Pub Date : 2022-06-01DOI: 10.1016/S2707-3688(23)00059-6
Yuanyuan XIE , Jiayi LUO , Yiming WANG , Guoan LUO , Shumei WANG
Traditional Chinese medicine is the treasure of Chinese traditional culture. A series of questions should be answered on the base of systematic, comprehensive and in-depth research according to the TCM theory and its own characteristics. For example, how to construct a research framework with clear guidance and route, as well as reasonable plan for elucidating the effective material basis of Traditional Chinese medicine (TCM) formula? How to discover and excavate the effective material basis of TCM, clarify and interpret the related action mechanism? How to clarify the reasonableness of compatibility, the scientificity for usage of medication, as well as the scientific connotation of TCM in preventing and curing disease? Aforementioned questions had been considered as the important frontier scientific problems to be solved in the field of TCM. According to the practice progress and thinking on research of effective material basis and related mechanism in Naomaitong Granule and Dengzhan Shengmai Capsule, a systematic strategy from the perspective of interaction between drug system, intestinal microecosystem and host living system had been proposed and elucidated in three aspects: (1) Discovering the TCM regulated genes and proteins dated from functional metabolites, in order to characterize the biological response of host systematically. (2) Deciphering the effective material basis and related acting mechanism on the base of interaction between TCM formula and intestinal microecosystem. (3) The systematic characterization of material basis of TCM formula by Chemomics.
{"title":"Research progress and prospect on the effective material basis and related mechanism of Traditional Chinese Medicine driven by systematic thinking","authors":"Yuanyuan XIE , Jiayi LUO , Yiming WANG , Guoan LUO , Shumei WANG","doi":"10.1016/S2707-3688(23)00059-6","DOIUrl":"10.1016/S2707-3688(23)00059-6","url":null,"abstract":"<div><p>Traditional Chinese medicine is the treasure of Chinese traditional culture. A series of questions should be answered on the base of systematic, comprehensive and in-depth research according to the TCM theory and its own characteristics. For example, how to construct a research framework with clear guidance and route, as well as reasonable plan for elucidating the effective material basis of Traditional Chinese medicine (TCM) formula? How to discover and excavate the effective material basis of TCM, clarify and interpret the related action mechanism? How to clarify the reasonableness of compatibility, the scientificity for usage of medication, as well as the scientific connotation of TCM in preventing and curing disease? Aforementioned questions had been considered as the important frontier scientific problems to be solved in the field of TCM. According to the practice progress and thinking on research of effective material basis and related mechanism in Naomaitong Granule and Dengzhan Shengmai Capsule, a systematic strategy from the perspective of interaction between drug system, intestinal microecosystem and host living system had been proposed and elucidated in three aspects: (1) Discovering the TCM regulated genes and proteins dated from functional metabolites, in order to characterize the biological response of host systematically. (2) Deciphering the effective material basis and related acting mechanism on the base of interaction between TCM formula and intestinal microecosystem. (3) The systematic characterization of material basis of TCM formula by Chemomics.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 2","pages":"Pages 206-216"},"PeriodicalIF":0.0,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000596/pdfft?md5=8a4c36e2a4e52965879e853a85ff9253&pid=1-s2.0-S2707368823000596-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87218734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01DOI: 10.1016/S2707-3688(23)00051-1
Yiqi YANG , Xiaowan WANG , Wanlin YU , Lihua HUANG , Yunshan WU , Xiaodong HAN , Fangfang XU , Wei MAO , Peng XU , Bo LIU
Objective
Triptriolide (T11) is isolated from the Chinese herb Tripterygium wilfordii Hook F (TwHF), which has been used as an immunosuppressive reagent for many years in China, while the role of this compound in TwHF is still unknown. In this study, we try to investigate the immune regulating effects of T11 on the proliferation and apoptosis of splenic lymphocyte in vivo and in vitro.
Methods
we pretreated the BALB/c mice with T11 (2.8, 14, 28 mg/kg) and prednisone acetate (PA, 1.3 mg/kg, positive control) for 7 days before lipopolysaccharides (LPS, 3 mg/kg) stimulation. Then the histopathologic change of spleen was examined by H&E method, and the apoptosis of splenic lymphocyte was assessed by flow cytometry method. In addition, we further studied the inhibition effect of T11 (50, 100, 150, 200 µg/mL) on the proliferation and activity of splenic lymphocyte, which was with or without the stimulation of concanavalin A (Con A, 5 µg/mL) and lipopolysaccharides (LPS, 5 µg/mL) in vitro, respectively.
Results
We found that T11 not only can restore the pathogenesis process in spleen, and boost the splenic lymphocyte apoptosis in vivo, but also can inhibit the splenic lymphocyte proliferation and affect its viability in vitro.
Conclusion
The results indicated that T11 might modulate the immune function of LPS-induced BALB/c mice by inhibiting the proliferation and inducing the apoptosis of splenic lymphocyte. Therefore, T11 could be a novel immunomodulator in autoimmune disease treatment in clinical.
