[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (157)

Search Parameters:
Keywords = NLR family

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 7020 KiB  
Article
Antidiabetic and Antihyperlipidemic Activities and Molecular Mechanisms of Phyllanthus emblica L. Extract in Mice on a High-Fat Diet
by Hsing-Yi Lin, Cheng-Hsiu Lin, Yueh-Hsiung Kuo and Chun-Ching Shih
Curr. Issues Mol. Biol. 2024, 46(9), 10492-10529; https://doi.org/10.3390/cimb46090623 - 20 Sep 2024
Viewed by 683
Abstract
We planned to explore the protective activities of extract of Phyllanthus emblica L. (EPE) on insulin resistance and metabolic disorders including hyperlipidemia, visceral obesity, and renal dysfunction in high-fat diet (HFD)-progressed T2DM mice. Mice treatments included 7 weeks of HFD induction followed by [...] Read more.
We planned to explore the protective activities of extract of Phyllanthus emblica L. (EPE) on insulin resistance and metabolic disorders including hyperlipidemia, visceral obesity, and renal dysfunction in high-fat diet (HFD)-progressed T2DM mice. Mice treatments included 7 weeks of HFD induction followed by EPE, fenofibrate (Feno), or metformin (Metf) treatment daily for another 4-week HFD in HFD-fed mice. Finally, we harvested blood to analyze some tests on circulating glycemia and blood lipid levels. Western blotting analysis was performed on target gene expressions in peripheral tissues. The present findings indicated that EPE treatment reversed the HFD-induced increases in blood glucose, glycosylated HbA1C, and insulin levels. Our findings proved that treatment with EPE in HFD mice effectively controls hyperglycemia and hyperinsulinemia. Our results showed that EPE reduced blood lipid levels, including a reduction in blood triglyceride (TG), total cholesterol (TC), and free fatty acid (FFA); moreover, EPE reduced blood leptin levels and enhanced adiponectin concentrations. EPE treatment in HFD mice reduced BUN and creatinine in both blood and urine and lowered albumin levels in urine; moreover, EPE decreased circulating concentrations of inflammatory NLR family pyrin domain containing 3 (NLRP3) and kidney injury molecule-1 (KIM-1). These results indicated that EPE displayed antihyperglycemic and antihyperlipidemic activities but alleviated renal dysfunction in HFD mice. The histology examinations indicated that EPE treatment decreased adipose hypertrophy and hepatic ballooning, thus contributing to amelioration of lipid accumulation. EPE treatment decreased visceral fat amounts and led to improved systemic insulin resistance. For target gene expression levels, EPE enhanced AMP-activated protein kinase (AMPK) phosphorylation expressions both in livers and skeletal muscles and elevated the muscular membrane glucose transporter 4 (GLUT4) expressions. Treatment with EPE reduced hepatic glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) expressions to suppress glucose production in the livers and decreased phosphorylation of glycogen synthase kinase 3β (GSK3β) expressions to affect hepatic glycogen synthesis, thus convergently contributing to an antidiabetic effect and improving insulin resistance. The mechanism of the antihyperlipidemic activity of EPE involved a decrease in the hepatic phosphorylation of mammalian target of rapamycin complex C1 (mTORC1) and p70 S6 kinase 1 (S6K1) expressions to improve insulin resistance but also a reduction in hepatic sterol regulatory element binding protein (SREBP)-1c expressions, and suppression of ACC activity, thus resulting in the decreased fatty acid synthesis but elevated hepatic peroxisome proliferator-activated receptor (PPAR) α and SREBP-2 expressions, resulting in lowering TG and TC concentrations. Our results demonstrated that EPE improves insulin resistance and ameliorates hyperlipidemia in HFD mice. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>The fruits of <span class="html-italic">Phyllanthus emblica</span> L.</p>
Full article ">Figure 2
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on the insulin (Ins)-stimulated expression levels of membrane glucose transporter type 4 (GLUT4), the ratio of phospho-5′-adenosine monophosphate kinase (p-AMPK) to total AMPK (t-AMPK), or phospho-Akt (p-Akt)/total Akt (t-Akt) in insulin-resistant C2C12 myotube cells induced by palmitate (Pal). The symbols “+++”, “###” and “***” represent <span class="html-italic">p</span> &lt; 0.001 as, respectively, compared to the value of the blank control, positive control (insulin) and negative control (insulin + palmitate) using analysis of variance (ANOVA) and with Dunnett’s tests. (<b>A</b>) Representative image, (<b>B</b>–<b>D</b>) quantification of the membrane GLUT4 expression levels, the ratio of p-AMPK to t-AMPK, or p-Akt/t-Akt expression levels. CON, the blank control; DMSO, a solvent control. All values are means ± SE (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on the insulin (Ins)-stimulated expression levels of membrane glucose transporter type 4 (GLUT4), the ratio of phospho-5′-adenosine monophosphate kinase (p-AMPK) to total AMPK (t-AMPK), or phospho-Akt (p-Akt)/total Akt (t-Akt) in insulin-resistant C2C12 myotube cells induced by palmitate (Pal). The symbols “+++”, “###” and “***” represent <span class="html-italic">p</span> &lt; 0.001 as, respectively, compared to the value of the blank control, positive control (insulin) and negative control (insulin + palmitate) using analysis of variance (ANOVA) and with Dunnett’s tests. (<b>A</b>) Representative image, (<b>B</b>–<b>D</b>) quantification of the membrane GLUT4 expression levels, the ratio of p-AMPK to t-AMPK, or p-Akt/t-Akt expression levels. CON, the blank control; DMSO, a solvent control. All values are means ± SE (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on the insulin (Ins)-stimulated expression levels of membrane glucose transporter type 4 (GLUT4), the ratio of phospho-5′-adenosine monophosphate kinase (p-AMPK) to total AMPK (t-AMPK), or phospho-Akt (p-Akt)/total Akt (t-Akt) in insulin-resistant C2C12 myotube cells induced by palmitate (Pal). The symbols “+++”, “###” and “***” represent <span class="html-italic">p</span> &lt; 0.001 as, respectively, compared to the value of the blank control, positive control (insulin) and negative control (insulin + palmitate) using analysis of variance (ANOVA) and with Dunnett’s tests. (<b>A</b>) Representative image, (<b>B</b>–<b>D</b>) quantification of the membrane GLUT4 expression levels, the ratio of p-AMPK to t-AMPK, or p-Akt/t-Akt expression levels. CON, the blank control; DMSO, a solvent control. All values are means ± SE (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) on (<b>A</b>) final body weight; (<b>B</b>) body weight gain over 4-week treatment; (<b>C</b>) absolute fat tissue weight; (<b>D</b>) absolute weights of spleen and pancreas; (<b>E</b>) blood glucose levels; (<b>F</b>) blood glycated hemoglobin (HbA1<sub>C</sub>) levels; (<b>G</b>) triglyceride levels; (<b>H</b>) total cholesterol levels; (<b>I</b>) insulin levels; (<b>J</b>) leptin levels; (<b>K</b>) adiponectin levels; (<b>L</b>) free fatty acid levels; (<b>M</b>) blood BUN levels; (<b>N</b>) plasma creatinine levels; (<b>O</b>) urine BUN levels; (<b>P</b>) urine creatinine levels; (<b>Q</b>) urine albumin levels; (<b>R</b>) blood kidney injury molecule-1 (KIM-1) levels; (<b>S</b>) blood CRP levels; and (<b>T</b>) blood NLRP3 levels in high-fat diet (HFD)-induced diabetic mice. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). The extracts of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Meft, metformin (300 mg/kg body weight). EWAT, epididymal white adipose tissue; RWAT, retroperioneal white adipose tissue; MWAT, mesenteric white adipose tissue.</p>
Full article ">Figure 4
<p>Histology examinations on (<b>A</b>) liver tissues (at 400×) and (<b>B</b>) white adipose tissues of mice (at 400×) in the control (CON), high-fat diet (HFD) plus vehicle (distilled water) (HF), HFD + EPE1, HFD + EPE2, HFD + EPE3, HFD + fenofibrate (Feno), or HFD + metformin (Metf) groups (<span class="html-italic">n</span> = 8 per group) by hematoxylin and eosin staining. The ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), EPE1, 100, EPE2, 200, EPE3, 400 mg/kg body weight; Feno, fenofibrate (250 mg/kg body weight); Metf, metformin (300 mg/kg body weight).</p>
Full article ">Figure 5
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The expression levels of (<b>A</b>,<b>B</b>) membrane GLUT4, p-AMPK (Thr<sup>172</sup>)/t-AMPK, (<b>C</b>,<b>D</b>) PEPCK, G6Pase, p-GSK3β/t-GSK3β, (<b>E</b>,<b>F</b>) p-mTORC1/t-mTORC1, p-p70S6K1/t-p70S6K1, p-ACC/t-ACC, (<b>G</b>–<b>K</b>) PPARα, FAS, PPARγ, SREBP1<sub>C</sub>, SREBP2 and (<b>L</b>,<b>M</b>) p-Akt (Ser<sup>473</sup>)/t-Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>)/t-FoxO1 (Ser<sup>256</sup>) in the skeletal muscles, livers, or adipose tissues of high-fat diet (HFD)-induced diabetic mice by oral gavage ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE). (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>J</b>,<b>L</b>) representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>I</b>,<b>K</b>,<b>M</b>) quantification of the p-AMPK to t-AMPK, p-GSK3β to t-GSK3β, p-mTORC1 to t-mTORC1, p-p70S6K1 to t-p70S6K1, p-ACC to t-ACC p-Akt (Ser<sup>473</sup>) to t- Akt (Ser<sup>473</sup>), and p-FoxO1 (Ser<sup>256</sup>) to t-FoxO1 (Ser<sup>256</sup>). Protein was separated by 12% SDS-PAGE detected by Western blot. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control (CON) group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the high-fat diet (HFD) plus vehicle (distilled water) (HF) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE1, EPE2, and EPE3, ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) (EPE1, EPE2, and EPE3, 100, 200, and 400 mg/kg body weight, resp.); fenofibrate (Feno; 250 mg/kg body weight); metformin (Metf, 300 mg/kg body weight).</p>
Full article ">Figure 6
<p>Effects of seven fractions of EPE (EA) on expression levels of phospho-AMPK/total-AMPK in C2C12 myoblasts cells by Western blotting analyses. C2C12 skeletal myoblast cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>,<b>B</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of phospho-AMPK (Thr<sup>172</sup>) in C2C12 myoblasts cells by Western blotting analysis. C2C12 skeletal myoblast cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>) Representative blots for 7 fractions in C2C12 myoblasts cells; (<b>B</b>) quantification of the expression levels of the ratio of phospho-AMPK to total-AMPK. All values are means ± S.E. <sup>+++</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the insulin group ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the palmitate+ insulin group. (<b>C</b>,<b>D</b>) High-performance liquid chromatography analysis of (<b>C</b>) 2500 ppm ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), (<b>D</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. [<a href="#B55-cimb-46-00623" class="html-bibr">55</a>].</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of phospho-AMPK/total-AMPK in C2C12 myoblasts cells by Western blotting analyses. C2C12 skeletal myoblast cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>,<b>B</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of phospho-AMPK (Thr<sup>172</sup>) in C2C12 myoblasts cells by Western blotting analysis. C2C12 skeletal myoblast cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>) Representative blots for 7 fractions in C2C12 myoblasts cells; (<b>B</b>) quantification of the expression levels of the ratio of phospho-AMPK to total-AMPK. All values are means ± S.E. <sup>+++</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the insulin group ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the palmitate+ insulin group. (<b>C</b>,<b>D</b>) High-performance liquid chromatography analysis of (<b>C</b>) 2500 ppm ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), (<b>D</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. [<a href="#B55-cimb-46-00623" class="html-bibr">55</a>].</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of phospho-AMPK/total-AMPK in C2C12 myoblasts cells by Western blotting analyses. C2C12 skeletal myoblast cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>,<b>B</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of phospho-AMPK (Thr<sup>172</sup>) in C2C12 myoblasts cells by Western blotting analysis. C2C12 skeletal myoblast cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for phospho-AMPK (Thr<sup>172</sup>). (<b>A</b>) Representative blots for 7 fractions in C2C12 myoblasts cells; (<b>B</b>) quantification of the expression levels of the ratio of phospho-AMPK to total-AMPK. All values are means ± S.E. <sup>+++</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the insulin group ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the palmitate+ insulin group. (<b>C</b>,<b>D</b>) High-performance liquid chromatography analysis of (<b>C</b>) 2500 ppm ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. (EPE), (<b>D</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">Phyllanthus emblica</span> L. [<a href="#B55-cimb-46-00623" class="html-bibr">55</a>].</p>
Full article ">Figure 7
<p>The graphic abstract of ethyl acetate extract of <span class="html-italic">Phyllanthus emblica</span> L. (EPE) in high-fat diet (HFD)-induced diabetic mice.</p>
Full article ">
20 pages, 5013 KiB  
Article
Examination of the Expression Profile of Resistance Genes in Yuanjiang Common Wild Rice (Oryza rufipogon)
by Wang Kan, Ling Chen, Bo Wang, Li Liu, Fuyou Yin, Qiaofang Zhong, Jinlu Li, Dunyu Zhang, Suqin Xiao, Yun Zhang, Cong Jiang, Tengqiong Yu, Yunyue Wang and Zaiquan Cheng
Genes 2024, 15(7), 924; https://doi.org/10.3390/genes15070924 - 16 Jul 2024
Viewed by 582
Abstract
The rice blight poses a significant threat to the rice industry, and the discovery of disease-resistant genes is a crucial strategy for its control. By exploring the rich genetic resources of Yuanjiang common wild rice (Oryza rufipogon) and analyzing their expression [...] Read more.
The rice blight poses a significant threat to the rice industry, and the discovery of disease-resistant genes is a crucial strategy for its control. By exploring the rich genetic resources of Yuanjiang common wild rice (Oryza rufipogon) and analyzing their expression patterns, genetic resources can be provided for molecular rice breeding. The target genes’ expression patterns, subcellular localization, and interaction networks were analyzed based on the annotated disease-resistant genes on the 9th and 10th chromosomes in the rice genome database using fluorescent quantitative PCR technology and bioinformatics tools. Thirty-three disease-resistant genes were identified from the database, including 20 on the 9th and 13 on the 10th. These genes were categorized into seven subfamilies of the NLR family, such as CNL and the G subfamily of the ABC family. Four genes were not expressed under the induction of the pathogen Y8, two genes were significantly down-regulated, and the majority were up-regulated. Notably, the expression levels of nine genes belonging to the ABCG, CN, and CNL classes were significantly up-regulated, yet the expression levels varied among roots, stems, and leaves; one was significantly expressed in the roots, one in the stems, and the remaining seven were primarily highly expressed in the leaves. Two interaction network diagrams were predicted based on the seven highly expressed genes in the leaves: complex networks regulated by CNL proteins and specific networks controlled by ABCG proteins. The disease-resistant genes on the 9th chromosome are actively expressed in response to the induction of rice blight, forming a critical gene pool for the resistance of Yuanjiang common wild rice (O. rufipogon) to rice blight. Meanwhile, the disease-resistant genes on the 10th chromosome not only participate in resisting the rice blight pathogen but may also be involved in the defense against other stem diseases. Full article
(This article belongs to the Section Plant Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>NLR and ABC family genes downloaded from rice chromosomes 9 and 10. Classification of 33 key genes (<b>A</b>). Distribution of genes on chromosomes 9 and 10 (<b>B</b>). Protein sequence length of 33 key genes (<b>C</b>). 33 protein sequence lengths (<b>D</b>).</p>
Full article ">Figure 2
<p>Expression of ABCG subfamily genes in response to the pathogen Y8. (<b>A</b>–<b>D</b>) represent genes <span class="html-italic">Os09g16330</span>, <span class="html-italic">Os09g16380</span>, <span class="html-italic">Os09g16449</span>, and <span class="html-italic">Os09g16458</span>.</p>
Full article ">Figure 3
<p>Expression of CC-type and CC-NBS-type NLR family genes in response to the pathogen Y8. (<b>A</b>–<b>E</b>) represent genes <span class="html-italic">Os09g15840</span>, <span class="html-italic">Os10g22290</span>, <span class="html-italic">Os10g04342</span>, <span class="html-italic">Os09g09490</span>, and <span class="html-italic">Os10g10360</span>.</p>
Full article ">Figure 4
<p>Expression of CC-NBS-LRR-type NLR family genes in response to the pathogen Y8. (<b>A</b>–<b>L</b>) represent genes <span class="html-italic">Os09g14060</span>, <span class="html-italic">Os10g33440</span>, <span class="html-italic">Os09g10054</span>, <span class="html-italic">Os09g30220</span>, <span class="html-italic">Os10g04674</span>, <span class="html-italic">Os09g20040</span>, <span class="html-italic">Os10g07400</span>, <span class="html-italic">Os09g34150</span>, <span class="html-italic">Os09g09750</span>, <span class="html-italic">Os09g14010</span>, <span class="html-italic">Os09g11020</span>, and <span class="html-italic">Os09g34160</span>.</p>
Full article ">Figure 4 Cont.
<p>Expression of CC-NBS-LRR-type NLR family genes in response to the pathogen Y8. (<b>A</b>–<b>L</b>) represent genes <span class="html-italic">Os09g14060</span>, <span class="html-italic">Os10g33440</span>, <span class="html-italic">Os09g10054</span>, <span class="html-italic">Os09g30220</span>, <span class="html-italic">Os10g04674</span>, <span class="html-italic">Os09g20040</span>, <span class="html-italic">Os10g07400</span>, <span class="html-italic">Os09g34150</span>, <span class="html-italic">Os09g09750</span>, <span class="html-italic">Os09g14010</span>, <span class="html-italic">Os09g11020</span>, and <span class="html-italic">Os09g34160</span>.</p>
Full article ">Figure 5
<p>Expression of NLR family genes with non-CC domains in response to the pathogen Y8. (<b>A</b>–<b>H</b>) represent genes <span class="html-italic">Os10g36270</span>, <span class="html-italic">Os09g14060</span>, <span class="html-italic">Os10g03100</span>, <span class="html-italic">Os10g25487</span>, <span class="html-italic">Os09g20020</span>, <span class="html-italic">Os10g07978</span>, <span class="html-italic">Os09g30230</span>, and <span class="html-italic">Os09g20030</span>.</p>
Full article ">Figure 6
<p>Tissue-specific expression of the top 10 genes highly induced by the pathogen Y8. The data are presented as the mean χ ± SE (<span class="html-italic">n</span> = 3) of three biological replicates. Analysis of variance was conducted using Bonferroni’s multiple comparisons test. ** means significant differences at the 0.05 level. <sup>ns</sup> means no significant differences.</p>
Full article ">Figure 7
<p>Construction and analysis of the ABCG protein regulatory network. Protein interactions mediated by ABCG-type proteins Os09g16458 and Os09g16449 (<b>A</b>). Interaction information between Os09g16458 and Os09g16449 with ten predicted proteins (<b>B</b>). COE: Co-expression. COS: Score of protein interactions predicted based on amino acid domain structures.</p>
Full article ">Figure 8
<p>Construction and analysis of the CNL protein regulatory network. Protein interactions mediated by CNL-type (<b>A</b>). Interaction information between CNL with ten predicted proteins (<b>B</b>). PHC: Phylogenetic co-occurrence. COE: Co-expression. EDI: Experimentally determined interaction. COS: Score of protein interactions predicted based on amino acid domain structures.</p>
Full article ">Figure 9
<p>Amino acid sequence comparison of <span class="html-italic">Os09g34150</span> and <span class="html-italic">Os09g34160.</span> Blue and white are differential amino acids; red is the same amino acid.</p>
Full article ">
22 pages, 1138 KiB  
Review
The Novel Role of the NLRP3 Inflammasome in Mycotoxin-Induced Toxicological Mechanisms
by Chengshui Liao, Fengru Xu, Zuhua Yu, Ke Ding and Yanyan Jia
Vet. Sci. 2024, 11(7), 291; https://doi.org/10.3390/vetsci11070291 - 28 Jun 2024
Cited by 1 | Viewed by 1029
Abstract
Mycotoxins are secondary metabolites produced by several fungi and moulds that exert toxicological effects on animals including immunotoxicity, genotoxicity, hepatotoxicity, teratogenicity, and neurotoxicity. However, the toxicological mechanisms of mycotoxins are complex and unclear. The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain [...] Read more.
Mycotoxins are secondary metabolites produced by several fungi and moulds that exert toxicological effects on animals including immunotoxicity, genotoxicity, hepatotoxicity, teratogenicity, and neurotoxicity. However, the toxicological mechanisms of mycotoxins are complex and unclear. The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome is a multimeric cytosolic protein complex composed of the NLRP3 sensor, ASC adapter protein, and caspase-1 effector. Activation of the NLRP3 inflammasome plays a crucial role in innate immune defence and homeostatic maintenance. Recent studies have revealed that NLRP3 inflammasome activation is linked to tissue damage and inflammation induced by mycotoxin exposure. Thus, this review summarises the latest advancements in research on the roles of NLRP3 inflammasome activation in the pathogenesis of mycotoxin exposure. The effects of exposure to multiple mycotoxins, including deoxynivalenol, aflatoxin B1, zearalenone, T-2 toxin, ochratoxin A, and fumonisim B1, on pyroptosis-related factors and inflammation-related factors in vitro and in vivo and the pharmacological inhibition of specific and nonspecific NLRP3 inhibitors are summarized and examined. This comprehensive review contributes to a better understanding of the role of the NLRP3 inflammasome in toxicity induced by mycotoxin exposure and provides novel insights for pharmacologically targeting NLRP3 as a novel anti-inflammatory agent against mycotoxin exposure. Full article
(This article belongs to the Special Issue Nutraceuticals to Mitigate the Secret Killers in Animals)
Show Figures

