1
|
Zhang B, Wang J, Li M, Wen J, Loor JJ, Wang S, Ji Z, Lv X, Wang G, Xia C, Yang W, Xu C. Calcium Release-Activated Calcium Modulator ORAI1-Sensitive Serine Dehydratase Regulates Fatty Acid-Induced CD4 + Th17/Treg Imbalance in Dairy Cows. Animals (Basel) 2025; 15:388. [PMID: 39943158 PMCID: PMC11815743 DOI: 10.3390/ani15030388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
High concentrations of free fatty acids (FFAs) caused by negative energy balance render the cow more prone to inflammatory diseases in part due to an imbalance in the types of immune cells and their specific functions. We previously demonstrated that ORAI calcium release-activated calcium modulator 1 (ORAI1) was associated with increased CD4+ Th17 content, but the precise mechanisms remain unclear. The purpose of this study was to evaluate the efficacy of FFAs on CD4+ T cell inflammatory response. High FFAs in dairy cows caused the transcript level of the pro-inflammatory factor IL-17A, plasma concentration of IL-17A, and amount of intracellular IL-17A to increase while the transcript levels and intracellular amount of the anti-inflammatory factor FOXP3 were downregulated. These changes indicated Th17/Treg imbalance and inflammation in dairy cows with high FFA. Moreover, ORAI1 and SDS abundance was elevated in dairy cows with high FFAs by transcriptomics, QPCR, and Western blot. Knockdown of SDS (siSDS) did not alter ORAI1 expression in CD4+ T cells from high-FFA cows, while it decreased the expression of inflammatory factors. Transfection of CD4+ T cells using siRNA knockdown for ORAI1 (siORAI1) revealed that SDS and inflammatory factor abundance decreased. Serine can be catabolized to pyruvate by the action of serine dehydratase (SDS). Data from this study suggested that high FFAs caused by negative energy balance after calving regulates the Th17/Treg balance via SDS, but SDS does not regulate ORAI1 abundance. The above data suggested a pro-inflammatory mechanism in CD4+ T cells regulated by the ORAI1-sensitive SDS pathway in early postpartum cows experiencing high-FFA conditions. Thus, targeting this pathway may represent a new therapeutic and interventional approach for preventing immune-related disorders around parturition.
Collapse
Affiliation(s)
- Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (B.Z.); (J.W.); (J.W.); (Z.J.); (G.W.)
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (B.Z.); (J.W.); (J.W.); (Z.J.); (G.W.)
- College of Animal Science, Ningxia University, Yinchuan 750021, China;
| | - Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (M.L.); (X.L.); (C.X.); (W.Y.)
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (B.Z.); (J.W.); (J.W.); (Z.J.); (G.W.)
| | - Juan J. Loor
- Mammalian Nutri Physio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA;
| | - Shuang Wang
- College of Animal Science, Ningxia University, Yinchuan 750021, China;
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (M.L.); (X.L.); (C.X.); (W.Y.)
| | - Ziwei Ji
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (B.Z.); (J.W.); (J.W.); (Z.J.); (G.W.)
| | - Xinquan Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (M.L.); (X.L.); (C.X.); (W.Y.)
| | - Guihua Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (B.Z.); (J.W.); (J.W.); (Z.J.); (G.W.)
| | - Cheng Xia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (M.L.); (X.L.); (C.X.); (W.Y.)
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (M.L.); (X.L.); (C.X.); (W.Y.)
| | - Chuang Xu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Zhu T, Du J, Song H, Lei C, Cen Y, Wang C, Li S. Whole genome resequencing reveals the correlation between selection signatures and adaptability of Micropterus salmoides to artificial fed. Sci Rep 2024; 14:30058. [PMID: 39627258 PMCID: PMC11614881 DOI: 10.1038/s41598-024-80904-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
Largemouth bass (Micropterus salmoides, LMB) is an important aquaculture species due to its excellent flesh quality and environmental adaptability. It has been continuously introduced to many countries and cultured for decades. Here, an LMB population was used for selective breeding to improve growth rate and feed adaptability. After five generations of breeding, the growth rate improved by 38%, and feed adaptability improved by 22% compared to the non-breeding population. To study the underlying genetic mechanism, 100 LMB from the breeding population and 100 from the non-breeding population were sampled for whole-genome resequencing. The population genetics analysis shows that the breeding population has a higher inbreeding coefficient and linkage disequilibrium (LD) level, a lower nucleic acid diversity and effective population size (Ne). Using [Formula: see text] (fixation index), we found that the average [Formula: see text] value between the two populations was 0.07, with the highest [Formula: see text] value reaching 0.38, which overlaps with the trypsin gene. Additionally, other genes exhibiting high [Formula: see text] values are associated with functions such as neural development, glucose metabolism, and growth. Using [Formula: see text] and nucleic acid diversity as criteria, we identified 698 genes that are positively selected in the breeding population, and gene functional enrichment analysis shows that 36 genes are related to the olfactory receptor pathway. Overall, our study found that multiple genes were selected in the LMB breeding population. These genes may be associated with adaptation and digestion of artificial feed in fish.
Collapse
Affiliation(s)
- Tao Zhu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Jinxing Du
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Hongmei Song
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Caixia Lei
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yingshen Cen
- Foshan Jiyurunda Fishery Technology Co. Ltd., Foshan, 528247, China
| | - Chenghui Wang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Shengjie Li
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China.
| |
Collapse
|
3
|
Zhao Q, Miao C, Chen YT, Zhu LY, Zhang YT, Luo SQ, Wang YL, Zhu ZM, Han X, Wen Y, Wu R, Du S, Yan QG, Huang X, Zhao S, Lang YF, Wang Y, Zheng Y, Zhao F, Cao SJ. Host Factor Rab4b Promotes Japanese Encephalitis Virus Replication. Microorganisms 2024; 12:1804. [PMID: 39338478 PMCID: PMC11433971 DOI: 10.3390/microorganisms12091804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Although the Japanese encephalitis virus (JEV) infects various cell types, its receptor molecules are still not clearly understood. In our laboratory's prior research, Rab4b was identified as a potential host factor that facilitates JEV infection in PK15 cells, utilizing a genome-wide CRISPR/Cas9 knockout library (PK-15-GeCKO). To further explore the effect of Rab4b on JEV replication, we used the Rab4b knockout PK15 cell line using the CRISPR/Cas9 technology and overexpressing the Rab4b PK15 cell line, with IFA, RT-qPCR, and Western blot to study the effect of Rab4b on viral replication in the whole life cycle of the JEV. The results show that the knockout of Rab4b inhibited the replication of the JEV in PK15 cells, and the overexpression of Rab4b promoted the replication of the JEV in PK15 cell lines. Furthermore, we demonstrated for the first time that host factor Rab4b facilitates the adsorption, internalization, assembly, and release of the JEV, thereby promoting JEV replication. This study enriches the regulatory network between the JEV and host factors and lays the experimental foundation for further understanding of the function of the Rab4b protein.