{"title":"Regulating effects of triptriolide on the proliferation and apoptosis of splenic lymphocyte in vivo and in vitro","authors":"Yiqi YANG , Xiaowan WANG , Wanlin YU , Lihua HUANG , Yunshan WU , Xiaodong HAN , Fangfang XU , Wei MAO , Peng XU , Bo LIU","doi":"10.1016/S2707-3688(23)00051-1","DOIUrl":"10.1016/S2707-3688(23)00051-1","url":null,"abstract":"<div><h3>Objective</h3><p>Triptriolide (T11) is isolated from the Chinese herb <em>Tripterygium wilfordii Hook</em> F (TwHF), which has been used as an immunosuppressive reagent for many years in China, while the role of this compound in TwHF is still unknown. In this study, we try to investigate the immune regulating effects of T11 on the proliferation and apoptosis of splenic lymphocyte <em>in vivo</em> and <em>in vitro.</em></p></div><div><h3>Methods</h3><p>we pretreated the BALB/c mice with T11 (2.8, 14, 28 mg/kg) and prednisone acetate (PA, 1.3 mg/kg, positive control) for 7 days before lipopolysaccharides (LPS, 3 mg/kg) stimulation. Then the histopathologic change of spleen was examined by H&E method, and the apoptosis of splenic lymphocyte was assessed by flow cytometry method. In addition, we further studied the inhibition effect of T11 (50, 100, 150, 200 µg/mL) on the proliferation and activity of splenic lymphocyte, which was with or without the stimulation of concanavalin A (Con A, 5 µg/mL) and lipopolysaccharides (LPS, 5 µg/mL) <em>in vitro,</em> respectively.</p></div><div><h3>Results</h3><p>We found that T11 not only can restore the pathogenesis process in spleen, and boost the splenic lymphocyte apoptosis <em>in vivo,</em> but also can inhibit the splenic lymphocyte proliferation and affect its viability <em>in vitro.</em></p></div><div><h3>Conclusion</h3><p>The results indicated that T11 might modulate the immune function of LPS-induced BALB/c mice by inhibiting the proliferation and inducing the apoptosis of splenic lymphocyte. Therefore, T11 could be a novel immunomodulator in autoimmune disease treatment in clinical.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 2","pages":"Pages 95-105"},"PeriodicalIF":0.0,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000511/pdfft?md5=2d6e564d5c54fd9fafc6ead09d38d6ce&pid=1-s2.0-S2707368823000511-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74150704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-06-01DOI: 10.1016/S2707-3688(23)00054-7
Chunmei LI , Zhe XIE , Siqing HE , Shumiao HE , Yongqi LI , Qun LU
Objective
Ligustilide (Lig), a major component of Radix Angelica Sinensis, exhibits a protective effect on many cardiovascular diseases. Our previous study showed that Lig inhibited the proliferation of vascular smooth muscle cells. In this study, we aimed to investigate the effect of Lig on the migration of vascular smooth muscle A7r5 cells induced by Angiotensin II (Ang II) and explore the mechanism underlying based on network pharmacology.
Methods
In this study, scratch-wound healing assay and transwell migration assay were used to assess the migration activity of Lig on Ang II-induced vascular smooth muscle A7r5 cells, and network pharmacology method was used to predict the possible target molecules. Western blot analysis, Gelatin zymography assay, and small interfering RNA (siRNA) were used to determine the underlying mechanism of Lig on the migration in Ang II-induced A7r5 cells.
Results
We found that Lig could prevented the migration induced by Ang II in A7r5 cells, which could be associated with c-Myc and MMPs from the results of network pharmacology. Our results revealed that Lig could significantly reduce the increase in the protein expression levels of c-Myc and MMP-2 in Ang II-induced A7r5 cells, but not affect the protein expression levels of MMP-9. Our data further showed that c-Myc siRNA, just as Lig, could obviously inhibit the cell migration accompanied by decreased MMP-2 expression.
Conclusion
These findings suggested the anti-migratory effect of Lig could be associated with the c-Myc/MMP-2 pathway, and Lig administration had a promising potential for preventing cardiovascular diseases.
