Next Article in Journal
Association Between Adherence to the Mediterranean Diet and Metabolic Syndrome and Its Components Among Polish Postmenopausal Women: A Cross-Sectional Study
Previous Article in Journal
Influence of Gestational Age on the Level of Functional Peptides (Peptidome) in Breast Milk
Previous Article in Special Issue
Gender Differences in Ultra-Processed Food Consumption and Its Association with Obesity Among Korean Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Effects of Low-Calorie Sweeteners on Human Health

1
Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
2
Department of Emergency Medicine, Chi Mei Medical Center, Tainan 710, Taiwan
3
Department of Nursing, National Tainan Junior College of Nursing, Tainan 700, Taiwan
4
Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
5
Tong-Yuan Diabetes Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2025, 17(17), 2726; https://doi.org/10.3390/nu17172726
Submission received: 10 June 2025 / Revised: 18 August 2025 / Accepted: 19 August 2025 / Published: 22 August 2025

Abstract

Low-calorie sweeteners (LCS) are widely utilized as sugar substitutes due to their intense sweetness, thermal stability, and applicability in weight management and diabetic-friendly products. However, increasing evidence has raised concerns about their potential long-term effects on metabolic health, glucose regulation, cardiovascular function, carcinogenicity, and gut microbiota composition. This review systematically evaluates the pharmacokinetics, metabolic effects, and associated health outcomes of major LCS. Mechanistically, LCS exert effects via sweet taste receptor-mediated pathways, altering glucose absorption, insulin secretion, and intracellular signaling cascades. Additionally, LCS influence gut microbiota composition, with certain agents promoting dysbiosis and glucose intolerance. While some findings support the metabolic benefits of selected LCS, others underscore potential risks, necessitating cautious interpretation. In conclusion, while LCS offer viable alternatives to sugar, their health effects are context-dependent and may vary across different sweeteners and populations. Long-term, high-quality clinical trials are essential to elucidate their safety and efficacy.

1. Introduction

The global prevalence of diabetes has reached alarming levels. According to the International Diabetes Federation, approximately 589 million adults aged 20–79 were living with diabetes in 2025, and this number is projected to rise to 853 million by 2050 if no effective preventive measures are implemented. The economic burden associated with diabetes is substantial. In the United States alone, diabetes-related healthcare expenditures exceeded USD 412.9 billion in 2022, a significant increase from USD 327 billion in the previous year. On a global scale, the total annual cost of diabetes management is estimated to exceed USD 760 billion. In response to the rising prevalence of diabetes and obesity, the global low-calorie sweetener market is expanding rapidly. Valued at USD 10–33 billion in 2025, it is projected to grow at a compound annual growth rate (CAGR) of 4.7–6.0%, reaching USD 14–52 billion by 2030–2035. Sucralose leads the market, while natural sweeteners like stevia are gaining momentum. These trends underscore the importance of understanding the metabolic effects of low-calorie sweeteners.
Low-calorie sweeteners (LCS) have become common substitutes to sucrose in modern diets, offering sweetness with fewer or no calories. Among these, substances can generally be divided into non-caloric artificial sweeteners, such as sucralose, acesulfame potassium (AceK), aspartame, and natural low-calorie sweeteners, including stevioside, and sorbitol. These sweeteners have gained widespread use in various food and beverage products due to their unique properties, including intense sweetness, stability under heat, and suitability for diabetic or weight management diets. However, concerns about their long-term health effects, including the impacts on glucose metabolism, body weight control, cardiovascular health, and potential carcinogenicity, have sparked significant scientific and public interest. Thus, we review the characteristics, benefits, and potential health risks associated with LCS to provide a comprehensive understanding of their role in dietary choices and public health.
We conducted the literature search in accordance with the PRISMA 2020 guidelines; however, since this review primarily aims to provide a broad synthesis of key aspects of commonly used artificial sweeteners, certain adjustments were made to the standard process. A total of 200 records were identified from electronic databases and additional sources, e.g., PubMed, Embase, and Cochrane, with key words of sucralose, aspartame, acesulfame potassium (AceK), stevioside, erythritol, xylitol, sorbitol, maltitol, saccharin, blood glucose, body weight, lipid, and disease risks. After removing duplicates and screening titles and abstracts, 59 full-text articles were assessed for eligibility. We subsequently categorized the retrieved articles by different types of artificial sweeteners and discussed them accordingly. The selection process is detailed in the diagram (Figure 1).
We have organized the relevant studies on various low-calorie sweeteners (LCS) into structured summary tables (Table 1, Table 2, Table 3, Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9), using the categories Category, Measured parameter, Conclusion, Author, and Year [ref.], to provide a concise and comparative overview of the key findings related to each sweetener. The “Measured parameter” column has been standardized to indicate the primary outcome or focus of each study (e.g., glycemic responses, body weight, serum lipid profile, disease risks), which corresponds directly to the structure and content of the respective sub-sections in the main text.
Figure 1. Study searching.
Figure 1. Study searching.
Nutrients 17 02726 g001

1.1. Sucralose

Sucralose is a commonly used artificial sweetener that is 600-fold sweeter than sucrose and is stable under heat. It is found in various products, including tabletop sweeteners and baked goods. The acceptable daily intake (ADI) of sucralose is set at 5 mg per kilogram of body weight per day by regulatory agencies, such as the U.S. Food and Drug Administration (FDA) and the European Food Safety Authority (EFSA) [1]. Sucralose is poorly absorbed in the small intestine, with about 15–27% entering the bloodstream, while the majority (~85%) passes through the digestive tract unchanged [2]. The absorbed portion is excreted unchanged via the kidneys. In addition, high doses may influence gut microbiota, but normal consumption levels (within safety limits) have minimal impact [3,4]. As shown in Table 1, we summarized the possible effects of sucralose on human health.
In several fundamental experimental studies, sucralose has been shown to exert measurable effects on metabolic processes. In mice, it has been associated with increased hepatic steatosis when combined with a high-fat diet [5]. Similarly, in rats, high-dose sucralose administration (4 g/kg/day) resulted in elevated triglyceride, cholesterol, and low-density lipoprotein cholesterol (LDL-c) levels, accompanied by marked hepatic histopathological alterations, including hemorrhagic lesions, chromatin condensation and loss, hepatocellular vacuolation, and infiltration of inflammatory cells [6]. Furthermore, another animal study demonstrated that long-term exposure to sucralose in drinking water for 12 weeks at varying concentrations (0.27, 0.33, 0.40, and 0.47 g/L) induced weight gain, impaired glucose homeostasis, and elevated blood glucose levels, potentially mediated through the upregulation of intestinal sweet taste receptors and glucose transporters [7,8].
In clinical investigations, similar phenomena have also been documented (Table 1). Although the substitution of sugar with LCS has traditionally been considered supportive of weight control, several studies have indicated that sucralose may worsen certain health outcomes, particularly those related to glycemic regulation [9]. In a controlled trial involving 45 healthy adults and a smaller cohort of adolescents, participants consumed beverages sweetened with either sucralose alone (LCS group), sucrose (Sugar group), or a combination of sucralose and maltodextrin (Combo group) over a two-week period, corresponding to a total of seven drinks and an average daily sucralose intake of approximately 0.43 mg/kg. The results showed that sucralose consumption reduced insulin sensitivity and altered the gut microbiota, potentially contributing to impaired glycemic responses [8,10]. By contrast, another short-term study in 17 healthy adults (10 females and 7 males) reported that daily supplementation with 136 mg of sucralose (~20% of the ADI) for two weeks did not significantly affect glucose metabolism [11].
The above findings from human and animal studies suggest sucralose may impair insulin sensitivity, alter gut microbiota, and affect liver and lipid metabolism, particularly at high doses or with long-term use. Regarding cardiovascular diseases, a study was conducted on 103,388 healthy participants in the web-based NutriNet cohort. Three non-consecutive days of 24 h dietary records were randomly collected over a two week period for two years that examined the associations among aspartame, AceK, sucralose, and cardiovascular disease risk, and it was found that total artificial sweetener intake was associated with an increased cardiovascular risk [12]. These findings highlight the need for further research to evaluate its long-term safety in typical dietary settings.
Table 1. Sucralose-related human studies.
Table 1. Sucralose-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Meta-analysisBody weightPositive association with weight gainLaviada-Molina et al., 2020 [9]
RCTGlycemic responsesImpaired insulin sensitivity and dysregulated glycemic responsesDalenberg, J.R., et al., 2020 [10]
Case controlDisease risksIncreased cardiovascular riskDebras, C., et al., 2022 [12]
RCT, randomized controlled trial.

1.2. Acesulfame Potassium (AceK)

AceK is a commonly used calorie-free sweetener, and the ADI of AceK is 15 mg per kilogram of body weight per day, established by the U.S. FDA. Its high sweetness intensity, stability under heat, and low cost make it a popular choice for manufacturers, especially in dietary and sugar-free products. AceK is rapidly absorbed in the small intestine and enters the bloodstream by prototype within 30–60 min after consumption [13]. AceK is not metabolized by the liver or any other organs in the body and is rapidly filtered by the kidneys and excreted through urine. Studies showed that over 95% of ingested AceK is excreted unchanged within 24 h [14].
On the aspect of whether AceK impacts blood sugar levels, public health recommendations are conflicting. Rathaus et al. reported that chronic AceK consumption exerted no significant effects on body weight, fat mass, or glucose metabolism in mice supplemented with AceK in drinking water in combination with either standard chow or a high fat diet over a 20-week period [15]. In contrast, Lin et al. found that concurrent intake of AceK exacerbated high cholesterol diet-induced dyslipidemia and atherosclerotic lesion formation in ApoE−/− mice during an 8-week study, in which animals received AceK supplementation at 15 mg/kg/day, a dose equivalent to the human ADI [16]. Furthermore, both sucralose and AceK have been reported to induce cardiotoxic and neurobehavioral effects in Daphnia [17]. Of particular concern, AceK has also been implicated in malignancy. Kim et al. demonstrated that AceK inhibited autophagic degradation of programmed death-ligand 1 (PD-L1) in hepatocellular carcinoma, thereby conferring resistance to T cell-mediated cytotoxicity. This suggests that AceK may promote liver cancer progression, as evidenced by experiments in mouse (RIL-175) and human (SK-Hep1) HCC cell lines treated with 1 mM AceK for 24 h [18].
As shown in Table 2, we summarized the possible effects of AceK on human health. The STOP Sugars NOW (Strategies To Oppose Sugars with Non-nutritive sweeteners Or Water) Trial is a pragmatic, randomized, open-label, crossover clinical study designed to assess whether replacing sugar-sweetened beverages (SSBs) with non-nutritive sweetened beverages (NSBs) or water improves metabolic outcomes and alters the gut microbiome. A total of 80 adults who were overweight or obese who habitually consumed SSBs were enrolled and completed a 4-week intervention period—continuing SSBs, substituting with NSBs, or substituting with water—with washout intervals between each phase. The complete results of this trial have not yet been fully published and are currently being presented at scientific meetings or undergoing journal submission [19]. With respect to cardiovascular diseases, a recent cohort study reported that AceK was associated with an increased risk of coronary heart disease [12], and patients with symptomatic carotid atherosclerosis were found to have elevated plasma levels of AceK and saccharin [20].
Overall, while some studies show no metabolic effects, others link AceK to impaired glucose tolerance, increased cardiovascular risk, dyslipidemia, atherosclerosis, and possible cancer-promoting effects. Further researches are needed to clarify its long-term safety.
Table 2. AceK-related human studies.
Table 2. AceK-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
RCTGlycemic responsesInterference of oral glucose toleranceAyoub-Charette et al., 2023 [19]
Case controlDisease risksIncreased coronary heart disease riskDebras, C., et al., 2022 [12]
Case controlDisease risksIncrease the risk of symptomatic carotid atherosclerosisStø, K., et al., 2023 [20]
RCT, randomized controlled trial.

