1
|
Pan Z, Gan C, Zhi S, Yang Y, Zhang Y, Li L, Zhang S, Huang Q. Gancao Xiexin decoction attenuated experimental colitis through suppressing ACSL4-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119532. [PMID: 39993549 DOI: 10.1016/j.jep.2025.119532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Gancao Xiexin decoction (GCXXD), comprising Glycyrrhiza glabra L., Pinellia ternata (Thunb.) Makino, Scutellaria baicalensis Georgi, Zingiber officinale Roscoe, Panax ginseng C.A.Mey. , Coptis chinensis Franch. , Ziziphus jujuba Mill. , represents a traditional Chinese medicinal formulation utilized for the treatment of ulcerative colitis (UC). Nevertheless, the potential mechanism behind GCXXD treatment for UC is not yet fully elucidated. AIM OF THE STUDY Ulcerative colitis is a chronic inflammatory disorder of the gastrointestinal system distinguished by intestinal barrier destruction. Previous studies have indicated that excessive ferroptosis activation in intestinal epithelial cells (IECs) can worsen damage and focal permeability abnormalities in the colon. One of the main mechanisms of ferroptosis is lipid peroxides, which are dependent on long-chain acyl-CoA synthetase 4 (ACSL4) for the synthesis of membrane phospholipids. Recent research findings have provided evidence that GCXXD significantly reduces the symptoms of ulcerative colitis (UC) by preserving the intestinal mucosal barrier. So, we aim to demonstrate that the pharmacological mechanism of GCXXD is related to ferroptosis mediated by ACSL4 in this research. MATERIALS AND METHODS In this investigation, we evaluated the GSE134025 datasets and established an experimental colitis model caused by DSS and treated with a 20 mg/kg ACSL4 inhibitor (rosiglitazone). Colon pathological alterations and Alcian blue staining were used to confirm ACSL4 inhibition as a possible therapy for UC. We then examined illness symptoms, intestinal mucosa repair, and ferroptosis markers in UC mice after treated with GCXXD (9,12,15 g/kg). Transcriptome study of colon tissues revealed more about the underlying mechanism of GCXXD in the treatment of UC. Finally, we co-administered the ACSL4 upstream agonist with GCXXD in the treatment of UC to show that GCXXD reduced inflammation in UC by modifying ACSL4-induced ferroptosis. RESULTS Through GSE134025 dataset analysis, we discovered that ACSL4 was substantially expressed in UC patients and that its inhibitors successfully reduced the clinical signs and symptoms of UC colon. Furthermore, we found that GCXXD improved colon length and body weight while increasing the expression of mucin, occuldin, and Claudin-1. It also lowered colon inflammatory cell infiltration and levels of IL-1β and TNF-α. In the meantime, GCXXD efficiently decreased ferroptosis-related indicators in colitis mice, such as MDA, Fe2+, COX2, and ACSL4, while also upregulated GPX4 expression. Using KEGG analysis of the genes that were differently expressed between the 3% DSS and GCXXD treatment group, we were able to discover important connections between the hippo signaling pathway, Arachidonic acid metabolism with GCXXD treatment. Due to the fact that TEAD4 functions as an upstream transcription factor for ACSL4, we combined GCXXD and Py-60, a YAP agonist in the treatment of UC. It was worth noting that GCXXD's inhibitory effect of on intestinal mucosa damage and ferroptosis was lessened when the ACSL4 upstream pathway was activated. CONCLUSION Gancao Xiexin decoction attenuated ferroptosis in UC which might through TEAD4/ACSL4 pathway.
Collapse
Affiliation(s)
- Zengfeng Pan
- Institute of Basic Medical Sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China; Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Caiyan Gan
- Institute of Basic Medical Sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China; Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Shaobin Zhi
- Institute of Cardiovascular Disease Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Yali Yang
- Institute of Hakka Medicinal Bio-resources, Medical College of Jiaying University, Meizhou, China
| | - Yinmei Zhang
- Institute of Basic Medical Sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China; Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Lihai Li
- Institute of Basic Medical Sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China; Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Shengyuan Zhang
- Institute of Hakka Medicinal Bio-resources, Medical College of Jiaying University, Meizhou, China.
| | - Qionghui Huang
- Institute of Cardiovascular Disease Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.