目的当归的主要成分——白芷内酯(Lig)对多种心血管疾病具有保护作用。我们之前的研究表明,Lig抑制血管平滑肌细胞的增殖。本研究旨在研究Lig对血管紧张素II (Angiotensin II, Ang II)诱导的血管平滑肌A7r5细胞迁移的影响,并基于网络药理学探讨其作用机制。方法本研究采用划伤愈合实验和跨井迁移实验评估Lig对Angⅱ诱导的血管平滑肌A7r5细胞的迁移活性,并采用网络药理学方法预测可能的靶分子。采用Western blot分析、明胶酶谱分析和小干扰RNA (siRNA)技术,探讨了Lig对Angⅱ诱导的A7r5细胞迁移的作用机制。结果网络药理学结果显示,Lig可抑制Angⅱ诱导的A7r5细胞的迁移,这可能与c-Myc和MMPs有关。我们的研究结果显示,Lig可以显著降低Ang ii诱导的A7r5细胞中c-Myc和MMP-2蛋白表达水平的升高,但不影响MMP-9蛋白表达水平。我们的数据进一步表明,c-Myc siRNA与Lig一样,可以明显抑制细胞迁移,同时降低MMP-2的表达。结论Lig的抗迁移作用可能与c-Myc/MMP-2通路有关,Lig在预防心血管疾病方面具有广阔的应用前景。
{"title":"Ligustilide from Radix Angelica Sinensis prevents the migration of vascular smooth muscle A7r5 cells based on network pharmacology and experimental verification","authors":"Chunmei LI , Zhe XIE , Siqing HE , Shumiao HE , Yongqi LI , Qun LU","doi":"10.1016/S2707-3688(23)00054-7","DOIUrl":"10.1016/S2707-3688(23)00054-7","url":null,"abstract":"<div><h3>Objective</h3><p>Ligustilide (Lig), a major component of Radix Angelica Sinensis, exhibits a protective effect on many cardiovascular diseases. Our previous study showed that Lig inhibited the proliferation of vascular smooth muscle cells. In this study, we aimed to investigate the effect of Lig on the migration of vascular smooth muscle A7r5 cells induced by Angiotensin II (Ang II) and explore the mechanism underlying based on network pharmacology.</p></div><div><h3>Methods</h3><p>In this study, scratch-wound healing assay and transwell migration assay were used to assess the migration activity of Lig on Ang II-induced vascular smooth muscle A7r5 cells, and network pharmacology method was used to predict the possible target molecules. Western blot analysis, Gelatin zymography assay, and small interfering RNA (siRNA) were used to determine the underlying mechanism of Lig on the migration in Ang II-induced A7r5 cells.</p></div><div><h3>Results</h3><p>We found that Lig could prevented the migration induced by Ang II in A7r5 cells, which could be associated with <em>c</em>-Myc and MMPs from the results of network pharmacology. Our results revealed that Lig could significantly reduce the increase in the protein expression levels of <em>c</em>-Myc and MMP-2 in Ang II-induced A7r5 cells, but not affect the protein expression levels of MMP-9. Our data further showed that <em>c</em>-Myc siRNA, just as Lig, could obviously inhibit the cell migration accompanied by decreased MMP-2 expression.</p></div><div><h3>Conclusion</h3><p>These findings suggested the anti-migratory effect of Lig could be associated with the <em>c</em>-Myc/MMP-2 pathway, and Lig administration had a promising potential for preventing cardiovascular diseases.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 2","pages":"Pages 139-152"},"PeriodicalIF":0.0,"publicationDate":"2022-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000547/pdfft?md5=1af797c959c4447c7675517e1d411b82&pid=1-s2.0-S2707368823000547-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72626870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00065-1
Kechun CHEN , Yating ZHAO , Lexun WANG , Yifan YIN , Ling YANG , Duosheng LUO
Objective
Aging is typically characterized by imbalanced gut microbiota and bile acid (BA) dyshomeostasis. However, features of the gut microbiota-regulated BA metabolism in the aging process remain unclear.
Aim
To establish a direct link between gut microbiota and BA profile changes in the liver, plasma, and intestinal contents and to further understand aging and aging-related diseases from the liver-gut axis.
Methods
The BA content in the liver, plasma, and intestine were determined using ultra-performance liquid chromatography-tandem mass spectrometry. Gut microbiota were sequenced by 16S rDNA, and the expression of the liver-gut axis-related proteins was detected.
Results
The plasma content of total cholesterol, triglycerides, and low-density lipoprotein cholesterol gradually increased during aging, and Lactobacillus, Bacteroides, Bacillus, Ruminococcus, Blautia, and Streptococcus, which can produce bile salt hydrolase, were increased. The concentration of total BAs was increased, especially that of unconjugated BAs, and the ratio of unconjugated BAs to conjugated BAs was also increased. The expression of bile acid transporter receptors and intestinal tight junction proteins was significantly decreased.
Conclusion
The presented data show that the changes in BA profile mediated by gut microbiota were closely related to aging and highlights that more attention should be paid to targeting gut microbiota-regulated BA metabolism to prevent and treat aging-related diseases, especially lipid metabolism disorders.
{"title":"Gut microbiota–regulated bile acids metabolism features in the aging process in mice","authors":"Kechun CHEN , Yating ZHAO , Lexun WANG , Yifan YIN , Ling YANG , Duosheng LUO","doi":"10.1016/S2707-3688(23)00065-1","DOIUrl":"10.1016/S2707-3688(23)00065-1","url":null,"abstract":"<div><h3>Objective</h3><p>Aging is typically characterized by imbalanced gut microbiota and bile acid (BA) dyshomeostasis. However, features of the gut microbiota-regulated BA metabolism in the aging process remain unclear.</p></div><div><h3>Aim</h3><p>To establish a direct link between gut microbiota and BA profile changes in the liver, plasma, and intestinal contents and to further understand aging and aging-related diseases from the liver-gut axis.</p></div><div><h3>Methods</h3><p>The BA content in the liver, plasma, and intestine were determined using ultra-performance liquid chromatography-tandem mass spectrometry. Gut microbiota were sequenced by 16S rDNA, and the expression of the liver-gut axis-related proteins was detected.</p></div><div><h3>Results</h3><p>The plasma content of total cholesterol, triglycerides, and low-density lipoprotein cholesterol gradually increased during aging, and <em>Lactobacillus, Bacteroides, Bacillus, Ruminococcus, Blautia,</em> and <em>Streptococcus,</em> which can produce bile salt hydrolase, were increased. The concentration of total BAs was increased, especially that of unconjugated BAs, and the ratio of unconjugated BAs to conjugated BAs was also increased. The expression of bile acid transporter receptors and intestinal tight junction proteins was significantly decreased.</p></div><div><h3>Conclusion</h3><p>The presented data show that the changes in BA profile mediated by gut microbiota were closely related to aging and highlights that more attention should be paid to targeting gut microbiota-regulated BA metabolism to prevent and treat aging-related diseases, especially lipid metabolism disorders.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 45-56"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000651/pdfft?md5=b95bf3970ded13867e15a81cecb3bb0c&pid=1-s2.0-S2707368823000651-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76158351","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qi-yin deficiency (QYD) and Yang-deficiency (YD) syndrome are two common syndromes with contrasting clinical manifestations of low fever and cold phobia. Different syndrome phenotypes must have distinct biological underpinnings in vivo. Based on serum metabolomics, this study aims to reveal the biological characteristics of the two diabetic coronary heart disease syndromes mentioned above.