Collapse
Affiliation(s)
- Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi-Ting Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Long-Yue Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ya-Ting Zhang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sai-Qi Luo
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu-Luo Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhu-Ming Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinfeng Han
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Qi-Gui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yi-Fei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Yi Zheng
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - Fei Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| | - San-Jie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- National Demonstration Center for Experimental Animal Education, Sichuan Agricultural University, Chengdu 611130, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Diagnostic Technique, Ministry of Agriculture and Rural Affairs, Chengdu 611130, China
| |
Collapse
|
4
|
Yue Y, Ren Y, Lu C, Li P, Zhang G. Epigenetic regulation of human FOXP3+ Tregs: from homeostasis maintenance to pathogen defense. Front Immunol 2024; 15:1444533. [PMID: 39144146 PMCID: PMC11323565 DOI: 10.3389/fimmu.2024.1444533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Regulatory T cells (Tregs), characterized by the expression of Forkhead Box P3 (FOXP3), constitute a distinct subset of T cells crucial for immune regulation. Tregs can exert direct and indirect control over immune homeostasis by releasing inhibitory factors or differentiating into Th-like Treg (Th-Treg), thereby actively contributing to the prevention and treatment of autoimmune diseases. The epigenetic regulation of FOXP3, encompassing DNA methylation, histone modifications, and post-translational modifications, governs the development and optimal suppressive function of Tregs. In addition, Tregs can also possess the ability to maintain homeostasis in diverse microenvironments through non-suppressive mechanisms. In this review, we primarily focus on elucidating the epigenetic regulation of Tregs as well as their multifaceted roles within diverse physiological contexts while looking forward to potential strategies involving augmentation or suppression of Tregs activity for disease management, particularly in light of the ongoing global COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 PMCID: PMC11694249 DOI: 10.1111/imr.13354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
6
|
Moreno-Corona NC, de León-Bautista MP, León-Juárez M, Hernández-Flores A, Barragán-Gálvez JC, López-Ortega O. Rab GTPases, Active Members in Antigen-Presenting Cells, and T Lymphocytes. Traffic 2024; 25:e12950. [PMID: 38923715 DOI: 10.1111/tra.12950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
Processes such as cell migration, phagocytosis, endocytosis, and exocytosis refer to the intense exchange of information between the internal and external environment in the cells, known as vesicular trafficking. In eukaryotic cells, these essential cellular crosstalks are controlled by Rab GTPases proteins through diverse adaptor proteins like SNAREs complex, coat proteins, phospholipids, kinases, phosphatases, molecular motors, actin, or tubulin cytoskeleton, among others, all necessary for appropriate mobilization of vesicles and distribution of molecules. Considering these molecular events, Rab GTPases are critical components in specific biological processes of immune cells, and many reports refer primarily to macrophages; therefore, in this review, we address specific functions in immune cells, concretely in the mechanism by which the GTPase contributes in dendritic cells (DCs) and, T/B lymphocytes.
Collapse
Affiliation(s)
| | - Mercedes Piedad de León-Bautista
- Escuela de Medicina, Universidad Vasco de Quiroga, Morelia, Mexico
- Human Health, Laboratorio de Enfermedades Infecciosas y Genómica (INEX LAB), Morelia, Mexico
| | - Moises León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | | | - Juan Carlos Barragán-Gálvez
- División de Ciencias Naturales y Exactas, Departamento de Farmacia, Universidad de Guanajuato, Guanajuato, Mexico
| | - Orestes López-Ortega
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institute Necker Enfants Malades, Paris, France
| |
Collapse
|
7
|
Engin A. Lipid Storage, Lipolysis, and Lipotoxicity in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:97-129. [PMID: 39287850 DOI: 10.1007/978-3-031-63657-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride (triacylglycerol: TAG) turnover. Hypoxia in obese adipose tissue due to hypertrophic adipocytes results in excess deposition of extracellular matrix (ECM) components. Cluster of differentiation (CD) 44, as the main receptor of the extracellular matrix component regulates cell-cell and cell-matrix interactions including diet-induced insulin resistance. Excess TAGs, sterols, and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets (LDs). Once LDs are formed, they grow up because of the excessive amount of intracellular FFA stored and reach a final size. The ratio of FFA turnover/lipolysis decreases significantly with increases in the degree of obesity. Dysfunctional adipose tissue is unable to expand further to store excess dietary lipids, increased fluxes of plasma FFAs lead to ectopic fatty acid deposition and lipotoxicity. Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. Adipocyte death is a prerequisite for the transition from hypertrophic to hyperplastic obesity. Increased adipocyte number in obesity has life-long effects on white adipose tissue mass. The positive correlation between the adipose tissue volume and magnetic resonance imaging proton density fat fraction estimation is used for characterization of the obesity phenotype, as well as the risk stratification and selection of appropriate treatment strategies. In obese patients with type 2 diabetes, visceral adipocytes exposed to chronic/intermittent hyperglycemia develop a new microRNAs' (miRNAs') expression pattern. Visceral preadipocytes memorize the effect of hyperglycemia via changes in miRNAs' expression profile and contribute to the progression of diabetic phenotype. Nonsteroidal anti-inflammatory drugs, metformin, and statins can be beneficial in treating the local or systemic consequences of white adipose tissue inflammation. Rapamycin inhibits leptin-induced LD formation. Collectively, in this chapter, the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of LD interactions with the other cellular organelles are reviewed. Furthermore, clinical perspective of fat cell turnover in obesity is also debated.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
8
|
Qi X, Li Z, Han J, Liu W, Xia P, Cai X, Liu X, Liu X, Zhang J, Yu P. Multifaceted roles of T cells in obesity and obesity-related complications: A narrative review. Obes Rev 2023; 24:e13621. [PMID: 37583087 DOI: 10.1111/obr.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 03/18/2023] [Accepted: 07/19/2023] [Indexed: 08/17/2023]
Abstract
Obesity is characterized by chronic low-grade inflammatory responses in the adipose tissue, accompanied by pronounced insulin resistance and metabolic anomalies. It affects almost all body organs and eventually leads to diseases such as fatty liver disease, type 2 diabetes mellitus, and atherosclerosis. Recently, T cells have emerged as interesting therapeutic targets because the dysfunction of T cells and their cytokines in the adipose tissue is implicated in obesity-induced inflammation and their complicated onset. Although several recent narrative reviews have provided a brief overview of related evidence in this area, they have mainly focused on either obesity-associated T cell metabolism or modulation of T cell activation in obesity. Moreover, at present, no published review has reported on the multifaceted roles of T cells in obesity and obesity-related complications, even though there has been a significant increase in studies on this topic since 2019. Therefore, this narrative review aims to comprehensively summarize current advances in the mechanistic roles of T cells in the development of obesity and its related complications. Further, we aim to discuss relevant drugs for weight loss as well as the contradictory role of T cells in the same disease so as to highlight key findings regarding this topic and provide a valid basis for future treatment strategies.