| |
Collapse
|
2
|
Lai CL, Santner-Nanan B, Maltese PJ, Ong CKS, Palmer DJ, Campbell DE, Makrides M, Gold M, Nanan R, Prescott SL, Hsu PS. Impaired calcium influx underlies skewed T helper cell differentiation in children with IgE-mediated food allergies. Allergy 2025; 80:513-524. [PMID: 39250135 DOI: 10.1111/all.16310] [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: 05/27/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Reasons for Th2 skewing in IgE-mediated food allergies remains unclear. Clinical observations suggest impaired T cell activation may drive Th2 responses evidenced by increased atopic manifestations in liver transplant patients on tacrolimus (a calcineurin inhibitor). We aimed to assess differentiation potential, T cell activation and calcium influx of naïve CD4+ T cells in children with IgE-mediated food allergies. METHODS Peripheral blood mononuclear cells from infants in the Starting Time for Egg Protein (STEP) Trial were analyzed by flow cytometry to assess Th1/Th2/Treg development. Naïve CD4+ T cells from children with and without food allergies were stimulated for 7 days to assess Th1/Th2/Treg transcriptional factors and cytokines. Store operated calcium entry (SOCE) was measured in children with and without food allergies. The effect of tacrolimus on CD4+ T cell differentiation was assessed by treating stimulated naïve CD4+ T cells from healthy volunteers with tacrolimus for 7 days. RESULTS Egg allergic infants had impaired development of IFNγ+ Th1 cells and FoxP3+ transitional CD4+ T cells compared with non-allergic infants. This parallels reduced T-bet, IFNγ and FoxP3 expression in naïve CD4+ T cells from food allergic children after in vitro culture. SOCE of naïve CD4+ T cells was impaired in food allergic children. Naïve CD4+ T cells treated with tacrolimus had reduced IFNγ, T-bet, and FoxP3, but preserved IL-4 expression. CONCLUSIONS In children with IgE-mediated food allergies, dysregulation of T helper cell development is associated with impaired SOCE, which underlies an intrinsic impairment in Th1 and Treg differentiation. Along with tacrolimus-induced Th2 skewing, this highlights an important role of SOCE/calcineurin pathway in T helper cell differentiation.
Collapse
Affiliation(s)
- C L Lai
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - B Santner-Nanan
- Sydney Medical School Nepean and Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales, Australia
| | - P J Maltese
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
| | - C K S Ong
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
| | - D J Palmer
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Medicine, The University of Western Australia, Crawley, Western Australia, Australia
| | - D E Campbell
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - M Makrides
- South Australian Health and Medical Research Institute, SAHMRI Women and Kids, Adelaide, South Australia, Australia
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - M Gold
- Discipline of Paediatrics, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - R Nanan
- Sydney Medical School Nepean and Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales, Australia
| | - S L Prescott
- School of Medicine, The University of Western Australia, Crawley, Western Australia, Australia
- The ORIGINS Project, Telethon Kids Institute, The University of Western Australia, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
| | - P S Hsu
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Kids Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Food Allergy Research (CFAR), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Yoodee S, Plumworasawat S, Malaitad T, Peerapen P, Thongboonkerd V. Differential structural characteristics, physicochemical properties, and calcium-binding capabilities of annexin A2 wild-type versus E53A, E96A, D162A, E247A and D322A mutants. Arch Biochem Biophys 2025; 764:110267. [PMID: 39674566 DOI: 10.1016/j.abb.2024.110267] [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: 09/30/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Annexin A2 (ANXA2) is a Ca2+-dependent multifunctional protein containing five Ca2+-binding domains, but their functional significance and difference remain unclear. Herein, glutamic acid (E) or aspartic acid (D) in five Ca2+-binding domains of canine ANXA2 (98.82 % and 96.76-99.41 % identical to ANXA2 from human and other mammals, respectively) was substituted by alanine (A) using site-directed mutagenesis. Recombinant ANXA2 wild-type (WT) and E53A, E96A, D162A, E247A and D322A mutants were constructed and expressed using a bacterial expression system followed by high-affinity purification using nickel-nitrilotriacetic acid (Ni-NTA) matrix. Efficacies of their expression and purification were confirmed by SDS-PAGE and Western blotting. Their amino acid sequences were verified by nanoLC-ESI-Qq-TOF tandem mass spectrometry. ATR-FTIR spectroscopy revealed that their secondary structure significantly differed (α-helix decreased but random coil increased in all mutants). Analyses of physicochemical properties revealed that molecular weight slightly decreased, whereas isoelectric point, aliphatic index, grand average of hydropathicity, electrostatic potential and molecular hydrophobicity potential slightly increased in all the mutants compared with WT. Interestingly, Ca2+-binding capability of these mutants (particularly E96A and D322A) significantly decreased from that of WT. In summary, secondary structure, physicochemical properties, and Ca2+-binding capability of E53A, E96A, D162A, E247A and D322A mutants of ANXA2 significantly differed from its WT, consistent with the loss of negatively charged E/D. In particular, E96A and D322A exhibited the lowest Ca2+-binding capability. These data and recombinant proteins would be useful for further investigations of the Ca2+-dependent functions of individual Ca2+-binding domains in ANXA2.
Collapse
Affiliation(s)
- Sunisa Yoodee
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thanyalak Malaitad
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Paleerath Peerapen
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
4
|
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
|
5
|
Hu Y, Tang J, Yu D, Su S, Fang J, Xia L, Xu W, Zhu W, Song N, Wang F, Diao D, Zhang W. Correlation and diagnostic significance of CD4 T cell subsets and NLRP3 inflammasome in ulcerative colitis: the role of the NLRP3/T-bet/GATA3 axis. BMC Gastroenterol 2025; 25:23. [PMID: 39833691 PMCID: PMC11748810 DOI: 10.1186/s12876-025-03603-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND AND AIM Ulcerative colitis (UC) is characterized by complex immunological interactions involving CD4 T cell subsets and the NLRP3 inflammasome, which influence inflammatory responses. This investigation focused on delineating the activation profiles of these components and their correlation with disease severity and activity, assessing their diagnostic implications in UC. METHODS We conducted immunohistochemistry and ELISA assays to measure markers expression of CD4 T cell subsets and the NLRP3 inflammasome in UC patients versus controls. Findings were validated using correlation analysis, molecular docking and ROC curves. RESULTS UC patients displayed increased Th1 (T-bet, TNF-α), Th2 (GATA3, IL-6), and Th17 (RORγt, IL-17, IL-22, IL-23) markers versus controls. Additionally, Th1 and Th2 cytokines (IL-2 and IL-4) were significantly elevated in severe UC, while Treg markers (FOXP3, IL-10, TGF-β1) were elevated only in mild-to-moderate UC. Enhanced NLRP3 inflammasome activation, indicated by elevated NLRP3, Caspase-1, and IL-1β levels. These molecular patterns, confirmed through correlation analysis and molecular docking, underscored strong correlations among NLRP3, T-bet, and GATA3, supporting the proposed NLRP3/T-bet/GATA3 axis. This axis, along with other biomarkers, showed strong associations with UC severity, Mayo score, UCEIS, demonstrated relatively high diagnostic value. CONCLUSION The NLRP3/T-bet/GATA3 axis provides a referable strategy for multi-targeted combined treatment of UC and may serve as potential biomarkers for enhancing diagnostic accuracy and guiding therapy.