Methods
A total of 33 diabetic coronary heart disease (DCHD) patients were included in the study, with 13 cases of QYD, 11 cases of YD syndrome, and 9 cases of Pinghe constitution (PH). Based on UPLC-Q/TOF MS, metabolomics was used to detect and analyze serum samples from the three groups mentioned above. To find differentiating metabolites, PLS-DA (Partial least squares discrimination analysis) and database searching were used. Meanwhile, the metabolic pathways of various metabolites were investigated.
Results
There were 17 different metabolites between QYD syndrome and YD syndrome of DCHD. Cyclic GMP, LysoPE (20:3), proline, isoleucine, N-lactoyl-tyrosine, 3-oxohexadecanoyl-CoA, and oxalosuccinic acid were lower in the (YD) group than those in the PH group, while galactitol, N-lactoyl-tyrosine, and oxalosuccinic acid were higher in the QYD group than in the PH group. The 17 differential metabolites mentioned above were linked to a variety of pathways, including amino acid metabolism, purine metabolism, fatty acid elongation in mitochondria, the tricarboxylic acid cycle, lipid metabolism, energy metabolism, etc.
Conclusion
Different DCHD syndromes, such as liver-kidney Yin deficiency and YD syndrome, have different biological bases for the same disease. To some extent, the findings of this study provide a scientific foundation for different approaches to treating the same disease in Traditional Chinese Medicine. It is also important for guiding the clinical use of herbal medicine.
{"title":"Serum metabolic profile analysis of syndrome of Qi-yin deficiency and Yang deficiency in diabetic coronary heart disease by UPLC-Q/TOF MS","authors":"Shenghua PIAO , Lei ZHANG , Ziqin ZHU , Huixia ZHAN , Xianglu RONG, Jiao GUO","doi":"10.1016/S2707-3688(23)00068-7","DOIUrl":"10.1016/S2707-3688(23)00068-7","url":null,"abstract":"<div><h3>Aims</h3><p>Qi-yin deficiency (QYD) and Yang-deficiency (YD) syndrome are two common syndromes with contrasting clinical manifestations of low fever and cold phobia. Different syndrome phenotypes must have distinct biological underpinnings <em>in vivo.</em> Based on serum metabolomics, this study aims to reveal the biological characteristics of the two diabetic coronary heart disease syndromes mentioned above.</p></div><div><h3>Methods</h3><p>A total of 33 diabetic coronary heart disease (DCHD) patients were included in the study, with 13 cases of QYD, 11 cases of YD syndrome, and 9 cases of Pinghe constitution (PH). Based on UPLC-Q/TOF MS, metabolomics was used to detect and analyze serum samples from the three groups mentioned above. To find differentiating metabolites, PLS-DA (Partial least squares discrimination analysis) and database searching were used. Meanwhile, the metabolic pathways of various metabolites were investigated.</p></div><div><h3>Results</h3><p>There were 17 different metabolites between QYD syndrome and YD syndrome of DCHD. Cyclic GMP, LysoPE (20:3), proline, isoleucine, <em>N</em>-lactoyl-tyrosine, 3-oxohexadecanoyl-CoA, and oxalosuccinic acid were lower in the (YD) group than those in the PH group, while galactitol, <em>N</em>-lactoyl-tyrosine, and oxalosuccinic acid were higher in the QYD group than in the PH group. The 17 differential metabolites mentioned above were linked to a variety of pathways, including amino acid metabolism, purine metabolism, fatty acid elongation in mitochondria, the tricarboxylic acid cycle, lipid metabolism, energy metabolism, etc.</p></div><div><h3>Conclusion</h3><p>Different DCHD syndromes, such as liver-kidney Yin deficiency and YD syndrome, have different biological bases for the same disease. To some extent, the findings of this study provide a scientific foundation for different approaches to treating the same disease in Traditional Chinese Medicine. It is also important for guiding the clinical use of herbal medicine.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 82-94"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000687/pdfft?md5=54c0c70739bfc42166bef526637b9512&pid=1-s2.0-S2707368823000687-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77217401","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00062-6
Zizhuo TU, Yaozu XIANG
Cardiovascular disease (CVD) is the leading cause of death worldwide. It has been recognized that specific regulatory factors from peripheral tissues can influence the pathological progression of atherosclerosis, myocardial infarction (MI) and heart failure (HF). Drugs targeting the regulation of pathways related to hepatic lipid metabolism, intestinal flora, renal sodium excretion and hypoglycemia, bone marrow clonal hematopoiesis and immune inflammation, such as Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, Sodium-glucose Cotransporter-2 (SGLT2) inhibitors, IL-1β monoclonal antibodies and Chimeric antigen receptor (CAR) -T therapy, have been shown to have significant protective effects in the prevention and treatment of cardiovascular disease, especially ischemic heart disease and HF. Various forms of interaction between cardiovascular system diseases and cerebrovascular, liver and intestinal metabolism, kidney and other functional disorders have been widely reported. In this review, we focus on the interactions between the cardiovascular system and other organs in ischemic heart disease and HF, highlight novel therapeutic strategies that modulate cardiometabolic and inflammatory processes.
