Next Article in Journal
Microbial Metabolism of Naringin and the Impact on Antioxidant Capacity
Previous Article in Journal
Commercial Complementary Food in Germany: A 2020 Market Survey
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Hot Water Extract of Curcuma longa L. Improves Fasting Serum Glucose Levels in Participants with Low-Grade Inflammation: Reanalysis of Data from Two Randomized, Double-Blind, Placebo-Controlled Trials

Research & Development Institute, House Wellness Foods Corp., 3-20 Imoji, Itami 664-0011, Japan
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(18), 3763; https://doi.org/10.3390/nu14183763
Submission received: 24 August 2022 / Revised: 8 September 2022 / Accepted: 8 September 2022 / Published: 13 September 2022
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
The dietary spice Curcuma longa L. (C. longa), also known as turmeric, has various biological effects. A hot water extract of C. longa was shown to have anti-inflammatory activities in preclinical and clinical studies. Chronic low-grade inflammation is associated with the disruption of glucose homeostasis, but the effect of C. longa extract on glucose metabolism in humans is poorly understood. Therefore, we investigated the effect of C. longa extracts on serum glucose levels in the presence of low-grade inflammation. We reanalyzed our published data from two randomized, double-blind, placebo-controlled trials in overweight participants aged 50 to 69 years and performed a stratified analysis using the inflammatory marker high-sensitivity C-reactive protein (hsCRP). In both studies, participants took a test food with a hot water extract of C. longa (C. longa extract group, n = 45 per study) or without C. longa extract (placebo group, n = 45 per study) daily for 12 weeks, and we measured the levels of serum hsCRP and fasting serum glucose. The mean baseline hsCRP value was used to stratify participants into two subgroups: a low-hsCRP subgroup (baseline mean hsCRP < 0.098 mg/dL) and a high-hsCRP subgroup (baseline mean hsCRP ≥ 0.098 mg/dL). In the low-hsCRP subgroup, we found no significant difference in fasting serum glucose levels between the two groups in either study, but in the high-hsCRP subgroup, the C. longa extract group had significantly lower levels of serum hsCRP (p < 0.05) and fasting serum glucose (p < 0.05) than the placebo group in both studies. In conclusion, a hot water extract of C. longa may help to improve systemic glucose metabolism in people with chronic low-grade inflammation.

1. Introduction

Glucose is a main source of cellular energy in mammalian cells and plays an important role in maintaining normal physiological function in humans [1]. Abnormally low levels of blood glucose can cause seizures, comas, and death. Islet tissue damage and various diseases such as type 2 diabetes cause elevated blood glucose levels [2]. Therefore, glucose homeostasis is tightly regulated through several processes, such as peripheral glucose uptake, gluconeogenesis, glycolysis, glycogenesis, and glycogenolysis [1]. These processes are controlled by glucoregulatory hormones, such as insulin, incretin, glucagon, glucocorticoid, and growth hormones released from some organs, including the pancreas, intestine, adrenal gland, and pituitary gland [3,4]. However, various factors related to aging, obesity, and an unhealthy lifestyle are known to induce pancreatic β-cell dysfunction, insulin resistance, and the disruption of systemic glucose homeostasis, resulting in the progression of type 2 diabetes [5,6].
Chronic low-grade inflammation is an ongoing inflammatory response. This type of inflammation is mediated by a long-lasting immune response and can cause tissue damage, inhibit wound healing, and impair glucose homeostasis [5,6]. A standard definition of chronic low-grade inflammation has not yet been determined, but various causes have been identified, including aging [7], obesity [8], and an unhealthy lifestyle [9] without any apparent infection or tissue injury [10]. These play a key role in the development of pancreatic β-cell dysfunction, insulin resistance, and type 2 diabetes. Anti-inflammatory drugs have been shown to improve insulin secretion, insulin resistance, and blood glucose levels in clinical intervention studies [11,12]. Recent research revealed that serum levels of high-sensitivity C-reactive protein (hsCRP) are only slightly elevated in chronic low-grade inflammation compared with acute inflammation [8,10]. However, previous reports indicated that participants with mildly elevated hsCRP may have a higher risk of impaired fasting glucose and type 2 diabetes [13,14].
The medicinal herb Curcuma longa L. (C. longa), also known as turmeric, is a member of the Zingiberaceae family and has various physiological activities [15]. Water extracts of C. longa have antioxidant and anti-inflammatory effects [16,17,18,19]; they can prevent skeletal muscle atrophy [20], improve the water content of skin [21], promote corneal wound healing [22], and show positive effects on fatigue [23]. In addition, in animal models, water extracts of C. longa were reported to prevent various chronic inflammatory diseases, including cotton pellet-induced granuloma [24], carbon tetrachloride-induced hepatitis [25], and non-alcoholic steatohepatitis [26], by reducing the production of inflammatory cytokines, chemokines, and cell adhesion molecules. Furthermore, previous intervention studies showed that daily intake of a hot water extract of C. longa improved levels of systemic inflammatory markers such as hsCRP, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 [27,28]. C. longa extract was recently shown to decrease blood glucose levels in an animal model of dietary-induced obesity and streptozotocin-induced diabetes [29,30]. However, the influence of water extract of C. longa on glucose metabolism in humans is not clearly understood.
Therefore, we reanalyzed our data from two randomized, double-blind, placebo-controlled trials to investigate the effects of C. longa extract on serum glucose levels in humans with low-grade inflammation. We then reanalyzed our data [27,28] and performed a stratified analysis by using the inflammatory marker hsCRP.

2. Materials and Methods

2.1. Study Design

We reanalyzed data from two randomized, double-blind, placebo-controlled trials (Nutrients, 11 (8): 1822, 2019 [study 1; [27]] and Nutr J, 20 (1): 91, 2021 [study 2; [28]]). All procedures in the two studies involving human participants were approved by the institutional review board of Chiyoda Paramedical Care Clinic (Tokyo, Japan). The studies were conducted according to the Declaration of Helsinki. Before enrollment in the study, all participants provided written informed consent. Both studies were performed by a contract research organization (CRO; CPCC Co., Ltd., Tokyo, Japan) at the Chiyoda Paramedical Care Clinic; study 1 was performed from June to December 2017 (University Hospital Medical Information Network registration number UMIN000029095), and study 2 was performed from June to December 2019 (UMIN000037370). Figure 1 and Figure 2 show the Consolidated Standards of Reporting Trials (CONSORT) 2010 diagrams of the flow of participants from enrollment to analysis in studies 1 and 2, respectively; the completed CONSORT checklists are provided in the original research papers [27,28].