Collapse
Affiliation(s)
- Yingnan Hu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Jingyi Tang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dian Yu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Su
- Department of Spleen and Stomach Diseases, Qujiang District Hospital of Traditional Chinese Medicine, Quzhou, China
| | - Jintao Fang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linying Xia
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenjun Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weihan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ninping Song
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318, Chaowang Road, Gongshu District, Hangzhou City, Zhejiang Province, CN310005, People's Republic of China
| | - Fengyong Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318, Chaowang Road, Gongshu District, Hangzhou City, Zhejiang Province, CN310005, People's Republic of China
| | - Dechang Diao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Wei Zhang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318, Chaowang Road, Gongshu District, Hangzhou City, Zhejiang Province, CN310005, People's Republic of China.
| |
Collapse
|
6
|
Liu Z, Wu S, Zhang W, Cui H, Zhang J, Yin X, Zheng X, Shen T, Ying H, Chen L, Wang H, Jiang J. Cordycepin mitigates dextran sulfate sodium-induced colitis through improving gut microbiota composition and modulating Th1/Th2 and Th17/Treg balance. Biomed Pharmacother 2024; 180:117394. [PMID: 39395256 DOI: 10.1016/j.biopha.2024.117394] [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: 06/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Imbalances in Th1/Th2 and Th17/Treg immune axes, coupled with disruptions in the gut microbiota (GM), play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). Cordycepin, a natural anti-inflammatory compound, holds promise in mitigating IBD by rebalancing these immune axes in conjunction with modulating the GM. The aim of this experiment is to investigate the potential of cordycepin in mitigating enteritis and elucidate the underlying mechanisms associated with its ameliorative effects on enteritis. METHODS On the day of inducing experimental colitis with Dextran Sulfate Sodium (DSS), mice in the DSS + Cordycepin and Cordycepin groups received 50 mg/kg/day Cordycepin via intra-gastric administration (i.g.) for seven consecutive days, respectively. Mice in the DSS and control groups were treated with equal volumes of saline. On day 8, all mice were euthanized under pentobarbital sodium anesthesia. RESULTS In a DSS-induced colitis mouse model, Cordycepin treatment led to a significant reduction in the disease activity index (DAI), splenic weight, and colonic pathological injury while simultaneously improving body weight and colonic length. Furthermore, it positively impacted GM composition, resulting in decreased Th1 and Th17 cells, alongside an increase in Th2 and Treg cells. The contents of the mouse colon were extracted for microbial community analysis. Mouse blood was prepared into a single-cell suspension, and flow cytometry was used to assess the expressio of Treg, Th17, Th1, and Th2 immune cells. CONCLUSIONS These results underscored the effective intervention of cordycepin in ameliorating DSS-induced colitis by harmonizing the interplay between GM and immune homeostasis.
Collapse
Affiliation(s)
- Zhilin Liu
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Wenting Zhang
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou Children's Hospital, Changzhou 213003, China
| | - Hengwei Cui
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jingfeng Zhang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xuan Yin
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Shen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, China
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Haitao Wang
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Institute of Cell Therapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| |
Collapse
|
7
|
Zhou S, Liu S, Jiang A, Li Z, Duan C, Li B. New insights into the stromal interaction molecule 2 function and its impact on the immunomodulation of tumor microenvironment. Cell Biosci 2024; 14:119. [PMID: 39272139 PMCID: PMC11395313 DOI: 10.1186/s13578-024-01292-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Immune cells-enhanced immunotherapy exhibits unprecedented overall survival-prolongation even curable in some cancer patients. Although so, most of the patients show no response. Tumor microenvironment (TME) where immune cells settle down has multi-faceted influences, but usually creates an immunosuppressive niche that facilitating tumor cells escape from immune attack. The metabolites and malnutrition of TME exert enormous effects on the resident immune cells, but the underlying mechanism is largely unknown. The stromal interaction molecules 2 (STIM2) is an endoplasmic reticulum (ER) calcium (Ca2+) sensor to maintain Ca2+ homeostasis. Notably, the cytosol STIM2 C-terminus is long with various domains that are available for the combination or/and molecular modification. This distinct structure endows STIM2 with a high susceptibility to numerous permeable physico-chemical molecules or protein interactions. STIM2 and its variants are extensively expressed in various immune cells, especially in T immune cells. STIM2 was reported closely correlated with the function of immune cells via regulating Ca2+ signaling, energy metabolism and cell fitness. Herein, we sum the latest findings on the STIM2 structure, focusing on its distinct characteristics and profound effect on the regulation of Ca2+ homeostasis and multi-talented functionality. We also outline the advancements on the underlying mechanism how STIM2 anomalies influence the function of immune cells and on the turbulent expression or/and amenably modification of STIM2 within the tumor niches. Then we discuss the translation of these researches into antitumor approaches, emphasizing the potential of STIM2 as a therapeutic target for direct inhibition of tumor cells or more activation towards immune cells driving to flare TME. This review is an update on STIM2, aiming to rationalize the potential of STIM2 as a therapeutic target for immunomodulation, engaging immune cells to exert the utmost anti-tumor effect.