{"title":"Heart crosstalk with other organs and pharmacological strategies for cardioprotection","authors":"Zizhuo TU, Yaozu XIANG","doi":"10.1016/S2707-3688(23)00062-6","DOIUrl":"10.1016/S2707-3688(23)00062-6","url":null,"abstract":"<div><p>Cardiovascular disease (CVD) is the leading cause of death worldwide. It has been recognized that specific regulatory factors from peripheral tissues can influence the pathological progression of atherosclerosis, myocardial infarction (MI) and heart failure (HF). Drugs targeting the regulation of pathways related to hepatic lipid metabolism, intestinal flora, renal sodium excretion and hypoglycemia, bone marrow clonal hematopoiesis and immune inflammation, such as Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, Sodium-glucose Cotransporter-2 (SGLT2) inhibitors, IL-1<em>β</em> monoclonal antibodies and Chimeric antigen receptor (CAR) -T therapy, have been shown to have significant protective effects in the prevention and treatment of cardiovascular disease, especially ischemic heart disease and HF. Various forms of interaction between cardiovascular system diseases and cerebrovascular, liver and intestinal metabolism, kidney and other functional disorders have been widely reported. In this review, we focus on the interactions between the cardiovascular system and other organs in ischemic heart disease and HF, highlight novel therapeutic strategies that modulate cardiometabolic and inflammatory processes.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 18-23"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000626/pdfft?md5=d3dd72dfc7e6736489f3c55a9ad319d8&pid=1-s2.0-S2707368823000626-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83556094","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00066-3
Zhican YANG , Xinyuan ZHAO , Ying LIU , Benchi ZHAO , Yi LUO , Jiansheng KANG , Qiaoping WANG
Objective
This study aimed to investigate the anti-obesity effects of the combination of metformin and epigallocatechin-3-gallate (EGCG) in a diet-induced obese (DIO) model.
Methods
Male C57BL/6J mice were fed with high-fat diet (HFD, 60% fat) to establish a DIO model. The DIO mice were administered intragastrically daily with vehicle, metformin (Met, 250 mg/kg), EGCG (200 mg/kg), or mixture of Met and EGCG (Met/EGCG; 250 and 200 mg/kg) for 32 days. Body weight was measured daily at a fixed time point. An intraperitoneal glucose tolerance test was performed on day 26 after treatments. Body composition was determined by using nuclear magnetic resonance technology. Adipose tissues and major organs were collected after culling. Gene expression in brown adipose tissue (BAT) and subcutaneous white adipose tissue (scWAT) were detected by quantitative polymerase chain reaction.
Results
Compared with the vehicle group, the body weight was significantly reduced in DIO mice in the Met and Met/EGCG groups. The weight loss rate in the Met and EGCG groups were 9.1% and 1.0%, respectively. However, Met/EGCG induced a 14.0% body weight loss, suggesting that the combination of Met and EGCG has a synergistic effect on reversing the body weight in DIO mice. Met/EGCG demonstrated beneficial effects on the subcutaneous fat, BAT, and glucose homeostasis in DIO mice. Furthermore, Met/EGCG did not significantly increase the thermogenic gene expression in BAT, whereas in scWAT, Met/EGCG showed moderate effects on the expression of specific markers of browning fat or beige fat.
Conclusion
Metformin and EGCG have combined treatment effect on the body weight of DIO mice. Their combination might be a promising treatment approach for obesity.