1.3. Aspartame

Aspartame is an artificial sweetener that is commonly used in sugar-free and “diet” food and beverages. It is approximately 200 times sweeter than sucrose [21]. The ADI of aspartame is 40 mg per kilogram of body weight per day per the EFSA and 50 mg per kilogram of body weight per day per the U.S. FDA. It was first discovered in 1965 by James M. Schlatter and has been the subject of numerous safety evaluations since its approval by the U.S. FDA in 1981. In contrast to sucralose and AceK, aspartame is metabolized in the body and broken down in the small intestine by digestive enzymes into phenylalanine, aspartic acid, and methanol.
Aspartame has long been a subject of criticism and controversy, particularly regarding its alleged associations with cancer and other adverse health outcomes. Owing to its low-calorie property, aspartame has been widely used for weight management and glycemic control. In animal and cell-based experiments, aspartame has been reported to alter gut microbial composition, including increases in Enterobacteriaceae and Clostridium leptum, thereby impairing glucose tolerance in rats administered 5–7 mg/kg/day for 8 weeks [22,23]. In response to growing concerns about aspartame safety, the Ramazzini Institute (RI) initiated a series of large-scale toxicological studies in 1997 to evaluate its potential carcinogenicity. In one study, Sprague-Dawley rats were exposed to estimated daily doses of 4–5000 mg/kg body weight until natural death (~159 weeks). A positive dose–response relationship was observed, with the highest incidence of malignancies occurring in animals exposed to the highest doses of aspartame [24,25,26,27]. Although the RI findings were controversial due to questions regarding the accuracy of histopathological diagnoses, a subsequent re-examination of hematopoietic and lymphoid tumors using immunohistochemistry and morphological reclassification confirmed that 92% of the lesions in aspartame-exposed animals were indeed malignant [28]. This reanalysis underscored the urgency of re-evaluating aspartame’s health risks, particularly those associated with prenatal exposure [29]. Overall, the available animal evidence indicates dose-related tumorigenicity and highlights the necessity of reassessing aspartame safety. Long-term studies remain essential to confirm its carcinogenic risk and clarify its safety profile.
As shown in Table 3, we summarized the potential effects of aspartame on human health. Several human studies have reported that aspartame provides no beneficial effects on the regulation of blood glucose homeostasis [30,31,32,33,34]. Similarly, a randomized controlled trial involving 100 healthy, lean adults who consumed aspartame at doses of 350–1050 mg/day for 12 weeks found no significant changes in body weight [32]. Investigations into the effects of non-nutritive sweeteners (NNS), including aspartame, on body weight also demonstrated no differences in weight change between NNS groups and placebo or water groups [9,35]. With regard to cardiovascular outcomes, aspartame intake has been linked to an increased risk of cerebrovascular disease [12,36]. The potential association between artificial sweeteners and cancer has also attracted growing attention. A large prospective cohort study of 102,865 French adults (2009–2021), with exposure assessed through repeated 24 h dietary records and quantitative estimates of artificial sweetener intake over a median follow-up of 7.8 years, reported that aspartame and AceK consumption were associated with an elevated cancer risk [37]. However, this association may be confounded by underlying characteristics of consumers—such as a higher prevalence of obesity, metabolic disorders, or specific dietary patterns—rather than indicating a direct carcinogenic effect of the sweeteners themselves.
Table 3. Aspartame-related human studies.
Table 3. Aspartame-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Case controlGlycemic responsesNo significant effectsBonnet, F., et al., 2018 [30]
CohortGlycemic responsesNo significant effectsBryant, C.E., et al., 2014 [31]
RCTGlycemic responsesNo significant effectsHiggins, K.A., R.V. Considine, and R.D. Mattes, 2018 [32]
RCTGlycemic responsesNo significant effectsOlalde-Mendoza, L. and Y.E. Moreno-González, 2013 [33]
RCTGlycemic responsesNo significant effectsTey, S.L., et al., 2017 [34]
Case controlDisease risksHigher risk of cerebrovascular diseaseDebras, C., et al., 2022 [12]
Case controlDisease risksHigher risk of cerebrovascular diseaseBernstein, A.M., et al., 2012 [36]
Case controlDisease risksIncreased cancer riskDebras, C., et al., 2022 [37]
Meta-analysisBody weightNo effect on body weight changes in the group of NNSRogers, P.J. and K.M. Appleton, 2021 [35]
RCT, randomized controlled trial; NNS, non-nutritive sweeteners.

1.4. Saccharin

Saccharin is one of the oldest artificial sweeteners, first discovered in 1879. The ADI of saccharin is 5 mg per kilogram of body weight per day by the U.S. FDA. Similar to sucralose and AceK, saccharin is rapidly absorbed in the small intestine after ingestion and excreted by prototype through the urine by the kidney, and more than 90% of absorbed saccharin is excreted within 24 h [38]. The small amount of saccharin that is not absorbed in the intestines is eliminated through feces.
An animal study demonstrated that long-term saccharin consumption did not significantly influence body weight, fat mass, or glucose metabolism, as compared with water intake [15]. However, saccharin has been the subject of considerable controversy due to early animal studies in rats, in which high dietary exposures—typically 5% saccharin in the diet (2–4 g/kg body weight/day)—were associated with potential cancer risk [39,40].
As shown in Table 4, we summarized the potential effects of saccharin on human health. Most studies have reported no significant association between saccharin consumption and blood glucose control [31,41]. Similarly, only limited effects on body weight reduction have been observed [15,35]. Substituting sugar with non-nutritive sweeteners, including saccharin, may represent the only favorable aspect of saccharin with respect to weight management [42]. In contrast, a randomized controlled trial with 12 weeks of saccharin intervention reported an incremental change in body weight [43]. Overall, these findings indicate limited metabolic benefits of saccharin. Regulatory authorities, including the U.S. FDA and the World Health Organization (WHO), have deemed saccharin safe for human consumption. Nevertheless, its health effects remain a matter of ongoing debate, and no global consensus has been established. Epidemiological evidence has suggested a significant association between saccharin intake and increased risks of mortality, particularly cardiovascular and cancer-related mortality, in diabetic and pre-diabetic populations [44]. Moreover, in a study including 15 patients with symptomatic carotid atherosclerosis, 18 patients with asymptomatic carotid atherosclerosis, and 15 healthy controls, elevated plasma saccharin levels (7.6 ± 6.8 ng/mL in symptomatic carotid atherosclerosis vs. 2.0 ± 2.2 ng/mL in healthy controls) were associated with greater carotid stenosis and higher weekly snack consumption, potentially implicating saccharin in the pathogenesis of cardiovascular disease [20,45]. Regarding malignancy, toxicological and epidemiological evidence has generally shown no association between saccharin or other NNS and overall cancer risk [46]. However, accumulating evidence from 12 observational studies (10 cohort, 2 case-control) involving 572,820 participants has indicated a potential association between saccharin intake and an increased risk of endometrial cancer [37,47,48]. Considering all the above, it can be inferred that possible associations with increased mortality and endometrial cancer warrant further investigation, especially in vulnerable populations.
Table 4. Saccharin-related human studies.
Table 4. Saccharin-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
CohortGlycemic responsesNo significant effectBryant, C.E., et al., 2014 [31]
RCTGlycemic responsesNo significant effectOrku, S.E., G. Suyen, and M. Bas, 2023 [41]
Meta-analysisBody weightNo significant effectRogers, P.J. and K.M. Appleton, 2021 [35]
RCT Body weightMay increase body weightHiggins, K.A. and R.D. Mattes, 2019 [43]
Case controlDisease risksIncrease the risk of mortality, particularly cardiovascular disease mortality and cancer mortalityGao, Y., et al., 2024 [44]
Case controlDisease risksAssociated with increased degree of carotid stenosisStø, K., et al., 2023 [20]
Systemic reviewDisease risksNo evidence of cancer risk associatedPavanello, S., et al., 2023 [46]
Meta-analysisDisease risksMay be associated with endometrial cancerLi, H., et al., 2024 [48]
RCT, randomized controlled trial.

1.5. Stevioside

Stevioside is derived from Stevia rebaudiana, native to South America where it has been used for its sweet properties for hundreds of years. It is a glycoside, 50–350 times sweeter than sucrose, chemically composed of steviol with attached glucose molecules. The ADI of steviol equivalents is 4 mg per kilogram of body weight per day per the EFSA and Joint FAO/WHO Expert Committee on Food Additives (JECFA), whereas stevioside is not permitted for use as a sweetener by the U.S. FDA. The ADI is expressed in steviol equivalents since stevioside and related compounds (such as rebaudioside A) are metabolized into steviol in human bodies [49]. Stevioside is not absorbed directly in the upper gastrointestinal tract; however, it is broken down into steviol by gut microbiota while it reaches the colon. The steviol, hydrolyzed from stevioside by gut bacteria, is absorbed into the bloodstream and transported to the liver. In the liver, steviol undergoes glucuronidation, forming steviol glucuronide, which is water-soluble and easier to be excreted in urine. The majority (90%) of steviol glucuronide is excreted unchanged via urine within 24–48 h, whereas a small portion of steviol is excreted in feces [50].
With regard to potential health effects, animal studies in rats have demonstrated that stevioside exerts hypolipidemic effects, characterized by reductions in cholesterol, triglycerides, and LDL-c, along with increases in HDL-c [51,52,53]. Furthermore, several in vitro studies have intriguingly suggested that stevioside possesses antitumor properties against bladder, colon, and breast cancer cells [54,55,56,57].
As shown in Table 5, we summarized the reported effects of stevioside in human studies. Stevioside appears to exert beneficial effects on glucose metabolism, particularly when used as a sugar substitute in younger, non-diabetic, and overweight/obese populations [58]. Several human studies have assessed this effect through single-meal evaluations, comparing reduced-sugar and calorie meals containing steviol glycosides with full-sugar, calorie-matched meals. In randomized controlled trials, one involving 31 healthy adults across three test days and another involving 12 patients with type 2 diabetes, significant reductions in postprandial blood glucose levels were observed when steviol glycosides were incorporated into reduced-sugar/calorie meals or administered as supplements in both healthy and diabetic individuals [59,60]. With respect to lipid regulation, a study in 20 women with elevated serum cholesterol demonstrated that daily consumption of 20 mL stevia extract (prepared from 3.3 g of stevia leaves) diluted in 200 mL water for one month significantly reduced cholesterol, triglyceride, and LDL-c levels, while increasing HDL-c [61]. Collectively, these findings suggested that stevioside may contribute to improved metabolic regulation. Overall, its favorable safety profile and natural origin highlight stevioside as a promising sugar substitute; however, further long-term clinical studies are warranted to confirm these benefits.
Table 5. Stevioside-related human studies.
Table 5. Stevioside-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Meta-analysisGlycemic responsesMay provide beneficial effects on glucose metabolismBai, X., et al., 2024 [58]
RCTGlycemic responsesSignificant decrease in postprandial blood glucose levelsAnton, S.D., et al., 2010 [59]
RCTGlycemic responsesSignificant decrease in postprandial blood glucose levelsGregersen, S., et al., 2004 [60]
RCT, randomized controlled trial.