Collapse
Affiliation(s)
- Shishan Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Shujie Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Anfeng Jiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Zhiyuan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
8
|
Hu Y, Tang J, Xie Y, Xu W, Zhu W, Xia L, Fang J, Yu D, Liu J, Zheng Z, Zhou Q, Shou Q, Zhang W. Gegen Qinlian decoction ameliorates TNBS-induced ulcerative colitis by regulating Th2/Th1 and Tregs/Th17 cells balance, inhibiting NLRP3 inflammasome activation and reshaping gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117956. [PMID: 38428658 DOI: 10.1016/j.jep.2024.117956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine Gegen Qinlian Decoction (GQD) has been clinically shown to be an effective treatment of ulcerative colitis (UC) in China. However, the underlying mechanism of GQD's anti-ulcerative colitis properties and its effect on gut microbiota still deserve further exploration. AIM OF THE STUDY This study observed the regulatory effects of GQD on Th2/Th1 and Tregs/Th17 cells balance, the NOD-like receptor family pyrin domain containing 3 (NLRP3) infammasome and gut microbiota in TNBS-induced UC in BALB/c mice. MATERIALS AND METHODS 61 main chemical compounds in the GQD were determined by UPLC-Q-TOF/MS. The UC BALB/c model was established by intrarectal administration of trinitrobenzene sulfonic acid (TNBS), and GQD was orally administered at low and high dosages of 2.96 and 11.83 g/kg/day, respectively. The anti-inflammatory effects of GQD for ulcerative colitis were evaluated by survival rate, body weight, disease activity index (DAI) score, colonic weight and index, spleen index, hematoxylin-eosin (HE) staining and histopathological scores. Flow cytometry was used to detect the percentage of CD4, Th1, Th2, Th17 and Tregs cells. The levels of Th1-/Th2-/Th17-/Tregs-related inflammatory cytokines and additional proinflammatory cytokines (IL-1β, IL-18) were detected by CBA, ELISA, and RT-PCR. The expressions of GATA3, T-bet, NLRP3, Caspase-1, IL-Iβ, Occludin and Zonula occludens-1 (ZO-1) on colon tissues were detected by Western blot and RT-PCR. Transcriptome sequencing was performed using colon tissue and 16S rRNA gene sequencing was performed on intestinal contents. Fecal microbiota transplantation (FMT) was employed to assess the contribution of intestinal microbiota and its correlation with CD4 T cells and the NLRP3 inflammasome. RESULTS GQD increased the survival rate of TNBS-induced UC in BALB/c mice, and significantly improved their body weight, DAI score, colonic weight and index, spleen index, and histological characteristics. The intestinal barrier dysfunction was repaired after GQD administration through promoting the expression of tight junction proteins (Occludin and ZO-1). GQD restored the balance of Th2/Th1 and Tregs/Th17 cells immune response of colitis mice, primarily inhibiting the increase in Th2/Th1 ratio and their transcription factor production (GATA3 and T-bet). Morever, GQD changed the secretion of Th1-/Th2-/Th17-/Tregs-related cytokines (IL-2, IL-12, IL-5, IL-13, IL-6, IL-10, and IL-17A) and reduced the expressions of IL-1β, IL-18. Transcriptome results suggested that GQD could also remodel the immune inflammatory response of colitis by inhibiting NOD-like receptor signaling pathway, and Western blot, immunohistochemistry and RT-PCR further revealed that GQD exerted anti-inflammatory effects by inhibiting the NLRP3 inflammasome, such as down-regulating the expression of NLRP3, Caspase-1 and IL-1β. More interestingly, GQD regulated gut microbiota dysbiosis, suppressed the overgrowth of conditional pathogenic gut bacteria like Helicobacter, Proteobacteria, and Mucispirillum, while the probiotic gut microbiota, such as Lactobacillus, Muribaculaceae, Ruminiclostridium_6, Akkermansia, and Ruminococcaceae_unclassified were increased. We further confirmed that GQD-treated gut microbiota was sufficient to relieve TNBS-induced colitis by FMT, involving the modulation of Th2/Th1 and Tregs/Th17 balance, inhibition of NLRP3 inflammasome activation, and enhancement of colonic barrier function. CONCLUSIONS GQD might alleviate TNBS-induced UC via regulating Th2/Th1 and Tregs/Th17 cells Balance, inhibiting NLRP3 inflammasome and reshaping gut microbiota, which may provide a novel strategy for patients with colitis.