{"title":"Treating obesity using the combination of metformin and epigallocatechin-3-gallate","authors":"Zhican YANG , Xinyuan ZHAO , Ying LIU , Benchi ZHAO , Yi LUO , Jiansheng KANG , Qiaoping WANG","doi":"10.1016/S2707-3688(23)00066-3","DOIUrl":"10.1016/S2707-3688(23)00066-3","url":null,"abstract":"<div><h3>Objective</h3><p>This study aimed to investigate the anti-obesity effects of the combination of metformin and epigallocatechin-3-gallate (EGCG) in a diet-induced obese (DIO) model.</p></div><div><h3>Methods</h3><p>Male C57BL/6J mice were fed with high-fat diet (HFD, 60% fat) to establish a DIO model. The DIO mice were administered intragastrically daily with vehicle, metformin (Met, 250 mg/kg), EGCG (200 mg/kg), or mixture of Met and EGCG (Met/EGCG; 250 and 200 mg/kg) for 32 days. Body weight was measured daily at a fixed time point. An intraperitoneal glucose tolerance test was performed on day 26 after treatments. Body composition was determined by using nuclear magnetic resonance technology. Adipose tissues and major organs were collected after culling. Gene expression in brown adipose tissue (BAT) and subcutaneous white adipose tissue (scWAT) were detected by quantitative polymerase chain reaction.</p></div><div><h3>Results</h3><p>Compared with the vehicle group, the body weight was significantly reduced in DIO mice in the Met and Met/EGCG groups. The weight loss rate in the Met and EGCG groups were 9.1% and 1.0%, respectively. However, Met/EGCG induced a 14.0% body weight loss, suggesting that the combination of Met and EGCG has a synergistic effect on reversing the body weight in DIO mice. Met/EGCG demonstrated beneficial effects on the subcutaneous fat, BAT, and glucose homeostasis in DIO mice. Furthermore, Met/EGCG did not significantly increase the thermogenic gene expression in BAT, whereas in scWAT, Met/EGCG showed moderate effects on the expression of specific markers of browning fat or beige fat.</p></div><div><h3>Conclusion</h3><p>Metformin and EGCG have combined treatment effect on the body weight of DIO mice. Their combination might be a promising treatment approach for obesity.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 57-67"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000663/pdfft?md5=b0ec30d75798a3b146cfceb07cb557f5&pid=1-s2.0-S2707368823000663-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79098597","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00067-5
Yubin YANG , Yunlong WANG , Zhengbang SUN , Ting DU , Ying YANG , Jiaojiao CHEN , Meiling WANG , Hairong LI , Jian QIN
Objective
To observe the effects of Lingguizhugan decoction on obesity and investigate its preliminary mechanism in activating fat mobilization based on gut microbiota.
Methods
In this prospective one-armed clinical research, we analyzed the data of 32 obese patients with Yang deficiency and phlegm-dampness syndrome treated by Lingguizhugan decoction combined with lifestyle intervention for 2 months. The anthropometric data, body composition, metabolic data, traditional Chinese medicine (TCM) syndrome scores, fecal 16S rDNA, core temperature, and visceral fat fractions of patients were analyzed. Animal experiments were conducted for further verification: ob/ob mice and pseudo-germ-free ob/ob mice (ABX) were given Lingguizhugan decoction by gavage for 2 months. The body mass, core temperature (rectum), respiratory exchange ratio (RER), micro-positron emission tomography/computed tomography (micro-PET/CT) results, and uncoupling protein 1 (UCP1) expression (including gene, protein, and immunohistochemistry staining results) in inguinal adipose tissue of mice were evaluated.
Results
The clinical effective rates of TCM and Western medicine were 96.87% and 93.75%, respectively. Compared with those before treatment, the body mass index (BMI), body fat ratio (BFR), waist-to-hip ratio, TCM syndrome scores, low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), triglyceride (TG), fasting plasma glucose (FPG), 2 h postprandial plasma glucose (2hPPG), glycosylated hemoglobin A1C (GHbA1c), and blood uric acid (UA) significantly decreased after treatment. Moreover, we randomly selected five patients to investigate their core temperature and visceral fat fractions. Compared with healthy volunteers, the five patients had higher core temperature. According to the 1.5 T magnetic resonance imaging (MRI) results, the fat fractions of the liver, pancreas, greater omentum, and subcutaneous abdominal wall were notably decreased after treatment. Fecal 16S rDNA principal component analysis and partial least squares discriminant analysis showed that the biodiversity and species abundance of gut microbiota were significantly different between pre- and post-treatment. The relative abundance of Bacteroidetes was remarkably elevated and that of Firmicutes was notably reduced by Lingguizhugan decoction. The animal experiments showed that the weight of ob/ob mice decreased significantly by gavage of Lingguizhugan decoction for 2 months. Compared with the model group, the Lingguizhugan group showed increased core temperature (rectum) and decreased RER. Micro-PET/CT showed that 18F-2-deoxyglucose-6-phosphate (18F-FDG) was enriched obviously and the SUV index was increased in the Lingguizhugan group. However, after eliminating the gut microbiota in the ABX group, the weight loss, core temperature, 18F-FDG enrichment, and SUV index decreased, and RER was similar to that