Collapse
Affiliation(s)
- Xinrui Qi
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiashu Han
- MD Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenqing Liu
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xia Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiao Liu
- Department of Cardiology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xu Liu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Kato Y, Shirai R, Ohbuchi K, Oizumi H, Yamamoto M, Miyata W, Iguchi T, Mimaki Y, Miyamoto Y, Yamauchi J. Hesperetin Ameliorates Inhibition of Neuronal and Oligodendroglial Cell Differentiation Phenotypes Induced by Knockdown of Rab2b, an Autism Spectrum Disorder-Associated Gene Product. Neurol Int 2023; 15:371-391. [PMID: 36976668 PMCID: PMC10057161 DOI: 10.3390/neurolint15010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a central nervous system (CNS) neurodevelopmental disorder that includes autism, pervasive developmental disorder, and Asperger’s syndrome. ASD is characterized by repetitive behaviors and social communication deficits. ASD is thought to be a multifactorial disorder with a range of genetic and environmental factors/candidates. Among such factors is the rab2b gene, although it remains unclear how Rab2b itself is related to the CNS neuronal and glial developmental disorganization observed in ASD patients. Rab2 subfamily members regulate intracellular vesicle transport between the endoplasmic reticulum and the Golgi body. To the best of our knowledge, we are the first to report that Rab2b positively regulates neuronal and glial cell morphological differentiation. Knockdown of Rab2b inhibited morphological changes in N1E-115 cells, which are often used as the neuronal cell differentiation model. These changes were accomplished with decreased expression levels of marker proteins in neuronal cells. Similar results were obtained for FBD-102b cells, which are used as the model of oligodendroglial cell morphological differentiation. In contrast, knockdown of Rab2a, which is another Rab2 family member not known to be associated with ASD, affected only oligodendroglial and not neuronal morphological changes. In contrast, treatment with hesperetin, a citrus flavonoid with various cellular protective effects, in cells recovered the defective morphological changes induced by Rab2b knockdown. These results suggest that knockdown of Rab2b inhibits differentiation in neuronal and glial cells and may be associated with pathological cellular phenotypes in ASD and that hesperetin can recover their phenotypes at the in vitro level at least.
Collapse
Affiliation(s)
- Yukino Kato
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Remina Shirai
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Wakana Miyata
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Tomoki Iguchi
- Department of Medicinal Pharmacognosy, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (T.I.); (Y.M.)
| | - Yoshihiro Mimaki
- Department of Medicinal Pharmacognosy, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (T.I.); (Y.M.)
| | - Yuki Miyamoto
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Tokyo, Japan
| | - Junji Yamauchi
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Tokyo, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya 156-8506, Tokyo, Japan
- Correspondence: ; Tel.: +81-42-676-7164; Fax: +81-42-676-8841
| |
Collapse
|
10
|
Zi C, Wang D, Gao Y, He L. The role of Th17 cells in endocrine organs: Involvement of the gut, adipose tissue, liver and bone. Front Immunol 2023; 13:1104943. [PMID: 36726994 PMCID: PMC9884980 DOI: 10.3389/fimmu.2022.1104943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
T Helper 17 (Th17) cells are adaptive immune cells that play myriad roles in the body. Immune-endocrine interactions are vital in endocrine organs during pathological states. Th17 cells are known to take part in multiple autoimmune diseases over the years. Current evidence has moved from minimal to substantial that Th17 cells are closely related to endocrine organs. Diverse tissue Th17 cells have been discovered within endocrine organs, including gut, adipose tissue, liver and bone, and these cells are modulated by various secretions from endocrine organs. Th17 cells in these endocrine organs are key players in the process of an array of metabolic disorders and inflammatory conditions, including obesity, insulin resistance, nonalcoholic fatty liver disease (NAFLD), primary sclerosing cholangitis (PSC), osteoporosis and inflammatory bowel disease (IBD). We reviewed the pathogenetic or protective functions played by Th17 cells in various endocrine tissues and identified potential regulators for plasticity of it. Furthermore, we discussed the roles of Th17 cells in crosstalk of gut-organs axis.
Collapse
Affiliation(s)
- Changyan Zi
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Die Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- School of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| | - Lisha He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Yongxiang Gao, ; Lisha He,
| |
Collapse
|
11
|
Abstract
The global prevalences of obesity and type 2 diabetes mellitus have reached epidemic status, presenting a heavy burden on society. It is therefore essential to find novel mechanisms and targets that could be utilized in potential treatment strategies and, as such, intracellular membrane trafficking has re-emerged as a regulatory tool for controlling metabolic homeostasis. Membrane trafficking is an essential physiological process that is responsible for the sorting and distribution of signalling receptors, membrane transporters and hormones or other ligands between different intracellular compartments and the plasma membrane. Dysregulation of intracellular transport is associated with many human diseases, including cancer, neurodegeneration, immune deficiencies and metabolic diseases, such as type 2 diabetes mellitus and its associated complications. This Review focuses on the latest advances on the role of endosomal membrane trafficking in metabolic physiology and pathology in vivo, highlighting the importance of this research field in targeting metabolic diseases.
Collapse
Affiliation(s)
- Jerome Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1065 C3M, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France.