Collapse
Affiliation(s)
- Yingnan Hu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingyi Tang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yongfeng Xie
- Department of Burn Plastic Surgery, Huai'an Hospital Affiliated to Xuzhou Medical University, Jiangsu, 223001, China
| | - Wenjun Xu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Weihan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Linying Xia
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jintao Fang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dian Yu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingjing Liu
- Department of General Surgery, Haining City Central Hospital, Jiaxing, 314408, China
| | - Zhipeng Zheng
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiujing Zhou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiyang Shou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Wei Zhang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China; The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
9
|
Palestra F, Memoli G, Ventrici A, Trocchia M, Galdiero M, Varricchi G, Loffredo S. Ca 2+-Dependent Processes of Innate Immunity in IBD. Cells 2024; 13:1079. [PMID: 38994933 PMCID: PMC11240513 DOI: 10.3390/cells13131079] [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: 05/22/2024] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
IBD is an uncontrolled inflammatory condition of the gastrointestinal tract, which mainly manifests in two forms: ulcerative colitis (UC) and Crohn's disease (CD). The pathogenesis of IBD appears to be associated with an abnormal response of innate and adaptive immune cells. Innate immunity cells, such as macrophages, mast cells, and granulocytes, can produce proinflammatory (e.g., TNF-α) and oxidative stress (ROS) mediators promoting intestinal damage, and their abnormal responses can induce an imbalance in adaptive immunity, leading to the production of inflammatory cytokines that increase innate immune damage, abate intestinal barrier functions, and aggravate inflammation. Considering that Ca2+ signalling plays a key role in a plethora of cellular functions, this review has the purpose of deepening the potential Ca2+ involvement in IBD pathogenesis.
Collapse
Affiliation(s)
- Francesco Palestra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Gina Memoli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Annagioia Ventrici
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Marialuisa Trocchia
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Mariarosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.P.); (G.M.); (A.V.); (M.T.); (M.G.); (G.V.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy
| |
Collapse
|
10
|
Cheng G, Zhao Y, Sun F, Zhang Q. Novel insights into STIM1's role in store-operated calcium entry and its implications for T-cell mediated inflammation in trigeminal neuralgia. Front Mol Neurosci 2024; 17:1391189. [PMID: 38962804 PMCID: PMC11221526 DOI: 10.3389/fnmol.2024.1391189] [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: 02/25/2024] [Accepted: 04/30/2024] [Indexed: 07/05/2024] Open
Abstract
This investigation aims to elucidate the novel role of Stromal Interaction Molecule 1 (STIM1) in modulating store-operated calcium entry (SOCE) and its subsequent impact on inflammatory cytokine release in T lymphocytes, thereby advancing our understanding of trigeminal neuralgia (TN) pathogenesis. Employing the Gene Expression Omnibus (GEO) database, we extracted microarray data pertinent to TN to identify differentially expressed genes (DEGs). A subsequent comparison with SOCE-related genes from the Genecards database helped pinpoint potential target genes. The STRING database facilitated protein-protein interaction (PPI) analysis to spotlight STIM1 as a gene of interest in TN. Through histological staining, transmission electron microscopy (TEM), and behavioral assessments, we probed STIM1's pathological effects on TN in rat models. Additionally, we examined STIM1's influence on the SOCE pathway in trigeminal ganglion cells using techniques like calcium content measurement, patch clamp electrophysiology, and STIM1- ORAI1 co-localization studies. Changes in the expression of inflammatory markers (TNF-α, IL-1β, IL-6) in T cells were quantified using Western blot (WB) and enzyme-linked immunosorbent assay (ELISA) in vitro, while immunohistochemistry and flow cytometry were applied in vivo to assess these cytokines and T cell count alterations. Our bioinformatic approach highlighted STIM1's significant overexpression in TN patients, underscoring its pivotal role in TN's etiology and progression. Experimental findings from both in vitro and in vivo studies corroborated STIM1's regulatory influence on the SOCE pathway. Furthermore, STIM1 was shown to mediate SOCE-induced inflammatory cytokine release in T lymphocytes, a critical factor in TN development. Supportive evidence from histological, ultrastructural, and behavioral analyses reinforced the link between STIM1-mediated SOCE and T lymphocyte-driven inflammation in TN pathogenesis. This study presents novel evidence that STIM1 is a key regulator of SOCE and inflammatory cytokine release in T lymphocytes, contributing significantly to the pathogenesis of trigeminal neuralgia. Our findings not only deepen the understanding of TN's molecular underpinnings but also potentially open new avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Guangyu Cheng
- Translational Medicine Research Center of Traditional Chinese Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Zhao
- Department of Acupuncture, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fujia Sun
- Department of Acupuncture, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhang
- Preventive Treatment Center, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
11
|