Figure 1

Figure 1
<p>Depiction of activation of an NLRP3 inflammasome by mycotoxin (DON, AFB1, ZEA, OTA, and FB1) exposure (created in Figdraw). In the case of DON exposure, the first signal comes from TLR4 and NF-κB and increases the intracellular transcripts of pro-IL-1β, pro-IL-18, and NLRP3. The NLRP3 inflammasome is activated by NLRP3 oligomerization with ASC and pro-caspase-1. Caspase-1 cleavage of GSDMD forms active N-termini, causeing cell membrane perforation. Active caspase-1 then converts pro-IL-1β and pro-IL-18 to their biologically active structures, causing the release of IL-1β and IL-18 into the extracellular matrix, amplifying inflammation, and inducing pyroptosis. DON, deoxynivalenol; AFB1, aflatoxin B1; ZEA, zearalenone; OTA, ochratoxin A; FB1, fumonisim B1; TLR4, toll-like receptor 4; Keap1, kelch-like ECH-associated protein 1; Nrf2, nuclear factor erythroid-derived 2-like 2; JAK2, Janus kinase 2; STAT3, signal transducer and activator of transcription 3; TXNIP, thioredoxin interacting protein; ROS, reactive oxygen species; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3, nucleotide-binding, oligomerization domain (NOD)-like receptor (NLR) family pyrin domain containing 3; ASC, apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain; GSDMD, gasdermin D; GSDMD-N, gasdermin D N-terminal; GSDME, gasdermin E; GSDME-N, gasdermin E N-terminal; IL, interleukin.</p>
Full article ">
18 pages, 6999 KiB  
Article
Integrated Transcriptome and Metabolome Analysis Reveals Molecular Mechanisms Underlying Resistance to Phytophthora Root Rot
by Ruidong Sun, Anan Han, Haitang Wang, Congcong Wang, Yang Lu, Danqing Ni, Na Guo, Han Xing and Jinming Zhao
Plants 2024, 13(12), 1705; https://doi.org/10.3390/plants13121705 - 19 Jun 2024
Viewed by 1076
Abstract
Soybean production is significantly impacted by Phytophthora root rot (PRR), which is caused by Phytophthora sojae. The nucleotide-binding leucine-rich repeat (NLR) gene family plays a crucial role in plant disease resistance. However, current understanding of the function of soybean NLR genes in [...] Read more.
Soybean production is significantly impacted by Phytophthora root rot (PRR), which is caused by Phytophthora sojae. The nucleotide-binding leucine-rich repeat (NLR) gene family plays a crucial role in plant disease resistance. However, current understanding of the function of soybean NLR genes in resistance to PRR is limited. To address this knowledge gap, transgenic soybean plants overexpressing the NLR gene (Glyma.18g283200) were generated to elucidate the molecular mechanism of resistance. Here, transcript changes and metabolic differences were investigated at three time points (12, 24, and 36 h) after P. sojae infection in hypocotyls of two soybean lines, Dongnong 50 (susceptible line, WT) and Glyma.18g283200 overexpression line (resistant line, OE). Based on the changes in differentially expressed genes (DEGs) in response to P. sojae infection in different lines and at different time points, it was speculated that HOPZ-ACTIVATED RESISTANCE 1 (ZAR1), valine, leucine, and isoleucine degradation, and phytohormone signaling may be involved in the defense response of soybean to P. sojae at the transcriptome level by GO term and KEGG pathway enrichment analysis. Differentially accumulated metabolites (DAMs) analysis revealed that a total of 223 and 210 differential metabolites were identified in the positive ion (POS) and negative ion (NEG) modes, respectively. An integrated pathway-level analysis of transcriptomics (obtained by RNA-seq) and metabolomics data revealed that isoflavone biosynthesis was associated with disease resistance. This work provides valuable insights that can be used in breeding programs aiming to enhance soybean resistance against PRR. Full article
(This article belongs to the Section Crop Physiology and Crop Production)
Show Figures