1.6. Erythritol

Erythritol is a naturally occurring polyol (sugar alcohol) with the chemical formula C4H10O4. It occurs naturally in small amounts in various fruits (e.g., grapes, melons, and pears), mushrooms, and fermented foods (e.g., soy sauce, wine, and cheese). Industrially, it is produced by the fermentation of glucose using osmophilic yeasts, mainly Moniliella pollinis or Trichosporonoides megachiliensis [62]. Erythritol exhibits a relative sweetness of approximately 60% to 80% that of sucrose, providing a comparable organoleptic profile with reduced caloric load. After ingestion, erythritol is rapidly absorbed in the small intestine via passive diffusion due to its small molecular weight. Unlike other polyols (e.g., sorbitol or xylitol) that undergo partial fermentation in the colon, erythritol is poorly metabolized by gut microbiota. About 90% is excreted unchanged in urine within 24 h; only about 10% reaches the colon and may be fermented slightly with minimal gas production. According to EFSA, its energy value is estimated at approximately 0.2 kcal per gram, primarily because more than 90% of ingested erythritol is absorbed in the small intestine and subsequently excreted unchanged in the urine, resulting in minimal caloric contribution. Erythritol does not have an officially established ADI. Erythritol has been granted Generally Recognized as Safe (GRAS) status by both the U.S. FDA and EFSA, reflecting its established safety for use as an LCS in various food and beverage applications. EFSA’s Scientific Committee on Food (SCF) concluded that erythritol’s absorption and rapid excretion make it unlikely to pose toxicological concerns at reasonable levels of intake, so a numerical ADI was not deemed necessary [63].
In an animal study with rats, erythritol administration resulted in significant reductions in glucose intolerance, blood glucose, and hemoglobin A1c (HbA1c) levels [64]. Moreover, erythritol may be associated with an increased risk of major adverse cardiovascular events. Mechanistic investigations further showed that erythritol at physiological concentrations enhanced platelet reactivity in vitro—manifested by increased aggregation, intracellular Ca2+ flux, P-selectin expression, and GP IIb/IIIa activation—and promoted platelet adhesion and thrombosis formation in mice [65].
As shown in Table 6, we summarized the possible effects of erythritol on human health. Erythritol does not increase plasma glucose or insulin levels, making it suitable for diabetic populations [66,67,68]. Munro et al. conducted an interpretive review of clinical findings and concluded that repeated daily consumption of erythritol at doses up to ~1 g/kg body weight for several weeks does not significantly affect markers of glycemic control, including HbA1c, even among individuals with impaired glucose metabolism [63]. With regard to body weight, only a limited number of clinical trials have directly assessed erythritol’s effects [66]. In one small study, 12 healthy men consumed 1 g/kg/day of erythritol for 7 days, with no significant changes in body weight observed [69]. Another small trial involving seven patients with type II diabetes reported an average weight reduction of ~2 kg after daily intake of 20 g erythritol for 2 weeks; however, this finding did not reach statistical significance, as the effect was primarily driven by three individuals [70]. Concerning lipid metabolism, a randomized controlled trial in 21 healthy adults demonstrated that erythritol did not significantly alter the blood lipid profile, including total cholesterol, LDL-c, HDL-c, and triglycerides [71]. These studies suggested that erythritol is a well-tolerated LCS with minimal impact on glucose and lipid metabolism. Recently, however, concerns have been raised regarding its cardiovascular safety. An untargeted metabolomics study in patients undergoing cardiac risk assessment (discovery cohort, n = 1157) revealed that higher plasma erythritol levels were strongly associated with an increased 3-year risk of major adverse cardiovascular events. In a pilot human-intervention study (n = 8), ingestion of 30 g erythritol rapidly elevated plasma concentrations to more than 1000-fold above baseline, remaining above pro-thrombotic thresholds for over 48 h [65]. Thus, further research is warranted to evaluate its long-term safety in human populations.
Table 6. Erythritol-related human studies.
Table 6. Erythritol-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Cohort Glycemic responsesDoes not increase plasma glucose or insulin levelsNoda, K., K. Nakayama, and T. Oku, 1994 [67]
RCTGlycemic responsesDoes not increase plasma glucose or insulin levelsWölnerhanssen, B.K., et al., 2016 [68]
Cohort Body weight No significant changes in body weightTetzloff, W., et al., 1996 [69]
Cohort Body weight No significant changes in body weightIshikawa, M., et al., 1996 [70]
RCTSerum lipid profileNo significant effects on lipid profileTeysseire, F., et al., 2023 [71]
CohortDisease risksStrongly associated with a higher 3-year risk of major adverse cardiovascular eventsWitkowski, M., et al., 2023 [65]
RCT, randomized controlled trial.

1.7. Xylitol

Xylitol is a naturally occurring five-carbon sugar alcohol (a polyol) with the chemical formula C5H12O5. It is found in small amounts in various fruits and vegetables, such as plums, strawberries, and cauliflower, and is commercially produced by the hydrogenation of xylose derived from birch wood or corncobs. Xylitol is approximately as sweet as sucrose but provides about 40% fewer calories (~2.4 kcal/g) [72]. To date, no numerical ADI for xylitol has been established by JECFA or EFSA due to its generally recognized safety when used in normal dietary amounts. Nonetheless, doses exceeding 20–40 g/day are more likely to cause laxative effects; therefore, divided intake is recommended to improve gastrointestinal tolerance [73]. Approximately 50% of ingested xylitol is absorbed in the small intestine via passive diffusion. Absorbed xylitol undergoes hepatic metabolism, entering the pentose phosphate pathway where it is converted into glucose and glycogen. The remaining unabsorbed fraction is fermented by colonic microbiota, which may lead to gastrointestinal discomfort (e.g., bloating, flatulence, osmotic diarrhea) if consumed in excessive amounts [74]. Unlike sucrose, xylitol has minimal effects on postprandial glucose and insulin levels, making it suitable for individuals with impaired glucose tolerance or diabetes [75]. Although xylitol provides approximately 40% fewer calories than sucrose (2.4 kcal/g vs. 4 kcal/g), there is currently insufficient high-quality clinical evidence to support a significant impact of xylitol consumption on long-term body weight reduction. Theoretically, partial substitution of sucrose with xylitol can contribute to overall caloric reduction, potentially aiding in weight management when combined with other dietary and lifestyle interventions [74]. However, excessive intake may lead to gastrointestinal disturbances due to partial fermentation by colonic microbiota. Therefore, such laxative effects are not a safe or sustainable weight control strategy [76].
On the aspect of the influence on the blood lipid profile, human data are limited and inconsistent at present. Several animal studies suggested that xylitol may have favorable effects on lipid metabolism. An animal study showed that plasma total cholesterol levels were significantly lower when fed with high dose xylitol (1.5 g/kg body weight/day of dietary) [77]. On the other hand, another animal study revealed that no significant changes in hepatic or serum triglycerides and cholesterol were seen with low/medium (40–194 mg/kg body weight per day) xylitol in normal diet mice [78]. Also, elevated plasma xylitol accelerated arterial thrombus formation in mice [79]. In aggregate, xylitol is an LCS that exerts minimal impact on glucose metabolism and may confer beneficial effects on lipid profiles.
As shown in Table 7, we summarized the possible effects of xylitol on human health. Regulatory assessments by WHO and EFSA, based on long-term safety data, found no carcinogenicity or chronic disease risks, approving xylitol as safe. However, in a June 2024 study in the European Heart Journal (by Cleveland Clinic researchers) with two large human cohorts (n ≈ 3300), higher fasting plasma xylitol levels were significantly associated with an increased 3-year risk of major adverse cardiovascular events (MACE), including heart attack, stroke, or death. Human blood studies showed that physiological and postprandial levels of xylitol enhanced platelet reactivity, making blood more prone to clot [79]. The observed association between elevated plasma xylitol levels and heightened cardiovascular risk highlights the importance of further studies to clarify its long-term safety profile.
Table 7. Xylitol-related human studies.
Table 7. Xylitol-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
CohortGlycemic responsesMinimal effects on postprandial glucose and insulin levelsNatah, S.S., et al., 1997 [75]
CohortDisease risksAssociated with an increased 3-year risk of major adverse cardiovascular eventsWitkowski, M., et al., 2024 [79]

1.8. Maltitol

Maltitol is a hydrogenated disaccharide polyol (sugar alcohol) derived from maltose, with the chemical formula C12H24O11. It provides approximately 75–90% of the sweetness of sucrose, while delivering fewer calories (approximately 2.1–2.4 kcal/g, compared to 4 kcal/g for sucrose). Due to its similar sweetness profile and functional properties, maltitol is widely used as a sugar substitute in various sugar-free or reduced-calorie products, such as chocolates, baked goods, chewing gum, and confectionery [80]. About 40–50% of ingested maltitol is absorbed in the small intestine via passive diffusion, then metabolized primarily in the liver to glucose and sorbitol, and further to fructose and glucose-6-phosphate. The remaining 50–60% reaches the colon, where it undergoes fermentation by gut microbiota, producing short-chain fatty acids (SCFAs), hydrogen, and methane gases [74]. Maltitol is classified as GRAS by the U.S. FDA and approved by EFSA for use in a variety of food products. No numerical ADI has been set by JECFA due to its generally low toxicity, but excessive intake may lead to gastrointestinal discomfort. Doses exceeding 40–50 g/day in adults may cause osmotic diarrhea, bloating, or flatulence due to colonic fermentation [81].
As shown in Table 8, we summarized the possible effects of maltitol on human health. A study showed that following an acute oral dose of 50 g maltitol, the glycemic and insulinemic responses were significantly lower, as compared with an equivalent dose of glucose or sucrose. In the chronic phase, daily intake of maltitol (10 g three times daily for five consecutive days) produced no significant changes in blood glucose or insulin levels relative to the same dose of sucrose [82]. These findings confirmed that maltitol elicits a reduced postprandial glycemic and insulinemic response, making it suitable for individuals who require blood glucose management, such as people with diabetes. To date, no randomized controlled trial has examined maltitol in isolation to determine if it leads to weight loss. The evidence relies primarily on indirect inferences from studies on polyols as a group or sugar substitutes more broadly. Overall, the evidence indicated that LCS, including polyols such as maltitol, exert modest or neutral effects on body weight when they are used as true substitutes for caloric sugars rather than simply added to the diet [83]. Collectively, these findings indicated that maltitol elicits lower glycemic and insulinemic responses than sucrose, supporting its suitability for blood glucose management, although evidence for its effects on weight loss remains limited. Meanwhile, animal and human studies have provided no indication that maltitol possesses carcinogenic, mutagenic, or genotoxic properties. The main health issue with maltitol is gastrointestinal discomfort when consumed in high doses (typically >40–50 g/day for adults). Unabsorbed maltitol can be fermented in the colon, producing gas and osmotic effects, leading to bloating, flatulence, or mild laxative effects [81].
Table 8. Maltitol-related human studies.
Table 8. Maltitol-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Cohort Glycemic responsesSignificantly lower blood sugar when following an acute oral doseSecchi, A., et al., 1986 [82]
Meta-analysisBody weight Modest or neutral effects on body weight when they are used as true substitutes for caloric sugars rather than simply added to the dietMiller, P.E. and V. Perez, 2014 [83]