Yu Z, Song W, Ren X, Chen J, Yao Q, Liu H, Wang X, Zhou J, Wang B, Chen X. Calcium deficiency is associated with malnutrition risk in patients with inflammatory bowel disease. Postgrad Med 2024; 136:456-467. [PMID: 38782760 DOI: 10.1080/00325481.2024.2359895] [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: 12/28/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND AIM Patients with inflammatory bowel disease (IBD) often have the condition of malnutrition, which can be presented as sarcopenia, micronutrient deficiencies, etc. Trace elements (magnesium, calcium, iron, copper, zinc, plumbum and manganese) belonging to micronutrients, are greatly vital for the assessment of nutritional status in humans. Trace element deficiencies are also the main manifestation of malnutrition. Calcium (Ca) has been proved to play an important part in maintaining body homeostasis and regulating cellular function. However, there are still a lack of studies on the association between malnutrition and Ca deficiency in IBD. This research aimed to investigate the role of Ca for malnutrition in IBD patients. METHODS We prospectively collected blood samples from 149 patients and utilized inductively coupled plasma mass spectrometry to examine their venous serum trace element concentrations. Logistic regression analyses were used to investigate the association between Ca and malnutrition. Receiver operating characteristic (ROC) curves were generated to calculate the cutoffs for determination of Ca deficiency. RESULTS Except Ca, the concentrations of the other six trace elements presented no statistical significance between non-malnutrition and malnutrition group. In comparison with the non-malnutrition group, the serum concentration of Ca decreased in the malnutrition group (89.36 vs 87.03 mg/L, p = 0.023). With regard to ROC curve, Ca < 87.21 mg/L showed the best discriminative capability with an area of 0.624 (95% CI: 0.520, 0.727, p = 0.023). Multivariate analyses demonstrated that Ca < 87.21 mg/L (OR = 3.393, 95% CI: 1.524, 7.554, p = 0.003) and age (OR = 0.958, 95% CI: 0.926, 0.990, p = 0.011) were associated with malnutrition risk. Serum Ca levels were significantly lower in the malnutrition group than those in the non-malnutrition group among UC patients, those with severe disease state or the female group. CONCLUSIONS In patients with IBD, Ca deficiency is an independent factor for high malnutrition risk.
Collapse
Affiliation(s)
- Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenxuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangfeng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hang Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinjie Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
12
|
Zhang Q, Wang C, He L. ORAI Ca 2+ Channels in Cancers and Therapeutic Interventions. Biomolecules 2024; 14:417. [PMID: 38672434 PMCID: PMC11048467 DOI: 10.3390/biom14040417] [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: 02/29/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
The ORAI proteins serve as crucial pore-forming subunits of calcium-release-activated calcium (CRAC) channels, pivotal in regulating downstream calcium-related signaling pathways. Dysregulated calcium homeostasis arising from mutations and post-translational modifications in ORAI can lead to immune disorders, myopathy, cardiovascular diseases, and even cancers. Small molecules targeting ORAI present an approach for calcium signaling modulation. Moreover, emerging techniques like optogenetics and optochemistry aim to offer more precise regulation of ORAI. This review focuses on the role of ORAI in cancers, providing a concise overview of their significance in the initiation and progression of cancers. Additionally, it highlights state-of-the-art techniques for ORAI channel modulation, including advanced optical tools, potent pharmacological inhibitors, and antibodies. These novel strategies offer promising avenues for the functional regulation of ORAI in research and may inspire innovative approaches to cancer therapy targeting ORAI.
Collapse
Affiliation(s)
| | | | - Lian He
- Department of Pharmacology, Joint Laboratory of Guangdong–Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| |
Collapse
|
13
|
Bacsa B, Hopl V, Derler I. Synthetic Biology Meets Ca 2+ Release-Activated Ca 2+ Channel-Dependent Immunomodulation. Cells 2024; 13:468. [PMID: 38534312 PMCID: PMC10968988 DOI: 10.3390/cells13060468] [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: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Many essential biological processes are triggered by the proximity of molecules. Meanwhile, diverse approaches in synthetic biology, such as new biological parts or engineered cells, have opened up avenues to precisely control the proximity of molecules and eventually downstream signaling processes. This also applies to a main Ca2+ entry pathway into the cell, the so-called Ca2+ release-activated Ca2+ (CRAC) channel. CRAC channels are among other channels are essential in the immune response and are activated by receptor-ligand binding at the cell membrane. The latter initiates a signaling cascade within the cell, which finally triggers the coupling of the two key molecular components of the CRAC channel, namely the stromal interaction molecule, STIM, in the ER membrane and the plasma membrane Ca2+ ion channel, Orai. Ca2+ entry, established via STIM/Orai coupling, is essential for various immune cell functions, including cytokine release, proliferation, and cytotoxicity. In this review, we summarize the tools of synthetic biology that have been used so far to achieve precise control over the CRAC channel pathway and thus over downstream signaling events related to the immune response.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Division of Medical Physics und Biophysics, Medical University of Graz, A-8010 Graz, Austria;
| | - Valentina Hopl
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
14
|
Piseddu I, Weidinger C, Mayerle J. Fine tuning calcium dynamics by inhibition of Store-operated Calcium Entry as a novel therapeutic approach for the treatment of chronic pancreatitis. Cell Calcium 2023; 116:102802. [PMID: 37757535 DOI: 10.1016/j.ceca.2023.102802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Chronic pancreatitis (CP) is a complex inflammatory disorder characterized by progressive fibrosis, leading to pancreatic dysfunction, reduced quality of life and an elevated pancreatic cancer risk. Current therapeutic options for CP are restricted to symptomatic treatment. Using ex vivo and in vivo preclinical disease models, Szabó et al. now explored for the first time the involvement of Store-operated Ca2+ entry (SOCE) in the progression of CP and propose that a selective pharmacological inhibition of the SOCE signaling component Orai1 might serve as specific treatment option for CP[1,2].