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
12
|
Ren HZ, Xia SZ, Qin XQ, Hu AY, Wang JL. FOXO1 Alleviates Liver Ischemia-reperfusion Injury by Regulating the Th17/Treg Ratio through the AKT/Stat3/FOXO1 Pathway. J Clin Transl Hepatol 2022; 10:1138-1147. [PMID: 36381102 PMCID: PMC9634774 DOI: 10.14218/jcth.2021.00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Hepatic ischemic reperfusion injury (IRI) occurring during surgery seriously affects patient prognosis. The specific mechanism of IRI has not been fully elucidated. The study aim was to explore the changes of inflammatory environment, and the relationship of the Th17/Treg cell ratio and FOXO1 expression in hepatic IRI. METHODS Liver samples at different ischemic times were collected from patients and mice. The expression of inflammatory markers and FOXO1 in the liver was detected by western blotting and qPCR. Phenotypic changes of liver lymphocytes were analyzed by flow cytometry. The AKT/Stat3/FOXO1 pathway was verified by targeting AKT with GSK2141795. The role of FOXO1 in liver inflammation and changes in lymphocyte phenotype was confirmed by upregulating FOXO1 with resveratrol. RESULTS Prolonged ischemic time aggravates liver injury in both humans and mouse models of hepatic IRI. IR-stress caused Th17/Treg imbalance and FOXO1 down-regulation by activating the AKT/Stat3/FOXO1 signaling pathway. Upregulation of FOXO1 reversed the Th17/Treg cytokine imbalance and altered the inflammation environment in the liver. CONCLUSIONS Liver IRI induced Th17/Treg imbalance. Upregulation of FOXO1 reversed the imbalance and alleviated liver inflammation.
Collapse
Affiliation(s)
- Hao-Zhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Sen-Zhe Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Xue-Qian Qin
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - An-Yin Hu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jing-Lin Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Ochsner SA, Pillich RT, Rawool D, Grethe JS, McKenna NJ. Transcriptional regulatory networks of circulating immune cells in type 1 diabetes: A community knowledgebase. iScience 2022; 25:104581. [PMID: 35832893 PMCID: PMC9272393 DOI: 10.1016/j.isci.2022.104581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Investigator-generated transcriptomic datasets interrogating circulating immune cell (CIC) gene expression in clinical type 1 diabetes (T1D) have underappreciated re-use value. Here, we repurposed these datasets to create an open science environment for the generation of hypotheses around CIC signaling pathways whose gain or loss of function contributes to T1D pathogenesis. We firstly computed sets of genes that were preferentially induced or repressed in T1D CICs and validated these against community benchmarks. We then inferred and validated signaling node networks regulating expression of these gene sets, as well as differentially expressed genes in the original underlying T1D case:control datasets. In a set of three use cases, we demonstrated how informed integration of these networks with complementary digital resources supports substantive, actionable hypotheses around signaling pathway dysfunction in T1D CICs. Finally, we developed a federated, cloud-based web resource that exposes the entire data matrix for unrestricted access and re-use by the research community. Re-use of transcriptomic type 1 diabetes (T1D) circulating immune cells (CICs) datasets We generated transcriptional regulatory networks for T1D CICs Use cases generate substantive hypotheses around signaling pathway dysfunction in T1D CICs Networks are freely accessible on the web for re-use by the research community
Collapse
Affiliation(s)
- Scott A. Ochsner
- Department of Molecular, Baylor College of Medicine, Houston, TX 77030, USA
- Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rudolf T. Pillich
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Deepali Rawool
- Center for Research in Biological Systems, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeffrey S. Grethe
- Center for Research in Biological Systems, University of California San Diego, La Jolla, CA 92093, USA
| | - Neil J. McKenna
- Department of Molecular, Baylor College of Medicine, Houston, TX 77030, USA
- Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Corresponding author
| |
Collapse
|
14
|
Vps33B controls Treg cell suppressive function through inhibiting lysosomal nutrient sensing complex-mediated mTORC1 activation. Cell Rep 2022; 39:110943. [PMID: 35705052 DOI: 10.1016/j.celrep.2022.110943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
The suppressive function of regulatory T (Treg) cells is tightly controlled by nutrient-fueled mechanistic target of rapamycin complex 1 (mTORC1) activation, yet its dynamics and negative regulation remain unclear. Here we show that Treg-specific depletion of vacuolar protein sorting 33B (Vps33B) in mice results in defective Treg cell suppressive function and acquisition of effector phenotype, which in turn leads to disturbed T cell homeostasis and boosted antitumor immunity. Mechanistically, Vps33B binds with lysosomal nutrient-sensing complex (LYNUS) and promotes late endosome and lysosome fusion and clearance of the LYNUS-containing late endosome/lysosome, and therefore suppresses mTORC1 activation. Vps33B deficiency in Treg cells results in disordered endosome lysosome fusion, which leads to accumulation of LYNUS that causes elevated mTORC1 activation and hyper-glycolytic metabolism. Taken together, our study reveals that Vps33B maintains Treg cell suppressive function through sustaining endolysosomal homeostasis and therefore restricting amino acid-licensed mTORC1 activation and metabolism.
Collapse
|
15
|
Liu Y, Kan Y, Huang Y, Jiang C, Zhao L, Hu J, Pang W. Physicochemical Characteristics and Antidiabetic Properties of the Polysaccharides from Pseudostellaria heterophylla. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123719. [PMID: 35744844 PMCID: PMC9227595 DOI: 10.3390/molecules27123719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
This study aimed to investigate the Pseudostellaria heterophylla polysaccharides (PF40) physicochemical and antidiabetic characteristics. The ultraviolet–visible (UV) spectra, Fourier transform infrared radiation (FT-IR) spectra, nuclear magnetic resonance (NMR) spectra, zeta potential, surface characteristics, and conformational and thermal stability properties of PF40 were characterized. X-ray diffraction (XRD) and scanning electron microscopy (SEM), combined with Congo red test, revealed that PF40 powder has mainly existed in amorphous form with triple-helix conformation. The single-molecular structure of PF40 exhibited a multi-branched structure extending from the center to the periphery by scanning probe microscopy (SPM) scanning. The monosaccharide residue of PF40 was an α-pyranoid ring and exhibits good stability below 168 °C. Experimental studies on antidiabetic characteristics found that PF40 could significantly improve STZ-induced intestinal mucosal damage and reduce the apoptosis of villus epithelial cells. PF40 combined with metformin could significantly improve the symptoms of insulin resistance in type 2 diabetes mellitus (T2DM) rats, the molecular mechanism might be through inhibiting the expression of RORγ protein and increasing Foxp3 protein in the jejunum of T2DM rats, and then restoring the STZ-induced imbalance of T helper 17(Th17)/ regulatory T cells (Treg) cells, thereby maintaining intestinal immune homeostasis. Results identified in this study provided important information regarding the structure and antidiabetic characteristics of Pseudostellaria heterophylla polysaccharides, which can contribute to the development of Pseudostellaria heterophylla polysaccharides for industrial purposes in the future.