Figure 1

Figure 1
<p>Differential analysis of gene expression between WT and OE groups. (<b>A</b>) Comparing the number of up- and down-regulated genes. (<b>B</b>–<b>D</b>) Volcano plots of differentially expressed genes at 12, 24, and 36 hpi between the WT and the OE plants as determined by RNA-seq. Red and yellow dots represent up- and down-regulated genes separately. Blue dots represent no differential genes. (<b>E</b>) Up-regulated DEGs at 12, 24, and 36 hpi compared by Venn diagram. (<b>F</b>) Down-regulated DEGs at 12, 24, and 36 hpi compared by Venn diagram.</p>
Full article ">Figure 2
<p>GO and KEGG enrichment analysis of DEGs between WT and OE plants after infection with <span class="html-italic">P. sojae</span>. The bubble chart of GO enrichment analysis of DEGs between WT and OE at 12 hpi (<b>A</b>), 24 hpi (<b>B</b>), and 36 hpi (<b>C</b>). The bubble size indicates the number of genes in the GO terms. Blue bubbles represent biological processes; orange bubbles represent molecular functions; green bubbles represent cellular components. The top 20 categories are shown on the right. KEGG enrichment analysis of DEGs between WT and OE at 12 hpi (<b>D</b>), 24 hpi (<b>E</b>), and 36 hpi (<b>F</b>). The x-axis indicates the rich factor, and the y-axis indicates the pathway name. The size of the dot represents the number of DEGs. The different colors of the dots represent different q values. Significantly enriched pathways (<span class="html-italic">p</span> &lt; 0.05) associated with disease resistance are underlined.</p>
Full article ">Figure 3
<p>GO and KEGG enrichment analysis of up-regulated DEGs between WT and OE plants at 12, 24, and 36 hpi. (<b>A</b>) GO enrichment analysis of up-regulated DEGs between WT12h and OE12h. (<b>B</b>) KEGG enrichment analysis of up-regulated DEGs between WT12h and OE12h. (<b>C</b>) GO enrichment analysis of up-regulated DEGs between WT24h and OE24h. (<b>D</b>) KEGG enrichment analysis of up-regulated DEGs between WT24h and OE24h. (<b>E</b>) GO enrichment analysis of up-regulated DEGs between WT36h and OE36h. (<b>F</b>) KEGG enrichment analysis of up-regulated DEGs between WT36h and OE36h. Significantly enriched pathways (<span class="html-italic">p</span> &lt; 0.05) associated with disease resistance are underlined.</p>
Full article ">Figure 4
<p>Dynamic transcriptome of WT and OE plants during <span class="html-italic">P. sojae</span> infection. (<b>A</b>) Gene expression patterns derived from short time-series expression miner (STEM) analysis. The number of genes in each profile is labeled in the lower left corner. The profiles are ordered based on the <span class="html-italic">p</span>-value significance of the number of genes assigned versus expected. The colored square frame indicates significant profiles (<span class="html-italic">p</span>-value &lt; 0.05). The black line represents the overall trend in each profile. The x-axis represents stages. From left to right, they represent 12 hpi, 24 hpi, and 36 hpi. The y-axis represents the log2-fold change in gene expression, log2 (fold-change) ≥ 1. (<b>B</b>) KEGG enrichment analysis of significantly enriched profiles in the OE group. (<b>C</b>) KEGG enrichment analysis of significantly enriched profiles in the WT group. Significantly enriched pathways (<span class="html-italic">p</span> &lt; 0.05) associated with disease resistance are underlined.</p>
Full article ">Figure 5
<p>Differential metabolites and their functional analysis. (<b>A</b>) Volcanic plot and quantitative statistical plot of differential metabolites in POS. The abscissa is the value of the difference multiple of metabolite abundance in each comparison group after log2, the ordinate is the value of <span class="html-italic">P</span> after the <span class="html-italic">t</span>-test after −log10, and the dotted line perpendicular to the y-axis is the threshold of the <span class="html-italic">p</span>-value for differential metabolite screening. The red dots indicate the differential metabolites with VIP ≥ 1 and <span class="html-italic">p</span> &lt; 0.05, which are up-regulated (FC &gt; 1); the blue dots indicate the differential metabolites with VIP ≥ 1 and <span class="html-italic">p</span> &lt; 0.05 which are down-regulated (FC &lt; −1). The larger the dot, the higher the VIP value of the metabolite. (<b>B</b>) Volcanic plot and quantitative statistical plot of differential metabolites in NEG. (<b>C</b>) KEGG enrichment analysis of the differences in the metabolite content between the WT and OE groups. The ordinate indicates the pathway, and the abscissa indicates the enrichment factor. The size represents the number, and the redder the color, the lower the Q value. (<b>D</b>) Diagram of the KEGG pathway interaction network in POS. (<b>E</b>) Diagram of the KEGG pathway interaction network in NEG. (<b>F</b>) MSEA enrichment plot in POS. The name of the enriched metabolic set is shown on the ordinate and the degree of enrichment is shown on the abscissa. (<b>G</b>) MSEA enrichment plot in NEG. Significantly enriched pathways (<span class="html-italic">p</span> &lt; 0.05) associated with disease resistance are underlined.</p>
Full article ">Figure 6
<p>Analysis of differential metabolites and isoflavonoid biosynthetic pathways. (<b>A</b>) Top 15 differential metabolites for VIP value in the POS mode. The horizontal coordinate is the VIP value; the higher the VIP value, the greater the contribution of the metabolite to the discrimination between the WT and OE groups. (<b>B</b>) Top 15 differential metabolites for VIP value in the NEG mode. (<b>C</b>) The relative metabolite content and gene expression were integrated to construct isoflavonoid biosynthesis pathways. The red circle-labeled metabolites are up-regulated, and the green circle-labeled metabolites are down-regulated. Differential metabolites are labeled with log2FC values. The heatmaps are drawn according to the transcriptome data. The horizontal row represents a DEG with its gene ID, and the vertical columns represent WT_12 hpi, WT_24h hpi, WT_36h hpi, OE 12 hpi, OE 24 hpi, and OE 36 hpi, from left to right.</p>
Full article ">
36 pages, 7923 KiB  
Article
The Ethyl Acetate Extract of Phyllanthus emblica L. Alleviates Diabetic Nephropathy in a Murine Model of Diabetes
by Cheng-Hsiu Lin and Chun-Ching Shih
Int. J. Mol. Sci. 2024, 25(12), 6686; https://doi.org/10.3390/ijms25126686 - 18 Jun 2024
Cited by 1 | Viewed by 1163
Abstract
Oil-Gan is the fruit of the genus Phyllanthus emblica L. The fruits have excellent effects on health care and development values. There are many methods for the management of diabetic nephropathy (DN). However, there is a lack of effective drugs for treating DN [...] Read more.
Oil-Gan is the fruit of the genus Phyllanthus emblica L. The fruits have excellent effects on health care and development values. There are many methods for the management of diabetic nephropathy (DN). However, there is a lack of effective drugs for treating DN throughout the disease course. The primary aim of this study was to examine the protective effects (including analyses of urine and blood, and inflammatory cytokine levels) and mechanisms of the ethyl acetate extract of P. emblica (EPE) on db/db mice, an animal model of diabetic nephropathy; the secondary aim was to examine the expression levels of p- protein kinase Cα (PKCα)/t-PKCα in the kidney and its downregulation of vascular endothelial growth factor (VEGF) and fibrosis gene transforming growth factor-β1 (TGF-β1) by Western blot analyses. Eight db/m mice were used as the control group. Forty db/db mice were randomly divided into five groups. Treatments included a vehicle, EPE1, EPE2, EPE3 (at doses of 100, 200, or 400 mg/kg EPE), or the comparative drug aminoguanidine for 8 weeks. After 8 weeks of treatment, the administration of EPE to db/db mice effectively controlled hyperglycemia and hyperinsulinemia by markedly lowering blood glucose, insulin, and glycosylated HbA1c levels. The administration of EPE to db/db mice decreased the levels of BUN and creatinine both in blood and urine and reduced urinary albumin excretion and the albumin creatine ratio (UACR) in urine. Moreover, EPE treatment decreased the blood levels of inflammatory cytokines, including kidney injury molecule-1 (KIM-1), C-reactive protein (CRP), and NLR family pyrin domain containing 3 (NLRP3). Our findings showed that EPE not only had antihyperglycemic effects but also improved renal function in db/db mice. A histological examination of the kidney by immunohistochemistry indicated that EPE can improve kidney function by ameliorating glomerular morphological damage following glomerular injury; alleviating proteinuria by upregulating the expression of nephrin, a biomarker of early glomerular damage; and inhibiting glomerular expansion and tubular fibrosis. Moreover, the administration of EPE to db/db mice increased the expression levels of p- PKCα/t-PKCα but decreased the expression levels of VEGF and renal fibrosis biomarkers (TGF-β1, collagen IV, p-Smad2, p-Smad3, and Smad4), as shown by Western blot analyses. These results implied that EPE as a supplement has a protective effect against renal dysfunction through the amelioration of insulin resistance as well as the suppression of nephritis and fibrosis in a DN model. Full article
(This article belongs to the Special Issue New Insights in Natural Bioactive Compounds: 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Fruits of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 2
<p>The targeted gene effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in human renal mesangial cell lysates by Western blot analyses with a primary antibody against TGF-β1 and Collagen IV. (<b>A</b>) Representative blots in human renal mesangial cells; (<b>B</b>,<b>C</b>) Quantification of the expression levels of TGF-β1 and Collagen IV. All values are means ± S.E. +++ <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the HG group. The β-actin was used as the internal standard in each sample. HG: high glucose. EPE: ethyl acetate extract of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 2 Cont.
<p>The targeted gene effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in human renal mesangial cell lysates by Western blot analyses with a primary antibody against TGF-β1 and Collagen IV. (<b>A</b>) Representative blots in human renal mesangial cells; (<b>B</b>,<b>C</b>) Quantification of the expression levels of TGF-β1 and Collagen IV. All values are means ± S.E. +++ <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the HG group. The β-actin was used as the internal standard in each sample. HG: high glucose. EPE: ethyl acetate extract of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 3
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 3 Cont.
<p>Effects of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group. EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 4
<p>Representative photographs of the kidney; ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice on (<b>A</b>) glomerular basement membrane in cortex and (<b>B</b>) mesangial matrix expansion in outer medulla by periodic acid Schiff (PAS) staining (magnification: 20 × 10); (<b>C</b>) expression levels of nephrin in the kidneys by immunohistochemical (IHC) staining; (<b>D</b>) Nephrin-positive area (%) score by IHC stains compared with the db/db group. EPE: EPE1, EPE2, and EPE3 (100, 200, and 400 mg/kg body). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group.</p>
Full article ">Figure 4 Cont.
<p>Representative photographs of the kidney; ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) in db/db mice on (<b>A</b>) glomerular basement membrane in cortex and (<b>B</b>) mesangial matrix expansion in outer medulla by periodic acid Schiff (PAS) staining (magnification: 20 × 10); (<b>C</b>) expression levels of nephrin in the kidneys by immunohistochemical (IHC) staining; (<b>D</b>) Nephrin-positive area (%) score by IHC stains compared with the db/db group. EPE: EPE1, EPE2, and EPE3 (100, 200, and 400 mg/kg body). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the db/m group; *** <span class="html-italic">p</span> &lt; 0.001 compared with the db/db + vehicle (distilled water) (db/db) group.</p>
Full article ">Figure 5
<p>The kidney target gene expression levels in db/db mice following treatment with ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) or aminoguanidine (AG, 20 mg/kg body weight) by Western blotting analysis on p-PKCα/t-PKCα, VEGF, nephrin, TGFβ1, collagen IV, fibronectin, Smad4, p-Smad2/t-Smad2, and p-Smad3/t-Smad3. (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) Representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) quantification of p-PKCα/t-PKCα, VEGF TGFβ1, collagen IV, Smad4, p-Smad2/t-Smad2, p-Smad3/t-Smad3, p-NLRP3/t-NLRP3, ICAM, and Capalase-1 to β-actin. Protein was separated by 12% SDS-PAG. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 as compared to the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to the db/db plus vehicle (distilled water) (db/db) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE: EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The kidney target gene expression levels in db/db mice following treatment with ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) or aminoguanidine (AG, 20 mg/kg body weight) by Western blotting analysis on p-PKCα/t-PKCα, VEGF, nephrin, TGFβ1, collagen IV, fibronectin, Smad4, p-Smad2/t-Smad2, and p-Smad3/t-Smad3. (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) Representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) quantification of p-PKCα/t-PKCα, VEGF TGFβ1, collagen IV, Smad4, p-Smad2/t-Smad2, p-Smad3/t-Smad3, p-NLRP3/t-NLRP3, ICAM, and Capalase-1 to β-actin. Protein was separated by 12% SDS-PAG. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 as compared to the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to the db/db plus vehicle (distilled water) (db/db) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE: EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The kidney target gene expression levels in db/db mice following treatment with ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) or aminoguanidine (AG, 20 mg/kg body weight) by Western blotting analysis on p-PKCα/t-PKCα, VEGF, nephrin, TGFβ1, collagen IV, fibronectin, Smad4, p-Smad2/t-Smad2, and p-Smad3/t-Smad3. (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) Representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) quantification of p-PKCα/t-PKCα, VEGF TGFβ1, collagen IV, Smad4, p-Smad2/t-Smad2, p-Smad3/t-Smad3, p-NLRP3/t-NLRP3, ICAM, and Capalase-1 to β-actin. Protein was separated by 12% SDS-PAG. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 as compared to the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to the db/db plus vehicle (distilled water) (db/db) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE: EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 5 Cont.
<p>The kidney target gene expression levels in db/db mice following treatment with ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) or aminoguanidine (AG, 20 mg/kg body weight) by Western blotting analysis on p-PKCα/t-PKCα, VEGF, nephrin, TGFβ1, collagen IV, fibronectin, Smad4, p-Smad2/t-Smad2, and p-Smad3/t-Smad3. (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) Representative image; (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>) quantification of p-PKCα/t-PKCα, VEGF TGFβ1, collagen IV, Smad4, p-Smad2/t-Smad2, p-Smad3/t-Smad3, p-NLRP3/t-NLRP3, ICAM, and Capalase-1 to β-actin. Protein was separated by 12% SDS-PAG. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 as compared to the db/m group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to the db/db plus vehicle (distilled water) (db/db) group. All values are means ± SE (<span class="html-italic">n</span> = 8 per group). EPE, ethyl acetate extract of <span class="html-italic">P. emblica</span> L. EPE: EPE1: 100, EPE2: 200, EPE3: 400 mg/kg body weight; AG: aminoguanidine (20 mg/kg body weight).</p>
Full article ">Figure 6
<p>Effects of seven fractions of EPE (EA) on expression levels of collagen IV, TGF-β1, VEGF, and KIM in human renal mesangial (HRM) cells by Western blotting analyses. Human renal mesangial (HRM) cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, VEGF, and KIM. (<b>A</b>–<b>E</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of Collagen IV, TGF-β1, KIM, and VEGF in HRM cells by Western blotting analysis. Human renal mesangial (HRM) cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, KIM, and VEGF. (<b>A</b>) Representative blots for 7 fractions in HRM cells; (<b>B</b>–<b>E</b>) Quantification of the expression levels to β-actin including (<b>B</b>) collagen IV, (<b>C</b>) TGF-β1, (<b>D</b>) KIM, and (<b>E</b>) VEGF. All values are means ± S.E. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the glucose group. (<b>F</b>,<b>G</b>) High-performance liquid chromatography analysis of (<b>F</b>) 2500 ppm ethyl acetate of <span class="html-italic">P. emblica</span> L. (EPE), (<b>G</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of collagen IV, TGF-β1, VEGF, and KIM in human renal mesangial (HRM) cells by Western blotting analyses. Human renal mesangial (HRM) cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, VEGF, and KIM. (<b>A</b>–<b>E</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of Collagen IV, TGF-β1, KIM, and VEGF in HRM cells by Western blotting analysis. Human renal mesangial (HRM) cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, KIM, and VEGF. (<b>A</b>) Representative blots for 7 fractions in HRM cells; (<b>B</b>–<b>E</b>) Quantification of the expression levels to β-actin including (<b>B</b>) collagen IV, (<b>C</b>) TGF-β1, (<b>D</b>) KIM, and (<b>E</b>) VEGF. All values are means ± S.E. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the glucose group. (<b>F</b>,<b>G</b>) High-performance liquid chromatography analysis of (<b>F</b>) 2500 ppm ethyl acetate of <span class="html-italic">P. emblica</span> L. (EPE), (<b>G</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of collagen IV, TGF-β1, VEGF, and KIM in human renal mesangial (HRM) cells by Western blotting analyses. Human renal mesangial (HRM) cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, VEGF, and KIM. (<b>A</b>–<b>E</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of Collagen IV, TGF-β1, KIM, and VEGF in HRM cells by Western blotting analysis. Human renal mesangial (HRM) cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, KIM, and VEGF. (<b>A</b>) Representative blots for 7 fractions in HRM cells; (<b>B</b>–<b>E</b>) Quantification of the expression levels to β-actin including (<b>B</b>) collagen IV, (<b>C</b>) TGF-β1, (<b>D</b>) KIM, and (<b>E</b>) VEGF. All values are means ± S.E. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the glucose group. (<b>F</b>,<b>G</b>) High-performance liquid chromatography analysis of (<b>F</b>) 2500 ppm ethyl acetate of <span class="html-italic">P. emblica</span> L. (EPE), (<b>G</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of collagen IV, TGF-β1, VEGF, and KIM in human renal mesangial (HRM) cells by Western blotting analyses. Human renal mesangial (HRM) cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, VEGF, and KIM. (<b>A</b>–<b>E</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of Collagen IV, TGF-β1, KIM, and VEGF in HRM cells by Western blotting analysis. Human renal mesangial (HRM) cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, KIM, and VEGF. (<b>A</b>) Representative blots for 7 fractions in HRM cells; (<b>B</b>–<b>E</b>) Quantification of the expression levels to β-actin including (<b>B</b>) collagen IV, (<b>C</b>) TGF-β1, (<b>D</b>) KIM, and (<b>E</b>) VEGF. All values are means ± S.E. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the glucose group. (<b>F</b>,<b>G</b>) High-performance liquid chromatography analysis of (<b>F</b>) 2500 ppm ethyl acetate of <span class="html-italic">P. emblica</span> L. (EPE), (<b>G</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 6 Cont.
<p>Effects of seven fractions of EPE (EA) on expression levels of collagen IV, TGF-β1, VEGF, and KIM in human renal mesangial (HRM) cells by Western blotting analyses. Human renal mesangial (HRM) cells were treated with seven fractions and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, VEGF, and KIM. (<b>A</b>–<b>E</b>) Effects of 7 fractions of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EtOAc soluble fractions) (EA-1, EA-2, EA-3, EA-A, EA-5, EA-6, and EA-7) on expression levels of Collagen IV, TGF-β1, KIM, and VEGF in HRM cells by Western blotting analysis. Human renal mesangial (HRM) cells were treated with 7 fractions as described in the experimental procedures and equal amounts of lysates were resolved by SDS-PAGE and blotted for collagen IV, TGF-β1, KIM, and VEGF. (<b>A</b>) Representative blots for 7 fractions in HRM cells; (<b>B</b>–<b>E</b>) Quantification of the expression levels to β-actin including (<b>B</b>) collagen IV, (<b>C</b>) TGF-β1, (<b>D</b>) KIM, and (<b>E</b>) VEGF. All values are means ± S.E. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the control group; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared with the glucose group. (<b>F</b>,<b>G</b>) High-performance liquid chromatography analysis of (<b>F</b>) 2500 ppm ethyl acetate of <span class="html-italic">P. emblica</span> L. (EPE), (<b>G</b>) 10.3 mg/5 mL EA-4 of ethyl acetate of <span class="html-italic">P. emblica</span> L.</p>
Full article ">Figure 7
<p>Graphic abstract of ethyl acetate extract of <span class="html-italic">P. emblica</span> L. (EPE) on diabetic nephropathy in a db/db mouse model.</p>
Full article ">
21 pages, 2901 KiB  
Review
CARD8: A Novel Inflammasome Sensor with Well-Known Anti-Inflammatory and Anti-Apoptotic Activity
by Tugay Karakaya, Marta Slaufova, Michela Di Filippo, Paulina Hennig, Thomas Kündig and Hans-Dietmar Beer
Cells 2024, 13(12), 1032; https://doi.org/10.3390/cells13121032 - 13 Jun 2024
Viewed by 1147
Abstract
Inflammasomes comprise a group of protein complexes with fundamental roles in the induction of inflammation. Upon sensing stress factors, their assembly induces the activation and release of the pro-inflammatory cytokines interleukin (IL)-1β and -18 and a lytic type of cell death, termed pyroptosis. [...] Read more.
Inflammasomes comprise a group of protein complexes with fundamental roles in the induction of inflammation. Upon sensing stress factors, their assembly induces the activation and release of the pro-inflammatory cytokines interleukin (IL)-1β and -18 and a lytic type of cell death, termed pyroptosis. Recently, CARD8 has joined the group of inflammasome sensors. The carboxy-terminal part of CARD8, consisting of a function-to-find-domain (FIIND) and a caspase activation and recruitment domain (CARD), resembles that of NLR family pyrin domain containing 1 (NLRP1), which is recognized as the main inflammasome sensor in human keratinocytes. The interaction with dipeptidyl peptidases 8 and 9 (DPP8/9) represents an activation checkpoint for both sensors. CARD8 and NLRP1 are activated by viral protease activity targeting their amino-terminal region. However, CARD8 also has some unique features compared to the established inflammasome sensors. Activation of CARD8 occurs independently of the inflammasome adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC), leading mainly to pyroptosis rather than the activation and secretion of pro-inflammatory cytokines. CARD8 was also shown to have anti-inflammatory and anti-apoptotic activity. It interacts with, and inhibits, several proteins involved in inflammation and cell death, such as the inflammasome sensor NLRP3, CARD-containing proteins caspase-1 and -9, nucleotide-binding oligomerization domain containing 2 (NOD2), or nuclear factor kappa B (NF-κB). Single nucleotide polymorphisms (SNPs) of CARD8, some of them occurring at high frequencies, are associated with various inflammatory diseases. The molecular mechanisms underlying the different pro- and anti-inflammatory activities of CARD8 are incompletely understood. Alternative splicing leads to the generation of multiple CARD8 protein isoforms. Although the functional properties of these isoforms are poorly characterized, there is evidence that suggests isoform-specific roles. The characterization of the functions of these isoforms, together with their cell- and disease-specific expression, might be the key to a better understanding of CARD8’s different roles in inflammation and inflammatory diseases. Full article
(This article belongs to the Special Issue Role of Inflammasome Activation in Innate and Adaptive Immunity)
Show Figures