1.9. Sorbitol

Sorbitol is a versatile sugar alcohol used extensively in food, pharmaceuticals, and cosmetics. The ADI for sorbitol has also not been specifically established. Although there is no official ADI and sorbitol is GRAS, excessive consumption of sorbitol may cause gastrointestinal issues, including bloating, gas, and diarrhea. A study of 70 healthy young adults suggested that consuming over 20–50 g of similar sugar alcohol, including erythritol and xylitol, per day may lead to laxative effects [73,74]. EFSA regulated that foods containing more than 10% sorbitol must carry a warning label in Europe. Its ability to provide sweetness without contributing significantly to caloric (around 2.6 kcal/gram) intake makes it popular for sugar-free and diabetic-friendly products. Sorbitol is slowly absorbed in the small intestine via passive diffusion. Due to its poor absorption, a portion of sorbitol remains in the intestines, where it can be fermented by gut bacteria, leading to gas production or retaining water in the intestine, leading to a laxative effect. Once absorbed, sorbitol is primarily metabolized in the liver via the polyol pathway and converts into fructose by sorbitol dehydrogenase.
Sorbitol may contribute to weight management as an LCS substitute; however, human studies directly linking its use to long-term weight control remain limited. In animal models, sorbitol has been shown to inhibit intestinal glucose absorption, enhance glucose uptake in muscle tissue, delay gastric emptying, and reduce blood glucose levels in both normoglycemic and diabetic rats treated with 0.4 g/kg sorbitol in combination with 2 g/kg glucose [84]. These findings suggested that sorbitol has potential as an anti-hyperglycemic sweetener for individuals with diabetes. However, a study indicated that long-term sorbitol consumption may induce glucose intolerance in mice through alterations in the gut microbiota [85].
As shown in Table 9, we summarized the potential effects of sorbitol on human health. In a clinical study, daily intake of 10 g sorbitol for one month produced no significant changes in blood glucose levels [86]. Sorbitol is, however, slowly absorbed in the small intestine, and excessive consumption (≥20–50 g/day) may lead to gastrointestinal discomfort and laxative effects. In severe cases, gastrointestinal complications such as perforated colonic ulcers, bleeding, ischemic colitis, and colonic necrosis have been reported [87]. Collectively, these findings suggested that sorbitol may provide glycemic benefits; nevertheless, the limited human evidence and the gastrointestinal risks associated with high doses warrant cautious use. Further investigations are required to elucidate its long-term effects on glucose metabolism and microbial composition.
Table 9. Sorbitol-related human studies.
Table 9. Sorbitol-related human studies.
CategoryMeasured ParameterConclusionAuthor, Year [Ref.]
Cohort Glycemic responsesNo significant effects on blood glucose levelsEllis, F.W., and JC, Jr Krantz., 1941 [86]

2. Possible Mechanisms of LCS in the Regulation of Metabolic Homeostasis

2.1. Receptor-Mediated Pathway

LCS are ligands of the sweet taste receptor. The sweet taste receptors (STRs), including taste receptor type 1 member 2 (TAS1R2) and taste receptor type 1 member 3 (TAS1R3), are G-protein-coupled receptors (GPCRs) that detect sweet stimuli on the tongue bud and in the gut, pancreas, and brain. STRs are accompanied by G-proteins, such as gustducin (Gt), Gs, and Gi/o, which regulate different metabolic processes [88]. Also, the STR plays a significant role in carbohydrate metabolism. Typically, the regulation of carbohydrates in the body relies on the pancreas detecting blood glucose levels, transporting and oxidizing glucose, and producing adenosine triphosphate, which subsequently triggers insulin release to maintain glycemic balance. However, the STR also contributes to several carbohydrate-related processes, including pre-absorptive cephalic phase insulin release [89], regulation of glucagon-like peptide 1 (GLP-1) secretion by enteroendocrine cells [90,91], upregulation and translocation of sodium-glucose cotransporter (SGLT) and glucose transporter 2 (GLUT2) in the intestine [92,93,94,95], and direct stimulation of insulin secretion from the pancreas [96,97]. Sweeteners interfere with glucose metabolism through their interaction with STR and downstream signaling pathways in gut enteroendocrine cells and pancreatic β-cells.
Activation of TAS1R2/TAS1R3 leads to upregulation of SGLT1 on gut enteroendocrine cells, further increasing glucose absorption. However, not all sweeteners activate the sweet taste receptor on enteroendocrine cells (EECs) [98]. When LCS bind to the STR, Gα-gustducin is then coupled, leading to increased intracellular cyclic adenosine monophosphate (cAMP) and the activation of phospholipase C [99]. This further results in the release of calcium (Ca2+) from intracellular stores to trigger downstream effects, including SGLT-1 upgrade expression; the promotion of GLUT2 translocation to the apical membrane, further increasing glucose uptake; and an increase in GLP-1 secretion and disruption of normal glucose-dependent insulinotropic polypeptide (GIP) signaling, potentially desensitizing insulin responses. TAS1R2/TAS1R3 is also present in pancreatic β-cells, and activation by sweeteners can enhance insulin secretion [90].
Oxidative stress is a key factor linking metabolic disorders such as obesity, diabetes, and hypertension. Reactive oxygen species (ROS) disrupt the insulin-signaling pathway to impair glucose uptake, leading to insulin resistance. In a normal insulin-signaling pathway, when insulin binds to the insulin receptor (IR), it triggers the autophosphorylation of IR and further phosphorylates insulin receptor substrate (IRS) at tyrosine residues. IRS-1 and IRS-2 activate phosphoinositide 3-kinase (PI3K) to recruit Akt, which plays a major role in glucose metabolism. ROS promote oxidation of cysteine residues in IR to change its conformation and impair the autophosphorylation of IR. As a result, IRS proteins are not effectively activated, and Akt phosphorylation is reduced, leading to reduced GLUT4 translocation consequently. This results in decreased glucose uptake into cells, leading to hyperglycemia [100].
An animal study revealed that repeated stimulation of STRs by sucralose causes increases in ROS [101], indicating the increase in ROS generation might participate in LCS-induced adverse effects. In addition, sucralose promotes ROS accumulation and enhances adipogenesis in human adipose tissue-derived mesenchymal stromal cells [101]. Also, the increment in ROS might further activate extracellular-regulated protein kinases (ERK1/2) pathways to disrupt insulin signaling [102]. These effects may contribute to insulin resistance and metabolic dysregulation with long-term use.
STRs in the brain and gut influence metabolic responses by modulating neural signals to the hypothalamus. AceK and sucralose activate brain reward pathways, potentially leading to increased food intake and altered glucose metabolism [103,104,105]. In contrast, steviol and steviol glucuronide exhibit anti-hyperglycemic effects by activating glucose-induced insulin secretion to enhance pancreatic β-cell function [106]. They may be beneficial for diabetes management in the future. Moreover, in addition to STRs, a previous study indicated that stevioside lowers plasma glucose via the opioid mu opioid receptor in animals in a dose-dependent manner [107].
Sorbitol is slowly converted into fructose and glucose in the liver and pancreatic cells via the sorbitol–fructose metabolic pathway. Since fructose can enhance insulin secretion via STRs [97], sorbitol may have an indirect effect once metabolized into fructose. However, unlike fructose, sorbitol does not rapidly stimulate insulin secretion due to its slower metabolism.