Collapse
Affiliation(s)
- Yingying Liu
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China;
| | - Yongjun Kan
- Institute of Materia, Fujian Academy of Chinese Medical Sciences, Fuzhou 350003, China; (Y.K.); (C.J.); (L.Z.)
| | - Yating Huang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Chang Jiang
- Institute of Materia, Fujian Academy of Chinese Medical Sciences, Fuzhou 350003, China; (Y.K.); (C.J.); (L.Z.)
| | - Li Zhao
- Institute of Materia, Fujian Academy of Chinese Medical Sciences, Fuzhou 350003, China; (Y.K.); (C.J.); (L.Z.)
| | - Juan Hu
- The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China;
- Institute of Materia, Fujian Academy of Chinese Medical Sciences, Fuzhou 350003, China; (Y.K.); (C.J.); (L.Z.)
- Correspondence: (J.H.); (W.P.); Tel.: +86-591-87878029 (J.H.); Tel.: +86-591-22861135 (W.P.)
| | - Wensheng Pang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
- Correspondence: (J.H.); (W.P.); Tel.: +86-591-87878029 (J.H.); Tel.: +86-591-22861135 (W.P.)
| |
Collapse
|
16
|
Zi C, He L, Yao H, Ren Y, He T, Gao Y. Changes of Th17 cells, regulatory T cells, Treg/Th17, IL-17 and IL-10 in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. Endocrine 2022; 76:263-272. [PMID: 35397088 DOI: 10.1007/s12020-022-03043-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/19/2022] [Indexed: 12/17/2022]
Abstract
PURPOSE The aim of this study was to investigate the changes of Helper T cells 17 (Th17 cells), Regulatory T cells (Treg cells), Treg/Th17, Interleukin-17 (IL-17) and Interleukin-10 (IL-10) in patients with type 2 diabetes mellitus (T2DM). METHODS Four electronic resource databases were searched from their inception to 1 August 2021. Case-control studies about changes of Th17 cells, Treg cells, Treg/Th17, IL-17 and IL-10 in patients with T2DM were retrieved. We performed this meta-analysis via RevMan V.5.3 and Stata14. RESULTS 20 studies with 1242 individuals were included in the meta-analysis. Compared with the controls, the patients with T2DM had significantly increased levels of percentage of Th17 cells (SMD, 1.74; 95% CI, 0.47-3.01; p < 0.001), IL-17 (SMD, 2.17; 95% CI, 0.06-4.28; p < 0.001), IL-10 (SMD, 1.20; 95% CI, 0.81-1.59; p = 0.003), but decreased levels of percentage of Treg cells (SMD, -1.17; 95% CI, -2.22 to -0.13; p < 0.001) and Treg/Th17 ratio (SMD, -4.43; 95% CI, -7.07 to -1.78; p < 0.001). Subgroup analysis showed that percentage of CD4+CD25+FOXP3+ Tregs (SMD, -2.36; 95% CI, -3.19 to -1.52; p = 0.003) in patients was notably lower than controls. While not significant changes were found in the percentage of CD4+CD25+Tregs (SMD, 0.03; 95% CI, -0.34-0.40; p = 0.63) between patients and controls. For plasma or serum IL-10, a higher plasma IL-10 level (SMD,1.37; 95% CI, 0.92-1.82; p = 0.01) was observed in T2DM. While serum IL-10 (SMD, 0.73; 95% CI, 0.35-1.12; p = 0.79) had no obvious difference between patients and controls. For ELISA or flow cytometry, IL-10 (SMD, 1.2; 95% CI, 0.71-1.70; p = 0.001) was higher in T2DM patients by using detection method of ELISA. Yet IL-10 using flow cytometry and subgroup analysis of IL-17 had no significant differences. CONCLUSIONS Adaptive immune system indeed plays an essential role in the process of T2DM. Imbalance between Th17 and Treg triggers pro-inflammatory environment in patients with T2DM.
Collapse
Affiliation(s)
- Changyan Zi
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, PR China.
| | - Lisha He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, PR China.
| | - Huan Yao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, PR China
| | - Yuan Ren
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, PR China
| | - Tingting He
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, PR China
| | - Yongxiang Gao
- School of International Education, Chengdu University of Traditional Chinese Medicine, 610075, Chengdu, PR China.
| |
Collapse
|
17
|
Zhang J, Chen J, Gao C, Sun X, Wang L, Hu Z, Li G, Wang J, Wang A. Maggot treatment promotes healing of diabetic foot ulcer wounds possibly by upregulating Treg levels. Diabetes Res Clin Pract 2022; 184:109187. [PMID: 35016990 DOI: 10.1016/j.diabres.2021.109187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 11/30/2021] [Accepted: 12/23/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE Through the study of regulatory T cells (Tregs), we found a possible way to promote the healing of diabetic foot ulcers (DFUs) with maggot treatment and investigated the associated mechanism. METHODS Immunohistochemistry was used to examinetissues from DFU patients treated with or without maggot debridement therapy (MDT). The expression of the signature Treg molecule Foxp3, interleukin-10 (IL-10), transforming growth factor-beta (TGF-β), and interferon regulatory factor 4 (IRF-4) in patients with DFU treated with or without MDT was tested by real-time PCR (RT-PCR). CD4+ T cells from mouse spleen cells were cocultured in vitro with maggot excretions/secretions (ES), and Foxp3, IL-10, TGF-β, and IRF-4 levels were measured by RT-PCR. RESULTS Foxp3 expression was obviously increased in DFU patients treated using MDT but less pronounced in those treated without MDT (P < 0.05). Foxp3, IL-10, TGF-β, and IRF-4 gene expression levels were higher in DFU patients treated with MDT than in those treated without MDT. Moreover, in vitro coculture of mouse spleen cells with ESs produced results consistent with the in vivo results (P < 0.001). CONCLUSION MDT/ESs can obviously upregulate the Treg level and may affect DFU healing in different ways, suggesting a new direction for the future treatment of DFU.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China; Nanjing Prevention and Treatment Center for Occupational Diseases, Nanjing, Jiangsu Province, China
| | - Jin'an Chen
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Chunchen Gao
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Xinjuan Sun
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Lei Wang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Zhiwei Hu
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Gai Li
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Jing Wang
- Translational Medicine Center, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China.
| | - Aiping Wang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China.
| |
Collapse
|
18
|
Liu L, Hu J, Wang Y, Lei H, Xu D. The role and research progress of the balance and interaction between regulatory T cells and other immune cells in obesity with insulin resistance. Adipocyte 2021; 10:66-79. [PMID: 33472506 PMCID: PMC7834085 DOI: 10.1080/21623945.2021.1876375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metabolic homoeostasis in adipose tissue plays a major role in obesity-related insulin resistance (IR). Regulatory T (Treg) cells have been recorded to regulate metabolic homoeostasis in adipose tissue. However, their specific mechanism is not yet known. This review aims to present the role of Treg cells and other immune cells in obesity-associated IR, focusing on the balance of numbers and functions of Treg cells and other immune cells as well as the crucial role of their interactions in maintaining adipose tissue homoeostasis. Th1 cells, Th17 cells, CD8+ T cells, and pro-inflammatory macrophages mediate the occurrence of obesity and IR by antagonizing Treg cells, while anti-inflammatory dendritic cells, eosinophils and type 2 innate lymphoid cells (ILC2s) regulate the metabolic homoeostasis of adipose tissue by promoting the proliferation and differentiation of Treg cells. γ δ T cells and invariant natural killer T (iNKT) cells have complex effects on Treg cells, and their roles in obesity-associated IR are controversial. The balance of Treg cells and other immune cells can help maintain the metabolic homoeostasis of adipose tissue. Further research needs to explore more specific molecular mechanisms, thus providing more precise directions for the treatment of obesity with IR.