Figure 1

Figure 1
<p><b>CARD8 has anti- and pro-inflammatory activity.</b> CARD8 can directly interact with NLRP1, NLRP3, and caspase-1, and inhibit the activation of the NLRP1, NLRP3, and AIM2 inflammasomes. CARD8 can negatively regulate NOD2, NF-κB, and apoptosis, the latter through an indirect or direct inhibition of caspase-8 or -9, respectively. Moreover, after autoproteolytic self-activation, CARD8 forms an ASC-independent inflammasome, activated by inhibition of the dipeptidyl peptidases DPP8/9, by talabostat or CQ31, or by enhanced proteasomal degradation of the inhibitory amino-terminus, mediated through proteolytic processing of its amino-terminal disordered region by viral proteases and potentiated by increased proteasome activity. Caspase-1 recruitment and activation by autocatalytic cleavage and release of the subunits p20 and p10 through the CARD8 inflammasome is mainly associated with pyroptosis.</p>
Full article ">Figure 2
<p><b>CARD8 and NLRP1 show high structural homology.</b> Side-by-side comparison of the longest isoforms of CARD8 and NLRP1 highlights the high structural similarities between the two proteins, especially regarding the FIIND and CARD.</p>
Full article ">Figure 3
<p><b>The <span class="html-italic">CARD8</span> gene, its most important SNPs, and protein isoforms.</b> (<b>a</b>) The <span class="html-italic">CARD8</span> gene locus, including introns, exons, and localization of selected disease-associated SNPs. (<b>b</b>) <span class="html-italic">CARD8</span> coding and non-coding exons and their SNPs. (<b>c</b>) <span class="html-italic">CARD8</span> coding sequences of all reported isoforms, their domains, and SNPs. (<b>d</b>) CARD8 protein sequences of all reported isoforms and consequences of the SNPs on the protein level. Results of the indel mutation rs140826611 on the protein level and color code of the Figure are shown on the left and right, respectively [<a href="#B168-cells-13-01032" class="html-bibr">168</a>,<a href="#B169-cells-13-01032" class="html-bibr">169</a>,<a href="#B170-cells-13-01032" class="html-bibr">170</a>].</p>
Full article ">
13 pages, 2573 KiB  
Article
The Anti-Inflammatory Effect of SDF-1 Derived Peptide on Porphyromonas gingivalis Infection via Regulation of NLRP3 and AIM2 Inflammasome
by Si Yeong Kim, Min Kee Son, Jung Hwa Park, Hee Sam Na and Jin Chung
Pathogens 2024, 13(6), 474; https://doi.org/10.3390/pathogens13060474 - 4 Jun 2024
Viewed by 941
Abstract
(1) Background: Peptides are appealing as pharmacological materials because they are easily produced, safe, and tolerable. Despite increasing gum-care awareness, periodontitis is still prevalent and is influenced by factors like high sugar consumption, smoking, and aging. Porphyromonas gingivalis is considered a major [...] Read more.
(1) Background: Peptides are appealing as pharmacological materials because they are easily produced, safe, and tolerable. Despite increasing gum-care awareness, periodontitis is still prevalent and is influenced by factors like high sugar consumption, smoking, and aging. Porphyromonas gingivalis is considered a major etiologic agent of periodontitis and activates the NLR family pyrin domain containing 3 (NLRP3) but is absent in melanoma 2 (AIM2) inflammasomes, resulting in pro-inflammatory cytokine release. (2) Methods: We examined the anti-inflammatory effects of 18 peptides derived from human stromal cell-derived factor-1 (SDF-1) on THP-1 macrophages. Inflammation was induced by P. gingivalis, and the anti-inflammatory effects were analyzed using molecular biological techniques. In a mouse periodontitis model, alveolar bone resorption was assessed using micro-CT. (3) Results: Of the 18 SDF-1-derived peptides, S10 notably reduced IL-1β and TNF-α secretion. S10 also diminished the P. gingivalis-induced expression of NLRP3, AIM2, ASC (apoptosis-associated speck-like protein), caspase-1, and IL-1β. Furthermore, S10 attenuated the enhanced TLR (toll-like receptor) signaling pathway and decreased the phosphorylation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs). In addition, S10 mitigated alveolar bone loss in our P. gingivalis-induced mouse model of periodontitis. (4) Conclusions: S10 suppressed TLR/NF-κB/NLRP3 inflammasome signaling and the AIM2 inflammasome in our P. gingivalis-induced murine periodontitis model, which suggests that it has potential use as a therapeutic treatment for periodontitis. Full article
(This article belongs to the Special Issue Nosocomial Infection and Antimicrobial Resistance)
Show Figures

Figure 1

Figure 1
<p>SDF-1-derived peptides suppressed the secretion of IL-1β and TNF-α. (<b>A</b>,<b>B</b>) THP-1 macrophages were pretreated with each SDF-1 derived peptide (1 μg/mL) for 30 min and were then infected with <span class="html-italic">P. gingivalis</span> (MOI 100). IL-1β (<b>A</b>) and TNF-α (<b>B</b>) secretion levels were measured by ELISA (<span class="html-italic">n =</span> 3). *** <span class="html-italic">p</span>-value &lt; 0.001 versus untreated group (Con); <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span>-value &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span>-value &lt; 0.001 versus <span class="html-italic">P. gingivalis</span> infection (Pg).</p>
Full article ">Figure 2
<p>S10 inhibited the activation of NLRP3 and AIM2 inflammasomes by <span class="html-italic">P. gingivalis</span>. (<b>A</b>) THP-1 macrophages were pretreated with S10 (1 μg/mL) for 30 min and were then infected with <span class="html-italic">P. gingivalis</span> (MOI 100) for 18 h. The secreted levels of IL-1β and TNF-α in the supernatant were determined by an ELISA (<span class="html-italic">n =</span> 3). (<b>B</b>) Real-time PCR was conducted to quantify its effects on NLRP3 and AIM2 inflammasome components; the results are presented in the graph (<span class="html-italic">n =</span> 3). (<b>C</b>) Cell lysates were subjected to Western blot analysis, and representative immunoblots and graphs of protein levels versus β-actin for each protein are shown (<span class="html-italic">n =</span> 3). Blot densities are expressed relative to treatment-naïve controls. (<b>D</b>) PMA-primed ASC-GFP-THP-1 cells were pretreated with S10 for 30 min and then infected with <span class="html-italic">P. gingivalis</span> for 18 h. “Pg” in the images indicates the ASC speck, indicated by yellow arrows (original magnification 200×). The graph shows the percentage of total cells containing ASC. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 3
<p>S10 suppressed the <span class="html-italic">P. gingivalis</span>-induced activation of the TLR2/4 and NF-κB/MAPK signaling pathways. (<b>A</b>) Cell lysates of THP-1-derived macrophages, pretreated with S10 (1 μg/mL) for 30 min and infected with <span class="html-italic">P. gingivalis</span> (MOI 100) for 18 h, were studied via Western blot analysis. TLR2/4 and their downstream signaling proteins were analyzed, and representative figures, along with densitometric graphs normalized versus β-actin, are shown (<span class="html-italic">n =</span> 3). (<b>B</b>,<b>C</b>) The phosphorylation levels of NF-Κb and MAPK were evaluated using dedicated antibodies targeting each phosphorylated site, and normalization was conducted versus total protein levels (<span class="html-italic">n =</span> 3). * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 4
<p>S10 alleviated the alveolar bone loss caused by <span class="html-italic">P. gingivalis</span> infection. (<b>A</b>) Details of the S10 treatment protocol. (<b>B</b>,<b>C</b>) Micro-CT images were used to measure alveolar bone areas after orally administering <span class="html-italic">P. gingivalis</span> with or without S10 treatment. Areas between the cementoenamel junctions and alveolar bone crests of the three molars in the captured images were measured using ImageJ. Images are representative of the three groups, and graphs summarize the measured alveolar bone areas (<span class="html-italic">n =</span> 6). * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01.</p>
Full article ">
17 pages, 12015 KiB  
Article
N-3 Polyunsaturated Fatty Acids Protect against Alcoholic Liver Steatosis by Activating FFA4 in Kupffer Cells
by Saeromi Kang, Jung-Min Koh and Dong-Soon Im
Int. J. Mol. Sci. 2024, 25(10), 5476; https://doi.org/10.3390/ijms25105476 - 17 May 2024
Viewed by 896
Abstract
Supplementation with fish oil rich in omega-3 polyunsaturated fatty acids (n-3 PUFAs) effectively reduces acute and chronic alcohol-induced hepatic steatosis. We aimed to find molecular mechanisms underlying the effects of n-3 PUFAs in alcohol-induced hepatic steatosis. Because free fatty acid receptor 4 (FFA4, [...] Read more.
Supplementation with fish oil rich in omega-3 polyunsaturated fatty acids (n-3 PUFAs) effectively reduces acute and chronic alcohol-induced hepatic steatosis. We aimed to find molecular mechanisms underlying the effects of n-3 PUFAs in alcohol-induced hepatic steatosis. Because free fatty acid receptor 4 (FFA4, also known as GPR120) has been found as a receptor for n-3 PUFAs in an ethanol-induced liver steatosis model, we investigated whether n-3 PUFAs protect against liver steatosis via FFA4 using AH7614, an FFA4 antagonist, and Ffa4 knockout (KO) mice. N-3 PUFAs and compound A (CpdA), a selective FFA4 agonist, reduced the ethanol-induced increase in lipid accumulation in hepatocytes, triglyceride content, and serum ALT levels, which were not observed in Ffa4 KO mice. N-3 PUFAs and CpdA also reduced the ethanol-induced increase in lipogenic sterol regulatory element-binding protein-1c expression in an FFA4-dependent manner. In Kupffer cells, treatment with n-3 PUFA and CpdA reversed the ethanol-induced increase in tumor necrosis factor-α, cyclooxygenase-2, and NLR family pyrin domain-containing 3 expression levels in an FFA4-dependent manner. In summary, n-3 PUFAs protect against ethanol-induced hepatic steatosis via the anti-inflammatory actions of FFA4 on Kupffer cells. Our findings suggest FFA4 as a therapeutic target for alcoholic hepatic steatosis. Full article
(This article belongs to the Special Issue Alcohol and Inflammation)
Show Figures

Figure 1

Figure 1
<p>N-3 polyunsaturated fatty acids (n-3 PUFAs) protect against ethanol-induced hepatic steatosis. C57BL/6 female mice were fed a diet containing 6.3% ethanol with or without n-3 PUFAs (7.5 g/kg) and AH7614 (10 mg/kg) for two weeks. Livers were collected from mice fed a control diet (CD), EtOH diet, EtOH diet + n-3 PUFAs (6–7 animals per group), and EtOH diet + n-3 PUFAs + AH7614. (<b>A</b>) Hematoxylin and eosin (H&amp;E)-stained liver sections showing liver morphology. Arrows indicate lesions of hepatic steatosis. (<b>B</b>) Oil red O staining. (<b>C</b>) Serum triglyceride levels. (<b>D</b>) Serum aspartate aminotransferase (AST) activities. (<b>E</b>) Serum alanine aminotransferase (ALT) activities. Data are presented as the mean ± standard error (SE) for 6 mice per experimental group. Statistical significance: ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; ** <span class="html-italic">p</span> &lt; 0.01 vs. EtOH-fed group; *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. EtOH- and n-3 PUFA-fed group contained in the second panel.</p>
Full article ">Figure 2
<p>Modulation of lipogenic gene expression in the liver by n-3 PUFAs. (<b>A</b>) Reverse transcription-polymerase chain reaction (RT-PCR) analysis of the sterol regulatory element-binding protein-1c (<span class="html-italic">Srebp-1c</span>), fatty acid synthase (<span class="html-italic">Fas</span>), and glycerol-phosphate acyltransferase (<span class="html-italic">Gpat</span>) levels. (<b>B</b>) Western blotting of preformed and mature SREBP-1c expression levels. Data are presented as the mean ± SE. Data are presented as the mean ± standard error (SE) for 6 mice per experimental group. Statistical significance: ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group; <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, and <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed and n-3 PUFA-fed group.</p>
Full article ">Figure 3
<p>Compound A (CpdA) protects against ethanol-induced hepatic steatosis. C57BL/6 female mice were administered CpdA (30 mg/kg) and AH7614 (10 mg/kg) for two weeks. Livers were collected from mice fed CD, EtOH diet, EtOH diet + CpdA, and EtOH diet + CpdA + AH7614. (<b>A</b>) H&amp;E-stained liver sections showing the liver morphology. Arrows indicate lesions of hepatic steatosis. (<b>B</b>) Oil red O staining. (<b>C</b>) Serum triglyceride levels. (<b>D</b>) Serum AST activities. (<b>E</b>) Serum ALT activities. Data are presented as the mean ± SE for 6 mice per experimental group. Statistical significance: ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group; <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.0001 vs. the EtOH-fed and CpdA-treated group.</p>
Full article ">Figure 4
<p>Modulation of lipogenic gene expression in the liver by CpdA. (<b>A</b>) RT-PCR analysis of <span class="html-italic">Srebp-1c</span>, <span class="html-italic">Fas</span>, and <span class="html-italic">Gpat</span> levels. (<b>B</b>) Western blotting of preformed and mature SREBP-1c expression levels. Data are presented as the mean ± SE for 6 mice per experimental group. Statistical significance: ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 and <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed and CpdA-treated group.</p>
Full article ">Figure 5
<p>Effects of n-3 PUFA and CpdA on ethanol-induced hepatic steatosis in free fatty acid receptor 4 (FFA4) knockout (KO) mice. C57BL/6 female mice were fed a 6.3% ethanol diet containing n-3 PUFAs (7.5 g/kg) and CpdA (30 mg/kg) for two weeks. Livers were collected from mice fed CD, EtOH diet, EtOH diet + n-3 PUFAs, and EtOH diet + CpdA. (<b>A</b>) H&amp;E-stained liver sections showing the liver morphology. Arrows indicate lesions of hepatic steatosis. (<b>B</b>) Oil red O staining. (<b>C</b>) Serum triglyceride levels. (<b>D</b>) Serum AST activities. (<b>E</b>) Serum ALT activities. Data are presented as the mean ± SE for 6 mice per experimental group. Statistical significance: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group. * <span class="html-italic">p</span> &lt; 0.1, *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group.</p>
Full article ">Figure 6
<p>Effects of n-3 PUFAs and CpdA on lipogenic gene expression levels in the liver of FFA4 KO mice. (<b>A</b>) RT-PCR analysis of <span class="html-italic">Srebp-1c</span>, <span class="html-italic">Fas</span>, and <span class="html-italic">Gpat</span> levels. (<b>B</b>) Western blotting of preformed and mature SREBP-1c expression levels. Data are presented as the mean ± SE for 6 mice per experimental group. Statistical significance: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group.</p>
Full article ">Figure 7
<p>Change in pro-inflammatory gene expression levels in Kupffer cells by n-3 PUFAs, CpdA, and AH7614. RT-PCR analysis of pro-inflammatory genes. (<b>A</b>) Kupffer cells were collected from mice fed CD, EtOH diet, EtOH diet + n-3 PUFAs, and EtOH diet + n-3 PUFAs + AH7614. (<b>B</b>) Kupffer cells were collected from mice fed CD diet, EtOH diet, EtOH diet + CpdA, and EtOH diet + CpdA + AH7614. (<b>C</b>) Kupffer cells were collected from FFA4 KO mice fed CD diet, EtOH diet, EtOH diet + n-3 PUFAs, and EtOH diet + CpdA. Data are presented as the mean ± SE for 6 mice per experimental group. Statistical significance: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001 vs. CD-fed group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. EtOH-fed group; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 and <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001 vs. EtOH + n-3 PUFA or CpdA group.</p>
Full article ">Figure 8
<p>Illustration of the action mechanism of n-3 PUFAs and CpdA via Ffa4. EtOH administration disrupts the integrity of the intestinal epithelial layer and increases gut permeability, resulting in the exposure of endotoxins to Kupffer cells. Activation of Kupffer cells leads to the release of pro-inflammatory cytokines like TNF-α. Then, TNF-α induces SREBP-1, resulting in hepatic steatosis. FFA4 activation by n-3 PUFAs or CpdA induces suppression of pro-inflammatory M1 Kupffer cells and activation of anti-inflammatory M2 Kupffer cells, resulting in protection against steatosis.</p>
Full article ">
19 pages, 2800 KiB  
Article
Nucleotide-Binding Oligomerization Domain 1 (NOD1) Agonists Prevent SARS-CoV-2 Infection in Human Lung Epithelial Cells through Harnessing the Innate Immune Response
by Edurne Garcia-Vidal, Ignasi Calba, Eva Riveira-Muñoz, Elisabet García, Bonaventura Clotet, Pere Serra-Mitjà, Cecilia Cabrera, Ester Ballana and Roger Badia
Int. J. Mol. Sci. 2024, 25(10), 5318; https://doi.org/10.3390/ijms25105318 - 13 May 2024
Viewed by 1120
Abstract
The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory [...] Read more.
The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory mucosa to trigger a potent immune response at the onset of infection could serve as a potential strategy for managing respiratory infections. This study focused on screening immunomodulators to enhance innate immune response in lung epithelial and immune cell models. Through testing various subfamilies and pathways of pattern recognition receptors (PRRs), the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family was found to selectively activate innate immunity in lung epithelial cells. Activation of NOD1 and dual NOD1/2 by the agonists TriDAP and M-TriDAP, respectively, increased the number of IL-8+ cells by engaging the NF-κB and interferon response pathways. Lung epithelial cells showed a stronger response to NOD1 and dual NOD1/2 agonists compared to control. Interestingly, a less-pronounced response to NOD1 agonists was noted in PBMCs, indicating a tissue-specific effect of NOD1 in lung epithelial cells without inducing widespread systemic activation. The specificity of the NOD agonist pathway was confirmed through gene silencing of NOD1 (siRNA) and selective NOD1 and dual NOD1/2 inhibitors in lung epithelial cells. Ultimately, activation induced by NOD1 and dual NOD1/2 agonists created an antiviral environment that hindered SARS-CoV-2 replication in vitro in lung epithelial cells. Full article
(This article belongs to the Special Issue Viral and Host Targets to Fight RNA Viruses)
Show Figures