2.2. LCS Alters Gut Microbiome

The gut microbiome is a critical modulator of host metabolism and T2DM risk. Emerging evidence indicates that alterations in the gut microbial composition—particularly a reduced Bacteroidetes-to-Firmicutes (B/F) ratio—are associated with increased energy harvest, metabolic dysregulation, chronic low-grade inflammation, and insulin resistance, all of which contribute to the development of T2DM. Key microbial metabolites, such as SCFAs, exert protective effects on gut barrier integrity and immune modulation, whereas elevated levels of branched-chain amino acids (BCAAs) are implicated in impaired insulin signaling. Experimental and clinical studies further demonstrate that modulation of the microbiome through dietary interventions, bariatric surgery, microbial transplantation, or pharmacological agents such as metformin can ameliorate metabolic dysfunction [108]. These findings underscore the significant impact of the gut microbiome on human health. However, recent studies have indicated that LCS may alter the composition and function of the gut microbiome, thereby potentially influencing host health.
Sucralose is poorly absorbed, and 85% reaches the colon unchanged, where it can interact with gut bacteria. However, studies revealed contradictory results. A randomized clinical trial (7 days) found no effect on gut microbiota and body weight; glycemic control and insulin resistance were not affected as well [4]. Another study (10 weeks) found minor reductions in Lactobacillus acidophilus and an increase in Blautia coccoides. Elevated insulin levels and increased glucose response (higher area under the curve for glucose during OGTT) was noted after sucralose intake. This suggests that sucralose may contribute to insulin resistance over time according to the study [109]. A recent study found that consumption of sucralose for two weeks significantly altered gut microbiota and increased glycemic response. Functional pathway changes were also noted in the study. Sucralose increased tricarboxylic acid cycle activity and reduced purine metabolism, which correlated with higher glycemic responses [45]. In addition, activation of TAS1R3 by sucralose promotes gut inflammation by activating mammalian target of rapamycin (mTOR) and suppressing peroxisome proliferator-activated receptor (PPAR)-γ, leading to increased inflammatory cytokines and intestinal barrier dysfunction. Wild-type mice had an overgrowth of inflammatory bacteria, including Enterobacteriaceae (associated with gut dysbiosis and inflammatory bowel disease), Paraprevotellaceae, and Prevotella (linked to gut inflammation). In contrast, Tas1r3 knockout mice had a significant increase in beneficial, butyrate-producing bacteria, such as Lachnospiraceae, Butyrivibrio, Roseburia, Faecalibacterium, and Butyricicoccus. Since butyrate is crucial for maintaining gut health and reducing inflammation, activation of TAS1R3 may result in dysbiosis of gut microbiota [110].
High doses of sucralose alter the structure of the T cell membrane, reducing membrane fluidity to impair T cell responses, particularly in CD8+ T cells. These changes make it harder for T cells to form effective immune synapses. In addition, T cells rely on calcium signaling for activation, and sucralose-treated T cells show reduced intracellular calcium release, leading to weakened immune activation [111]. In summary, exposure to sucralose may reduce the efficiency of T cell receptor signaling, which is essential for T cell activation and further results in gut microbiota dysbiosis. AceK is rapidly absorbed and excreted unchanged in urine; however, a human study found no significant differences in gut microbiota between AceK consumers and non-consumers [112].
Aspartame is broken down in the small intestine; therefore, aspartame itself might not be directly exposed to the gut microbiota in its intact form, whereas its metabolites may influence gut bacteria and metabolism; however, clinical studies revealed mixed results. A controlled trial found no effects on gut microbiota or metabolic markers after 2 weeks of aspartame consumption [113], whereas another trial showed that aspartame decreases the abundance of Porphyromonas and Prevotella nanceiensis in the oral microbiota.
Saccharin is partially absorbed, with some remaining in the gut, where it interacts with microbiota. A small human study (6 days, 5 mg/kg/day) demonstrated that four out of seven participants developed glucose intolerance, and their gut microbiota changed significantly, increasing Bacteroides fragilis and Weissella cibaria and decreasing Candidatus Arthromitus. Fecal microbiota transplantation from these human participants to germ-free mice resulted in similar glucose intolerance [114]. A recent study showed a significant increase in Prevotella copri in the saccharin group, accompanied with higher glycemic responses. Functional pathway changes with an increased uridine monophosphate (UMP) biosynthesis pathway were also noted. The clinical outcome of the study revealed that saccharin raised a glycemic response compared to glucose (p = 0.042) and no supplement control groups (p = 0.018) [45].
There is limited research on the effects of stevioside on human gut microbiota. A recent 12-week human study found that regular stevia consumption did not significantly alter the composition of the gut microbiota [115]. Sorbitol has notable effects on the human gut microbiota. A study indicated that extended sorbitol consumption could decrease the relative abundances of beneficial bacteria such as Bifidobacterium and certain members of the Lachnospiraceae family, while increasing the presence of genera like Helicobacter and Prevotella. These microbial shifts were associated with the development of glucose intolerance in the study subjects [85].

3. Conclusions

LCS are widely used as sugar alternatives in food and beverages, providing sweetness without the caloric burden of sucrose. However, while LCS serve as useful sugar substitutes, their long-term health effects remain a subject of debate (Table 10). While some sweeteners, such as stevioside, may provide health benefits, others, particularly certain artificial sweeteners, raise concerns regarding metabolic disturbances, cardiovascular risk, possible associations with cancer, and gut microbiota changes. More long-term, large-scale clinical studies are needed to fully understand their implications for human health. Until then, moderation remains key, and consumers should make informed choices based on their health needs and dietary preferences.
Table 10. Comparative summary of each LCS.
Table 10. Comparative summary of each LCS.
Low-Calorie SweetenersCurrent ConsumptionSummary
SucraloseMost frequentlyMay impair insulin sensitivity, alter gut microbiota, and affect liver and lipid metabolism
AceKMost frequentlyMay associate with impaired glucose tolerance, cardiovascular risk, and possible cancer-promoting effects
AspartameDeclining in new products but still widely presentMay impair glucose tolerance, alter gut microbiota, and increas risks of cardiovascular disease and cancer
SaccharinLessLimited metabolic benefits but rising concerns about its potential effects on cardiovascular health and cancer risk
SteviosideRisingPotential metabolic benefits, including improved glycemic control and lipid regulation, with preliminary evidence suggesting antitumor properties
ErythritolRisingMinimal impact on glucose and lipid metabolism, but may increase cardiovascular risk
XylitolCommonly consumed in daily life, particularly in chewing gum and toothpasteMinimal effects on glucose metabolism, potential lipid benefits, but may increase cardiovascular risk
MaltitolWidely present in food formulations but lacking specific statistical dataReduced glycemic effects, no known toxicological risks, but high doses may cause gastrointestinal discomfort
SorbitolWidely present in food formulations but lacking specific statistical dataPotential glycemic benefits, but limited human data and gastrointestinal risks at high doses