2.2. Enrolment of Participants

2.2.1. Study 1

Among people attending the Chiyoda Paramedical Care Clinic, 297 potential participants were consecutively assessed for eligibility. The study included men and menopausal women aged 50 to 69 years with a body mass index (BMI) between overweight and obesity class 1 (≥23 to <30 kg/m2) [31] or blood pressure between prehypertension and grade 1 (mild) hypertension (systolic blood pressure ≥120 to <160 mmHg or diastolic blood pressure ≥80 to <100 mmHg). The exclusion criteria were as follows: (1) positive for hepatitis C virus antibody or hepatitis B surface antigen, (2) use of medications or health foods that could possibly influence the results of this study, (3) history of heart, liver, kidney, or gastrointestinal disease, (4) history of cardiovascular disease, (5) excessive alcohol intake (mean daily consumption of 60 g or more) or smoking (mean daily consumption of two packs), (6) extremely irregular dietary habits, (7) allergies to medications or foods, (8) participation in another trial, either currently or in the past 4 weeks, or a plan to participate in another trial during the scheduled study period, (9) donation of blood within 1 month before the study, (10) in men, donation of 400 mL of blood within 3 months before the study, (11) in women, donation of 400 mL of blood within 4 months before the study, (12) in men, donation of more than 1200 mL of blood minus the estimated volume of blood collected during the study within 1 year before the study, (13) in women, donation of more than 800 mL of blood minus the estimated volume of blood collected during the study within 1 year before the study, and (14) judged to be unsuitable for the study for other reasons by the investigators.

2.2.2. Study 2

Among people attending the Chiyoda Paramedical Care Clinic, 339 potential participants were consecutively assessed for eligibility. The study included overweight men and menopausal women aged 50 to 69 years (BMI ≥ 23 to <30 kg/m2) [31], with blood glucose less than 126 mg/dL and high-density blood cholesterol greater than or equal to 35 mg/dL. The exclusion criteria were as follows: (1) positive for hepatitis C virus antibody or hepatitis B surface antigen, (2) use of medications or health foods that could possibly influence the results of this study, (3) history of heart, liver, kidney, or gastrointestinal disease, (4) history of circulatory disease, (5) excessive alcohol intake (mean daily consumption of 60 g or more), (6) excessive smoking (mean daily consumption of two packs or more), (7) extremely irregular dietary habits, (8) allergies to medications or foods (especially soybeans or gelatin), (9) participation in another trial, either currently or in the past 4 weeks, or a plan to participate in another trial during the scheduled study period, (10) donation of blood within 1 month before the study, (11) in men, donation of 400 mL of blood within 3 months before the study, (12) in women, donation of 400 mL of blood within 4 months before the study, (13) in men, donation of more than 1200 mL of blood minus the estimated volume of blood collected during the study within 1 year before the study, (14) in women, donation of more than 800 mL of blood minus the estimated volume of blood collected during the study within 1 year before the study, and (15) judged to be unsuitable for the study for other reasons by the investigators.

2.3. Study Agent

The composition of the test diets used in the two studies is shown in Table 1. The turmeric diets contained a hot water extract of C. longa (House Wellness Foods) and were prepared according to the method previously described in [27,28]. The placebo diets contained coloring agents to match the turmeric diet’s color.

2.4. Intervention

The intervention was conducted according to the procedures described in the publications of the two studies [27,28]. Briefly, the CRO selected 90 participants who satisfied the inclusion and exclusion criteria for the respective study (Figure 1 and Figure 2). The test diets and the selected participants were randomly assigned numbers, and the assignment list was stored carefully until the database was locked. Throughout the study, all participants and investigators were blind to the treatment provided. The participants were randomly allocated into two groups by stratified randomization. Each participant consumed a test diet with C. longa extract (C. longa extract group, n = 45) or without (placebo group, n = 45) once daily for 12 weeks. Participants visited the study center during weeks 0, 4, 8, and 12 to undergo an interview conducted by an experienced physician, physiological measurements, hematology and biochemistry tests, and urinalysis, and to complete questionnaires. The hematology and biochemistry tests and urinalysis were performed by a contracted laboratory company (LSI Medience Co., Ltd., Tokyo, Japan). The participants were asked to daily record the following information in a diary until the end of the study: occurrence of diseases and symptoms, intake of the study diet, healthy foods, medications, reasons for taking medications, the dosage, and the duration of use.

2.5. Measurement of Serum hsCRP Level

The serum levels of hsCRP were measured by an immunonephelometric method with an upper detection limit of 0.500 mg/dL (LSI Medience) [32]. For samples exceeding this limit, a remeasurement was performed by standard latex agglutination turbidimetry using N-assay LA CRP-T kits (Nittobo Medical Co., Ltd., Tokyo, Japan) [33].

2.6. Measurement of Fasting Serum Glucose Level

The level of fasting serum glucose was measured by commercial glucose assay kits based on the glucokinase method (LSI Medience) [32,34].

2.7. Sample Size

To calculate the minimum number of participants required for adequate statistical power, we used the G Power 3.1.9 program (University of Düsseldorf, Düsseldorf, Germany). In a previous clinical study, a green tea extract with anti-inflammatory activity reduced the serum CRP level by about 30% from the baseline [35]. Therefore, a sample size of 45 participants per group was estimated to be sufficient for the present study based on the following assumptions: a 30% reduction of serum CRP by C. longa extract, Cohen’s d-value of 0.60, a statistical power of 80%, and type I error of 5% (two-tailed).

2.8. Stratified Analysis

To investigate the effect of a C. longa extract on serum glucose levels in the presence of low-grade inflammation, we stratified the participants according to the baseline mean value of hsCRP (0.098 mg/dL) in study 1 (Table S1) and divided them into two subgroups: a low-hsCRP subgroup (baseline mean hsCRP < 0.098 mg/dL) and a high-hsCRP subgroup (baseline mean hsCRP ≥ 0.098 mg/dL).

2.9. Statistical Analysis

Statistical analysis was performed in the intention-to-treat (ITT) population, which was defined as all randomized participants. The full analysis set (FAS) was used to assess safety, and the per-protocol set (PPS) was used to evaluate efficacy. In the efficacy assessment of inflammatory markers, we excluded the data of participants who were suspected of having acute inflammation because of a markedly increasing hsCRP level or symptoms associated with acute inflammation as diagnosed by a physician. All statistical analyses were performed with the IBM SPSS statistical software package (version 26) for Windows (IBM Corp., Armonk, NY, USA). Results are presented as the mean (standard deviation [SD]). Baseline characteristics were compared between the two groups by the two-tailed unpaired Student’s t-test when variance was homogeneous or the Aspin-Welch t-test when variance was heterogeneous, except for sex-related results, which were analyzed by the two-tailed Mann-Whitney U-test. Changes from the baseline were analyzed by repeated measures of two-way analysis of variance (ANOVA; two groups × three-time points) with the SPSS general linear model for determining the main effects of the group and time and their interaction, followed by a comparison between the C. longa extract and placebo groups at each time point with simple tests for main effect [27,36,37,38]. A probability (p) value less than 0.05 was considered to indicate statistical significance.