Figure 1

Figure 1
<p>NLR agonists induce innate immune activation in in vitro lung epithelial and myeloid models. (<b>A</b>) Workflow to screen for potential immunomodulators of the innate immune system in A549 lung epithelial and THP-1 myeloid cell lines. (<b>B</b>) Library classification of tested compounds according to their reported target. (<b>C</b>) Heatmap illustrates the immune activation induced by immunomodulators targeting PRR subfamilies in lung epithelial A549 and myeloid THP-1 cells, as determined by the intracellular staining of IL-8 by flow cytometry. (<b>D</b>) Representative dot-plots showing IL-8+ intracellular staining of lung epithelial A549 (<b>left panel</b>) and myeloid THP-1 (<b>right panel</b>) cells upon treatment with NLR agonists, as determined by flow cytometry compared to untreated (UN) cells.</p>
Full article ">Figure 2
<p>Cytokine response is preferentially triggered by NOD1 and dual NOD1/2 agonists in lung epithelial cells. (<b>A</b>) Cytokine response to NLR agonists triggered by NOD1-, NOD1/2- and NOD2-specific agonists in lung epithelial A549-Dual cells. Immune response was determined by the percentage of intracellular IL-8+ (<b>left</b>) and TNFα+ (<b>right</b>) cell quantification by flow cytometry after 24 h of treatment, using LPS (1 µg/mL, yellow bar) non-treated condition (UN, black bar) as controls. (<b>B</b>) Induction of the proinflammatory response upon treatment with increasing concentrations of NOD1, NOD1/2 and NOD2 ligands in lung epithelial A549-Dual cells after 24 h of treatment. The intracellular stainings of IL-8 and TNFα were determined by flow cytometry as subrogate representative markers of the proinflammatory response, using LPS and UN as controls. Mean ± SD of three independent experiments is shown. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>NOD1 and NOD1/2 agonists induce innate immune activation in vitro in lung epithelial through the NF-κB and ISRE pathways. (<b>A</b>) Induction of the NF-κB activity triggered by NLR agonists upon recognition by the NOD1, NOD1/2 and NOD2 receptors in lung epithelial A549-Dual cells after 24 h of treatment. LPS (yellow bar) and Poly(I:C) (grey bar) were used as controls for NF-κB activation. (<b>B</b>) Assessment of NLR agonist activity on type I IFN response signaling by the quantification of interferon-stimulated response element (ISRE)-dependent gene expression in lung epithelial A549-Dual cells after 24 h of treatment. Values were relativized to the untreated (ND, black bar) condition. (<b>C</b>) Relative mRNA expression of IL-8, CXCL10 and ISG15 in A549-Dual treated cells with 50 µM of selected NOD agonists for 8 h measured by qPCR (normalized to GAPDH expression). Mean ± SD of three independent experiments is shown. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Activity of NOD1 and dual NOD1/2 agonists is specific in lung epithelial cells. (<b>A</b>) Gene expression of NOD1 receptor in A549-Dual cells transiently silenced with siRNA targeting NOD1 (siNOD1). Mock and non-specific siRNA (siNT) were used as controls. (<b>B</b>) Cell viability of A549-Dual cells treated with siNOD1 and siNT, using mock condition as control. Cell viability was determined by LIVE/DEAD staining and measured by flow cytometry. (<b>C</b>) Activity of NOD1 agonists (TriDAP and C12-iE-DAP), dual NOD1/2 (M-TriDAP) and NOD2 (MDP) in A549 cells treated with siNOD1. Intracellular staining of proinflammatory IL-8+ cells was determined by flow cytometry using the mock and siNT conditions, respectively. (<b>D</b>) Induction of the NF-κB and ISRE (<b>E</b>) activation pathways in A549-Dual cells treated with NOD1, dual NOD1/2 or NOD2 agonists with 50 µM of selective NOD1 inhibitor ML130, 50 µM of NOD1/2 inhibitor NOD-IN-1 or untreated (UNT), respectively. Red dotted line indicates the basal NF-κB (<b>left</b>) or ISRE activity (<b>right</b>) in A549-Dual cells. Mean ± SD of three independent experiments is shown. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>NLR agonist-induced cytokine response is preferentially triggered by NOD2 in PBMCs. (<b>A</b>) Assessment of the cytokine response to NLR agonists triggered by specific NOD1, dual NOD1/2 and NOD2 agonists in PBMCs. The percentages of intracellular IL-1β+, TNFα+ and IL-6+ cells were measured as representative markers of the proinflammatory response. Values were relativized to the non-treated condition (ND, black bar). LPS (1 µg/mL, yellow bar) and PMA (50 ng/mL) + ionomycin (1 µM) were used as positive controls. (<b>B</b>) Dose-response induction of proinflammatory cytokines IL-1β+, TNFα+ and IL-6+ in PBMCs treated with increasing concentrations of TriDAP (NOD1), M-TriDAP (dual NOD1/2) and MDP (NOD2) agonists. Mean ± SD of three independent experiments is shown. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>NOD1 and dual NOD1/2 agonists impair SARS-CoV-2 replication in lung epithelial cells. Pretreatment of lung epithelial A549-Dual cells for 3 h with increasing concentrations of NOD1 and dual NOD1/2 agonists preferentially inhibits SARS-CoV-2 replication. (<b>A</b>) Representative dot-plots of infected cells treated with NOD agonists as measured by flow cytometry. (<b>B</b>) Quantification of viral replication measured as the percentage of SARS-CoV-2-GFP+ cells determined by flow cytometry after 48 h of infection. Values were relativized to the untreated condition (INF, black bar). TLR3 agonist Poly(I:C) (light gray bars) was used as control for the induction of the innate immune response. Mean ± SD of three independent experiments is shown. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; EC<sub>50</sub>: half maximal effective concentration.</p>
Full article ">
15 pages, 1170 KiB  
Article
Grape/Blueberry Anthocyanins and Their Gut-Derived Metabolites Attenuate LPS/Nigericin-Induced Inflammasome Activation by Inhibiting ASC Speck Formation in THP-1 Monocytes
by Inken Behrendt, Isabella Röder, Frank Will, Gabriela Michel, Elvira Friedrich, Daniela Grote, Zoe Martin, Hanna Pauline Dötzer, Mathias Fasshauer, Martin Speckmann and Sabine Kuntz
Metabolites 2024, 14(4), 203; https://doi.org/10.3390/metabo14040203 - 3 Apr 2024
Cited by 1 | Viewed by 1278
Abstract
Inflammasomes are multi-protein complexes, which are formed in response to tissue injury, infections, and metabolic stress. However, aberrant inflammasome activation has been linked to several inflammatory diseases. Anthocyanins have been reported to attenuate NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation, but the [...] Read more.
Inflammasomes are multi-protein complexes, which are formed in response to tissue injury, infections, and metabolic stress. However, aberrant inflammasome activation has been linked to several inflammatory diseases. Anthocyanins have been reported to attenuate NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation, but the influence of grape/blueberry anthocyanins and especially their gut-derived metabolites on NLRP3 inflammasome activation in human monocytes remains unclear. Therefore, human leukemic monocytes (THP-1 cells, Tohoku Hospital Pediatrics-1 cells) were preincubated with different concentrations of grape/blueberry anthocyanins, homovanillyl alcohol, or 2,4,6-trihydroxybenzaldehyde (THBA) before the NLRP3 inflammasome was activated by lipopolysaccharide and/or nigericin. Apoptosis-associated speck-like protein containing a CARD (ASC) speck formation, as well as ASC and NLRP3 protein expression, were determined using flow cytometry. Caspase-1 activity was measured in cultured cells, and pro-inflammatory cytokine secretion was determined using enzyme-linked immunosorbent assays. Anthocyanins and their metabolites had no effect on ASC or NLRP3 protein expression. However, THBA significantly inhibited ASC speck formation in primed and unprimed THP-1 monocytes, while caspase-1 activity was significantly declined by grape/blueberry anthocyanins. Furthermore, reduced inflammasome activation resulted in lower pro-inflammatory cytokine secretion. In conclusion, our results show for the first time that grape/blueberry anthocyanins and their gut-derived metabolites exert anti-inflammatory effects by attenuating NLRP3 inflammasome activation in THP-1 monocytes. Full article
(This article belongs to the Special Issue Role of Gut-Derived Metabolites in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Effect of grape/blueberry anthocyanins and their gut-derived metabolites on THP-1 cell viability. THP-1 monocytes were incubated with the indicated concentrations of (<b>a</b>) the GBE, (<b>b</b>) HVA, or (<b>c</b>) THBA for 24 h before cell viability was measured by flow cytometry. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). Significant differences compared to the untreated control were calculated using one-way ANOVA followed by Dunnett’s multiple comparisons test. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Detection of ASC speck formation in THP-1 cells using flow cytometry. THP-1 monocytes were either left untreated or primed with LPS followed by activation of the NLRP3 inflammasome with nigericin as mentioned in the methods section. Flow analysis was performed and the percentage of ASC speck-positive cells was quantified. Data are presented as (<b>a</b>) representative dot plots and (<b>b</b>) column bars with mean ± SD (<span class="html-italic">n</span> = 3). Significant differences compared to the untreated control were calculated using one-way ANOVA followed by Dunnett’s multiple comparisons test. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Effect of grape/blueberry anthocyanins and their gut-derived metabolites on ASC speck formation in THP-1 cells. THP-1 monocytes were preincubated with the indicated concentrations of grape/blueberry anthocyanins and their gut-derived metabolites before the NLRP3 inflammasome was activated as mentioned in the methods section. Cells were flow cytometrically analyzed and the percentage of ASC speck-positive cells was quantified. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). Significant differences compared to (<b>a</b>,<b>c</b>) LPS- and nigericin-stimulated cells or (<b>b</b>,<b>d</b>) cells treated only with nigericin were calculated using one-way ANOVA followed by Dunnett’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Effect of grape/blueberry anthocyanins and their gut-derived metabolites on caspase-1 activity in THP-cells. THP-1 monocytes were preincubated with the indicated concentrations of grape/blueberry anthocyanins and their gut-derived metabolites before the NLRP3 inflammasome was activated as mentioned in the methods section. Caspase-1 activity was measured by using the Caspase-Glo<sup>®</sup> 1 Inflammasome Assay and luminescence was measured. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3). Significant differences compared to (<b>a</b>,<b>c</b>) LPS- and nigericin-stimulated cells or (<b>b</b>,<b>d</b>) cells treated only with nigericin were calculated using one-way ANOVA followed by Dunnett’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001. RLU, relative light unit.</p>
Full article ">Figure 5
<p>Effect of grape/blueberry anthocyanins and their gut-derived metabolites on inflammatory cytokine secretion in THP-1 cells. THP-1 monocytes were preincubated with the indicated concentrations of grape/blueberry anthocyanins and their gut-derived metabolites before the NLRP3 inflammasome was activated as mentioned in the methods section. The release of (<b>a</b>,<b>c</b>) IL-1β and (<b>b</b>,<b>d</b>) IL-18 into the cell-culture supernatant was measured using ELISA. Data are presented as mean ± SD of at least three replicated experiments. Significant differences compared to LPS- and nigericin-stimulated cells were calculated using one-way ANOVA followed by Dunnett’s multiple comparisons test. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
12 pages, 256 KiB  
Article
Pan-Immune-Inflammation Value Could Be a New Marker to Predict Amyloidosis and Disease Severity in Familial Mediterranean Fever
by Tuğba Ocak, Ahmet Görünen, Belkıs Nihan Coşkun, Burcu Yağız, Sebnem Ozemri Sağ, Gökhan Ocakoğlu, Ediz Dalkılıç and Yavuz Pehlivan
Diagnostics 2024, 14(6), 634; https://doi.org/10.3390/diagnostics14060634 - 16 Mar 2024
Viewed by 1340
Abstract
Familial Mediterranean fever (FMF) is characterized by recurrent episodes of fever and serositis. Blood-based biomarkers determined in FMF patients during attack-free periods could be used to predict the risk of amyloidosis and the severity of the disease. The recently defined pan-immune-inflammation value (PIV) [...] Read more.
Familial Mediterranean fever (FMF) is characterized by recurrent episodes of fever and serositis. Blood-based biomarkers determined in FMF patients during attack-free periods could be used to predict the risk of amyloidosis and the severity of the disease. The recently defined pan-immune-inflammation value (PIV) comprises four distinct subsets of blood cells and serves as an easily accessible and cost-effective marker. The objective of this study was to assess the role of PIV in predicting amyloidosis and moderate-to-severe disease. Clinical characteristics and laboratory values during the attack-free period were retrospectively analyzed in 321 patients over 18 years of age diagnosed with familial Mediterranean fever (FMF). In our tertiary adult rheumatology outpatient clinic, disease severity and laboratory markers were evaluated during the first attack-free interval. At baseline, patients with amyloidosis were excluded. Patients were categorized based on the presence of amyloidosis and the severity of the disease. When focusing on amyloidosis in receiver operating characteristic (ROC) analysis, optimal cut-off values for pan-immune-inflammation value (PIV), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio were determined as ≥518.1, ≥2.3, and ≥127.2, respectively. In multivariate analysis, PIV, C-reactive protein (CRP), and the presence of the M694V homozygous mutation emerged as independent risk factors for both amyloidosis and moderate-to-severe disease. Additionally, NLR was identified as an independent risk factor for amyloidosis, while red blood cell distribution width was associated with moderate-to-severe disease. In patients with FMF, especially in the presence of the M694V homozygous mutation, CRP and PIV may be useful in predicting both amyloidosis and moderate-to-severe disease. Full article
(This article belongs to the Special Issue A Useful Diagnostic Method: Blood Test)
22 pages, 23951 KiB  
Article
Development of an NLR-ID Toolkit and Identification of Novel Disease-Resistance Genes in Soybean
by Wei Shao, Gongfu Shi, Han Chu, Wenjia Du, Zikai Zhou and Hada Wuriyanghan
Plants 2024, 13(5), 668; https://doi.org/10.3390/plants13050668 - 28 Feb 2024
Cited by 1 | Viewed by 1462
Abstract
The recognition of pathogen effectors through the nucleotide-binding leucine-rich repeat receptor (NLR) family is an important component of plant immunity. In addition to typical domains such as TIR, CC, NBS, and LRR, NLR proteins also contain some atypical integrated domains (IDs), the roles [...] Read more.
The recognition of pathogen effectors through the nucleotide-binding leucine-rich repeat receptor (NLR) family is an important component of plant immunity. In addition to typical domains such as TIR, CC, NBS, and LRR, NLR proteins also contain some atypical integrated domains (IDs), the roles of which are rarely investigated. Here, we carefully screened the soybean (Glycine max) genome and identified the IDs that appeared in the soybean TNL-like proteins. Our results show that multiple IDs (36) are widely present in soybean TNL-like proteins. A total of 27 Gm-TNL-ID genes (soybean TNL-like gene encoding ID) were cloned and their antiviral activity towards the soybean mosaic virus (SMV)/tobacco mosaic virus (TMV) was verified. Two resistance (R) genes, SRA2 (SMV resistance gene contains AAA_22 domain) and SRZ4 (SMV resistance gene contains zf-RVT domain), were identified to possess broad-spectrum resistance characteristics towards six viruses including SMV, TMV, plum pox virus (PPV), cabbage leaf curl virus (CaLCuV), barley stripe mosaic virus (BSMV), and tobacco rattle virus (TRV). The effects of Gm-TNL-IDX (the domain of the Gm-TNL-ID gene after the TN domain) on the antiviral activity of a R protein SRC7TN (we previously reported the TN domain of the soybean broad-spectrum resistance gene SRC7) were validated, and most of Gm-TNL-IDX inhibits antiviral activity mediated by SRC7TN, possibly through intramolecular interactions. Yeast-two-hybrid (Y2H) and bimolecular fluorescence complementation (BiFC) assays showed that seven Gm-TNL-IDX interacted with SMV-component proteins. Truncation analysis on a broad-spectrum antiviral protein SRZ4 indicated that SRZ4TIR is sufficient to mediate antiviral activity against SMV. Soybean cDNA library screening on SRZ4 identified 48 interacting proteins. In summary, our results indicate that the integration of IDs in soybean is widespread and frequent. The NLR-ID toolkit we provide is expected to be valuable for elucidating the functions of atypical NLR proteins in the plant immune system and lay the foundation for the development of engineering NLR for plant-disease control in the future. Full article
(This article belongs to the Special Issue Molecular Genetics and Breeding of Oilseed Crops)
Show Figures