Funding

This study was funded by the Ministry of Science and Technology (MOST 114-2314-B-006-095-MY3) and National Cheng Kung University Hospital (NCKUH-11404023, NCKUH-11404021, NCKUH-11403038, NCKUH-11403037 and NCKUH-11404071).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Evaluation of Certain Food Additives and Contaminants. Thirty-Seventh Report of the Joint FAO/WHO Expert Committee on Food Additives; World Health Organization Technical Report Series 806; World Health Organization: Geneva, Switzelrand, 1991; pp. 1–52.
  2. Grotz, V.L.; Munro, I.C. An overview of the safety of sucralose. Regul. Toxicol. Pharmacol. 2009, 55, 1–5. [Google Scholar] [CrossRef]
  3. Ruiz-Ojeda, F.J.; Plaza-Díaz, J.; Sáez-Lara, M.J.; Gil, A. Effects of Sweeteners on the Gut Microbiota: A Review of Experimental Studies and Clinical Trials. Adv. Nutr. 2019, 10 (Suppl. S1), S31–S48. [Google Scholar] [CrossRef]
  4. Thomson, P.; Santibañez, R.; Aguirre, C.; Galgani, J.E.; Garrido, D. Short-term impact of sucralose consumption on the metabolic response and gut microbiome of healthy adults. Br. J. Nutr. 2019, 122, 856–862. [Google Scholar] [CrossRef]
  5. Wu, H.T.; Lin, C.H.; Pai, H.L.; Chen, Y.C.; Cheng, K.P.; Kuo, H.Y.; Li, C.H.; Ou, H.Y. Sucralose, a Non-nutritive Artificial Sweetener Exacerbates High Fat Diet-Induced Hepatic Steatosis Through Taste Receptor Type 1 Member 3. Front. Nutr. 2022, 9, 823723. [Google Scholar] [CrossRef]
  6. Khamise, N.A.; Tayel, D.I.; Helmy, M.W.; Aborhyem, S. Effect of Aspartame and Sucralose Artificial Sweeteners on Weight and Lipid Profile of Male Albino Rats. J. High. Inst. Public Health 2020, 50, 87–100. [Google Scholar] [CrossRef]
  7. Shi, Q.; Xu, L.; Cai, L.; Deng, S.; Qi, X. Sucralose regulates postprandial blood glucose in mice through intestinal sweet taste receptors Tas1r2/Tas1r3. J. Sci. Food Agric. 2024, 104, 2233–2244. [Google Scholar] [CrossRef]
  8. Singh, S.A.; Singh, S.; Begum, R.F.; Vijayan, S.; Vellapandian, C. Unveiling the profound influence of sucralose on metabolism and its role in shaping obesity trends. Front. Nutr. 2024, 11, 1387646. [Google Scholar] [CrossRef] [PubMed]
  9. Laviada-Molina, H.; Molina-Segui, F.; Pérez-Gaxiola, G.; Cuello-García, C.; Arjona-Villicaña, R.; Espinosa-Marrón, A.; Martinez-Portilla, R.J. Effects of nonnutritive sweeteners on body weight and BMI in diverse clinical contexts: Systematic review and meta-analysis. Obes. Rev. 2020, 21, e13020. [Google Scholar] [CrossRef]
  10. Dalenberg, J.R.; Patel, B.P.; Denis, R.; Veldhuizen, M.G.; Nakamura, Y.; Vinke, P.C.; Luquet, S.; Small, D.M. Short-Term Consumption of Sucralose with, but Not without, Carbohydrate Impairs Neural and Metabolic Sensitivity to Sugar in Humans. Cell Metab. 2020, 31, 493–502.e7. [Google Scholar] [CrossRef]
  11. Ahmad, S.Y.; Friel, J.K.; MacKay, D.S. The effect of the artificial sweeteners on glucose metabolism in healthy adults: A randomized, double-blinded, crossover clinical trial. Appl. Physiol. Nutr. Metab. 2020, 45, 606–612. [Google Scholar] [CrossRef] [PubMed]
  12. Debras, C.; Chazelas, E.; Sellem, L.; Porcher, R.; Druesne-Pecollo, N.; Esseddik, Y.; de Edelenyi, F.S.; Agaësse, C.; De Sa, A.; Lutchia, R.; et al. Artificial sweeteners and risk of cardiovascular diseases: Results from the prospective NutriNet-Santé cohort. BMJ 2022, 378, e071204. [Google Scholar] [CrossRef]
  13. Renwick, A.G. The metabolism of intense sweeteners. Xenobiotica 1986, 16, 1057–1071. [Google Scholar] [CrossRef]
  14. Schiffman, S.S. Rationale for Further Medical and Health Research on High-Potency Sweeteners. Chem. Senses 2012, 37, 671–679. [Google Scholar] [CrossRef]
  15. Rathaus, M.; Azem, L.; Livne, R.; Ron, S.; Ron, I.; Hadar, R.; Efroni, G.; Amir, A.; Braun, T.; Haberman, Y.; et al. Long-term metabolic effects of non-nutritive sweeteners. Mol. Metab. 2024, 88, 101985. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, C.H.; Li, H.Y.; Wang, S.H.; Chen, Y.H.; Chen, Y.C.; Wu, H.T. Consumption of Non-Nutritive Sweetener, Acesulfame Potassium Exacerbates Atherosclerosis through Dysregulation of Lipid Metabolism in ApoE−/− Mice. Nutrients 2021, 13, 3984. [Google Scholar] [CrossRef]
  17. Wiklund, A.E.; Guo, X.; Gorokhova, E. Cardiotoxic and neurobehavioral effects of sucralose and acesulfame in Daphnia: Toward understanding ecological impacts of artificial sweeteners. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2023, 273, 109733. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, D.H.; Kwon, E.J.; Park, K.G.; Jin, J.; Byun, J.K. Acesulfame potassium upregulates PD-L1 in HCC cells by attenuating autophagic degradation. Biochem. Biophys. Res. Commun. 2024, 711, 149921. [Google Scholar] [CrossRef]
  19. Ayoub-Charette, S.; McGlynn, N.D.; Lee, D.; Khan, T.A.; Blanco Mejia, S.; Chiavaroli, L.; Kavanagh, M.E.; Seider, M.; Taibi, A.; Chen, C.T.; et al. Rationale, Design and Participants Baseline Characteristics of a Crossover Randomized Controlled Trial of the Effect of Replacing SSBs with NSBs versus Water on Glucose Tolerance, Gut Microbiome and Cardiometabolic Risk in Overweight or Obese Adult SSB Consumer: Strategies to Oppose SUGARS with Non-Nutritive Sweeteners or Water (STOP Sugars NOW) Trial and Ectopic Fat Sub-Study. Nutrients 2023, 15, 1238. [Google Scholar] [CrossRef]
  20. Stø, K.; Skagen, K.R.; Valeur, J.; Yang, K.; Bjerkeli, V.; Aukrust, P.; Skjelland, M.; Halvorsen, B. Increased plasma levels of non-sugar sweeteners in patients with symptomatic carotid atherosclerosis. Scand. Cardiovasc. J. 2023, 57, 2205068. [Google Scholar] [CrossRef]
  21. Czarnecka, K.; Pilarz, A.; Rogut, A.; Maj, P.; Szymańska, J.; Olejnik, Ł.; Szymański, P. Aspartame-True or False? Narrative Review of Safety Analysis of General Use in Products. Nutrients 2021, 13, 1957. [Google Scholar] [CrossRef] [PubMed]
  22. Palmnäs, M.S.; Cowan, T.E.; Bomhof, M.R.; Su, J.; Reimer, R.A.; Vogel, H.J.; Hittel, D.S.; Shearer, J. Low-dose aspartame consumption differentially affects gut microbiota-host metabolic interactions in the diet-induced obese rat. PLoS ONE 2014, 9, e109841. [Google Scholar] [CrossRef]
  23. Nettleton, J.E.; Cho, N.A.; Klancic, T.; Nicolucci, A.C.; Shearer, J.; Borgland, S.L.; Johnston, L.A.; Ramay, H.R.; Noye Tuplin, E.; Chleilat, F.; et al. Maternal low-dose aspartame and stevia consumption with an obesogenic diet alters metabolism, gut microbiota and mesolimbic reward system in rat dams and their offspring. Gut 2020, 69, 1807–1817. [Google Scholar] [CrossRef]
  24. Soffritti, M.; Belpoggi, F.; Degli Esposti, D.; Lambertini, L.; Tibaldi, E.; Rigano, A. First experimental demonstration of the multipotential carcinogenic effects of aspartame administered in the feed to Sprague-Dawley rats. Environ. Health Perspect. 2006, 114, 379–385. [Google Scholar] [CrossRef] [PubMed]
  25. Soffritti, M.; Belpoggi, F.; Tibaldi, E.; Esposti, D.D.; Lauriola, M. Life-span exposure to low doses of aspartame beginning during prenatal life increases cancer effects in rats. Environ. Health Perspect. 2007, 115, 1293–1297. [Google Scholar] [CrossRef]
  26. Soffritti, M.; Belpoggi, F.; Manservigi, M.; Tibaldi, E.; Lauriola, M.; Falcioni, L.; Bua, L. Aspartame administered in feed, beginning prenatally through life span, induces cancers of the liver and lung in male Swiss mice. Am. J. Ind. Med. 2010, 53, 1197–1206. [Google Scholar] [CrossRef] [PubMed]
  27. Belpoggi, F.; Soffritti, M.; Padovani, M.; Degli Esposti, D.; Lauriola, M.; Minardi, F. Results of long-term carcinogenicity bioassay on Sprague-Dawley rats exposed to aspartame administered in feed. Ann. N. Y. Acad. Sci. 2006, 1076, 559–577. [Google Scholar] [CrossRef]
  28. Tibaldi, E.; Gnudi, F.; Panzacchi, S.; Mandrioli, D.; Vornoli, A.; Manservigi, M.; Sgargi, D.; Falcioni, L.; Bua, L.; Belpoggi, F. Identification of aspartame-induced haematopoietic and lymphoid tumours in rats after lifetime treatment. Acta Histochem. 2020, 122, 151548. [Google Scholar] [CrossRef]
  29. Landrigan, P.J.; Straif, K. Aspartame and cancer—New evidence for causation. Environ. Health 2021, 20, 42. [Google Scholar] [CrossRef] [PubMed]
  30. Bonnet, F.; Tavenard, A.; Esvan, M.; Laviolle, B.; Viltard, M.; Lepicard, E.M.; Lainé, F. Consumption of a Carbonated Beverage with High-Intensity Sweeteners Has No Effect on Insulin Sensitivity and Secretion in Nondiabetic Adults. J. Nutr. 2018, 148, 1293–1299. [Google Scholar] [CrossRef]
  31. Bryant, C.E.; Wasse, L.K.; Astbury, N.; Nandra, G.; McLaughlin, J.T. Non-nutritive sweeteners: No class effect on the glycaemic or appetite responses to ingested glucose. Eur. J. Clin. Nutr. 2014, 68, 629–631. [Google Scholar] [CrossRef]
  32. Higgins, K.A.; Considine, R.V.; Mattes, R.D. Aspartame Consumption for 12 Weeks Does Not Affect Glycemia, Appetite, or Body Weight of Healthy, Lean Adults in a Randomized Controlled Trial. J. Nutr. 2018, 148, 650–657. [Google Scholar] [CrossRef]
  33. Olalde-Mendoza, L.; Moreno-González, Y.E. Modification of fasting blood glucose in adults with diabetes mellitus type 2 after regular soda and diet soda intake in the State of Querétaro, Mexico. Arch. Latinoam. Nutr. 2013, 63, 142–147. [Google Scholar]
  34. Tey, S.L.; Salleh, N.B.; Henry, C.J.; Forde, C.G. Effects of non-nutritive (artificial vs. natural) sweeteners on 24-h glucose profiles. Eur. J. Clin. Nutr. 2017, 71, 1129–1132. [Google Scholar] [CrossRef]
  35. Rogers, P.J.; Appleton, K.M. The effects of low-calorie sweeteners on energy intake and body weight: A systematic review and meta-analyses of sustained intervention studies. Int. J. Obes. 2021, 45, 464–478. [Google Scholar] [CrossRef] [PubMed]
  36. Bernstein, A.M.; de Koning, L.; Flint, A.J.; Rexrode, K.M.; Willett, W.C. Soda consumption and the risk of stroke in men and women. Am. J. Clin. Nutr. 2012, 95, 1190–1199. [Google Scholar] [CrossRef]
  37. Debras, C.; Chazelas, E.; Srour, B.; Druesne-Pecollo, N.; Esseddik, Y.; Szabo de Edelenyi, F.; Agaesse, C.; De Sa, A.; Lutchia, R.; Gigandet, S.; et al. Artificial sweeteners and cancer risk: Results from the NutriNet-Sante population-based cohort study. PLoS Med. 2022, 19, e1003950. [Google Scholar] [CrossRef]