{"title":"Effect of Lingguizhugan decoction in activating fat mobilization in obesity","authors":"Yubin YANG , Yunlong WANG , Zhengbang SUN , Ting DU , Ying YANG , Jiaojiao CHEN , Meiling WANG , Hairong LI , Jian QIN","doi":"10.1016/S2707-3688(23)00067-5","DOIUrl":"10.1016/S2707-3688(23)00067-5","url":null,"abstract":"<div><h3>Objective</h3><p>To observe the effects of Lingguizhugan decoction on obesity and investigate its preliminary mechanism in activating fat mobilization based on gut microbiota.</p></div><div><h3>Methods</h3><p>In this prospective one-armed clinical research, we analyzed the data of 32 obese patients with Yang deficiency and phlegm-dampness syndrome treated by Lingguizhugan decoction combined with lifestyle intervention for 2 months. The anthropometric data, body composition, metabolic data, traditional Chinese medicine (TCM) syndrome scores, fecal 16S rDNA, core temperature, and visceral fat fractions of patients were analyzed. Animal experiments were conducted for further verification: ob/ob mice and pseudo-germ-free ob/ob mice (ABX) were given Lingguizhugan decoction by gavage for 2 months. The body mass, core temperature (rectum), respiratory exchange ratio (RER), micro-positron emission tomography/computed tomography (micro-PET/CT) results, and uncoupling protein 1 (UCP1) expression (including gene, protein, and immunohistochemistry staining results) in inguinal adipose tissue of mice were evaluated.</p></div><div><h3>Results</h3><p>The clinical effective rates of TCM and Western medicine were 96.87% and 93.75%, respectively. Compared with those before treatment, the body mass index (BMI), body fat ratio (BFR), waist-to-hip ratio, TCM syndrome scores, low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), triglyceride (TG), fasting plasma glucose (FPG), 2 h postprandial plasma glucose (2hPPG), glycosylated hemoglobin A1C (GHbA1c), and blood uric acid (UA) significantly decreased after treatment. Moreover, we randomly selected five patients to investigate their core temperature and visceral fat fractions. Compared with healthy volunteers, the five patients had higher core temperature. According to the 1.5 T magnetic resonance imaging (MRI) results, the fat fractions of the liver, pancreas, greater omentum, and subcutaneous abdominal wall were notably decreased after treatment. Fecal 16S rDNA principal component analysis and partial least squares discriminant analysis showed that the biodiversity and species abundance of gut microbiota were significantly different between pre- and post-treatment. The relative abundance of Bacteroidetes was remarkably elevated and that of Firmicutes was notably reduced by Lingguizhugan decoction. The animal experiments showed that the weight of ob/ob mice decreased significantly by gavage of Lingguizhugan decoction for 2 months. Compared with the model group, the Lingguizhugan group showed increased core temperature (rectum) and decreased RER. Micro-PET/CT showed that <sup>18</sup>F-2-deoxyglucose-6-phosphate (<sup>18</sup>F-FDG) was enriched obviously and the SUV index was increased in the Lingguizhugan group. However, after eliminating the gut microbiota in the ABX group, the weight loss, core temperature, <sup>18</sup>F-FDG enrichment, and SUV index decreased, and RER was similar to that","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 68-81"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000675/pdfft?md5=628b4ef2b6b53825db0d1efb6ddbd850&pid=1-s2.0-S2707368823000675-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87330475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00061-4
Dianhui ZHANG , Zhenni GUO , Yi YANG , Junlei CHANG
The blood-brain barrier (BBB) prevents unregulated substance exchange between the central nervous system and the blood, while providing highly regulated transport of nutrients and tonic factors essential to brain metabolism. A group of carriers, transporters, and receptors is utilized by endothelial cells of the BBB to aid the influx and efflux of nutrients and metabolic wastes, and their function is subject to changes during metabolic disorders such as diabetes mellitus and obesity. This regulated barrier function of BBB is essential for maintaining the normal metabolism of the brain and transduction of metabolic signals from the periphery. As such, disruption of the BBB nutrient/hormone transport system has been proposed to be major contributors of many neurological diseases.
{"title":"Regulation of brain metabolism by peripheral nutritional signals: the role of blood-brain barrier in health and disease","authors":"Dianhui ZHANG , Zhenni GUO , Yi YANG , Junlei CHANG","doi":"10.1016/S2707-3688(23)00061-4","DOIUrl":"10.1016/S2707-3688(23)00061-4","url":null,"abstract":"<div><p>The blood-brain barrier (BBB) prevents unregulated substance exchange between the central nervous system and the blood, while providing highly regulated transport of nutrients and tonic factors essential to brain metabolism. A group of carriers, transporters, and receptors is utilized by endothelial cells of the BBB to aid the influx and efflux of nutrients and metabolic wastes, and their function is subject to changes during metabolic disorders such as diabetes mellitus and obesity. This regulated barrier function of BBB is essential for maintaining the normal metabolism of the brain and transduction of metabolic signals from the periphery. As such, disruption of the BBB nutrient/hormone transport system has been proposed to be major contributors of many neurological diseases.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 7-17"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2707368823000614/pdfft?md5=41eed719dbf7eeb1bef7a57a8f1ac7e9&pid=1-s2.0-S2707368823000614-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79637100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-03-01DOI: 10.1016/S2707-3688(23)00064-X
Yiqi YANG , Haibo TAN , Xiaoyu ZHANG, Minyi HUANG, Weijian BEI, Jiao GUO
Objective
To explore the renal protection effect and mechanism of Fufang Zhenzhu Tiaozhi capsule (FTZ) in diabetic db/db mice in vivo and in vitro.