Figure 1

Figure 1
<p>Overview of ID in soybean TNL-like proteins. (<b>A</b>) Integrated domain (ID) locations, indicated by black rectangles, are shown within soybean TNL-like proteins relative to protein length (0–1); (<b>B</b>) IDs were grouped into functional categories based on their potential involvement in nucleic acid-binding, other signaling activity pathways, or unknown functional pathways (shown in light purple, light blue, and light yellow, respectively); (<b>C</b>) word cloud analysis of the putative IDs found in fusion to soybean TNL-like proteins. The word cloud represents relative abundance of different domains found in fusion.</p>
Full article ">Figure 2
<p>Antiviral activity of these <span class="html-italic">Gm-TNL-ID</span> genes in <span class="html-italic">Nicotiana benthamiana</span>. (<b>A</b>,<b>B</b>) Transient expression assay for antiviral activity. <span class="html-italic">N. benthamiana</span> leaves were infiltrated with <span class="html-italic">A. tumefaciens</span> inocula carrying different overexpression vectors for <span class="html-italic">Gm-TNL-ID</span> genes, and co-infected with SMV-GFP (<b>A</b>) or TMV-GFP (<b>B</b>). GFP was visualized under hand-held UV lamp (wavelength = 365 nm) at 4 dpi. SRC7 and SRC7<sup>TN</sup>: full-length SRC7 and truncations for SRC7 were used as a positive control. EV, empty vector. Each experiment was repeated three times with at least 20–30 technical replicates; (<b>C</b>,<b>D</b>) fluorescence intensity quantification of virus spread through Image J v1.8.0. The fluorescence diffusion of leaf infiltration sites under UV light after 4 dpi of mixed infiltration of SMV-GFP (<b>C</b>) and TMV-GFP (<b>D</b>), with the target gene was counted using software Image J v1.8.0 and analyzed for differences. <span class="html-italic">Y</span>-axis represents the mean fluorescence intensity of leaf infiltration region and <span class="html-italic">X</span>-axis represents the target gene. The fluorescence intensity of each gene was subjected to 10 technical replicates; (<b>E</b>,<b>F</b>) virus titer detection via RT-qPCR. Total RNA was extracted from a mixed sample of 10 leaves. <span class="html-italic">Y</span>-axis represents the mean virus titers of leaf infiltration region and <span class="html-italic">X</span>-axis represents the target gene. In <span class="html-italic">t</span>-test, bars represent SD of three independent biological replicates, each with three repeats, ns: no significant difference, *: <span class="html-italic">p &lt;</span> 0.05, **: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Identification of broad-spectrum resistance of <span class="html-italic">Gm-TNL-ID</span> genes in <span class="html-italic">Nicotiana benthamiana</span>. <span class="html-italic">N. benthamiana</span> leaves were infiltrated with <span class="html-italic">A. tumefaciens</span> inocula carrying different overexpression vectors and co-infected with PPV, CaLCuV, BSMV, or TRV. Tissue RNA was extracted from infiltrated region and was used for quantitative analysis of PPV (<b>A</b>), CaLCuV (<b>B</b>), BSMV (<b>C</b>), and TRV (<b>D</b>) virus titers. <span class="html-italic">Y</span>-axis represents the mean virus titers of leaf infiltration region and <span class="html-italic">X</span>-axis represents the target gene. In <span class="html-italic">t</span>-test, bars represent SD of three independent biological replicates, each with three repeats, ns: no significant difference, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>The effect of Gm-TNL-ID<sup>X</sup> domain on antiviral resistance of SRC7<sup>TN</sup> and its interaction with SRC7<sup>TN</sup>. (<b>A</b>,<b>B</b>) Transient expression assay for antiviral activity. <span class="html-italic">N. benthamiana</span> leaves were infiltrated with <span class="html-italic">A. tumefaciens</span> inocula carrying different overexpression vectors for SRC7<sup>TN</sup>-ID<sup>X</sup> and co-infected with SMV-GFP (<b>A</b>) or TMV-GFP (<b>B</b>). GFP was visualized under hand-held UV lamp (wavelength = 365 nm) at 4 dpi. SRC7 and SRC7<sup>TN</sup>: full-length SRC7 and truncations for SRC7 were used as a positive control. EV, empty vector. Each experiment was repeated three times with at least 20–30 technical replicates; (<b>C</b>) Y2H assay. The yeast cells containing pGADT7 (AD) -SRC7<sup>TN</sup> and different pGBKT7 (BD) -Gm-TNL-ID<sup>X</sup> constructs were grown on different selective media, and 100 μL of X-α-Gal was added and visualized the reporter expression. The yeast concentration gradients were 10<sup>−1</sup>, 10<sup>−2</sup> and 10<sup>−3</sup>, respectively. -L-T = yeast growth on medium lacking Leu and Trp, -L-W-H = yeast growth on medium lacking Leu, Trp, and His, -L-W-H-A = yeast growth on medium lacking Leu, Trp, His, and Ade.</p>
Full article ">Figure 5
<p>Interaction assay for Gm-TNL-ID<sup>X</sup> domain and SMV-component proteins. (<b>A</b>) Y2H assay. The yeast cells containing ten AD-SMV-component proteins and different BD-Gm-TNL-ID<sup>X</sup> constructs were grown on different selective media, and 100 μL of X-α-Gal was added and visualized the reporter expression. The yeast concentration gradients were 10<sup>−1</sup>, 10<sup>−2</sup>, and 10<sup>−3</sup>, respectively. (<b>B</b>) BiFC further validates the protein–protein interaction between the Gm-TNL-ID<sup>X</sup> domain and SMV components in <span class="html-italic">N. benthamiana</span>. The Gm-TNL-ID<sup>X</sup> and SMV-component proteins’ constructs were transiently expressed by co-infiltration of cVenus and nVenus fusions into <span class="html-italic">N. benthamiana</span> leaves. At 3 dpi, yellow fluorescent protein (YFP) fluorescence imaging was performed using confocal laser scanning microscopy.</p>
Full article ">Figure 6
<p>Subcellular localization and identification of antiviral functional domains of SRZ4. (<b>A</b>) Subcellular localization of SRZ4. The SRZ4–GFP fusion protein was transiently co-expressed with membrane marker (mCherry) in <span class="html-italic">N. benthamiana</span> leaf epidermis cells. The fluorescence signals were observed using confocal microscopy at 3 dpi. Scale bars = 20 µm; (<b>B</b>) transient expression assay for antiviral activity. <span class="html-italic">N. benthamiana</span> leaves were infiltrated with <span class="html-italic">A. tumefaciens</span> inocula carrying different SRZ4 truncated vectors, and co-infected with SMV-GFP. Each experiment was repeated three times with at least 20–30 technical replicates. GFP was visualized under hand-held UV lamp (wavelength = 365 nm) at 4 dpi. EV, empty vector.</p>
Full article ">Figure 7
<p>Interaction protein analysis of SRZ4. (<b>A</b>) Schematic diagram for screening and subculture of cDNA library. Using the mating method, BD-SRZ4 and soybean cDNA library were co-transformed into yeast cells and coated on 150 mm SD/-L/-W/-H plates. We selected colonies with good growth conditions and inoculated them onto SD/-L/-W/-H/-A plates. After 5–7 days, a total of 126 single colonies were obtained on SD/-L/-W/-H/-A plates. After three generations, selected single colonies that can turn blue and grow well in SD/-L/-W/-H/-A/X-α-Gal plate; (<b>B</b>) yeast colony PCR. PCR amplification was performed on each colony using universal primers from the yeast library, and PCR products with different band sizes were selected for sequencing; (<b>C</b>) GO enrichment analysis of SRZ4-interacting proteins. <span class="html-italic">X</span>-axis represents GO analysis annotation, where BP: biology process, CC: cellular component, and MF: molecular function. The <span class="html-italic">Y</span>-axis represents the number of SRZ4-interacting proteins. The black arrows represent the biological functions that interest us.</p>
Full article ">
20 pages, 3753 KiB  
Article
Implications of NLRP3 Suppression Using Glibenclamide and miR-223 against Colorectal Cancer
by Shaimaa Hamza, Ekaterina E. Garanina, Layaly Shkair, Mohammad Alsaadi, Svetlana F. Khaiboullina and Gulcin Tezcan
Pharmaceuticals 2024, 17(3), 299; https://doi.org/10.3390/ph17030299 - 26 Feb 2024
Cited by 1 | Viewed by 1557
Abstract
The NLR family pyrin domain containing 3 (NLRP3) promotes the growth of colorectal cancer (CRC). However, the therapeutic effect of NLRP3 inhibition on CRC cell progression is controversial. This study comparatively investigated the therapeutic effect of a pharmacological NLRP3 inhibitor, glibenclamide (gli), and [...] Read more.
The NLR family pyrin domain containing 3 (NLRP3) promotes the growth of colorectal cancer (CRC). However, the therapeutic effect of NLRP3 inhibition on CRC cell progression is controversial. This study comparatively investigated the therapeutic effect of a pharmacological NLRP3 inhibitor, glibenclamide (gli), and the post-translational suppression of NLRP3 by miR-223 on CRC cell progression in HCT-116 and HCT-15 cells. LPS and ATP were used to activate Gli-treated and LSB-hsa-miR-223-3p (WTmiR-223)-expressing HCT-116 cells. NLRP3.AB.pCCL.sin.cPPT.U6.miR-223-Decoy.hPGK.GFP.WPRE plasmid (DmiR-223) was the negative control for miR-223 expression. NLRP3, gasdermin D, and BAX expressions were analyzed using western blotting. Real-time PCR detected the RNA expression of autophagy-related genes ATG5, BECN1, and miR-223 in non-transfected cells. ELISA analyzed IL-1β and IL-18 in the medium. MTS-1, annexin V, wound-healing, and sphere-invasion assays were used to assess cell viability and progression. A multiplex cytokine assay detected proinflammatory cytokine secretion. LPS–ATP-activated NLRP3 produced gasdermin D cleavage, released IL-1b and IL-18, and activated cell migration and sphere invasion. In contrast, reduced cell growth, miR-223 expression, IFN-γ, CXCL10, and LIF secretion were found in cells after inflammasome activation. Both gli and WTmiR-223 induced autophagy genes ATG5 and BECN1 and reduced the NLRP3 activation and its downstream proteins. However, while gli had a limited effect on the production of IFN-γ, CXCL10, and LIF, WTmiR-223 increased the release of those cytokines. In addition, gli did not suppress cell growth, while WTmiR-223 promoted apoptosis. Notably, neither gli nor WTmiR-223 effectively prevented sphere invasion. These data suggest that, while WTmiR-223 could have a better anticancer effect in CRC compared to gli, the sole usage of miR-223-mediated NLRP3 suppression may not be sufficient to prevent CRC metastasis. Full article
(This article belongs to the Special Issue Inflammasomes as the Target of Pharmacotherapy)
Show Figures