3. Results

3.1. Subjects

3.1.1. Study 1

The flow of participants through the study is shown in Figure 1. The 90 participants were randomly allocated to the C. longa extract or placebo group (n = 45 per group), and 87 participants completed the study: two participants dropped out before completing the study because of cerebral infarction (n = 1 in the placebo group) and ovarian cystectomy (n = 1 in the C. longa extract group); one participant did not consume the test agent in compliance with the protocol (n = 1 in the C. longa extract group) and was therefore excluded from the efficacy analysis (PPS analysis). The baseline characteristics showed no significant differences between the two groups (Table S1). The mean intake of the test diets also showed no significant differences between the two groups. In the stratified analysis, the baseline characteristics showed no significant differences between the C. longa extract and placebo groups in either the low- or high-hsCRP subgroup (Table 2).

3.1.2. Study 2

The flow of participants through the study is shown in Figure 2. The 90 participants were randomly allocated to the C. longa extract group or the placebo group (n = 45 per group), and 79 participants completed the study: two participants dropped out before completing the study because they declined to continue participation because of a marked reduction of body weight (n = 1 in the placebo group) and a bone fracture resulting from a slip-and-fall accident while walking (n = 1 in the C. longa extract group); nine participants were excluded from the efficacy assessment (PPS analysis) because they did not comply with the protocol, i.e., they changed their dietary habits (n = 4 in the C. longa extract group and n = 4 in the placebo group) or fractured a bone as a result of a slip-and-fall accident while cycling (n = 1 in the C. longa extract group). Baseline characteristics showed no significant differences between the C. longa extract and placebo groups, except for significantly higher serum glucose levels in the placebo group compared with the C. longa extract group (Table S2). The mean intake of the test diets showed no significant differences between the two groups. In the stratified analysis, baseline characteristics showed no significant differences between the C. longa extract and placebo groups in the low-hsCRP subgroup, but in the high-hsCRP subgroup, LDL cholesterol was significantly lower at baseline in the C. longa extract group than in the placebo group (Table 3).

3.2. Effect of C. longa Extract on Serum hsCRP Levels

3.2.1. Study 1

In the low-hsCRP subgroup, the change of hsCRP from baseline was significantly lower in the C. longa extract group than in the placebo group throughout the study period (p = 0.013 by r-ANOVA); in addition, the hsCRP level tended to be lower in week 8 (p = 0.084) and significantly lower in week 12 (p = 0.006) in the C. longa extract group than in the placebo group (Table 4). In the high-hsCRP subgroup, the change of hsCRP from baseline showed no significant differences between the two groups throughout the study period (r-ANOVA); however, the hsCRP level was significantly lower in week 8 (p = 0.007) and tended to be lower in week 12 (p = 0.095) in the C. longa extract group than in the placebo group (Table 4).

3.2.2. Study 2

In the low-hsCRP subgroup, the change of hsCRP from baseline did not show any significant differences between the two groups (Table 5). In the high-hsCRP subgroup, the change of hsCRP from baseline was significantly lower in the C. longa extract group than in the placebo group (p = 0.001 by r-ANOVA). Furthermore, in the high-hsCRP subgroup, the hsCRP level tended to be lower in week 4 (p = 0.076) and significantly lower in week 8 (p = 0.037) and week 12 (p = 0.014) in the C. longa extract group than in the placebo group (Table 5).

3.3. Effect of C. longa Extract on Fasting Serum Glucose Levels

3.3.1. Study 1

In the low-hsCRP subgroup, the change in glucose from baseline did not show any significant differences between the two groups (Table 4). In the high-hsCRP subgroup, the change of glucose from baseline tended to be lower in the C. longa extract group than in the placebo group throughout the study period (p = 0.052 by r-ANOVA), and the serum glucose level was significantly lower in week 4 (p < 0.001), week 8 (p = 0.006), and week 12 (p < 0.001) in the C. longa extract group than in the placebo group (Table 4).

3.3.2. Study 2

In the low-hsCRP subgroup, the change of glucose from baseline showed no significant differences between the two groups throughout the study period (r-ANOVA), but the serum glucose level was significantly higher in week 4 (p = 0.003) in the C. longa extract group than in the placebo group (Table 5). In the high-hsCRP subgroup, the change of glucose from baseline showed no significant differences between the two groups throughout the study period (r-ANOVA), but the serum glucose level was significantly lower in week 12 (p = 0.022) in the C. longa extract group than in the placebo group (Table 5).

3.4. Safety of the Intervention

Adverse events were assessed in the ITT population (C. longa extract group, n = 45; placebo group, n = 45), and safety parameters were assessed in the FAS population (Figure 1 and Figure 2). These results were described in detail in the original research papers [27,28]. In brief, some adverse events were observed in both studies, but they were mild, and an experienced physician judged that they were unrelated to the dietary intervention. In study 1, the male participants showed elevated levels of aspartate aminotransferase and creatinine above the corresponding reference ranges, but the results of the other hematology and biochemistry tests, urinalysis, and physiological tests did not differ significantly between the two groups. In study 2, the safety parameters did not show any significant differences between the two groups.

