which itself is a multi-hit metabolic disorder involving gut dysbiosis,  
oxidative stress, inflammation, and impaired lipid metabolism.  
The proposed schematic consolidates four major pathways consistently identified in in vitro, in vivo, and  
clinical studies:  
(1) inhibition of digestive enzymes (α-amylase and α-glucosidase),  
(2) modulation of gut microbiota and intestinal barrier integrity,  
(3) attenuation of oxidative stress, and  
(4) regulation of metabolic pathways via SIRT1AMPK activation and lipogenic gene suppression.  
The hepatoprotective mechanisms of Caulerpa spp. against Non-Alcoholic Fatty Liver Disease (NAFLD) and  
its inflammatory progression, Non-Alcoholic Steatohepatitis (NASH), are comprehensive, aligning with the  
hypothesis which posits that the disease arises from numerous, parallel insults [23]. Caulerpa exerts its  
protective effects not through a single target, but by simultaneously modulating four distinct, yet interconnected,  
pathways.  
First, as an upstream intervention, Caulerpa addresses the initial metabolic burden originating from the diet.  
Bioactive compounds, including phenolics and polysaccharides, have been shown to effectively inhibit key  
digestive enzymes, specifically -amylase and -glucosidase [24]. This action slows the breakdown of complex  
carbohydrates in the gastrointestinal tract, leading to a blunted postprandial glucose spike. The immediate  
consequence is a reduced demand for insulin, which in turn mitigates a primary driver for de novo lipogenesis  
(the creation of new fat) in the liver.  
Second, Caulerpa plays a crucial role in modulating the gut-liver axis, a critical communication highway that is  
often compromised in NAFLD [27]. Evidence from studies on C. lentillifera reveals a dual-action benefit: it  
enhances intestinal barrier integrity by upregulating the expression of tight-junction proteins like Occludin and  
ZO-1, and it concurrently fosters a healthier gut microbiome by enriching beneficial populations, such  
as Akkermansia [26]. This fortified barrier function significantly reduces the translocation of bacterial  
endotoxins (LPS) from the gut into the portal circulation. By preventing this "leaky gut"  
phenomenon, Caulerpa effectively diminishes the primary trigger for hepatic inflammation, dampening  
the TLR4/NF-B signaling cascade within liver cells.  
Third, Caulerpa provides direct hepatocellular protection by combating oxidative stress, a key factor that propels  
the progression from simple fatty liver (steatosis) to the more dangerous inflammatory state of NASH [25]. The  
seaweed's compounds bolster the liver's endogenous antioxidant defense system, demonstrably increasing the  
activity of crucial enzymes like Superoxide Dismutase (SOD), Catalase, and Glutathione. Simultaneously, it  
significantly lowers levels of malondialdehyde (MDA), a key marker of lipid peroxidation and cellular  
damage [22]. This powerful antioxidant effect shields hepatocytes from damage by Reactive Oxygen Species  
(ROS), preserving cellular integrity and reducing a major source of inflammation.  
Finally, at the core of liver metabolism, Caulerpa directly addresses the accumulation of fat by  
promoting metabolic regulation at the intracellular level. This is primarily achieved through the activation of  
the AMPK and SIRT1 signaling pathways, which act as master regulators of cellular energy homeostasis [28].  
This activation initiates a critical metabolic shift: it actively suppresses de novo lipogenesis by downregulating  
the key transcription factors and enzymes responsible for fat synthesis (e.g., SREBP-1c, FAS, ACC), while  
it simultaneously promotes fatty acid oxidation (the "burning" of fat for energy).  
Page 3709  
In conclusion, the therapeutic potential of Caulerpa spp. for NAFLD/NASH lies in this synergistic, multi-target  
approach. By combining upstream digestive enzyme inhibition, restoration of the gut-liver axis, potent  
antioxidant defense, and a direct reprogramming of hepatic lipid metabolism, these pathways converge. The  
unified outcome is a significant reduction in lipid accumulation (steatosis), decreased hepatic inflammation, and  
improved systemic insulin sensitivity, all of which contribute to robust protection against the progression of  
NAFLD to NASH [23].  
Limitation  
Although the existing preclinical evidence is promising, several significant limitations must be addressed before  
the clinical potential of Caulerpa can be realized. First, the clinical data gap is substantial; this review identified  
only one human clinical trial , which focused on obesity rather than diagnosed NAFLD or NASH patients.  