Figure 1

Figure 1
<p>Effect of gli on NLRP3 function in HCT-116 cells. LPS (1 µg/mL, 3 h) was used to prime NLRP3, followed by ATP (5 mM ATP; 25 min) to activate the inflammasome. Gli (50 ug/mL) was added 24 h before NLRP3 induction to inhibit the inflammasome. (<b>A</b>) Western blot of NLRP3 and gasdermin D proteins. (<b>B</b>) ELISA assesses IL-1β and (<b>C</b>) IL-18 secretion. (<b>D</b>) The MTS assay values of treated cells were normalized to the untreated sample. (<b>E</b>) The Annexin V assay of cell viability. (<b>F</b>) ATG5 and BECN1 RNA expression levels. Data represent three biological replicates, and <span class="html-italic">p</span>-value was calculated using the one-way ANOVA model with Tukey’s post hoc tests. * <span class="html-italic">p</span> &lt; 0.05. U: Untreated, L: LPS only, LA: LPS–ATP, GLA: Gli–LPS–ATP.</p>
Full article ">Figure 2
<p>The effect of gli inhibition of NLRP3 on migration and sphere formation of HCT-116 cells. LPS (1 µg/mL, 3 h) was used to prime NLRP3, followed by ATP (5 mM ATP; 25 min) to activate the inflammasome. Gli (50 ug/mL) inhibited NLRP3 24 h before LPR–ATP induction. (<b>A</b>) Wound-healing of HCT-116 cells. Images were captured before LPS treatment (time 0) and 24 h after ATP treatment. (<b>B</b>) Sphere formation and invading capacity of HCT-116 cells. Images were captured 24–72 h after ATP treatment. Images were analyzed using Image J 1.53s software (NIH, Bethesda, MD, USA). Data represent three biological replicates. The <span class="html-italic">p</span>-value was calculated using an independent sample <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05 U: Untreated, LA: LPS–ATP, GLA: Gli–LPS–ATP, WSI: Wound size at the initiation.</p>
Full article ">Figure 3
<p>The effect of gli on cytokine secretion in LPS–ATP-induced HCT-116 cells. (<b>A</b>) The heat map shows changes in cytokine secretion patterns in HCT-116 cells after LPS–ATP and gli–LPS–ATP treatments. Cytokines with concentrations below the detection level were excluded from the heatmap graphs. (<b>B–G</b>) The cytokines, which were affected by LPS–ATP and gli–LPS–ATP. Data represent three biological replicates. The <span class="html-italic">p</span>-value was calculated using a Kruskal–Wallis test. H: Test statistic for the Kruskal–Wallis test. * <span class="html-italic">p</span> &lt; 0.05 U: Untreated, LA: LPS–ATP, GLA: Gli–LPS–ATP.</p>
Full article ">Figure 4
<p>The effect of miR-223 on NLRP3 in HCT-116 cells. (<b>A</b>) The interaction of has-miR-223-3p with the 3′UTS of NLRP3 gene (Targetscan); (<b>B</b>) Effect of LPS–ATP and gli–LPS–ATP on miR-223 expression of HCT-116 cells; (<b>C</b>) Immunofluorescence (IF) analysis of the cells transfected with WT<sup>miR-223</sup> (blue) and D<sup>miR-223</sup> (green). WT<sup>miR-223</sup> expressed cells were selected through puromycin treatment, while D<sup>miR-223</sup> expressed cells were selected with flow sorting. (<b>D</b>) Western blotting of NLRP3 and gasdermin D proteins; (<b>E</b>) ELISA assessment of IL-1β; (<b>F</b>) IL-18 secretion. (<b>G</b>) The MTS assay values of D<sup>miR-223</sup> and WT<sup>miR-223</sup> expressing cells; (<b>H</b>) Western blotting of BAX protein. (<b>I</b>) ATG5 and BECN1 RNA expression levels in untreated and (<b>J</b>) LPS–ATP treated D<sup>miR-223</sup> and WT<sup>miR-223</sup> expressing cells. n = 2 for each experiment. * <span class="html-italic">p</span>-value was calculated using an independent sample <span class="html-italic">t</span>-test for (<b>B</b>) and a one-way Anova and Tukey Test for (<b>D</b>–<b>H</b>). * <span class="html-italic">p</span> value &lt; 0.05. U: Untreated, LA: LPS–ATP.</p>
Full article ">Figure 5
<p>The effect of miR-223 on migration and sphere formation of HCT-116 cells. LPS (1 µg/mL, 3 h) was used to prime NLRP3, followed by ATP (5 mM ATP; 25 min) to activate the inflammasome. (<b>A</b>) Wound healing of HCT-116 cells. Images were captured before LPS treatment (time 0) and 24 h after ATP treatment. (<b>B</b>) Sphere-forming and invading capacity of HCT-116 cells. Images were captured 24–72 h after ATP treatment. Images were analyzed using Image J 1.53s software (NIH, Bethesda, MD, USA). Data represent three biological replicates. <span class="html-italic">p</span>-value was calculated using an independent sample <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05 D: Decoy, WT: Wild type, U: Untreated, LA: LPS–ATP, WSI: Wound size at initiation.</p>
Full article ">Figure 6
<p>The effect miR-223 on cytokine secretion in inflammasome induced by LPS–ATP HCT-116 cells. (<b>A</b>)The heat map shows the changes in the levels of cytokine secretion patterns in HCT-116 cells after LPS–ATP and gli–LPS–ATP treatments. (<b>B–D</b>) The cytokines, which were affected by LPS–ATP. Data represent three biological replicates. The <span class="html-italic">p</span>-value was calculated using a Kruskal–Wallis test. H: Test statistic for the Kruskal–Wallis test. * <span class="html-italic">p</span> &lt; 0.05 D: Decoy, WT: Wild type, U: Untreated, LA: LPS–ATP.</p>
Full article ">
16 pages, 2693 KiB  
Article
Stefin B Inhibits NLRP3 Inflammasome Activation via AMPK/mTOR Signalling
by Mojca Trstenjak-Prebanda, Monika Biasizzo, Klemen Dolinar, Sergej Pirkmajer, Boris Turk, Veronique Brault, Yann Herault and Nataša Kopitar-Jerala
Cells 2023, 12(23), 2731; https://doi.org/10.3390/cells12232731 - 29 Nov 2023
Cited by 1 | Viewed by 1382
Abstract
Stefin B (cystatin B) is an inhibitor of lysosomal and nuclear cysteine cathepsins. The gene for stefin B is located on human chromosome 21 and its expression is upregulated in the brains of individuals with Down syndrome. Biallelic loss-of-function mutations in the stefin [...] Read more.
Stefin B (cystatin B) is an inhibitor of lysosomal and nuclear cysteine cathepsins. The gene for stefin B is located on human chromosome 21 and its expression is upregulated in the brains of individuals with Down syndrome. Biallelic loss-of-function mutations in the stefin B gene lead to Unverricht–Lundborg disease-progressive myoclonus epilepsy type 1 (EPM1) in humans. In our past study, we demonstrated that mice lacking stefin B were significantly more sensitive to sepsis induced by lipopolysaccharide (LPS) and secreted higher levels of interleukin 1-β (IL-1β) due to increased inflammasome activation in bone marrow-derived macrophages. Here, we report lower interleukin 1-β processing and caspase-11 expression in bone marrow-derived macrophages prepared from mice that have an additional copy of the stefin B gene. Increased expression of stefin B downregulated mitochondrial reactive oxygen species (ROS) generation and lowered the NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in macrophages. We determined higher AMP-activated kinase phosphorylation and downregulation of mTOR activity in stefin B trisomic macrophages—macrophages with increased stefin B expression. Our study showed that increased stefin B expression downregulated mitochondrial ROS generation and increased autophagy. The present work contributes to a better understanding of the role of stefin B in regulation of autophagy and inflammasome activation in macrophages and could help to develop new treatments. Full article
(This article belongs to the Section Autophagy)
Show Figures