4. Discussion

To investigate the effects of a hot water extract of C. longa on fasting serum glucose levels and assess the influence of low-grade inflammation, we reanalyzed our published data from two randomized, double-blind, placebo-controlled trials in overweight participants aged 50 to 69 [27,28]. We performed a stratified analysis by dividing participants into two groups based on baseline serum hsCRP level. In the low-hsCRP subgroup, we found no significant differences in serum glucose levels between the C. longa extract and placebo groups, but in the high-hsCRP subgroup, we found that intake of the C. longa extracts significantly improved levels of serum hsCRP and fasting serum glucose. These results suggest that intake of C. longa extract could potentially improve systemic glucose metabolism in people with low-grade inflammation.
Glucose metabolism is tightly regulated by various hormones, such as insulin and glucagon [1,3]. However, chronic low-grade inflammation is known to induce β-cell dysfunction and insulin resistance and disrupt glucose homeostasis [6]. The standard test for CRP cannot detect the small increases in CRP levels seen in low-grade inflammation, but the hsCRP assay can be used as a marker of such inflammation [8,10]. Epidemiological studies have reported that participants with a slight elevation of hsCRP levels (≥0.10 mg/dL) have a potential risk of impaired fasting glucose (a marker of prediabetes) and type 2 diabetes [13,14]. In previous large-scale observational studies, the risk of type 2 diabetes has been shown to be lower in people with anti-inflammatory agents such as aspirin than in people without these agents [39,40]. In the present study, the C. longa extract significantly improved the levels of serum hsCRP and fasting serum glucose in participants with mildly elevated hsCRP (≥0.098 mg/dL) in both clinical studies (Table 4 and Table 5). Therefore, these results suggest that C. longa extract may improve systemic glucose metabolism associated with chronic low-grade inflammation and could possibly decrease the risk of prediabetes and type 2 diabetes.
TNF-α and IL-1β are pro-inflammatory cytokines and are known to have negative effects on the action of insulin, such as impairing glucose uptake by inhibiting the phosphatidylinositol-3-kinase (PI3K) and protein kinase B (Akt) pathway and promoting the de-elopement of insulin resistance [41]. The transcription nuclear factor kappa B (NF-kB) is known to be activated by various inflammatory stimuli and induce the expression of inflammatory-associated genes, including TNF-α and IL-1β. Inhibition of NF-kB activation improved insulin resistance in leptin-deficient obese mouse models [42]. In addition, anti-inflammatory drugs targeting TNF-α, IL-1β, and NF-kB improved insulin resistance and fasting blood glucose levels in clinical studies [11,12]. In a study in a non-alcoholic steatohepatitis mouse model, the hot water extract of C. longa inhibited the hepatic production of TNF-α, IL-1β, and IL-6 [26], and in our earlier clinical studies, C. longa extract improved systemic inflammatory markers, including hsCRP, TNF-α, and IL-6 [27,28]. Some components in aqueous extracts of C. longa have anti-inflammatory activity. For example, turmeronol A and B derived from a water extract of C. longa were reported to inhibit the production of TNF-α, IL-1β, and IL-6 and the activation of NF-kB in activated macrophages [43]. Bisacurone, another component of C. longa extracts, also suppressed the NF-kB signaling pathway by inhibiting phosphorylation of NF-kB inhibitor protein alpha (IkBα) and nuclear translocation of NF-kB in endothelial cells stimulated with TNF-α [44]. Our results suggest that C. longa extract may improve systemic glucose levels. We hypothesize that it does so by suppressing the activation of the NF-kB signaling pathway and the production of inflammatory cytokines.
Peripheral tissues such as the pancreas, skeletal muscle, and liver are important organs for regulating systemic glucose homeostasis [3,45]. In response to hyperglycemia, pancreatic β-cells secrete insulin, which binds to its receptor, activates PI3/Akt signaling, induces the translocation of glucose transporter 4 to the plasma membrane, and promotes glucose uptake in various cells, such as skeletal muscle cells [3,45]. Hepatic glucose production contributes to regulating glucose homeostasis, and insulin decreases the systemic glucose level by inhibiting the production of glucose by the liver [46]. However, chronic inflammation is known to induce pancreatic β-cell injury and dysfunction, inhibit insulin-mediated glucose uptake and lead to hepatic insulin resistance, as characterized by impaired insulin-induced suppression of hepatic gluconeogenesis [8,46]. An aqueous extract of C. longa was reported to decrease lipid peroxidation-induced DNA damage [47], and an herb mixture containing C. longa extract was shown to protect pancreatic β-cell injury from an inflammatory cytokine cocktail consisting of TNF-α, IL-1β, and interferon-γ [48]. In addition, an ex vivo study found that an aqueous extract of C. longa increased the secretion of insulin in pancreatic tissue and promoted insulin-induced glucose uptake in muscle tissue by activating PI3K/Akt signaling [49,50]. Bisacurone also was shown to induce hepatic activation of AMP-activated kinase (AMPK) and peroxisome proliferator-activated receptor (PPAR)-α proteins in in vitro and in vivo studies [51]. Furthermore, in animal models, the administration of fenofibrate or aminoimidazole-4-carboxamide riboside (AICAR), activators of PPAR-α and AMPK, respectively, improved hepatic insulin resistance [52,53]. Thus, C. longa extract may reduce cell injury and dysfunction in pancreatic β-cells and improve glucose uptake and insulin resistance in peripheral tissues, resulting in the improvement of systemic glucose levels.

5. Conclusions

To investigate the effects a hot water extract of C. longa on fasting serum glucose levels and assess the influence of low-grade inflammation, we reanalyzed data from two randomized, double-blind, placebo-controlled trials in overweight participants aged 50 to 69 and performed a stratified analysis by dividing participants into two subgroups based on baseline hsCRP levels. In the low-hsCRP subgroup, we found no significant differences in the serum glucose level between the two groups. In contrast, in the high-hsCRP subgroup, the C. longa extract group showed significantly lower levels of serum hsCRP and fasting serum glucose than the placebo group. These results suggest that daily intake of a hot water extract of C. longa may have the potential to improve systemic glucose metabolism in people with chronic low-grade inflammation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu14183763/s1. Table S1: Baseline characteristics of the participants in study 1. Table S2: Baseline characteristics of the participants in study 2.

Author Contributions

Conceptualization, R.U., K.K., K.M., Y.Y., S.M., Y.Y. and Y.H.; methodology, R.U., K.K., K.M., Y.Y., S.M., Y.Y. and Y.H.; formal analysis, R.U., C.O.-H. and R.S.; investigation, R.U.; data curation, R.U., C.O.-H. and R.S.; writing—original draft preparation, R.U.; writing—review and editing, R.U., Y.H. and S.M.; visualization, R.U.; supervision, K.K., K.M., S.M., Y.Y. and Y.H.; project administration, K.K., K.M. and Y.H.; Y.H. had primary responsibility for the final content of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

These studies received no external funding and were conducted by CRO (CPCC Co., Ltd., Tokyo, Japan) with financial support from House Wellness Foods Corp (Itami, Japan). The article processing charges was founded by House Wellness Foods Corp.

Institutional Review Board Statement

The two studies were performed in accordance with the Declaration of Helsinki, were approved by the institutional review board of Chiyoda Paramedical Care Clinic, and registered with the UMIN (Registration number of study 1: UMIN000029095 and study 2: UMIN000037370).

Informed Consent Statement

All participants provided written informed consent.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

This research received no specific grant from any funding agency or from the commercial or not-for-profit sectors. The authors thank Ryuji Fukushima from ESUMI Co., Ltd. (Tokyo, Japan), a statistical consulting company, for providing helpful advice about statistical analysis; Fukushima was not involved in the study. In addition, the authors thank Jacquie Klesing (who was not involved in this study), Board-certified Editor in the Life Sciences (ELS) from Yamada Translation Bureau, Inc. (Osaka, Japan), for checking the English grammar of the manuscript.

Conflicts of Interest

All authors are employees of House Wellness Foods Corp., which markets health food products.