Second, pharmacokinetic and bioavailability (PK/PD) challenges were unexplored in the existing studies. Key  
bioactive compounds, such as sulfated polysaccharides, are large macromolecules generally known for poor oral  
bioavailability. Future studies must quantify the absorption of these compounds and their concentrations in liver  
tissue to validate that effective doses are systemically achievable. Third, the extreme dose heterogeneity in in  
vivo studies (ranging from 65 mg/kg to 50 g/kg diets) makes the determination of a human-equivalent dose  
(HED) nearly impossible. The high doses used in some animal models may not be practical or tolerable for  
chronic supplementation in humans. Finally, toxicological and long-term safety considerations are entirely  
unaddressed. While Caulerpa is edible, some species (particularly invasive strains) are known to accumulate  
heavy metals or produce toxic secondary metabolites like caulerpicin and caulerpenyne as defense mechanisms.  
The lack of formal long-term toxicology studies is a major barrier to regulatory approval as a nutraceutical.  
Therefore, extraction standardization, phytochemical profiling, and rigorous safety assessments are urgently  
required.  
Future Research  
To translate these promising preclinical findings into viable nutraceuticals or therapeutics, several key industrial  
and research challenges must be addressed.  
Extraction Standardization and Quality Control  
A primary challenge is the lack of standardization. As seen in the reviewed studies, extraction methods vary  
widely (e.g., different solvent macerations vs. Soxhlet), directly impacting the bioactive profile and potency.  
Commercial development demands the establishment of validated, standardized green extraction protocols.  
Furthermore, phytochemical fingerprinting (e.g., using HPLC or GC-MS) is essential to establish a consistent  
compound profile and ensure batch-to-batch quality.  
Scalability and Sustainability  
Reliance on wild harvesting of Caulerpa is neither sustainable nor scalable for mass production. Industrial  
scalability is entirely dependent on optimizing aquaculture techniques. Future research should focus on  
improving cultivation yields while rigorously monitoring for potential contaminant accumulation (like heavy  
metals) from seawater to ensure product safety.  
Formulation and Delivery  
Overcoming poor bioavailability (as discussed in Limitations) is critical for efficacy. Research must focus on  
advanced formulations. Technologies such as nanoencapsulation or biopolymer delivery systems are crucial for  
protecting bioactive compounds from degradation, enhancing solubility, and ensuring targeted delivery, thereby  
improving overall bioefficacy.  
Clinical Validation  
Ultimately, all these efforts must culminate in larger, well-designed, multi-center randomized controlled trials  
Page 3710  
(RCTs) in diagnosed NAFLD and NASH patient populations to validate safety, efficacy, and optimal dosage in  
the target population.  
Industrial Implications: Scalability, Standardization, and Sustainability  
The therapeutic potential of Caulerpa spp. for metabolic liver disease must also be evaluated through the lens of  
industrial feasibility. In order for Caulerpa-derived extracts to progress from preclinical studies to commercial  
nutraceutical or pharmaceutical products, several large-scale challenges including extraction standardization,  
production scalability, and sustainability must be systematically addressed. These considerations are essential to  
ensure consistency, safety, and economic viability for future clinical and industrial applications.  
Extraction Standardization and Quality Control  
Current findings reveal substantial heterogeneity in extraction methods across studies, resulting in wide variation  
in phytochemical profiles and bioactivity. Most research employs different solvents (water, ethanol, methanol,  
ethyl acetate), extraction times, or temperatures, leading to inconsistent concentrations of key bioactive  
constituents such as sulfated polysaccharides, carotenoids, peptides, and polyphenols. This variability poses a  
major barrier to reproducibility and regulatory validation. Industrial translation therefore requires the  
establishment of standardized extraction protocols based on Good Manufacturing Practices (GMP) and green  
extraction technologies, including supercritical CO₂ extraction, microwave-assisted extraction, or ultrasound-  
assisted extraction. These approaches improve yield, reduce solvent waste, and preserve thermolabile  
compounds, making them ideal for large-scale production. Robust quality control must also be integrated using  
analytical techniques such as HPLC, LCMS/MS, or GCMS to generate a stable phytochemical fingerprint that  
ensures batch-to-batch consistency. This step aligns with regulatory expectations for botanical drug  
development, where marker compounds must be identified and quantified to guarantee product reliability [21].  
Scalability and Aquaculture Requirements  
Large-scale production of Caulerpa cannot depend solely on wild harvesting, as this raises concerns about  
ecological disturbance, seasonality, and variability in nutrient composition. Moreover, wild Caulerpa may  
accumulate environmental contaminants such as heavy metals, pesticides, or microplastics, which compromise  
extract safety and require extensive purification. To achieve industrial scalability, commercial development must  
shift toward controlled aquaculture systems, including tank-based cultivation, integrated multi-trophic  
aquaculture (IMTA), or long-line marine farming. These approaches allow for standardized nutrient conditions,  
controlled light and salinity, and continuous monitoring to avoid contaminant accumulation. Aquaculture-based  
production also ensures stable biomass supply while reducing ecological pressures on coastal ecosystems.  