Collapse
Affiliation(s)
- Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Abstract
Sjögren's syndrome (SjS) is a systemic autoimmune disease marked by xerostomia (dry mouth), keratoconjunctivitis sicca (eye dryness), and other systematic disorders. Its pathogenesis involves an inflammatory process that is characterized by lymphocytic infiltration into exocrine glands and other tissues. Although the development of ectopic lymphoid tissue and overproduction of autoantibodies by hyperactive B cells suggest that they may promote SjS development, treatment directed towards them fails to induce significant laboratory or clinical improvement. T cells are overwhelming infiltrators in most phases of the disease, and the involvement of multiple T cell subsets of suggests the extraordinary complexity of SjS pathogenesis. The factors, including various cellular subtypes and molecules, regulate the activation and suppression of T cells. T cell activation induces inflammatory cell infiltration, B cell activation, tissue damage, and metabolic changes in SjS. Knowledge of the pathways that link these T cell subtypes and regulation of their activities are not completely understood. This review comprehensively summarizes the research progress and our understanding of T cells in SjS, including CD4+ T cells, CD8+ TRM cells, and innate T cells, to provide insights into for clinical treatment.
Collapse
|
20
|
Zhang S, Gang X, Yang S, Cui M, Sun L, Li Z, Wang G. The Alterations in and the Role of the Th17/Treg Balance in Metabolic Diseases. Front Immunol 2021; 12:678355. [PMID: 34322117 PMCID: PMC8311559 DOI: 10.3389/fimmu.2021.678355] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/22/2021] [Indexed: 12/17/2022] Open
Abstract
Chronic inflammation plays an important role in the development of metabolic diseases. These include obesity, type 2 diabetes mellitus, and metabolic dysfunction-associated fatty liver disease. The proinflammatory environment maintained by the innate immunity, including macrophages and related cytokines, can be influenced by adaptive immunity. The function of T helper 17 (Th17) and regulatory T (Treg) cells in this process has attracted attention. The Th17/Treg balance is regulated by inflammatory cytokines and various metabolic factors, including those associated with cellular energy metabolism. The possible underlying mechanisms include metabolism-related signaling pathways and epigenetic regulation. Several studies conducted on human and animal models have shown marked differences in and the important roles of Th17/Treg in chronic inflammation associated with obesity and metabolic diseases. Moreover, Th17/Treg seems to be a bridge linking the gut microbiota to host metabolic disorders. In this review, we have provided an overview of the alterations in and the functions of the Th17/Treg balance in metabolic diseases and its role in regulating immune response-related glucose and lipid metabolism.
Collapse
Affiliation(s)
- Siwen Zhang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xiaokun Gang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Shuo Yang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Mengzhao Cui
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Lin Sun
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Zhuo Li
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Adipose Tissue Immunomodulation and Treg/Th17 Imbalance in the Impaired Glucose Metabolism of Children with Obesity. CHILDREN-BASEL 2021; 8:children8070554. [PMID: 34199040 PMCID: PMC8305706 DOI: 10.3390/children8070554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
In the last few decades, obesity has increased dramatically in pediatric patients. Obesity is a chronic disease correlated with systemic inflammation, characterized by the presence of CD4 and CD8 T cell infiltration and modified immune response, which contributes to the development of obesity related diseases and metabolic disorders, including impaired glucose metabolism. In particular, Treg and Th17 cells are dynamically balanced under healthy conditions, but imbalance occurs in inflammatory and pathological states, such as obesity. Some studies demonstrated that peripheral Treg and Th17 cells exhibit increased imbalance with worsening of glucose metabolic dysfunction, already in children with obesity. In this review, we considered the role of adipose tissue immunomodulation and the potential role played by Treg/T17 imbalance on the impaired glucose metabolism in pediatric obesity. In the patient care, immune monitoring could play an important role to define preventive strategies of pediatric metabolic disease treatments.
Collapse
|
22
|
Raad G, Serra F, Martin L, Derieppe MA, Gilleron J, Costa VL, Pisani DF, Amri EZ, Trabucchi M, Grandjean V. Paternal multigenerational exposure to an obesogenic diet drives epigenetic predisposition to metabolic diseases in mice. eLife 2021; 10:61736. [PMID: 33783350 PMCID: PMC8051948 DOI: 10.7554/elife.61736] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/28/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is a growing societal scourge. Recent studies have uncovered that paternal excessive weight induced by an unbalanced diet affects the metabolic health of offspring. These reports mainly employed single-generation male exposure. However, the consequences of multigenerational unbalanced diet feeding on the metabolic health of progeny remain largely unknown. Here, we show that maintaining paternal Western diet feeding for five consecutive generations in mice induces an enhancement in fat mass and related metabolic diseases over generations. Strikingly, chow-diet-fed progenies from these multigenerational Western-diet-fed males develop a 'healthy' overweight phenotype characterized by normal glucose metabolism and without fatty liver that persists for four subsequent generations. Mechanistically, sperm RNA microinjection experiments into zygotes suggest that sperm RNAs are sufficient for establishment but not for long-term maintenance of epigenetic inheritance of metabolic pathologies. Progressive and permanent metabolic deregulation induced by successive paternal Western-diet-fed generations may contribute to the worldwide epidemic of metabolic diseases.