References

  1. Petersen, M.C.; Vatner, D.F.; Shulman, G.I. Regulation of hepatic glucose metabolism in health and disease. Nat. Rev. Endocrinol. 2017, 13, 572–587. [Google Scholar] [CrossRef] [PubMed]
  2. Szablewski, L. Glucose Homeostasis–Mechanism and Defects. Diabetes-Damages and Treatments; IntechOpen Ltd.: London, UK, 2011; Volume 2. [Google Scholar] [CrossRef]
  3. Röder, P.V.; Wu, B.; Liu, Y.; Han, W. Pancreatic regulation of glucose homeostasis. Exp. Mol. Med. 2016, 48, e219. [Google Scholar] [CrossRef] [PubMed]
  4. Roh, E.; Kim, M.-S. Emerging role of the brain in the homeostatic regulation of energy and glucose metabolism. Exp. Mol. Med. 2016, 48, e216. [Google Scholar] [CrossRef] [PubMed]
  5. Netea, M.G.; Balkwill, F.; Chonchol, M.; Cominelli, F.; Donath, M.Y.; Giamarellos-Bourboulis, E.J.; Golenbock, D.; Gresnigt, M.S.; Heneka, M.T.; Hoffman, H.M.; et al. A guiding map for inflammation. Nat. Immunol. 2017, 18, 826–831. [Google Scholar] [CrossRef]
  6. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
  7. Calder, P.C.; Bosco, N.; Bourdet-Sicard, R.; Capuron, L.; Delzenne, N.; Dore, J.; Franceschi, C.; Lehtinen, M.J.; Recker, T.; Salvioli, S.; et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res. Rev. 2017, 40, 95–119. [Google Scholar] [CrossRef]
  8. Leon-Pedroza, J.I.; Gonzalez-Tapia, L.A.; del Olmo-Gil, E.; Castellanos-Rodriguez, D.; Escobedo, G.; Gonzalez-Chavez, A. Low-grade systemic inflammation and the development of metabolic diseases: From the molecular evidence to the clinical practice. Cirugia Y Cirujanos 2015, 83, 543–551. [Google Scholar] [CrossRef]
  9. Kolb, H.; Mandrup-Poulsen, T. The global diabetes epidemic as a consequence of lifestyle-induced low-grade inflammation. Diabetologia 2010, 53, 10–20. [Google Scholar] [CrossRef]
  10. Kushner, I.; Samols, D.; Magrey, M. A unifying biologic explanation for “high-sensitivity” C-reactive protein and “low-grade” inflammation. Arthritis Care Res. 2010, 62, 442–446. [Google Scholar] [CrossRef]
  11. Donath, M.Y.; Shoelson, S.E. Type 2 diabetes as an inflammatory disease. Nat. Rev. Immunol. 2011, 11, 98–107. [Google Scholar] [CrossRef]
  12. Donath, M.Y. Targeting inflammation in the treatment of type 2 diabetes: Time to start. Nat. Rev. Drug Discov. 2014, 13, 465–476. [Google Scholar] [CrossRef]
  13. Matsuo, Y.; Hashizume, T.; Shioji, S.; Akasaka, T. Metabolic syndrome is strongly associated with chronic subclinical inflammation in patients achieving optimal low-density lipoprotein-cholesterol levels in secondary prevention of cardiovascular disease. Circ. J. 2008, 72, 2046. [Google Scholar] [CrossRef]
  14. Tamakoshi, K.; Yatsuya, H.; Kondo, T.; Hori, Y.; Ishikawa, M.; Zhang, H.; Murata, C.; Otsuka, R.; Zhu, S.; Toyoshima, H. The metabolic syndrome is associated with elevated circulating C-reactive protein in healthy reference range, a systemic low-grade inflammatory state. Int. J. Obes. 2003, 27, 443–449. [Google Scholar] [CrossRef]
  15. Amalraj, A.; Pius, A.; Gopi, S.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives–A review. J. Tradit. Complement. Med. 2017, 7, 205–233. [Google Scholar] [CrossRef]
  16. Aggarwal, B.B.; Yuan, W.; Li, S.; Gupta, S.C. Curcumin-free turmeric exhibits anti-inflammatory and anticancer activities: Identification of novel components of turmeric. Mol. Nutr. Food Res. 2013, 57, 1529–1542. [Google Scholar] [CrossRef]
  17. Kawasaki, K.; Muroyama, K.; Yamamoto, N.; Murosaki, S. A hot water extract of Curcuma longa inhibits adhesion molecule protein expression and monocyte adhesion to TNF-alpha-stimulated human endothelial cells. Biosci. Biotechnol. Biochem. 2015, 79, 1654–1659. [Google Scholar] [CrossRef]
  18. Kawasaki, K.; Okuda-Hanafusa, C.; Aoyagi, M.; Taoka, K.; Yamamoto, N.; Muroyama, K.; Murosaki, S.; Yamamoto, Y. Inhibitory effect of the compounds from the water extract of Curcuma longa on the production of PGE2 and NO in a macrophage cell line stimulated by LPS. Biosci. Biotechnol. Biochem. 2018, 82, 2109–2117. [Google Scholar] [CrossRef]
  19. Uchio, R.; Higashi, Y.; Kohama, Y.; Kawasaki, K.; Hirao, T.; Muroyama, K.; Murosaki, S. A hot water extract of turmeric (Curcuma longa) suppresses acute ethanol-induced liver injury in mice by inhibiting hepatic oxidative stress and inflammatory cytokine production. J. Nutr. Sci. 2017, 6, e3. [Google Scholar] [CrossRef]
  20. Kim, S.; Kim, K.; Park, J.; Jun, W. Curcuma longa L. Water Extract Improves Dexamethasone-Induced Sarcopenia by Modulating the Muscle-Related Gene and Oxidative Stress in Mice. Antioxidants 2021, 10, 1000. [Google Scholar] [CrossRef]
  21. Asada, K.; Ohara, T.; Muroyama, K.; Yamamoto, Y.; Murosaki, S. Effects of hot water extract of Curcuma longa on human epidermal keratinocytes in vitro and skin conditions in healthy participants: A randomized, double-blind, placebo-controlled trial. J. Cosmet. Dermatol. 2019, 18, 1866–1874. [Google Scholar] [CrossRef]
  22. Mehra, K.S.; Mikuni, I.; Gupta, U.; Gode, K.D. Curcuma longa (Linn) drops in corneal wound healing. Tokai J. Exp. Clin. Med. 1984, 9, 27–31. [Google Scholar]
  23. Kawasaki, K.; Muroyama, K.; Murosaki, S. Effect of a water extract of Curcuma longa on emotional states in healthy participants. Biosci. Microbiota Food Health 2018, 37, 25–29. [Google Scholar] [CrossRef]
  24. Anandakumar, S.; Joseph, J.A.; Bethapudi, B.; Agarwal, A.; Jung, E.-B. Anti-inflammatory effects of turmeric (Curcuma longa L.) extract on acute and chronic inflammation models. J. Korean Soc. Food Sci. Nutr. 2014, 43, 612–617. [Google Scholar] [CrossRef]