Figure 1

Figure 1
<p><b>Decreased caspase-11 mRNA levels in StB3n BMDMs following LPS stimulation.</b> BMDMs were challenged with LPS (100 ng/mL) for 4 h and total RNA was then isolated. (<b>A</b>–<b>D</b>) Relative mRNA expression was measured, normalised to <span class="html-italic">Gapdh</span> and <span class="html-italic">B2m</span> reference genes, and shown as fold increases compared with control (unstimulated) samples. Three independent experiments were performed in triplicate, and the results are presented as mean ± S.D. *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p><b>NLPR3 inflammasome activation in StB3n BMDMs.</b> a(<b>A</b>) Cell lysates were blotted with the indicated antibodies. (<b>B</b>) BMDMs were seeded on 96-well plates and stimulated, as indicated. IL-1β was determined in cell culture supernatants using an enzyme-linked immunosorbent assay (ELISA). (<b>C</b>) BMDMs viability was determined using LDH release into the cell culture media. Cytotoxicity is represented as a percentage of total LDH released. Relative caspase-11, caspase-1, cleaved caspase-1, pro-IL-1<span class="html-italic">β</span>, pro-IL-18, and IL-18 protein band intensities were quantified using ImageJ software, and the quantities of the target proteins were normalised to those of <span class="html-italic">β</span>-actin. The results are representative of three independent experiments. Bars represent mean ± S.E.M. *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p><b>Decreased nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression in stefin B 3n BMDMs.</b> (<b>A</b>) NO release in the BMDM supernatants was measured via the Griess reagent. Macrophages were cultured in 96-well plates and stimulated with IFN-γ (100 U/mL) and LPS (100 ng/mL) for 18 h or treated overnight with IFN-γ (100 U/mL) and then stimulated with LPS (100 ng/mL) for 18 h. Absorbance was measured at 545 nm. Data were obtained from three independent experiments performed in triplicate, with three different biological samples, and the results are presented as mean ± S.D. **, <span class="html-italic">p</span> &lt; 0.01. (<b>B</b>) Cells were left untreated or primed with IFN-γ (100 U/mL) and then stimulated with LPS (100 U/mL). Whole cell lysates were analysed via Western blotting with iNOS-specific antibodies. Band intensities were quantified and iNOS quantities were normalised to those of <span class="html-italic">β</span>-actin. The results are representative of three independent experiments. Bars represent mean ± S.E.M. **, <span class="html-italic">p</span> &lt; 0.01. (<b>C</b>,<b>D</b>) Cysteine cathepsin activities in RAW 264.7 mouse macrophages and RAW 264.7 mouse macrophages stably transfected with stefin B (RMA cells). Cells were treated for 2 h with E64d (20 µM), stimulated for 4 h with LPS (100 ng/mL), and treated for 20 min with ATP (5 mM). (<b>C</b>,<b>D</b>) Cells were lysed with digitonin (200 µg/mL) and enzyme activity was measured using fluorogenic substrates: cathepsin L-like activity (zFR–AMC); cathepsin B-like activity (zRR–AMC). Three independent experiments were performed in triplicate; the results are presented as mean ± S.D. **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p><b>Increased stefin B expression prevents accumulation of dysfunctional mitochondria.</b> RAW 264.7 mouse macrophages transfected with an empty plasmid (RAW R0 cells) or with stefin B (RAW RMA cells) were stimulated with 100 ng/mL LPS for 4 and 24 h and further labelled with MitoTracker Green (total mitochondrial mass) and MitoSOX Red (in order to determine mitochondrial ROS). Three independent experiments were performed in duplicate; the results are shown as means ± S.D. (*, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01. ***, <span class="html-italic">p</span> &lt; 0.005).</p>
Full article ">Figure 5
<p><b>RAW 264.7 mouse macrophages with increased expression of stefin B (RMA cells) exhibit altered metabolic profiles following LPS stimulation.</b> R0 and RMA RAW 264.7 cells were treated with LPS (100 ng/mL) or vehicle (control) for 4 h or 12 h. Mito stress test was then performed in Seahorse Analyzer using oligomycin (OM) (1.5 μM), FCCP (1.5 μM), and rotenone + antimycin A (R + AA) (each 0.5 μM). Graphs show group means and SEM (n = 10; 2 experiments with 5 replicates per group in each experiment). Graphs on the left: ECAR and OCR curves. Graphs on the right: OCR and ECAR after adding OM, FCCP, and R + AA, normalised to the respective basal measurement. Each data point represents the average of three consecutive normalised measurements from each well taken during each phase of mito stress. *, <span class="html-italic">p</span> &lt; 0.05, R0 LPS vs. R0 control and RMA LPS vs. RMA control in the same phase of mito stress test; #, <span class="html-italic">p</span> &lt; 0.05, RMA control vs R0 control and RMA LPS vs R0 LPS in the same phase of mito stress test; two-way ANOVA with Bonferroni’s test.</p>
Full article ">Figure 6
<p><b>Stefin B promotes the induction of autophagy.</b> (<b>A</b>) Mitochondria (red) were labelled with MitoTracker Red CMXROS, treated as written in the Materials and Methods section. Cells were further incubated with anti-LC3 antibodies (green) and labelled with Alexa conjugated secondary antibodies (Alexa 488). Co-localisation of LC3 vesicles and mitochondria was determined in merged images. The image is representative of three independent experiments. Upon carbonyl cyanide m-chlorophenylhydrazone (CCCP) treatment (20 μM for 4 h), cells were fixed and stained for LC3 and MitoTracker Red in order to evaluate mitophagy. Scale bar 10 μm. (<b>B</b>) WT and StB3n BMDMs were pretreated with Bafilomycin A1 (80 nM) for 1 h and stimulated with LPS (100 ng/mL) for the indicated times. Cell lysate proteins were analysed via immunoblotting using anti-LC3 antibodies (<b>A</b>) and P62 antibodies. Images are representative of three independent experiments. Relative LC3-II and p62 protein levels were quantified using ImageJ software and the values were normalised to those of <span class="html-italic">β</span>-actin. The results are representative of three independent experiments. Bars represent mean ± S.E.M. *, <span class="html-italic">p</span> &lt; 0.05. **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p><b>Stefin B influences autophagy and mitochondrial integrity by inhibiting mTOR.</b> (<b>A</b>) BMDMs were treated with LPS (100 ng/mL) for 4 h or 24 h. (<b>B</b>) BMDMs were treated with LPS (100 ng/mL) for the indicated times. Cell lysates were immunoblotted with specific antibodies, as indicated. Images are representative of three independent experiments. Relative pUlk, pAMPK, and p-70 phosphorylated protein band intensities were quantified using ImageJ software and the values were normalised to those of <span class="html-italic">β</span>-actin. The results are representative of three independent experiments. Bars represent mean ± S.E.M. **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
25 pages, 6338 KiB  
Article
Glycine-Alanine Dipeptide Repeat Protein from C9-ALS Interacts with Sulfide Quinone Oxidoreductase (SQOR) to Induce the Activity of the NLRP3 Inflammasome in HMC3 Microglia: Irisflorentin Reverses This Interaction
by Ru-Huei Fu, Hui-Jye Chen and Syuan-Yu Hong
Antioxidants 2023, 12(10), 1896; https://doi.org/10.3390/antiox12101896 - 23 Oct 2023
Cited by 4 | Viewed by 1926
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal rare disease of progressive degeneration of motor neurons. The most common genetic mutation in ALS is the hexanucleotide repeat expansion (HRE) located in the first intron of the C9orf72 gene (C9-ALS). HRE can produce dipeptide repeat [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a fatal rare disease of progressive degeneration of motor neurons. The most common genetic mutation in ALS is the hexanucleotide repeat expansion (HRE) located in the first intron of the C9orf72 gene (C9-ALS). HRE can produce dipeptide repeat proteins (DPRs) such as poly glycine-alanine (GA) in a repeat-associated non-ATG (RAN) translation. GA-DPR has been shown to be toxic to motor neurons in various biological models. However, its effects on microglia involved in C9-ALS have not been reported. Here, we show that GA-DPR (GA50) activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome in a human HMC3 microglia model. MCC950 (specific inhibitor of the NLRP3) treatment can abrogate this activity. Next, using yeast two-hybrid screening, we identified sulfide quinone oxidoreductase (SQOR) as a GA50 interacting protein. SQOR knockdown in HMC3 cells can significantly induce the activity of the NLRP3 inflammasome by upregulating the level of intracellular reactive oxygen species and the cytoplasmic escape of mitochondrial DNA. Furthermore, we obtained irisflorentin as an effective blocker of the interaction between SQOR and GA50, thus inhibiting NLRP3 inflammasome activity in GA50-expressing HMC3 cells. These results imply the association of GA-DPR, SQOR, and NLRP3 inflammasomes in microglia and establish a treatment strategy for C9-ALS with irisflorentin. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Intracellular expression of GA50 in human HMC3 microglia leads to NLRP3 inflammasome activation. The control vector or GA<sub>50</sub> plasmid was transfected into HMC3 cells for 24 h. (<b>A</b>) Immunofluorescence staining of GA<sub>50</sub> using anti-Myc-tag antibody (green). The location of the nuclei was confirmed by DAPI staining. The magnification of a microscope is 400X. GA<sub>50</sub> mainly forms aggregates in the cytoplasm (red arrows). (<b>B</b>) The amount of NF-κB p65 in the nucleus was evaluated by Western blotting and quantified using ImageJ software (version 1.53). TBP protein is an internal loading control. (<b>C</b>) ELISA analysis quantifies the transcriptional activity of NF-κB p65. (<b>D</b>) Expression of GA<sub>50</sub> (Myc), NLRP3, ASC, caspase-1, IL-1β, and IL-18 in cell lysates was analyzed by Western blot and quantified using ImageJ software (version 1.53). GAPDH was employed as an internal loading control. (<b>E</b>) The amount of IL-1β and IL-18 secreted in conditional media was determined using ELISA assay. (<b>F</b>) The number of cells with chromosomal breaks was estimated using TUNEL assay. The magnification of a microscope is 100X. (<b>G</b>) Ratios of pyroptosis cell were determined by flow cytometry with annexin v-FITC and propidium iodide (PI) staining. The histogram shows the pyroptosis rate [(Q1 + Q2)/(Q1 + Q2 + Q3 + Q4) × 100%].</p>
Full article ">Figure 1 Cont.
<p>Intracellular expression of GA50 in human HMC3 microglia leads to NLRP3 inflammasome activation. The control vector or GA<sub>50</sub> plasmid was transfected into HMC3 cells for 24 h. (<b>A</b>) Immunofluorescence staining of GA<sub>50</sub> using anti-Myc-tag antibody (green). The location of the nuclei was confirmed by DAPI staining. The magnification of a microscope is 400X. GA<sub>50</sub> mainly forms aggregates in the cytoplasm (red arrows). (<b>B</b>) The amount of NF-κB p65 in the nucleus was evaluated by Western blotting and quantified using ImageJ software (version 1.53). TBP protein is an internal loading control. (<b>C</b>) ELISA analysis quantifies the transcriptional activity of NF-κB p65. (<b>D</b>) Expression of GA<sub>50</sub> (Myc), NLRP3, ASC, caspase-1, IL-1β, and IL-18 in cell lysates was analyzed by Western blot and quantified using ImageJ software (version 1.53). GAPDH was employed as an internal loading control. (<b>E</b>) The amount of IL-1β and IL-18 secreted in conditional media was determined using ELISA assay. (<b>F</b>) The number of cells with chromosomal breaks was estimated using TUNEL assay. The magnification of a microscope is 100X. (<b>G</b>) Ratios of pyroptosis cell were determined by flow cytometry with annexin v-FITC and propidium iodide (PI) staining. The histogram shows the pyroptosis rate [(Q1 + Q2)/(Q1 + Q2 + Q3 + Q4) × 100%].</p>
Full article ">Figure 1 Cont.
<p>Intracellular expression of GA50 in human HMC3 microglia leads to NLRP3 inflammasome activation. The control vector or GA<sub>50</sub> plasmid was transfected into HMC3 cells for 24 h. (<b>A</b>) Immunofluorescence staining of GA<sub>50</sub> using anti-Myc-tag antibody (green). The location of the nuclei was confirmed by DAPI staining. The magnification of a microscope is 400X. GA<sub>50</sub> mainly forms aggregates in the cytoplasm (red arrows). (<b>B</b>) The amount of NF-κB p65 in the nucleus was evaluated by Western blotting and quantified using ImageJ software (version 1.53). TBP protein is an internal loading control. (<b>C</b>) ELISA analysis quantifies the transcriptional activity of NF-κB p65. (<b>D</b>) Expression of GA<sub>50</sub> (Myc), NLRP3, ASC, caspase-1, IL-1β, and IL-18 in cell lysates was analyzed by Western blot and quantified using ImageJ software (version 1.53). GAPDH was employed as an internal loading control. (<b>E</b>) The amount of IL-1β and IL-18 secreted in conditional media was determined using ELISA assay. (<b>F</b>) The number of cells with chromosomal breaks was estimated using TUNEL assay. The magnification of a microscope is 100X. (<b>G</b>) Ratios of pyroptosis cell were determined by flow cytometry with annexin v-FITC and propidium iodide (PI) staining. The histogram shows the pyroptosis rate [(Q1 + Q2)/(Q1 + Q2 + Q3 + Q4) × 100%].</p>
Full article ">Figure 2
<p>The activity of the NLRP3 inflammasome in GA<sub>50</sub>-expressing HMC3 microglia can be inhibited by MCC950. HMC3 cells that expressed GA<sub>50</sub> were treated with 0, 25, 50, or 100 nM MCC950 for 24 h. (<b>A</b>) Protein levels of GA<sub>50</sub> (Myc) and NLRP3 inflammasome-associated components in cell lysates detected by Western blot analysis. The internal loading control was GAPDH protein. (<b>B</b>) The conditional medium was used to measure the secretion of mature IL-1β and mature IL-18 by ELISA.</p>
Full article ">Figure 2 Cont.
<p>The activity of the NLRP3 inflammasome in GA<sub>50</sub>-expressing HMC3 microglia can be inhibited by MCC950. HMC3 cells that expressed GA<sub>50</sub> were treated with 0, 25, 50, or 100 nM MCC950 for 24 h. (<b>A</b>) Protein levels of GA<sub>50</sub> (Myc) and NLRP3 inflammasome-associated components in cell lysates detected by Western blot analysis. The internal loading control was GAPDH protein. (<b>B</b>) The conditional medium was used to measure the secretion of mature IL-1β and mature IL-18 by ELISA.</p>
Full article ">Figure 3
<p>GA-DPR specifically interacts with sulfide quinone oxidoreductase (SQOR) in HMC3 cells. (<b>A</b>) Flowchart showing the yeast two-hybrid screening strategy, using GA<sub>50</sub> as a bait to search for potential interacting proteins in a human HMC3 microglia cDNA library. (<b>B</b>) The reporter gene activated by the interaction induces the growth of blue diploid yeast containing positive clones of the <span class="html-italic">SQOR</span> cDNA fragment (red arrows). (<b>C</b>) Schematic representation of three cDNA clones spanning the coding sequence of the <span class="html-italic">SQOR</span> gene (NCBI reference sequence: NP_067022.1). SQOR comprises the FadH2 domain: sulfur reductase. (<b>D</b>) The interaction of GA<sub>50</sub> with full-length SQOR was confirmed using the yeast two-hybrid assay. The positive control group was diploid yeast expressing BD-p53 and AD-T. (<b>E</b>) Confirmation of GA<sub>50</sub> interaction with SQOR using co-immunoprecipitation analysis. 293T cells transfected with only one plasmid were used as a negative control. (<b>F</b>) Analysis using immunofluorescence staining confirmed that GA<sub>50</sub> and SQOR are partially colocalized in the cytoplasm of HMC3 cells (white arrows). DAPI was used to stain the nuclei.</p>
Full article ">Figure 3 Cont.
<p>GA-DPR specifically interacts with sulfide quinone oxidoreductase (SQOR) in HMC3 cells. (<b>A</b>) Flowchart showing the yeast two-hybrid screening strategy, using GA<sub>50</sub> as a bait to search for potential interacting proteins in a human HMC3 microglia cDNA library. (<b>B</b>) The reporter gene activated by the interaction induces the growth of blue diploid yeast containing positive clones of the <span class="html-italic">SQOR</span> cDNA fragment (red arrows). (<b>C</b>) Schematic representation of three cDNA clones spanning the coding sequence of the <span class="html-italic">SQOR</span> gene (NCBI reference sequence: NP_067022.1). SQOR comprises the FadH2 domain: sulfur reductase. (<b>D</b>) The interaction of GA<sub>50</sub> with full-length SQOR was confirmed using the yeast two-hybrid assay. The positive control group was diploid yeast expressing BD-p53 and AD-T. (<b>E</b>) Confirmation of GA<sub>50</sub> interaction with SQOR using co-immunoprecipitation analysis. 293T cells transfected with only one plasmid were used as a negative control. (<b>F</b>) Analysis using immunofluorescence staining confirmed that GA<sub>50</sub> and SQOR are partially colocalized in the cytoplasm of HMC3 cells (white arrows). DAPI was used to stain the nuclei.</p>
Full article ">Figure 4
<p>Confirmation of the C-terminus of SQOR as the major interaction region of GA<sub>50</sub> by the yeast two-hybrid test. (<b>A</b>) Schematic showing that SQOR is divided into three fragments for the yeast two-hybrid test. (<b>B</b>–<b>D</b>) GA<sub>50</sub> only interacts with the S3 fragment of SQOR in the yeast two-hybrid assay. The positive control was diploid yeast expressing BD-p53 and AD-T.</p>
Full article ">Figure 5
<p>SQOR knockdown in HMC3 cells directly activates NLRP3 inflammasome but fails to enhance the effect of GA<sub>50</sub>. We used siRNA to knockdown the expression of SQOR in non-expressing and GA50-expressing HMC3 cells for 24 h. (<b>A</b>) Cell lysates were detected by Western blot for the level of GA50, SQOR, and NLRP3 inflammasome-associated components and quantified using ImageJ software (revision 1.53). GAPDH was used as an internal loading control. (<b>B</b>) Exocrine levels of mature IL-1β and mature IL-18 in conditional media were assessed by ELISA.</p>
Full article ">Figure 6
<p>GA<sub>50</sub> expression or SQOR knockdown increased the generation of ROS and the cytoplasmic escape of mitochondrial DNA in HMC3 cells. (<b>A</b>) ROS levels in HMC3 cells with GA<sub>50</sub> expression or SQOR knockdown were measured by H2DCFDA probe and flow cytometry. (<b>B</b>) RT-qPCR was used to analyze the relative changes in the content of mitochondrial ND-1 DNA in the cytoplasm (normalized to ND-1 DNA of total lysates) to determine the degree of cytoplasmic escape of mitochondrial DNA.</p>
Full article ">Figure 7
<p>Irisflorentin (IFT) blocks the interaction of GA<sub>50</sub> and SQOR in a yeast two-hybrid model. (<b>A</b>) Schematic showing the strategy based on the yeast two-hybrid principle to screen inhibitors of the interaction between GA<sub>50</sub> and SQOR. (<b>B</b>–<b>I</b>) Diploid yeast harboring plasmids encoding BD-/AD-, BD-p53/AD-T, BD-GA<sub>50</sub>/AD-SQOR (S3), or BD-S3/AD-GA<sub>50</sub> grow to logarithmic phase in non-selective broth (SD/-Leu/-Trp) containing serially diluted IFT. (<b>B</b>–<b>E</b>) In yeast spot analysis, the amount of diploid yeast in each group was normalized and serially diluted, then spotted on non-selective and selective (SD/-Leu/-Trp/-Ade/-His) culture plates. Spots were incubated at 30°C for 3 days. (<b>F</b>–<b>I</b>) Diploid yeasts from each group were normalized (OD<sub>600</sub> = 0.2) and cultured in non-selective and selective broth for 2 days for optical density measurements at 12 h intervals.</p>
Full article ">Figure 7 Cont.
<p>Irisflorentin (IFT) blocks the interaction of GA<sub>50</sub> and SQOR in a yeast two-hybrid model. (<b>A</b>) Schematic showing the strategy based on the yeast two-hybrid principle to screen inhibitors of the interaction between GA<sub>50</sub> and SQOR. (<b>B</b>–<b>I</b>) Diploid yeast harboring plasmids encoding BD-/AD-, BD-p53/AD-T, BD-GA<sub>50</sub>/AD-SQOR (S3), or BD-S3/AD-GA<sub>50</sub> grow to logarithmic phase in non-selective broth (SD/-Leu/-Trp) containing serially diluted IFT. (<b>B</b>–<b>E</b>) In yeast spot analysis, the amount of diploid yeast in each group was normalized and serially diluted, then spotted on non-selective and selective (SD/-Leu/-Trp/-Ade/-His) culture plates. Spots were incubated at 30°C for 3 days. (<b>F</b>–<b>I</b>) Diploid yeasts from each group were normalized (OD<sub>600</sub> = 0.2) and cultured in non-selective and selective broth for 2 days for optical density measurements at 12 h intervals.</p>
Full article ">Figure 7 Cont.
<p>Irisflorentin (IFT) blocks the interaction of GA<sub>50</sub> and SQOR in a yeast two-hybrid model. (<b>A</b>) Schematic showing the strategy based on the yeast two-hybrid principle to screen inhibitors of the interaction between GA<sub>50</sub> and SQOR. (<b>B</b>–<b>I</b>) Diploid yeast harboring plasmids encoding BD-/AD-, BD-p53/AD-T, BD-GA<sub>50</sub>/AD-SQOR (S3), or BD-S3/AD-GA<sub>50</sub> grow to logarithmic phase in non-selective broth (SD/-Leu/-Trp) containing serially diluted IFT. (<b>B</b>–<b>E</b>) In yeast spot analysis, the amount of diploid yeast in each group was normalized and serially diluted, then spotted on non-selective and selective (SD/-Leu/-Trp/-Ade/-His) culture plates. Spots were incubated at 30°C for 3 days. (<b>F</b>–<b>I</b>) Diploid yeasts from each group were normalized (OD<sub>600</sub> = 0.2) and cultured in non-selective and selective broth for 2 days for optical density measurements at 12 h intervals.</p>
Full article ">Figure 8
<p>Irisflorentin (IFT) reverses NLRP3 inflammasome activity in GA<sub>50</sub>-expressing HMC3 microglia. (<b>A</b>) Cell viability was determined by MTT assay. HMC3 cells were analyzed after treatment with DMSO, 2.5, 5, 10, 20, 40, or 80 μM IFT for 24 h. (<b>B</b>) IFT inhibits the interaction of GA<sub>50</sub> and SQOR in a co-immunoprecipitation assay of 293T cells expressing GA<sub>50</sub>-Myc and SQOR-HA. (<b>C</b>) Expression of GA<sub>50</sub> (Myc) and NLRP3 inflammasome-associated components in cell lysates was quantified by Western blotting and ImageJ software (version 1.53). The internal loading control protein is GAPDH. (<b>D</b>) The exocrine levels of mature IL-1β and mature IL-18 in conditioned media were determined by ELISA. (<b>E</b>) Western blot analysis of the expression of SQOR and NLRP3 inflammasome-associated components in SQOR-knockdown HMC3 cells (<b>top</b> panel). GAPDH served as an internal loading control. Mature IL-1β activity in the conditional medium was detected by ELISA (<b>bottom</b> panel).</p>
Full article ">Figure 8 Cont.
<p>Irisflorentin (IFT) reverses NLRP3 inflammasome activity in GA<sub>50</sub>-expressing HMC3 microglia. (<b>A</b>) Cell viability was determined by MTT assay. HMC3 cells were analyzed after treatment with DMSO, 2.5, 5, 10, 20, 40, or 80 μM IFT for 24 h. (<b>B</b>) IFT inhibits the interaction of GA<sub>50</sub> and SQOR in a co-immunoprecipitation assay of 293T cells expressing GA<sub>50</sub>-Myc and SQOR-HA. (<b>C</b>) Expression of GA<sub>50</sub> (Myc) and NLRP3 inflammasome-associated components in cell lysates was quantified by Western blotting and ImageJ software (version 1.53). The internal loading control protein is GAPDH. (<b>D</b>) The exocrine levels of mature IL-1β and mature IL-18 in conditioned media were determined by ELISA. (<b>E</b>) Western blot analysis of the expression of SQOR and NLRP3 inflammasome-associated components in SQOR-knockdown HMC3 cells (<b>top</b> panel). GAPDH served as an internal loading control. Mature IL-1β activity in the conditional medium was detected by ELISA (<b>bottom</b> panel).</p>
Full article ">
Back to TopTop