Collapse
Affiliation(s)
- Georges Raad
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France.,Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Fabrizio Serra
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | - Luc Martin
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | - Jérôme Gilleron
- Université Côte d'Azur, Inserm, C3M, Team Cellular and Molecular Pathophysiology of Obesity and Diabetes (7), Nice, France
| | - Vera L Costa
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | | | | | - Michele Trabucchi
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| | - Valerie Grandjean
- Université Côte d'Azur, Inserm, C3M, TeamControl of Gene Expression (10), Nice, France
| |
Collapse
|
23
|
Hu H, Garcia-Barrio M, Jiang ZS, Chen YE, Chang L. Roles of Perivascular Adipose Tissue in Hypertension and Atherosclerosis. Antioxid Redox Signal 2021; 34:736-749. [PMID: 32390459 PMCID: PMC7910418 DOI: 10.1089/ars.2020.8103] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Perivascular adipose tissue (PVAT), which is present surrounding most blood vessels, from the aorta to the microvasculature of the dermis, is mainly composed of fat cells, fibroblasts, stem cells, mast cells, and nerve cells. Although the PVAT is objectively present, its physiological and pathological significance has long been ignored. Recent Advances: PVAT was considered as a supporting component of blood vessels and a protective cushion to the vessel wall from the neighboring tissues during relaxation and contraction. Nonetheless, further extensive research found that PVAT actively regulates blood vessel tone through PVAT-derived vasoactive factors, including both relaxing and contracting factors. In addition, PVAT contributes to atherosclerosis through paracrine secretion of a large number of bioactive factors such as adipokines and cytokines. Thereby, PVAT regulates the functions of blood vessels through various mechanisms operating directly on PVAT or on the underlying vessel layers, including vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). Critical Issues: PVAT is a unique adipose tissue that plays an essential role in maintaining the vascular structure and regulating vascular function and homeostasis. This review focuses on recent updates on the various PVAT roles in hypertension and atherosclerosis. Future Directions: Future studies should further investigate the actual contribution of alterations in PVAT metabolism to the overall systemic outcomes of cardiovascular disease, which remains largely unknown. In addition, the messengers and underlying mechanisms responsible for the crosstalk between PVAT and ECs and VSMCs in the vascular wall should be systematically addressed, as well as the contributions of PVAT aging to vascular dysfunction.
Collapse
Affiliation(s)
- Hengjing Hu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Minerva Garcia-Barrio
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Yuqing Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Homma Y, Hiragi S, Fukuda M. Rab family of small GTPases: an updated view on their regulation and functions. FEBS J 2021; 288:36-55. [PMID: 32542850 PMCID: PMC7818423 DOI: 10.1111/febs.15453] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
The Rab family of small GTPases regulates intracellular membrane trafficking by orchestrating the biogenesis, transport, tethering, and fusion of membrane-bound organelles and vesicles. Like other small GTPases, Rabs cycle between two states, an active (GTP-loaded) state and an inactive (GDP-loaded) state, and their cycling is catalyzed by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Because an active form of each Rab localizes on a specific organelle (or vesicle) and recruits various effector proteins to facilitate each step of membrane trafficking, knowing when and where Rabs are activated and what effectors Rabs recruit is crucial to understand their functions. Since the discovery of Rabs, they have been regarded as one of the central hubs for membrane trafficking, and numerous biochemical and genetic studies have revealed the mechanisms of Rab functions in recent years. The results of these studies have included the identification and characterization of novel GEFs, GAPs, and effectors, as well as post-translational modifications, for example, phosphorylation, of Rabs. Rab functions beyond the simple effector-recruiting model are also emerging. Furthermore, the recently developed CRISPR/Cas technology has enabled acceleration of knockout analyses in both animals and cultured cells and revealed previously unknown physiological roles of many Rabs. In this review article, we provide the most up-to-date and comprehensive lists of GEFs, GAPs, effectors, and knockout phenotypes of mammalian Rabs and discuss recent findings in regard to their regulation and functions.
Collapse
Affiliation(s)
- Yuta Homma
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Shu Hiragi
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking MechanismsDepartment of Integrative Life SciencesGraduate School of Life SciencesTohoku UniversitySendaiJapan
| |
Collapse
|
25
|
Xiong J, Hu H, Guo R, Wang H, Jiang H. Mesenchymal Stem Cell Exosomes as a New Strategy for the Treatment of Diabetes Complications. Front Endocrinol (Lausanne) 2021; 12:646233. [PMID: 33995278 PMCID: PMC8117220 DOI: 10.3389/fendo.2021.646233] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease, now prevalent worldwide, which is characterized by a relative or absolute lack of insulin secretion leading to chronically increased blood glucose levels. Diabetic patients are often accompanied by multiple macrovascular complications, such as coronary heart disease, hypertension, macrovascular arteriosclerosis, and microvascular complications. Microvascular complications include diabetic kidney injury, diabetic encephalopathy, and diabetic foot, which reduce the quality of life and survival status of patients. Mesenchymal stem cell exosomes (MSC-Exos) possess repair functions similar to MSCs, low immunogenicity, and ease of storage and transport. MSC-Exos have been proven to possess excellent repair effects in repairing various organ damages. This study reviews the application of MSC-Exos in the treatment of DM and its common complications. MSC-Exos may be used as an effective treatment for DM and its complications.
Collapse
Affiliation(s)
| | | | | | - Hui Wang
- *Correspondence: Hui Wang, ; Hua Jiang,
| | - Hua Jiang
- *Correspondence: Hui Wang, ; Hua Jiang,
| |
Collapse
|
26
|
Cai H, Li M, Jian W, Song C, Huang Y, Lan X, Lei C, Chen H. A novel lncRNA BADLNCR1 inhibits bovine adipogenesis by repressing GLRX5 expression. J Cell Mol Med 2020; 24:7175-7186. [PMID: 32449295 PMCID: PMC7339203 DOI: 10.1111/jcmm.15181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 01/14/2023] Open
Abstract
Adipogenesis is a complex cellular process, which needs a series of molecular events, including long non‐coding RNA (lncRNA). In the present study, a novel lncRNA named BADLNCR1 was identified as a regulator during bovine adipocyte differentiation, which plays an inhibitory role in lipid droplet formation and adipogenic marker gene expression. CHIPR‐seq data demonstrated a potential competitive binding motif between BADLNCR1 and sterol regulatory element‐binding proteins 1 and 2 (SREBP1/2). Dual‐luciferase reporter assay indicated target relationship between KLF2 and BADLNCR1. Moreover, after the induction of KLF2, the expression of adipogenic gene reduced, while the expression of BADLNCR1 increased. Real‐time quantitative PCR (qPCR) showed that BADLNCR1 negatively regulated mRNA expression of GLRX5 gene, a stimulator of genes that promoted formation of lipid droplets and expression of adipogenic genes. GLRX5 could partially reverse the effect of BADLNCR1 in bovine adipocyte differentiation. Dual‐luciferase reporter assay stated that BADLNCR1 significantly reduced the enhancement of C/EBPα on promoter activity of GLRX5 gene. Furthermore, CHIP‐PCR and CHIRP‐PCR confirmed the suppressing effect of BADLNCR1 on binding of C/EBPα to GLRX5 promoter. Collectively, this study revealed the molecular mechanisms underlying the negative regulation of BADLNCR1 in bovine adipogenic differentiation.