  25. Sengupta, M.; Sharma, G.D.; Chakraborty, B. Hepatoprotective and immunomodulatory properties of aqueous extract of Curcuma longa in carbon tetra chloride intoxicated Swiss albino mice. Asian Pac. J. Trop. Biomed. 2011, 1, 193–199. [Google Scholar] [CrossRef]
  26. Uchio, R.; Murosaki, S.; Ichikawa, H. Hot water extract of turmeric (Curcuma longa) prevents non-alcoholic steatohepatitis in mice by inhibiting hepatic oxidative stress and inflammation. J. Nutr. Sci. 2018, 7, e36. [Google Scholar] [CrossRef]
  27. Uchio, R.; Muroyama, K.; Okuda-Hanafusa, C.; Kawasaki, K.; Yamamoto, Y.; Murosaki, S. Hot Water Extract of Curcuma longa L. Improves Serum Inflammatory Markers and General Health in Subjects with Overweight or Prehypertension/Mild Hypertension: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2019, 11, 1822. [Google Scholar] [CrossRef]
  28. Uchio, R.; Kawasaki, K.; Okuda-Hanafusa, C.; Saji, R.; Muroyama, K.; Murosaki, S.; Yamamoto, Y.; Hirose, Y. Curcuma longa extract improves serum inflammatory markers and mental health in healthy participants who are overweight: A randomized, double-blind, placebo-controlled trial. Nutr. J. 2021, 20, 91. [Google Scholar] [CrossRef]
  29. Hussain, H.E.M.A. Hypoglycemic, hypolipidemic and antioxidant properties of combination of Curcumin from Curcuma longa, Linn, and partially purified product from Abroma augusta, Linn in streptozotocin induced diabetes. Indian J. Clin. Biochem. 2002, 17, 33–43. [Google Scholar] [CrossRef]
  30. Lee, S.-J.; Han, J.-M.; Lee, J.-S.; Son, C.-G.; Im, H.-J.; Jo, H.-K.; Yoo, H.-R.; Kim, Y.-S.; Seol, I.-C. ACE reduces metabolic abnormalities in a high-fat diet mouse model. Evid. Based Complement. Alternat. Med. 2015, 2015, 352647. [Google Scholar] [CrossRef]
  31. Shiwaku, K.; Anuurad, E.; Enkhmaa, B.; Nogi, A.; Kitajima, K.; Shimono, K.; Yamane, Y.; Oyunsuren, T. Overweight Japanese with body mass indexes of 23.0-24.9 have higher risks for obesity-associated disorders: A comparison of Japanese and Mongolians. Int. J. Obes. 2004, 28, 152–158. [Google Scholar] [CrossRef] [Green Version]
  32. Katabami, T.; Tsukiyama, H.; Tanabe, M.; Matsuba, R.; Murakami, M.; Nishine, A.; Shimizu, S.; Sakai, K.; Tanaka, Y.; Yanase, T. Development of a simple prediction model for adrenal crisis diagnosis. Sci. Rep. 2020, 10, 13546. [Google Scholar] [CrossRef]
  33. Muramoto, A.; Tsushita, K.; Kato, A.; Ozaki, N.; Tabata, M.; Endo, M.; Oike, Y.; Oiso, Y. Angiopoietin-like protein 2 sensitively responds to weight reduction induced by lifestyle intervention on overweight Japanese men. Nutr. Diabetes 2011, 1, e20. [Google Scholar] [CrossRef] [PubMed]
  34. Watanabe, A.; Tochio, T.; Kadota, Y.; Takahashi, M.; Kitaura, Y.; Ishikawa, H. Supplementation of 1-Kestose Modulates the Gut Microbiota Composition to Ameliorate Glucose Metabolism in Obesity-Prone Hosts. Nutrients 2021, 13, 2983. [Google Scholar] [CrossRef] [PubMed]
  35. Bogdanski, P.; Suliburska, J.; Szulinska, M.; Stepien, M.; Pupek-Musialik, D.; Jablecka, A. Green tea extract reduces blood pressure, inflammatory biomarkers, and oxidative stress and improves parameters associated with insulin resistance in obese, hypertensive patients. Nutr. Res. 2012, 32, 421–427. [Google Scholar] [CrossRef]
  36. Howell, D.C. Statistical Methods for Psychology; Cengage Learning: Belmont, CA, USA, 2009; pp. 461–483. [Google Scholar]
  37. Paschalis, V.; Theodorou, A.A.; Kyparos, A.; Dipla, K.; Zafeiridis, A.; Panayiotou, G.; Vrabas, I.S.; Nikolaidis, M.G. Low vitamin C values are linked with decreased physical performance and increased oxidative stress: Reversal by vitamin C supplementation. Eur. J. Nutr. 2016, 55, 45–53. [Google Scholar] [CrossRef]
  38. Matsumoto, M.; Kitada, Y.; Naito, Y. Endothelial function is improved by inducing microbial polyamine production in the gut: A randomized placebo-controlled trial. Nutrients 2019, 11, 1188. [Google Scholar] [CrossRef]
  39. Hu, F.B.; Meigs, J.B.; Li, T.Y.; Rifai, N.; Manson, J.E. Inflammatory markers and risk of developing type 2 diabetes in women. Diabetes 2004, 53, 693–700. [Google Scholar] [CrossRef]
  40. Hayashino, Y.; Hennekens, C.H.; Kurth, T. Aspirin use and risk of type 2 diabetes in apparently healthy men. Am. J. Med. 2009, 122, 374–379. [Google Scholar] [CrossRef]
  41. McArdle, M.A.; Finucane, O.M.; Connaughton, R.M.; McMorrow, A.M.; Roche, H.M. Mechanisms of obesity-induced inflammation and insulin resistance: Insights into the emerging role of nutritional strategies. Front. Endocrinol. 2013, 4, 52. [Google Scholar] [CrossRef]
  42. Chen, L.; Chen, R.; Wang, H.; Liang, F. Mechanisms Linking Inflammation to Insulin Resistance. Int. J. Endocrinol. 2015, 2015, 508409. [Google Scholar] [CrossRef] [Green Version]
  43. Okuda-Hanafusa, C.; Uchio, R.; Fuwa, A.; Kawasaki, K.; Muroyama, K.; Yamamoto, Y.; Murosaki, S. Turmeronol A and turmeronol B from Curcuma longa prevent inflammatory mediator production by lipopolysaccharide-stimulated RAW264. 7 macrophages, partially via reduced NF-κB signaling. Food Funct. 2019, 10, 5779–5788. [Google Scholar] [CrossRef]
  44. Sun, D.I.; Nizamutdinova, I.T.; Kim, Y.M.; Cai, X.F.; Lee, J.J.; Kang, S.S.; Kim, Y.S.; Kang, K.M.; Chai, G.Y.; Chang, K.C.; et al. Bisacurone inhibits adhesion of inflammatory monocytes or cancer cells to endothelial cells through down-regulation of VCAM-1 expression. Int. Immunopharmacol. 2008, 8, 1272–1281. [Google Scholar] [CrossRef]
  45. Merz, K.E.; Thurmond, D.C. Role of Skeletal Muscle in Insulin Resistance and Glucose Uptake. Compr. Physiol. 2020, 10, 785–809. [Google Scholar] [CrossRef]
  46. Samuel, V.T.; Shulman, G.I. Mechanisms for insulin resistance: Common threads and missing links. Cell 2012, 148, 852–871. [Google Scholar] [CrossRef]