Collapse
Affiliation(s)
- Ignazio Piseddu
- Department of Medicine II, University Hospital, LMU, Munich, Germany; Gene Center and Department of Biochemistry, LMU, Munich, Germany
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, LMU, Munich, Germany
| |
Collapse
|
15
|
Lee H, Jeon JH, Kim ES. Mitochondrial dysfunctions in T cells: focus on inflammatory bowel disease. Front Immunol 2023; 14:1219422. [PMID: 37809060 PMCID: PMC10556505 DOI: 10.3389/fimmu.2023.1219422] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Mitochondria has emerged as a critical ruler of metabolic reprogramming in immune responses and inflammation. In the context of colitogenic T cells and IBD, there has been increasing research interest in the metabolic pathways of glycolysis, pyruvate oxidation, and glutaminolysis. These pathways have been shown to play a crucial role in the metabolic reprogramming of colitogenic T cells, leading to increased inflammatory cytokine production and tissue damage. In addition to metabolic reprogramming, mitochondrial dysfunction has also been implicated in the pathogenesis of IBD. Studies have shown that colitogenic T cells exhibit impaired mitochondrial respiration, elevated levels of mROS, alterations in calcium homeostasis, impaired mitochondrial biogenesis, and aberrant mitochondria-associated membrane formation. Here, we discuss our current knowledge of the metabolic reprogramming and mitochondrial dysfunctions in colitogenic T cells, as well as the potential therapeutic applications for treating IBD with evidence from animal experiments.
Collapse
Affiliation(s)
- Hoyul Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Han Jeon
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Eun Soo Kim
- Division of Gastroenterology, Department of Internal Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
16
|
Song WX, Yu ZH, Ren XF, Chen JH, Chen X. Role of micronutrients in inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2023; 31:711-731. [DOI: 10.11569/wcjd.v31.i17.711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an autoimmune intestinal disease that includes ulcerative colitis, Crohn's disease, and indeterminate colitis. Patients with IBD are often at risk for malnutrition, including micronutrient deficiencies, due to dietary restrictions and poor intestinal absorption. Micronutrients, including vitamins and minerals, play an important role in the human body's metabolism and maintenance of tissue functions. This article reviews the role of micronutrients in IBD. Micronutrients can affect the occurrence and progression of IBD by regulating immunity, intestinal flora, oxidative stress, intestinal barrier function, and other aspects. Monitoring and timely supplementation of micronutrients are important to delay progression and improve clinical symptoms in IBD patients.
Collapse
Affiliation(s)
- Wen-Xuan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zi-Han Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiang-Feng Ren
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ji-Hua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
17
|
Berry V, Ionides A, Georgiou M, Quinlan RA, Michaelides M. Multimorbidity due to novel pathogenic variants in the WFS1/RP1/NOD2 genes: autosomal dominant congenital lamellar cataract, retinitis pigmentosa and Crohn's disease in a British family. BMJ Open Ophthalmol 2023; 8:e001252. [PMID: 37493686 PMCID: PMC10351282 DOI: 10.1136/bmjophth-2023-001252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/08/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND A five generation family has been analysed by whole exome sequencing (WES) for genetic associations with the multimorbidities of congenital cataract (CC), retinitis pigmentosa (RP) and Crohn's disease (CD). METHODS WES was performed for unaffected and affected individuals within the family pedigree followed by bioinformatic analyses of these data to identify disease-causing variants with damaging pathogenicity scores. RESULTS A novel pathogenic missense variant in WFS1: c.1897G>C; p.V633L, a novel pathogenic nonsense variant in RP1: c.6344T>G; p.L2115* and a predicted pathogenic missense variant in NOD2: c.2104C>T; p.R702W are reported. The three variants cosegregated with the phenotypic combinations of autosomal dominant CC, RP and CD within individual family members. CONCLUSIONS Here, we report multimorbidity in a family pedigree listed on a CC register, which broadens the spectrum of potential cataract associated genes to include both RP1 and NOD2.
Collapse
Affiliation(s)
- Vanita Berry
- Genetics, UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK, London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, EC1V 2PD, UK, London, UK
| | - Alexander Ionides
- Moorfields Eye Hospital NHS Foundation Trust, London, EC1V 2PD, UK, London, UK
| | - Michalis Georgiou
- Genetics, UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK, London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, EC1V 2PD, UK, London, UK
| | - Roy A Quinlan
- Department of Biosciences, University of Durham, Upper Mountjoy Science Site, Durham DH1 3LE, UK, Durham, UK
| | - Michel Michaelides
- Genetics, UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK, London, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, EC1V 2PD, UK, London, UK
| |
Collapse
|
18
|
Kongkatitham V, Dehlinger A, Wang M, Poldorn P, Weidinger C, Letizia M, Chaotham C, Otto C, Ruprecht K, Paul F, Rungrotmongkol T, Likhitwitayawuid K, Böttcher C, Sritularak B. Immunomodulatory Effects of New Phenanthrene Derivatives from Dendrobium crumenatum. JOURNAL OF NATURAL PRODUCTS 2023; 86:1294-1306. [PMID: 37140218 DOI: 10.1021/acs.jnatprod.3c00107] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Three new phenanthrene derivatives (1, 2, 4), one new fluorenone (3), and four known compounds (5-8) were isolated from the ethyl acetate extract of Dendrobium crumenatum Sw. stems using column chromatography. The chemical structures were elucidated by analysis of spectroscopic data. The absolute configuration of 4 was determined by electronic circular dichroism calculation. We also evaluated the immunomodulatory effects of compounds isolated from D. crumenatum in human peripheral blood mononuclear cells from healthy individuals and those from patients with multiple sclerosis in vitro. Dendrocrumenol B (2) and dendrocrumenol D (4) showed strong immunomodulatory effects on both CD3+ T cells and CD14+ monocytes. Compounds 2 and 4 could reduce IL-2 and TNF production in T cells and monocytes that were treated with phorbol-12-myristate-13-acetate and ionomycin (PMA/Iono). Deep immune profiling using high-dimensional single-cell mass cytometry could confirm immunomodulatory effects of 4, quantified by the reduction of activated T cell population under PMA/Iono stimulation, in comparison to the stimulated T cells without treatment.