Methods
Male db/m mice were considered as control group, and male db/db mice were considered as diabetic kidney disease (DKD) group, which were administered with different doses of FTZ (FTZ-L, 1 g•kg-1•d-1; FTZ-H, 2 g•kg-1•d-1) and losartan (positive control, 30 mg•kg-1•d-1) treatment for 12 weeks respectively, and then all mice were sacrificed. The high glucose-induced Jurkat T cell was used to evaluate the immune regulatory effect of FTZ in vitro. The renal pathological change was evaluated by H&E method. The levels of serum creatinine (Scr), proteinuria, and urinary albumin-to-creatinine ratio (UACR) were detected by biochemical parameter test kits. The expression levels of fibronectin and collagen IV were detected by immunofluorescence (IF) and immunohistochemistry (IHC). The inflammation factors including TNF-α, TGF-β, IL-6, IL-8, and IL-10 were detected by ELISA kits. The protein expression levels of IL-17A, RORγt, Foxp3, p-NF-κB, p-IκB, NF-κB, and IκB were detected by western blot. The percentage of T helper 17 (Th17) cells and regulatory T (Treg) cells were measured by flow cytometry.
Results
FTZ could significantly alleviate the renal pathological changes and decrease the levels of renal function parameters including Scr, proteinuria, UACR levels, and attenuate the renal inflammation injury by reducing the inflammatory cells infiltration and cytokines (TNF-α, TGF-β, IL-6, and IL-8) secretion, as well as inhibit NF-κB signaling pathway activation and restore the Th17/Treg balance in diabetic db/db mice. Furthermore, FTZ treatment could also decrease the protein levels of RORγt and IL-17A in high glucose-induced Jurkat T cells.
Conclusion
FTZ exerted protective effects on the kidney in diabetic db/db mice, whose mechanism might be related to regulating the Th17/Treg balance and inhibiting NF-κB signaling pathway.
{"title":"Fufang Zhenzhu Tiaozhi capsule attenuates renal damage by restoring Th17/Treg balance in diabetic db/db mice","authors":"Yiqi YANG , Haibo TAN , Xiaoyu ZHANG, Minyi HUANG, Weijian BEI, Jiao GUO","doi":"10.1016/S2707-3688(23)00064-X","DOIUrl":"10.1016/S2707-3688(23)00064-X","url":null,"abstract":"<div><h3>Objective</h3><p>To explore the renal protection effect and mechanism of Fufang Zhenzhu Tiaozhi capsule (FTZ) in diabetic <em>db/db</em> mice <em>in vivo</em> and <em>in vitro.</em></p></div><div><h3>Methods</h3><p>Male <em>db/m</em> mice were considered as control group, and male <em>db/db</em> mice were considered as diabetic kidney disease (DKD) group, which were administered with different doses of FTZ (FTZ-L, 1 g•kg<sup>-1</sup>•d<sup>-1</sup>; FTZ-H, 2 g•kg<sup>-1</sup>•d<sup>-1</sup>) and losartan (positive control, 30 mg•kg<sup>-1</sup>•d<sup>-1</sup>) treatment for 12 weeks respectively, and then all mice were sacrificed. The high glucose-induced Jurkat T cell was used to evaluate the immune regulatory effect of FTZ <em>in vitro.</em> The renal pathological change was evaluated by H&E method. The levels of serum creatinine (Scr), proteinuria, and urinary albumin-to-creatinine ratio (UACR) were detected by biochemical parameter test kits. The expression levels of fibronectin and collagen IV were detected by immunofluorescence (IF) and immunohistochemistry (IHC). The inflammation factors including TNF-<em>α</em>, TGF-<em>β</em>, IL-6, IL-8, and IL-10 were detected by ELISA kits. The protein expression levels of IL-17A, ROR<em>γ</em>t, Foxp3, <em>p</em>-NF-<em>κ</em>B, <em>p-</em>I<em>κ</em>B, NF-<em>κ</em>B, and I<em>κ</em>B were detected by western blot. The percentage of T helper 17 (Th17) cells and regulatory T (Treg) cells were measured by flow cytometry.</p></div><div><h3>Results</h3><p>FTZ could significantly alleviate the renal pathological changes and decrease the levels of renal function parameters including Scr, proteinuria, UACR levels, and attenuate the renal inflammation injury by reducing the inflammatory cells infiltration and cytokines (TNF-<em>α</em>, TGF-<em>β</em>, IL-6, and IL-8) secretion, as well as inhibit NF-<em>κ</em>B signaling pathway activation and restore the Th17/Treg balance in diabetic <em>db/db</em> mice. Furthermore, FTZ treatment could also decrease the protein levels of ROR<em>γ</em>t and IL-17A in high glucose-induced Jurkat T cells.</p></div><div><h3>Conclusion</h3><p>FTZ exerted protective effects on the kidney in diabetic <em>db/db</em> mice, whose mechanism might be related to regulating the Th17/Treg balance and inhibiting NF-<em>κ</em>B signaling pathway.</p></div>","PeriodicalId":100787,"journal":{"name":"Journal of Holistic Integrative Pharmacy","volume":"3 1","pages":"Pages 32-44"},"PeriodicalIF":0.0,"publicationDate":"2022-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S270736882300064X/pdfft?md5=bc974d4ab5f3ca5e234af086092cd401&pid=1-s2.0-S270736882300064X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90349953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}