  47. Shalini, V.; Srinivas, L. Lipid peroxide induced DNA damage: Protection by turmeric (Curcuma longa). Mol. Cell Biochem. 1987, 77, 3–10. [Google Scholar] [CrossRef]
  48. Damame, H.; Rooge, S.; Patil, R.; Garad, C.; Arvindekar, A. Beta cell protective effect of Curcuma longa and Piper nigrum in cytokine cocktail induced apoptosis in Min6 pancreatic beta cells. Phytomed. Plus 2021, 1, 100072. [Google Scholar] [CrossRef]
  49. Mohankumar, S.; McFarlane, J.R. An aqueous extract of Curcuma longa (turmeric) rhizomes stimulates insulin release and mimics insulin action on tissues involved in glucose homeostasis in vitro. Phytother. Res. 2011, 25, 396–401. [Google Scholar] [CrossRef]
  50. Kasabri, V.; Flatt, P.R.; Abdel-Wahab, Y.H. In vitro modulation of pancreatic insulin secretion, extrapancreatic insulin action and peptide glycation by Curcuma longa aqueous extracts. J. Exp. Integr. Med. 2014, 4, 187. [Google Scholar] [CrossRef]
  51. Ashida, H.; Tian, X.; Kitakaze, T.; Yamashita, Y. Bisacurone suppresses hepatic lipid accumulation through inhibiting lipogenesis and promoting lipolysis. J. Clin. Biochem. Nutr. 2020, 67, 43–52. [Google Scholar] [CrossRef]
  52. Iglesias, M.A.; Ye, J.M.; Frangioudakis, G.; Saha, A.K.; Tomas, E.; Ruderman, N.B.; Cooney, G.J.; Kraegen, E.W. AICAR administration causes an apparent enhancement of muscle and liver insulin action in insulin-resistant high-fat-fed rats. Diabetes 2002, 51, 2886–2894. [Google Scholar] [CrossRef] [Green Version]
  53. Chan, S.M.; Sun, R.Q.; Zeng, X.Y.; Choong, Z.H.; Wang, H.; Watt, M.J.; Ye, J.M. Activation of PPARα ameliorates hepatic insulin resistance and steatosis in high fructose-fed mice despite increased endoplasmic reticulum stress. Diabetes 2013, 62, 2095–2105. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Study flow diagram (CONSORT 2010) in study 1.
Figure 1. Study flow diagram (CONSORT 2010) in study 1.
Nutrients 14 03763 g001
Figure 2. Study flow diagram (CONSORT 2010) in study 2.
Figure 2. Study flow diagram (CONSORT 2010) in study 2.
Nutrients 14 03763 g002
Table 1. Composition of test diets.
Table 1. Composition of test diets.
Study 1Study 2
Placebo
(0.90 g/3 Tablets)
C. longa Extract
(0.90 g/3 Tablets)
Placebo
(0.97 g/2 Capsules)
C. longa Extract
(0.97 g/2 Capsules)
Energy, Kcal3.43.46.15.5
Carbohydrate, g0.820.760.240.23
Protein, g00.020.200.29
Lipid, g0.010.030.470.38
Sodium chloride, mg0.740.450.660.13
Bisacurone, μg04000400
Turmeronol A, μg0800100
Turmeronol B, μg0200100
Table 2. Baseline characteristics of the participants stratified by serum hsCRP levels in study 1 1.
Table 2. Baseline characteristics of the participants stratified by serum hsCRP levels in study 1 1.
Low-hsCRP Subgroup
(<0.098 mg/dL)
High-hsCRP Subgroup
(≥0.098 mg/dL)
Placebo
(n = 26)
C. longa Extract
(n = 29)
Placebo
(n = 18)
C. longa Extract
(n = 14)
MeanSDMeanSDMeanSDMeanSD
Sex, male/female, n16/1017/126/126/8
Age, y57.85.459.05.559.65.658.35.1
Physical measurements and tests
Height, cm163.89.1163.16.8159.75.8162.211.0
Body weight, kg67.410.865.47.365.06.968.78.6
BMI, kg/m225.12.924.51.525.41.926.12.0
SBP, mmHg134.618.3128.614.3125.713.2130.618.0
DBP, mmHg84.112.480.510.480.110.383.010.5
Serum inflammatory markers
hsCRP, mg/dL0.0550.0190.0590.0260.1820.0790.1650.068
Metabolic markers
Glucose, mg/dL83.25.885.84.785.76.286.610.4
HbA1c, %5.390.185.500.275.600.265.520.30
Triglyceride, mg/dL106.848.1118.679.5120.766.9148.262.0
Total cholesterol, mg/dL214.737.1210.133.2228.040.4234.130.0
LDL-cholesterol, mg/dL133.030.8126.130.1145.438.2144.426.0
HDL-cholesterol, mg/dL55.518.155.712.552.68.756.512.2
BMI, body mass index; C. longa, Curcuma longa L.; DBP, diastolic blood pressure; HbA1c, hemoglobin A1c; HDL, high-density lipoprotein; hsCRP, high-sensitivity C-reactive protein; LDL, low-density lipoprotein; SBP, systolic blood pressure. 1 Values represent the means and standard deviations for n = 26 (placebo group) or n = 29 (C. longa extract group) in the participants with a low-hsCRP level (<0.098 mg/dL). n = 18 (placebo group) or n = 14 (C. longa extract group) in the participants with a high-hsCRP level (≥0.098 mg/dL). Results in men and women were compared with the two-tailed Mann Whitney U-test, and physical measurements and tests, serum inflammatory markers, and metabolic markers were compared with the two-tailed unpaired Student’s t-test when variance was homogeneous or the Aspin-Welch t-test when variance was heterogeneous.
Table 3. Baseline characteristics of the participants stratified by serum hsCRP protein levels in study 2 1.
Table 3. Baseline characteristics of the participants stratified by serum hsCRP protein levels in study 2 1.
Low-hsCRP Subgroup
(<0.098 mg/dL)
High-hsCRP Subgroup
(≥0.098 mg/dL)
Placebo
(n = 19)
C. longa Extract
(n = 26)
Placebo
(n = 21)
C. longa Extract
(n = 13)
MeanSDMeanSDMeanSDMeanSD
Sex, male/female, n8/1111/158/134/9
Age, y56.54.456.14.656.74.457.95.7
Physical measurements and tests
Height, cm164.48.7162.87.8160.88.1161.910.1
Body weight, kg70.57.771.06.969.48.468.99.5
BMI, kg/m226.11.826.81.526.81.726.21.6
SBP, mmHg120.814.0121.314.1124.811.6116.516.1
DBP, mmHg76.911.678.39.478.29.076.210.1
Serum inflammatory markers
hsCRP, mg/dL0.0420.0250.0490.0310.1260.0160.1420.078
Metabolic markers
Glucose, mg/dL93.510.888.06.991.16.389.55.5
HbA1c, %5.630.225.580.265.630.195.480.26
Triglyceride, mg/dL125.261.0128.775.1116.745.5109.145.5
Total cholesterol, mg/dL229.846.6223.438.4232.826.1216.836.8
LDL-cholesterol, mg/dL145.940.5142.233.6151.122.3130.2 *27.5
HDL-cholesterol, mg/dL59.711.255.712.056.712.763.323.0