Collapse
Affiliation(s)
- Hanfang Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingxun Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wang Jian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengchuang Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
27
|
Li C, Spallanzani RG, Mathis D. Visceral adipose tissue Tregs and the cells that nurture them. Immunol Rev 2020; 295:114-125. [PMID: 32162339 DOI: 10.1111/imr.12850] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Visceral adipose tissue (VAT) is a primary site for storage of excess energy, but it also serves as an important endocrine organ that impacts organismal metabolism. Chronic, low-grade inflammation of VAT, and eventually systemically, is one of the major drivers of obesity-associated insulin resistance and metabolic abnormalities. A unique population of regulatory T cells (Tregs), with a distinct transcriptional profile and antigen receptor repertoire resides in VAT, keeps inflammation in check and regulates organismal metabolism. Accumulation of these cells depends on interactions with other local immunocytes and, importantly, subtypes of VAT mesenchymal stromal cells (VmSCs) that are either immunomodulators or adipogenic. We summarize our current understanding of the phenotype, function, dependencies, derivation, and modulations of VAT Tregs, and review the heterogeneity and regulation of VmSCs as well as their cross talk with VAT Tregs. Lastly, we discuss imperative questions remaining to be answered.
Collapse
Affiliation(s)
- Chaoran Li
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Raul German Spallanzani
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Gong Y, Liu H, Tao L. Cajanonic acid A regulates the ratio of Th17/Treg via inhibition of expression of IL-6 and TGF-β in insulin-resistant HepG2 cells. Biosci Rep 2019; 39:BSR20181716. [PMID: 31729529 PMCID: PMC6928521 DOI: 10.1042/bsr20181716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 07/01/2019] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The objectives of the present study are to investigate whether cajanonic acid A (CAA) can reduce insulin resistance (IR) in HepG2 cells and to gain a preliminary understanding of the mechanisms underlying this effect. METHODS Following induction of IR in HepG2 cells, we tested the regulatory effect of CAA on glucose consumption and evaluated hepatocyte production of IL-6, TGF-β, and key molecules in the insulin transduction pathway. A transwell co-culturing system was used to assess the effect of CAA on IR in HepG2 cells during the differentiation of CD4+ T cells by calculating the ratio of (Th17)/regulatory T cell (Treg). We evaluated the effect of CAA on the expression of IL-17RC cells and HepG2 cell apoptosis by immunofluorescence and flow cytometry assay. RESULTS CAA improved dexamethasone-induced reduction in glucose consumption in HepG2 cells, inhibited hepatocyte production of IL-6 and TGF-β, increased the expression of IL-17RC cell, and increased cellular apoptosis in insulin-resistant HepG2 cells. When co-cultured with CD4+ T cells, insulin-resistant HepG2 cells induced a decrease in the ratio of Th17/Treg, but CAA dampened the effect. Application of IL-6 and TGF-β, together with CAA, reversed the effect of CAA on insulin-resistant HepG2 cells. Overexpression of IL17R, however, counteracted the effect of IL-6 neutralizing antibody within the culture system. CONCLUSION CAA can regulate the ratio of Th17/Treg by mediating the expression of IL-6 and TGF-β in insulin-resistant HepG2 cells.
Collapse
Affiliation(s)
- Yanfeng Gong
- Department of Geriatrics, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330006, Jiangxi, China
| | - Huanbing Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330006, Jiangxi, China
| | - Liming Tao
- Department of Geriatrics, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang 330006, Jiangxi, China
| |
Collapse
|
29
|
Gilleron J, Gerdes JM, Zeigerer A. Metabolic regulation through the endosomal system. Traffic 2019; 20:552-570. [PMID: 31177593 PMCID: PMC6771607 DOI: 10.1111/tra.12670] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
The endosomal system plays an essential role in cell homeostasis by controlling cellular signaling, nutrient sensing, cell polarity and cell migration. However, its place in the regulation of tissue, organ and whole body physiology is less well understood. Recent studies have revealed an important role for the endosomal system in regulating glucose and lipid homeostasis, with implications for metabolic disorders such as type 2 diabetes, hypercholesterolemia and non-alcoholic fatty liver disease. By taking insights from in vitro studies of endocytosis and exploring their effects on metabolism, we can begin to connect the fields of endosomal transport and metabolic homeostasis. In this review, we explore current understanding of how the endosomal system influences the systemic regulation of glucose and lipid metabolism in mice and humans. We highlight exciting new insights that help translate findings from single cells to a wider physiological level and open up new directions for endosomal research.
Collapse
Affiliation(s)
- Jerome Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (INSERM), Mediterranean Center of Molecular Medicine (C3M)NiceFrance
| | - Jantje M. Gerdes
- Institute for Diabetes and RegenerationHelmholtz Center MunichNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Anja Zeigerer
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Institute for Diabetes and CancerHelmholtz Center MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes ProgramHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
30
|
Liu R, Nikolajczyk BS. Tissue Immune Cells Fuel Obesity-Associated Inflammation in Adipose Tissue and Beyond. Front Immunol 2019; 10:1587. [PMID: 31379820 PMCID: PMC6653202 DOI: 10.3389/fimmu.2019.01587] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-associated inflammation stems from a combination of cell-intrinsic changes of individual immune cell subsets and the dynamic crosstalk amongst a broad array of immune cells. Although much of the focus of immune cell contributions to metabolic disease has focused on adipose tissue-associated cells, these potent sources of inflammation inhabit other metabolic regulatory tissues, including liver and gut, and recirculate to promote systemic inflammation and thus obesity comorbidities. Tissue-associated immune cells, especially T cell subpopulations, have become a hotspot of inquiry based on their contributions to obesity, type 2 diabetes, non-alcoholic fatty liver diseases and certain types of cancers. The cell-cell interactions that take place under the stress of obesity are mediated by intracellular contact and cytokine production, and constitute a complicated network that drives the phenotypic alterations of immune cells and perpetuates a feed-forward loop of metabolic decline. Herein we discuss immune cell functions in various tissues and obesity-associated cancers from the viewpoint of inflammation. We also emphasize recent advances in the understanding of crosstalk amongst immune cell subsets under obese conditions, and suggest future directions for focused investigations with clinical relevance.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|