Collapse
Affiliation(s)
- Virunh Kongkatitham
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Adeline Dehlinger
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
| | - Meng Wang
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
| | - Preeyaporn Poldorn
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 12203, Germany
- Clinician Scientist Program, Berlin Institute of Health, Berlin 10117, Germany
| | - Marilena Letizia
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin 12203, Germany
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Carolin Otto
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Klemens Ruprecht
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Thanyada Rungrotmongkol
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kittisak Likhitwitayawuid
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
19
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
20
|
Anthocyanin extract from black rice attenuates chronic inflammation in DSS-induced colitis mouse model by modulating the gut microbiota. OPEN CHEM 2023. [DOI: 10.1515/chem-2022-0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Abstract
There is substantial evidence for the probiotic activity of anthocyanins, but the relationship between anthocyanins involved in the regulation of microbiota and intestinal inflammation has not been fully elucidated. The aim of this study was to investigate the regulatory effects of black rice anthocyanin extract (BRAE) on intestinal microbiota imbalance in mice with dextran sulfate sodium (DSS)-induced chronic colitis. DSS was added into drinking water to induce a mouse model of chronic experimental colitis, and BRAE was given by gavage (200 mg/kg/day) for 4 weeks. Body weight, fecal viscosity, and hematochezia were monitored during administration. After mice were sacrificed, the serum concentrations of TNF-α and IL-6 were detected by enzyme-linked immunosorbent assay, and the composition of intestinal flora was analyzed by 16S rDNA sequencing. The results showed that BRAE significantly suppressed DSS-induced colonic inflammatory phenotypes and maintained colon length in mice. In addition, BRAE reduced intestinal permeability and improved intestinal barrier dysfunction in mice with colitis. Gut microbiota analysis showed that BRAE significantly improved the imbalance of intestinal microecological diversity caused by DSS, inhibited the increase in the relative abundance of inflammatory bacteria, and promoted the abundance of anti-inflammatory probiotics including Akkermansia spp.
Collapse
|
21
|
Feng J, Xie Z, Hu H. Ion channel regulation of gut immunity. J Gen Physiol 2022; 155:213734. [PMID: 36459135 PMCID: PMC9723512 DOI: 10.1085/jgp.202113042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/15/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Mounting evidence indicates that gastrointestinal (GI) homeostasis hinges on communications among many cellular networks including the intestinal epithelium, the immune system, and both intrinsic and extrinsic nerves innervating the gut. The GI tract, especially the colon, is the home base for gut microbiome which dynamically regulates immune function. The gut's immune system also provides an effective defense against harmful pathogens entering the GI tract while maintaining immune homeostasis to avoid exaggerated immune reaction to innocuous food and commensal antigens which are important causes of inflammatory disorders such as coeliac disease and inflammatory bowel diseases (IBD). Various ion channels have been detected in multiple cell types throughout the GI tract. By regulating membrane properties and intracellular biochemical signaling, ion channels play a critical role in synchronized signaling among diverse cellular components in the gut that orchestrates the GI immune response. This work focuses on the role of ion channels in immune cells, non-immune resident cells, and neuroimmune interactions in the gut at the steady state and pathological conditions. Understanding the cellular and molecular basis of ion channel signaling in these immune-related pathways and initial testing of pharmacological intervention will facilitate the development of ion channel-based therapeutic approaches for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China,Correspondence to Jing Feng:
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Hongzhen Hu:
| |
Collapse
|
22
|
Kappel S, Peinelt C. Targeting CRAC channels in inflammatory bowel disease. EMBO Mol Med 2022; 14:e16489. [PMID: 35969215 PMCID: PMC9449586 DOI: 10.15252/emmm.202216489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a collective term for inflammatory diseases of the human gastrointestinal (GI) tract that are characterized by perturbations in the intestinal immune responses. In their study, Letizia et al (2022) found an enrichment of CD4+ effector T cells, interferon gamma (IFNγ) producing CD8+ T cells, regulatory T cells, and innate lymphoid cells (ILC) in the lamina propria (LP) of IBD patients. In these cells, pharmacological inhibition of store-operated calcium entry (SOCE) reduced cytokine production. In addition, in a murine IBD model, systemic SOCE inhibition reduced IBD severity and weight loss.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|