  38. Sweatman, T.W.; Renwick, A.G.; Burgess, C.D. The pharmacokinetics of saccharin in man. Xenobiotica 1981, 11, 531–540. [Google Scholar] [CrossRef]
  39. Whysner, J.; Williams, G.M. Saccharin mechanistic data and risk assessment: Urine composition, enhanced cell proliferation, and tumor promotion. Pharmacol. Ther. 1996, 71, 225–252. [Google Scholar] [CrossRef] [PubMed]
  40. Arnold, D.L. Toxicology of saccharin. Fundam. Appl. Toxicol. 1984, 4, 674–685. [Google Scholar] [CrossRef] [PubMed]
  41. Orku, S.E.; Suyen, G.; Bas, M. The effect of regular consumption of four low- or no-calorie sweeteners on glycemic response in healthy women: A randomized controlled trial. Nutrition 2023, 106, 111885. [Google Scholar] [CrossRef]
  42. Wilk, K.; Korytek, W.; Pelczyńska, M.; Moszak, M.; Bogdański, P. The Effect of Artificial Sweeteners Use on Sweet Taste Perception and Weight Loss Efficacy: A Review. Nutrients 2022, 14, 1261. [Google Scholar] [CrossRef]
  43. Higgins, K.A.; Mattes, R.D. A randomized controlled trial contrasting the effects of 4 low-calorie sweeteners and sucrose on body weight in adults with overweight or obesity. Am. J. Clin. Nutr. 2019, 109, 1288–1301. [Google Scholar] [CrossRef]
  44. Gao, Y.; Yin, L.; Zhang, Y.; Li, X.; Liu, L. Associations of saccharin intake with all-cause, cardiovascular and cancer mortality risk in USA adults. Br. J. Nutr. 2024, 132, 1205–1213. [Google Scholar] [CrossRef] [PubMed]
  45. Suez, J.; Cohen, Y.; Valdés-Mas, R.; Mor, U.; Dori-Bachash, M.; Federici, S.; Zmora, N.; Leshem, A.; Heinemann, M.; Linevsky, R.; et al. Personalized microbiome-driven effects of non-nutritive sweeteners on human glucose tolerance. Cell 2022, 185, 3307–3328.e19. [Google Scholar] [CrossRef]
  46. Pavanello, S.; Moretto, A.; La Vecchia, C.; Alicandro, G. Non-sugar sweeteners and cancer: Toxicological and epidemiological evidence. Regul. Toxicol. Pharmacol. 2023, 139, 105369. [Google Scholar] [CrossRef]
  47. Palomar-Cros, A.; Straif, K.; Romaguera, D.; Aragonés, N.; Castaño-Vinyals, G.; Martin, V.; Moreno, V.; Gómez-Acebo, I.; Guevara, M.; Aizpurua, A.; et al. Consumption of aspartame and other artificial sweeteners and risk of cancer in the Spanish multicase-control study (MCC-Spain). Int. J. Cancer 2023, 153, 979–993. [Google Scholar] [CrossRef] [PubMed]
  48. Li, H.; Zhang, Y.; He, Y.; Huang, J.; Yao, J.; Zhuang, X. Association between consumption of sweeteners and endometrial cancer risk: A systematic review and meta-analysis of observational studies. Br. J. Nutr. 2024, 131, 63–72. [Google Scholar] [CrossRef]
  49. Younes, M.; Aquilina, G.; Degen, G.; Engel, K.H.; Fowler, P.; Frutos Fernandez, M.J.; Fürst, P.; Gundert-Remy, U.; Gürtler, R.; Husøy, T.; et al. Safety evaluation of the food additive steviol glycosides, predominantly Rebaudioside M, produced by fermentation using Yarrowia lipolytica VRM. EFSA J. 2023, 21, e8387. [Google Scholar] [CrossRef] [PubMed]
  50. Geuns, J.M.; Buyse, J.; Vankeirsbilck, A.; Temme, E.H. Metabolism of stevioside by healthy subjects. Exp. Biol. Med. 2007, 232, 164–173. [Google Scholar]
  51. Hossain, M.S.; Alam, M.B.; Asadujjaman, M.; Islam, M.M.; Rahman, M.A.; Islam, M.A.; Islam, A. Antihyperglycemic and antihyperlipidemic effects of different fractions of Stevia rebaudiana leaves in alloxan induced diabetic rats. Int. J. Pharm. Sci. Res. 2011, 2, 1722–1729. [Google Scholar]
  52. Ahmad, U.; Ahmad, R.S.; Arshad, M.S.; Mushtaq, Z.; Hussain, S.M.; Hameed, A. Antihyperlipidemic efficacy of aqueous extract of Stevia rebaudiana Bertoni in albino rats. Lipids Health Dis. 2018, 17, 175. [Google Scholar] [CrossRef]
  53. Assaei, R.; Mokarram, P.; Dastghaib, S.; Darbandi, S.; Darbandi, M.; Zal, F.; Akmali, M.; Ranjbar Omrani, G.H. Hypoglycemic Effect of Aquatic Extract of Stevia in Pancreas of Diabetic Rats: PPARγ-dependent Regulation or Antioxidant Potential. Avicenna J. Med. Biotechnol. 2016, 8, 65–74. [Google Scholar]
  54. Chen, C.; Na, X.; Wang, L.; Yu, R. High-throughput screening identifies stevioside as a potent agent to induce apoptosis in bladder cancer cells. Biochem. Pharmacol. 2022, 203, 115166. [Google Scholar] [CrossRef]
  55. Ren, H.P.; Yin, X.Y.; Yu, H.Y.; Xiao, H.F. Stevioside induced cytotoxicity in colon cancer cells via reactive oxygen species and mitogen-activated protein kinase signaling pathways-mediated apoptosis. Oncol. Lett. 2017, 13, 2337–2343. [Google Scholar] [CrossRef]
  56. Khare, N.; Chandra, S. Stevioside mediated chemosensitization studies and cytotoxicity assay on breast cancer cell lines MDA-MB-231 and SKBR3. Saudi J. Biol. Sci. 2019, 26, 1596–1601. [Google Scholar] [CrossRef] [PubMed]
  57. Paul, S.; Sengupta, S.; Bandyopadhyay, T.K.; Bhattacharyya, A. Stevioside induced ROS-mediated apoptosis through mitochondrial pathway in human breast cancer cell line MCF-7. Nutr. Cancer 2012, 64, 1087–1094. [Google Scholar] [CrossRef] [PubMed]
  58. Bai, X.; Qu, H.; Zhang, J.; Li, L.; Zhang, C.; Li, S.; Li, G. Effect of steviol glycosides as natural sweeteners on glucose metabolism in adult participants. Food Funct. 2024, 15, 3908–3919. [Google Scholar] [CrossRef]
  59. Anton, S.D.; Martin, C.K.; Han, H.; Coulon, S.; Cefalu, W.T.; Geiselman, P.; Williamson, D.A. Effects of stevia, aspartame, and sucrose on food intake, satiety, and postprandial glucose and insulin levels. Appetite 2010, 55, 37–43. [Google Scholar] [CrossRef]
  60. Gregersen, S.; Jeppesen, P.B.; Holst, J.J.; Hermansen, K. Antihyperglycemic effects of stevioside in type 2 diabetic subjects. Metabolism 2004, 53, 73–76. [Google Scholar] [CrossRef] [PubMed]
  61. Sharma, N.; Mogra, R.; Upadhyay, B. Effect of stevia extract intervention on lipid profile. Stud. Ethno-Med. 2009, 3, 137–140. [Google Scholar] [CrossRef]
  62. Regnat, K.; Mach, R.L.; Mach-Aigner, A.R. Erythritol as sweetener-wherefrom and whereto? Appl. Microbiol. Biotechnol. 2018, 102, 587–595. [Google Scholar] [CrossRef]
  63. Munro, I.C.; Berndt, W.O.; Borzelleca, J.F.; Flamm, G.; Lynch, B.S.; Kennepohl, E.; Bär, E.A.; Modderman, J. Erythritol: An interpretive summary of biochemical, metabolic, toxicological and clinical data. Food Chem. Toxicol. 1998, 36, 1139–1174. [Google Scholar] [CrossRef]
  64. Soetan, O.A.; Ajao, F.O.; Ajayi, A.F. Blood glucose lowering and anti-oxidant potential of erythritol: An in vitro and in vivo study. J. Diabetes Metab. Disord. 2023, 22, 1217–1229. [Google Scholar] [CrossRef] [PubMed]
  65. Witkowski, M.; Nemet, I.; Alamri, H.; Wilcox, J.; Gupta, N.; Nimer, N.; Haghikia, A.; Li, X.S.; Wu, Y.; Saha, P.P.; et al. The artificial sweetener erythritol and cardiovascular event risk. Nat. Med. 2023, 29, 710–718. [Google Scholar] [CrossRef] [PubMed]
  66. Mazi, T.A.; Stanhope, K.L. Erythritol: An In-Depth Discussion of Its Potential to Be a Beneficial Dietary Component. Nutrients 2023, 15, 204. [Google Scholar] [CrossRef]
  67. Noda, K.; Nakayama, K.; Oku, T. Serum glucose and insulin levels and erythritol balance after oral administration of erythritol in healthy subjects. Eur. J. Clin. Nutr. 1994, 48, 286–292. [Google Scholar]
  68. Wölnerhanssen, B.K.; Cajacob, L.; Keller, N.; Doody, A.; Rehfeld, J.F.; Drewe, J.; Peterli, R.; Beglinger, C.; Meyer-Gerspach, A.C. Gut hormone secretion, gastric emptying, and glycemic responses to erythritol and xylitol in lean and obese subjects. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E1053–E1061. [Google Scholar] [CrossRef]
  69. Tetzloff, W.; Dauchy, F.; Medimagh, S.; Carr, D.; Bär, A. Tolerance to subchronic, high-dose ingestion of erythritol in human volunteers. Regul. Toxicol. Pharmacol. 1996, 24, S286–S295. [Google Scholar] [CrossRef]
  70. Ishikawa, M.; Miyashita, M.; Kawashima, Y.; Nakamura, T.; Saitou, N.; Modderman, J. Effects of oral administration of erythritol on patients with diabetes. Regul. Toxicol. Pharmacol. 1996, 24, S303–S308. [Google Scholar] [CrossRef] [PubMed]
  71. Teysseire, F.; Bordier, V.; Budzinska, A.; Van Oudenhove, L.; Weltens, N.; Beglinger, C.; Wölnerhanssen, B.K.; Meyer-Gerspach, A.C. Metabolic Effects and Safety Aspects of Acute D-allulose and Erythritol Administration in Healthy Subjects. Nutrients 2023, 15, 458. [Google Scholar] [CrossRef]
  72. Salli, K.; Lehtinen, M.J.; Tiihonen, K.; Ouwehand, A.C. Xylitol’s Health Benefits beyond Dental Health: A Comprehensive Review. Nutrients 2019, 11, 1813. [Google Scholar] [CrossRef]
  73. Storey, D.; Lee, A.; Bornet, F.; Brouns, F. Gastrointestinal tolerance of erythritol and xylitol ingested in a liquid. Eur. J. Clin. Nutr. 2007, 61, 349–354. [Google Scholar] [CrossRef] [PubMed]
  74. Livesey, G. Health potential of polyols as sugar replacers, with emphasis on low glycaemic properties. Nutr. Res. Rev. 2003, 16, 163–191. [Google Scholar] [CrossRef] [PubMed]
  75. Natah, S.S.; Hussien, K.R.; Tuominen, J.A.; Koivisto, V.A. Metabolic response to lactitol and xylitol in healthy men. Am. J. Clin. Nutr. 1997, 65, 947–950. [Google Scholar] [CrossRef] [PubMed]
  76. Mäkinen, K.K. Gastrointestinal Disturbances Associated with the Consumption of Sugar Alcohols with Special Consideration of Xylitol: Scientific Review and Instructions for Dentists and Other Health-Care Professionals. Int. J. Dent. 2016, 2016, 5967907. [Google Scholar] [CrossRef]
  77. Tamura, M.; Hoshi, C.; Hori, S. Xylitol affects the intestinal microbiota and metabolism of daidzein in adult male mice. Int. J. Mol. Sci. 2013, 14, 23993–24007. [Google Scholar] [CrossRef]
  78. Uebanso, T.; Kano, S.; Yoshimoto, A.; Naito, C.; Shimohata, T.; Mawatari, K.; Takahashi, A. Effects of Consuming Xylitol on Gut Microbiota and Lipid Metabolism in Mice. Nutrients 2017, 9, 756. [Google Scholar] [CrossRef]
  79. Witkowski, M.; Nemet, I.; Li, X.S.; Wilcox, J.; Ferrell, M.; Alamri, H.; Gupta, N.; Wang, Z.; Tang, W.H.W.; Hazen, S.L. Xylitol is prothrombotic and associated with cardiovascular risk. Eur. Heart J. 2024, 45, 2439–2452. [Google Scholar] [CrossRef]
  80. Nabors, L.O. Sweet choices: Sugar replacements for foods and beverages. Food Technol. 2002, 56, 28–34. [Google Scholar]
  81. Lenhart, A.; Chey, W.D. A Systematic Review of the Effects of Polyols on Gastrointestinal Health and Irritable Bowel Syndrome. Adv. Nutr. 2017, 8, 587–596. [Google Scholar] [CrossRef]