BMI, body mass index; C. longa, Curcuma longa L.; DBP, diastolic blood pressure; HbA1c, hemo-globin A1c; HDL, high-density lipoprotein; hsCRP, high-sensitivity C-reactive protein; LDL, low-density lipoprotein; SBP, systolic blood pressure. 1 Values represent the means and standard deviations for n = 19 (placebo group) or n = 26 (C. longa extract group) in the participants with a low-hsCRP level (<0.098 mg/dL) and n = 21 (placebo group) or n = 13 (C. longa extract group) in the participants with a high-hsCRP level (≥0.098 mg/dL). * p < 0.05 vs. placebo group. Results in men and women were compared with the two-tailed Mann Whitney U-test, and physical measurements and tests, serum inflammatory markers, and metabolic markers were compared with the two-tailed unpaired Student’s t-test when variance was homogeneous or the Aspin-Welch t-test when variance was heterogeneous.
Table 4. Effect of C. longa extract on serum hsCRP and glucose levels stratified by baseline hsCRP levels in the study 1 1.
Table 4. Effect of C. longa extract on serum hsCRP and glucose levels stratified by baseline hsCRP levels in the study 1 1.
Change from BaselineRepeated Measures Two-Way ANOVA
Week 4Week 8Week 12GroupTimeInteraction
MeanSDMeanSDMeanSD
All study participants
hsCRP, mg/dL
Placebo0.0160.0720.0360.0920.0570.1380.0770.0980.243
C. longa extract0.0060.086−0.001 *0.0710.015 **0.068
Glucose, mg/dL
Placebo2.24.62.74.72.05.00.0700.1530.900
C. longa extract0.1 **6.51.0 *5.5−0.1 **6.6
Stratified analysis
Low-hsCRP subgroup (<0.098 mg/dL)
hsCRP, mg/dL
Placebo0.0420.0640.0550.0800.0830.1500.0130.2810.387
C. longa extract0.0230.0640.0200.0480.026 **0.049
Glucose, mg/dL
Placebo1.24.62.44.71.45.30.5760.3960.829
C. longa extract1.06.11.34.50.85.3
High-hsCRP subgroup (≥0.098 mg/dL)
hsCRP, mg/dL
Placebo−0.0260.0650.0070.1030.0170.1110.3060.2070.233
C. longa extract−0.0410.124−0.056 **0.091−0.0170.101
Glucose, mg/dL
Placebo3.64.43.14.82.94.40.0520.1800.193
C. longa extract−1.4 **7.30.4 **7.6−2.0 **8.8
ANOVA, analysis of variance; C. longa, Curcuma longa L.; hsCRP, high-sensitivity C-reactive protein. 1 Values represent the means and standard deviations for n = 44 (placebo group) or n = 43 (C. longa extract group) in all study participants, n = 26 (placebo group) or n = 29 (C. longa extract group) in the participants with a low-hsCRP level (<0.098 mg/dL) and n = 18 (placebo group) or n = 14 (C. longa extract group) in the participants with a high-hsCRP level (≥0.098 mg/dL). * p < 0.05 and ** p < 0.01 vs. placebo group, as assessed by repeated measures two-way ANOVA followed by a simple test of main effect.
Table 5. Effect of C. longa extract on serum hsCRP and glucose levels stratified by baseline hsCRP levels in study 2 1.
Table 5. Effect of C. longa extract on serum hsCRP and glucose levels stratified by baseline hsCRP levels in study 2 1.
Change from BaselineRepeated Measures Two-Way ANOVA
Week 4Week 8Week 12GroupTimeInteraction
MeanSDMeanSDMeanSD
All study participants
hsCRP, mg/dL
Placebo0.0190.0440.0360.0910.0160.0470.0570.3630.302
C. longa extract0.0210.0890.002 *0.032−0.0070.035
Glucose, mg/dL
Placebo−2.46.3−1.36.3−0.76.10.9670.1890.433
C. longa extract−1.45.5−1.87.2−0.96.3
Stratified analysis
Low-hsCRP subgroup (<0.098 mg/dL)
hsCRP, mg/dL
Placebo0.0110.0450.0390.1030.0190.0500.1010.0600.831
C. longa extract0.0330.0950.0100.0280.0080.022
Glucose, mg/dL
Placebo−3.95.5−1.56.0−1.35.60.2350.0660.193
C. longa extract−0.6 *5.8−1.06.00.46.8
High-hsCRP subgroup (≥0.098 mg/dL)
hsCRP, mg/dL
Placebo0.0310.0410.0310.0760.0100.0430.0010.3340.924
C. longa extract−0.0240.039−0.019 *0.035−0.047 *0.034
Glucose, mg/dL
Placebo−1.06.8−1.06.7−0.16.60.1870.9490.790
C. longa extract−3.24.8−3.49.2−3.6 *4.3
ANOVA, analysis of variance; C. longa, Curcuma longa L.; hsCRP, high-sensitivity C-reactive protein. 1 Values represent the means and standard deviations for n = 40 (placebo group) or n = 39 (C. longa extract group) in all study participants, n = 19 (placebo group) or n = 26 (C. longa extract group) in the participants with a low-hsCRP level (<0.098 mg/dL) and n = 21 (placebo group) or n = 13 (C. longa extract group) in the participants with a high-hsCRP level (≥0.098 mg/dL). * p < 0.05 vs. placebo group, as assessed by repeated measures two-way ANOVA followed by a simple test of main effect.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Uchio, R.; Okuda-Hanafusa, C.; Saji, R.; Kawasaki, K.; Muroyama, K.; Murosaki, S.; Yamamoto, Y.; Hirose, Y. A Hot Water Extract of Curcuma longa L. Improves Fasting Serum Glucose Levels in Participants with Low-Grade Inflammation: Reanalysis of Data from Two Randomized, Double-Blind, Placebo-Controlled Trials. Nutrients 2022, 14, 3763. https://doi.org/10.3390/nu14183763

AMA Style

Uchio R, Okuda-Hanafusa C, Saji R, Kawasaki K, Muroyama K, Murosaki S, Yamamoto Y, Hirose Y. A Hot Water Extract of Curcuma longa L. Improves Fasting Serum Glucose Levels in Participants with Low-Grade Inflammation: Reanalysis of Data from Two Randomized, Double-Blind, Placebo-Controlled Trials. Nutrients. 2022; 14(18):3763. https://doi.org/10.3390/nu14183763

Chicago/Turabian Style

Uchio, Ryusei, Chinatsu Okuda-Hanafusa, Ryosuke Saji, Kengo Kawasaki, Koutarou Muroyama, Shinji Murosaki, Yoshihiro Yamamoto, and Yoshitaka Hirose. 2022. "A Hot Water Extract of Curcuma longa L. Improves Fasting Serum Glucose Levels in Participants with Low-Grade Inflammation: Reanalysis of Data from Two Randomized, Double-Blind, Placebo-Controlled Trials" Nutrients 14, no. 18: 3763. https://doi.org/10.3390/nu14183763

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