Studies in seaweed biotechnology demonstrate that optimized aquaculture can increase biomass yield by 40–  
60% while improving polysaccharide uniformity and reducing heavy-metal uptake [29] Such models are directly  
applicable to Caulerpa spp. and necessary for its future commercialization.  
Sustainability and Environmental Considerations  
Sustainable production is especially important because certain Caulerpa species (e.g., C. taxifolia) are known to  
behave as invasive organisms in non-native waters. Large-scale harvesting without proper ecological assessment  
may risk habitat disturbance or biodiversity loss. Therefore, environmental risk assessment and Life Cycle  
Assessment (LCA) must be incorporated into cultivation planning to ensure low carbon footprint, low nutrient  
discharge, and minimal ecological disturbance. Additionally, valorization of residual biomass such as converting  
spent seaweed pulp into animal feed, compost, or biodegradable packaging may enhance sustainability and  
economic value, aligning with circular bioeconomy principles recommended by United Nations Environment  
Programme [30].  
Formulation, Delivery, and Bioavailability Challenges  
Even with scalable biomass production, formulation challenges remain. Bioactive polysaccharides from  
Page 3711  
Caulerpa generally possess large molecular weights and limited gastrointestinal absorption. To improve oral  
bioavailability and therapeutic potency, advanced delivery systems including nanoencapsulation, liposomal  
carriers, or biopolymer-based hydrogels should be explored. These technologies can protect compounds from  
degradation, enhance solubility, and enable targeted hepatic delivery, improving overall clinical efficacy.  
Translational and Regulatory Pathways  
Finally, translational success will depend on rigorous toxicological evaluation, standardized dosage  
determination, and multi-center clinical trials. Regulatory agencies such as the FDA and EMA require  
comprehensive safety, pharmacokinetic, and manufacturing data before approving botanical therapeutics.  
Establishing standardized cultivation, extraction, and quality-control systems is therefore not only scientifically  
necessary but also essential for meeting regulatory benchmarks for future Caulerpa-based interventions.  
CONCLUSION  
This review demonstrates that Caulerpa spp. contains bioactive compounds with strong hepatoprotective and  
antihyperglycemic effects supported by in vitro, in vivo, and early clinical evidence. Through antioxidant  
activity, metabolic regulation, enzyme inhibition, and gutliver axis modulation, Caulerpa effectively reduces  
oxidative stress, lipid accumulation, and glucose dysregulation, key of NAFLD and NASH progression. While  
findings are consistently positive, clinical research remains limited and extract standardization is still required.  
Future clinical trials are essential to evaluate long-term safety, establish clinically relevant therapeutic dosages,  
and determine if these preclinical findings can be effectively translated into nutraceuticals for metabolic liver  
disease.  
Conflicts of Interest  
The Authors declare no potential conflict of interest concerning the contents, authorship, and/or publication of  
this article.  
Funding  
The authors declared that this study has received no financial support.  
Data Availability Statement  
The data supporting this paper is available in the cited references.  
Ethical Approvals  
Not applicable  
REFERENCES  
1. Lin, X., Liu, Z., Xiao, Y., Xie, X., Wang, Y., Li, H., Wang, R., Xie, X., Zhang, Y., Song, Y., & Hu, W.  
(2024). Metabolomics provides insights into the alleviating effect of dietary Caulerpa lentillifera on  
diquat-induced oxidative damage in zebrafish (Danio rerio) liver. Aquaculture, 584, 740630.  
doi:10.1016/j.aquaculture.2024.740630  
2. Kurniawan, R., Nurkolis, F., Bukhari, A., Syauki, A. Y., Bahar, B., Aman, A. M., & Taslim, N. A. (2025).  
Carotenoid and peptide supplementation from Caulerpa sp. (sea grapes) extract mitigate metabolic  
syndrome in cholesterol-enriched diet rats via modulation of gut microbiota. Diabetology & Metabolic  
Syndrome, 17, Article 333. doi:10.1186/s13098-025-01869-4  
3. Liu, S., Qin, L., Li, D., Lu, F., Liang, M., & Hao, J. (2025). A sulfated polysaccharide from the green  
alga Caulerpa taxifolia: Characteristics of its structure and anti-diabetic activity. Marine Drugs, 23(10),  
Article 374. doi:10.3390/md23100374  
4. Ouahabi, S., Daoudi, N. E., Chebaibi, M., Mssillou, I., Rahhou, I., Bnouham, M., Hammouti, B.,  
Fauconnier, M.-L., Ayerdi Gotor, A., Rhazi, L., & Ramdani, M. (2025). A comparative study of the  
Page 3712  
phytochemical composition, antioxidant properties, and in vitro anti-diabetic efficacy of different  
extracts of Caulerpa prolifera. Marine Drugs, 23(7), Article 259. doi:10.3390/md23070259  
5. Lin, K.-Y., Yang, H.-Y., Yang, S.-C., Chen, Y.-L., Watanabe, Y., & Chen, J.-R. (2023). Caulerpa  
lentillifera improves ethanol-induced liver injury and modulates the gut microbiota in rats. Current  
Research in Food Science, 7, 100546. doi:10.1016/j.crfs.2023.100546  
6. Sangpairoj, K., Pranweerapaiboon, K., Saengkhae, C., Meemon, K., Niamnont, N., Tamtin, M., Sobhon,  
P., Yisarakun, W., & Siangcham, T. (2024). Extracts of tropical green seaweed Caulerpa  
lentillifera reduce hepatic lipid accumulation by modulating lipid metabolism molecules in HepG2  
cells. Heliyon, 10(5), e27635. doi:10.1016/j.heliyon.2024.e27635  
7. Mayulu, N., Gunawan, W. B., Park, M. N., Chung, S., Suh, J. Y., Song, H., Kusuma, R. J., Taslim, N.  
A., Kurniawan, R., Kartawidjajaputra, F., Nurkolis, F., & Kim, B. (2023). Sulfated polysaccharide  
from Caulerpa racemosa attenuates the obesity-induced cardiometabolic syndrome via regulating the  
PRMT1-DDAH-ADMA  
with  
mTOR-SIRT1-AMPK  
pathways  
and  
gut  
microbiota  
modulation. Antioxidants, 12(8), Article 1555. doi:10.3390/antiox12081555  
8. Dissanayake, I. H., Bandaranayake, U., Keerthirathna, L. R., Manawadu, C., Silva, R. M., Mohamed, B.,  
Ali, R., & Peiris, D. C. (2022). Integration of in vitro and in silico analysis of Caulerpa racemosa against  
antioxidant, antidiabetic, and anticancer activities. Scientific Reports, 12, Article 20848.  
doi:10.1038/s41598-022-24021-y  
9. Nurkolis, F., Taslim, N. A., Subali, D., Kurniawan, R., Hardinsyah, H., Gunawan, W. B., Kusuma, R. J.,  
Yusuf, V. M., Pramono, A., Kang, S., Mayulu, N., Syauki, A. Y., Tallei, T. E., Tsopmo, A., & Kim, B.  
(2023). Dietary supplementation of Caulerpa racemosa ameliorates cardiometabolic syndrome via  
regulation of PRMT-1/DDAH/ADMA pathway and gut microbiome in mice. Nutrients, 15(4), Article  
909. doi:10.3390/nu15040909  
10. Manoppo, J. I. C., Kartawidjajaputra, F., Gunawan, W., Kurniawan, R., Nurkolis, F., Suharman, F., &  
Kim, B. (2022). Amelioration of obesity-related metabolic disorders via supplementation of Caulerpa  
lentillifera in rats fed with a high-fat and high-cholesterol diet. Frontiers in Nutrition, 9, Article 1010867.  
doi:10.3389/fnut.2022.1010867  
11. Permatasari, H. K., Kusuma, R. J., Nurkolis, F., Pratama, M. Y., Nugraha, A. S., Apriliana, H. E.,  
Wibowo, A. M., & Kim, B. (2022). Metabolomic assay, computational screening, and pharmacological  
evaluation of Caulerpa racemosa as an anti-obesity with anti-aging by altering lipid profile and  
peroxisome proliferator-activated receptor-γ coactivator 1-α levels. Frontiers in Nutrition, 9, Article  
939073. doi:10.3389/fnut.2022.939073  
12. Kuswari, M., Nurkolis, F., Mayulu, N., Kartawidjajaputra, F., Gunawan, W. B., Tallei, T. E., & Kim, B.  
(2021). Sea grapes extract improves blood glucose, total cholesterol, and PGC-1α in rats fed on  
cholesterol- and fat-enriched diet. F1000Research, 10, Article 718. doi:10.12688/f1000research.54952.2  
13. Cao, M., Li, Y., Famurewa, A. C., & Olatunji, O. J. (2021). Antidiabetic and nephroprotective effects of  
polysaccharide extract from the seaweed Caulerpa racemosa in high fructose-streptozotocin-induced  
diabetic nephropathy. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy, 14, 21212131.  
doi:10.2147/DMSO.S302748  
14. du Preez, R., Majzoub, M. E., Thomas, T., Panchal, S. K., & Brown, L. (2020). Caulerpa lentillifera (sea  
grapes) improves cardiovascular and metabolic health of rats with diet-induced metabolic  
syndrome. Metabolites, 10(12), Article 500. doi:10.3390/metabo10120500  
15. Sharma, B. R., Kim, H. J., & Rhyu, D. Y. (2015). Caulerpa lentillifera extract ameliorates insulin  
resistance and regulates glucose metabolism in C57BL/KsJ-db/db mice via PI3K/AKT signaling pathway  
in myocytes. Journal of Translational Medicine, 13, Article 62. doi:10.1186/s12967-015-0412-5  
16. Friedman, S. L., Neuschwander-Tetri, B. A., Rinella, M., & Sanyal, A. J. (2018). Mechanisms of NAFLD  
development  
and  
therapeutic  
strategies.  
Nature  
Medicine,  
24(7),  
908922.  
17. Cotter, T. G., & Rinella, M. (2020). NAFLD 2020: The state of the disease. Nature Reviews  
Gastroenterology & Hepatology, 17(4), 236250. https://doi.org/10.1038/s41575-019-0247-7 [  
18. Tilg, H., Effenberger, M., & Stadlmann, S. (2021). Gutliver axis in nonalcoholic fatty liver disease:  
Pathophysiological insights and therapeutic implications. Nature Reviews Gastroenterology &  
19. Buzzetti, E., Pinzani, M., & Tsochatzis, E. A. (2016). The multiple-hit pathogenesis of non-alcoholic  
Page 3713  
fatty  
liver  
disease  
(NAFLD).  
Metabolism,  
65(8),  
10381048.  
20. Page, M. J., Moher, D., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021).  
PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic  
21. U.S. Food and Drug Administration. (2016, December). Botanical Drug Development: Guidance for  
Industry.pdf  
22. Chen, J., Xu, Y., Wang, Y., Zhang, X., Li, Z., & Chen, J. (2022). Antioxidant and hepatoprotective effects  
of polysaccharides from Caulerpa lentillifera on nonalcoholic fatty liver disease in mice. Journal of  
Functional Foods, 90, 104975.  
23. Friedman, S. L., Neuschwander-Tetri, B. A., Rinella, M., & Sanyal, A. J. (2018). Mechanisms of NAFLD  
development and therapeutic strategies. Nature Medicine, 24(7), 908922.  
24. Kusuma, I. W., Hidayati, N., & Murwanti, R. (2020). -Amylase and -Glucosidase inhibitory activities  
of Caulerpa racemosa extracts. Pharmacognosy Journal, 12(4), 819824.  
25. Li, S., Tan, H. Y., Wang, N., Zhang, Z. J., Lao, L., Wong, C. W., & Feng, Y. (2015). The role of oxidative  
stress and antioxidants in nonalcoholic fatty liver disease. International Journal of Molecular  
Sciences, 16(11), 2738527405.  
26. Sall, F. N., Tbin, M., Yim, J., Ahn, G., Park, S. Y., Lee, K. W., & Lee, H. J. (2021). Caulerpa  
lentillifera extract improves intestinal barrier function and modulates gut microbiota in high-fat diet-fed  
mice. Marine Drugs, 19(12), 675.  
27. Tilg, H., & Adolph, T. E. (2020). Gut-liver axis: A 'GI' new home for microbes. Gut, 69(8), 13691370.  
28. Zhao, P., Wang, H., Wu, Y., Liu, X., & Zhang, Y. (2021). Natural compounds as activators of the  
AMPK/SIRT1 pathway for the treatment of non-alcoholic fatty liver disease. Current Medicinal  
Chemistry, 28(28), 57855807  
29. Buschmann, A. H., Camus, C., Infante, J., Neori, A., Israel, Á., Hernández-González, M. C., Pereda, S.  
V., Gomez-Pinchetti, J. L., Golberg, A., Tadmor-Shalev, N., & Critchley, A. T. (2017). Seaweed  
production: Overview of the global state of exploitation, farming and emerging research  
activity. European Journal of Phycology, 52(4), 391406  
30. United Nations Environment Assembly. (2022). Enhancing circular economy as a contribution to  
achieving sustainable consumption and production (Resolution 11). United Nations Environment  
Page 3714