  82. Secchi, A.; Pontiroli, A.E.; Cammelli, L.; Bizzi, A.; Cini, M.; Pozza, G. Effects of oral administration of maltitol on plasma glucose, plasma sorbitol, and serum insulin levels in man. Klin. Wochenschr. 1986, 64, 265–269. [Google Scholar] [CrossRef]
  83. Miller, P.E.; Perez, V. Low-calorie sweeteners and body weight and composition: A meta-analysis of randomized controlled trials and prospective cohort studies. Am. J. Clin. Nutr. 2014, 100, 765–777. [Google Scholar] [CrossRef] [PubMed]
  84. Chukwuma, C.I.; Islam, M.S. Sorbitol increases muscle glucose uptake ex vivo and inhibits intestinal glucose absorption ex vivo and in normal and type 2 diabetic rats. Appl. Physiol. Nutr. Metab. 2017, 42, 377–383. [Google Scholar] [CrossRef]
  85. Li, C.H.; Wang, C.T.; Lin, Y.J.; Kuo, H.Y.; Wu, J.S.; Hong, T.C.; Chang, C.J.; Wu, H.T. Long-term consumption of the sugar substitute sorbitol alters gut microbiome and induces glucose intolerance in mice. Life Sci. 2022, 305, 120770. [Google Scholar] [CrossRef] [PubMed]
  86. Ellis, F.W.; Krantz, J.C., Jr. Sugar alcohols. 22. Metabolism and toxicity studies with mannitol and sorbitol in man and animals. J. Biol. Chem. 1941, 141, 147–154. [Google Scholar] [CrossRef]
  87. Godswill, A.C. Sugar alcohols: Chemistry, production, health concerns and nutritional importance of mannitol, sorbitol, xylitol, and erythritol. Int. J. Adv. Acad. Res. 2017, 3, 31–66. [Google Scholar]
  88. Kim, S.K.; Guthrie, B.; Goddard, W.A., 3rd. Ligand-Dependent and G Protein-Dependent Properties for the Sweet Taste Heterodimer, TAS1R2/1R3. J. Phys. Chem. B 2024, 128, 8927–8932. [Google Scholar] [CrossRef]
  89. Just, T.; Pau, H.W.; Engel, U.; Hummel, T. Cephalic phase insulin release in healthy humans after taste stimulation? Appetite 2008, 51, 622–627. [Google Scholar] [CrossRef]
  90. Kokrashvili, Z.; Mosinger, B.; Margolskee, R.F. T1r3 and alpha-gustducin in gut regulate secretion of glucagon-like peptide-1. Ann. N. Y. Acad. Sci. 2009, 1170, 91–94. [Google Scholar] [CrossRef]
  91. Daly, K.; Al-Rammahi, M.; Arora, D.K.; Moran, A.W.; Proudman, C.J.; Ninomiya, Y.; Shirazi-Beechey, S.P. Expression of sweet receptor components in equine small intestine: Relevance to intestinal glucose transport. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2012, 303, R199–R208. [Google Scholar] [CrossRef] [PubMed]
  92. Margolskee, R.F.; Dyer, J.; Kokrashvili, Z.; Salmon, K.S.; Ilegems, E.; Daly, K.; Maillet, E.L.; Ninomiya, Y.; Mosinger, B.; Shirazi-Beechey, S.P. T1R3 and gustducin in gut sense sugars to regulate expression of Na+-glucose cotransporter 1. Proc. Natl. Acad. Sci. USA 2007, 104, 15075–15080. [Google Scholar] [CrossRef]
  93. Mace, O.J.; Affleck, J.; Patel, N.; Kellett, G.L. Sweet taste receptors in rat small intestine stimulate glucose absorption through apical GLUT2. J. Physiol. 2007, 582, 379–392. [Google Scholar] [CrossRef]
  94. Smith, K.; Karimian Azari, E.; LaMoia, T.E.; Hussain, T.; Vargova, V.; Karolyi, K.; Veldhuis, P.P.; Arnoletti, J.P.; de la Fuente, S.G.; Pratley, R.E.; et al. T1R2 receptor-mediated glucose sensing in the upper intestine potentiates glucose absorption through activation of local regulatory pathways. Mol. Metab. 2018, 17, 98–111. [Google Scholar] [CrossRef]
  95. Mace, O.J.; Lister, N.; Morgan, E.; Shepherd, E.; Affleck, J.; Helliwell, P.; Bronk, J.R.; Kellett, G.L.; Meredith, D.; Boyd, R.; et al. An energy supply network of nutrient absorption coordinated by calcium and T1R taste receptors in rat small intestine. J. Physiol. 2009, 587, 195–210. [Google Scholar] [CrossRef]
  96. Nakagawa, Y.; Nagasawa, M.; Yamada, S.; Hara, A.; Mogami, H.; Nikolaev, V.O.; Lohse, M.J.; Shigemura, N.; Ninomiya, Y.; Kojima, I. Sweet taste receptor expressed in pancreatic beta-cells activates the calcium and cyclic AMP signaling systems and stimulates insulin secretion. PLoS ONE 2009, 4, e5106. [Google Scholar] [CrossRef] [PubMed]
  97. Kyriazis, G.A.; Soundarapandian, M.M.; Tyrberg, B. Sweet taste receptor signaling in beta cells mediates fructose-induced potentiation of glucose-stimulated insulin secretion. Proc. Natl. Acad. Sci. USA 2012, 109, E524–E532. [Google Scholar] [CrossRef] [PubMed]
  98. Teysseire, F.; Bordier, V.; Budzinska, A.; Weltens, N.; Rehfeld, J.F.; Holst, J.J.; Hartmann, B.; Beglinger, C.; Van Oudenhove, L.; Wölnerhanssen, B.K.; et al. The Role of D-allulose and Erythritol on the Activity of the Gut Sweet Taste Receptor and Gastrointestinal Satiation Hormone Release in Humans: A Randomized, Controlled Trial. J. Nutr. 2022, 152, 1228–1238. [Google Scholar] [CrossRef] [PubMed]
  99. Kojima, I.; Nakagawa, Y. The Role of the Sweet Taste Receptor in Enteroendocrine Cells and Pancreatic β-Cells. Diabetes Metab. J. 2011, 35, 451–457. [Google Scholar] [CrossRef]
  100. Masenga, S.K.; Kabwe, L.S.; Chakulya, M.; Kirabo, A. Mechanisms of Oxidative Stress in Metabolic Syndrome. Int. J. Mol. Sci. 2023, 24, 7898. [Google Scholar] [CrossRef]
  101. Kundu, N.; Domingues, C.C.; Patel, J.; Aljishi, M.; Ahmadi, N.; Fakhri, M.; Sylvetsky, A.C.; Sen, S. Sucralose promotes accumulation of reactive oxygen species (ROS) and adipogenesis in mesenchymal stromal cells. Stem Cell Res. Ther. 2020, 11, 250. [Google Scholar] [CrossRef]
  102. Tsai, M.J.; Li, C.H.; Wu, H.T.; Kuo, H.Y.; Wang, C.T.; Pai, H.L.; Chang, C.J.; Ou, H.Y. Long-Term Consumption of Sucralose Induces Hepatic Insulin Resistance through an Extracellular Signal-Regulated Kinase 1/2-Dependent Pathway. Nutrients 2023, 15, 2814. [Google Scholar] [CrossRef]
  103. Van Opstal, A.M.; Hafkemeijer, A.; van den Berg-Huysmans, A.A.; Hoeksma, M.; Mulder, T.P.J.; Pijl, H.; Rombouts, S.; van der Grond, J. Brain activity and connectivity changes in response to nutritive natural sugars, non-nutritive natural sugar replacements and artificial sweeteners. Nutr. Neurosci. 2021, 24, 395–405. [Google Scholar] [CrossRef]
  104. Dore, R.; Krotenko, R.; Reising, J.P.; Murru, L.; Sundaram, S.M.; Di Spiezio, A.; Müller-Fielitz, H.; Schwaninger, M.; Jöhren, O.; Mittag, J.; et al. Nesfatin-1 decreases the motivational and rewarding value of food. Neuropsychopharmacology 2020, 45, 1645–1655. [Google Scholar] [CrossRef] [PubMed]
  105. Jiang, L.; Yu, Z.; Zhao, Y.; Yin, D. Obesogenic potentials of environmental artificial sweeteners with disturbances on both lipid metabolism and neural responses. Sci. Total Environ. 2024, 919, 170755. [Google Scholar] [CrossRef]
  106. Li, Y.; Zhu, W.; Cai, J.; Liu, W.; Akihisa, T.; Li, W.; Kikuchi, T.; Xu, J.; Feng, F.; Zhang, J. The role of metabolites of steviol glycosides and their glucosylated derivatives against diabetes-related metabolic disorders. Food Funct. 2021, 12, 8248–8259. [Google Scholar] [CrossRef]
  107. Yang, P.S.; Lee, J.J.; Tsao, C.W.; Wu, H.T.; Cheng, J.T. Stimulatory effect of stevioside on peripheral mu opioid receptors in animals. Neurosci. Lett. 2009, 454, 72–75. [Google Scholar] [CrossRef] [PubMed]
  108. Sikalidis, A.K.; Maykish, A. The Gut Microbiome and Type 2 Diabetes Mellitus: Discussing a Complex Relationship. Biomedicines 2020, 8, 8. [Google Scholar] [CrossRef] [PubMed]
  109. Méndez-García, L.A.; Bueno-Hernández, N.; Cid-Soto, M.A.; De León, K.L.; Mendoza-Martínez, V.M.; Espinosa-Flores, A.J.; Carrero-Aguirre, M.; Esquivel-Velázquez, M.; León-Hernández, M.; Viurcos-Sanabria, R.; et al. Ten-Week Sucralose Consumption Induces Gut Dysbiosis and Altered Glucose and Insulin Levels in Healthy Young Adults. Microorganisms 2022, 10, 434. [Google Scholar] [CrossRef] [PubMed]
  110. Shon, W.J.; Song, J.W.; Oh, S.H.; Lee, K.H.; Seong, H.; You, H.J.; Seong, J.K.; Shin, D.M. Gut taste receptor type 1 member 3 is an intrinsic regulator of Western diet-induced intestinal inflammation. BMC Med. 2023, 21, 165. [Google Scholar] [CrossRef]
  111. Zani, F.; Blagih, J.; Gruber, T.; Buck, M.D.; Jones, N.; Hennequart, M.; Newell, C.L.; Pilley, S.E.; Soro-Barrio, P.; Kelly, G.; et al. The dietary sweetener sucralose is a negative modulator of T cell-mediated responses. Nature 2023, 615, 705–711. [Google Scholar] [CrossRef]
  112. Frankenfeld, C.L.; Sikaroodi, M.; Lamb, E.; Shoemaker, S.; Gillevet, P.M. High-intensity sweetener consumption and gut microbiome content and predicted gene function in a cross-sectional study of adults in the United States. Ann. Epidemiol. 2015, 25, 736–742.e4. [Google Scholar] [CrossRef] [PubMed]
  113. Ahmad, S.Y.; Friel, J.; Mackay, D. The Effects of Non-Nutritive Artificial Sweeteners, Aspartame and Sucralose, on the Gut Microbiome in Healthy Adults: Secondary Outcomes of a Randomized Double-Blinded Crossover Clinical Trial. Nutrients 2020, 12, 3408. [Google Scholar] [CrossRef] [PubMed]
  114. Suez, J.; Korem, T.; Zeevi, D.; Zilberman-Schapira, G.; Thaiss, C.A.; Maza, O.; Israeli, D.; Zmora, N.; Gilad, S.; Weinberger, A.; et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014, 514, 181–186. [Google Scholar] [CrossRef]
  115. Singh, G.; McBain, A.J.; McLaughlin, J.T.; Stamataki, N.S. Consumption of the Non-Nutritive Sweetener Stevia for 12 Weeks Does Not Alter the Composition of the Human Gut Microbiota. Nutrients 2024, 16, 296. [Google Scholar] [CrossRef] [PubMed]
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

Chen, H.-P.; Kao, Y.; Lin, M.-W.; Lee, C.-T.; Wu, H.-T.; Kuo, H.-Y. Potential Effects of Low-Calorie Sweeteners on Human Health. Nutrients 2025, 17, 2726. https://doi.org/10.3390/nu17172726

AMA Style

Chen H-P, Kao Y, Lin M-W, Lee C-T, Wu H-T, Kuo H-Y. Potential Effects of Low-Calorie Sweeteners on Human Health. Nutrients. 2025; 17(17):2726. https://doi.org/10.3390/nu17172726

Chicago/Turabian Style

Chen, Huang-Pin, Yuan Kao, Meng-Wei Lin, Chun-Te Lee, Hung-Tsung Wu, and Hsin-Yu Kuo. 2025. "Potential Effects of Low-Calorie Sweeteners on Human Health" Nutrients 17, no. 17: 2726. https://doi.org/10.3390/nu17172726

APA Style

Chen, H.-P., Kao, Y., Lin, M.-W., Lee, C.-T., Wu, H.-T., & Kuo, H.-Y. (2025). Potential Effects of Low-Calorie Sweeteners on Human Health. Nutrients, 17(17), 2726. https://doi.org/10.3390/nu17172726

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop